University of Calgary PRISM: University of Calgary's Digital Repository

Graduate Studies The Vault: Electronic Theses and Dissertations

2020-07-21 Pathophysiology and Proteogenomics of Post-infectious and Post-hemorrhagic Hydrocephalus in Infants

Isaacs, Albert M.

Isaacs, A. M. (2020). Pathophysiology and Proteogenomics of Post-infectious and Post-hemorrhagic Hydrocephalus in Infants (Unpublished doctoral thesis). University of Calgary, Calgary, AB. http://hdl.handle.net/1880/112344 doctoral thesis

University of Calgary graduate students retain copyright ownership and moral rights for their thesis. You may use this material in any way that is permitted by the Copyright Act or through licensing that has been assigned to the document. For uses that are not allowable under copyright legislation or licensing, you are required to seek permission. Downloaded from PRISM: https://prism.ucalgary.ca UNIVERSITY OF CALGARY

Pathophysiology and Proteogenomics of Post-infectious and Post-hemorrhagic Hydrocephalus in Infants

by

Albert M. Isaacs

A THESIS

SUBMITTED TO THE FACULTY OF GRADUATE STUDIES

IN PARTIAL FULFILMENT OF THE REQUIREMENTS FOR THE

DEGREE OF DOCTOR OF PHILOSOPHY

GRADUATE PROGRAM IN NEUROSCIENCE

CALGARY, ALBERTA

JULY, 2020

© Albert M. Isaacs 2020

Abstract

Post-infectious (PIH) and post-hemorrhagic (PHH) hydrocephalus occur as sequalae of neonatal sepsis or intraventricular hemorrhage (IVH) of prematurity, respectively. Together, PIH and PHH represent the most common form of infantile hydrocephalus, the most common indication for neurosurgery in children globally, and the leading cause of neurological morbidity and mortality worldwide. The lack of understanding of the pathophysiology of PIH and PHH, particularly with regards to the host central nervous system response to the antecedent infection and hemorrhage, perturbation of differentiating neural stems in the ventricular (VZ) and subventricular (SVZ) zones, and damage to periventricular white matter (PVWM) tracts carrying sensorimotor fibers, has hindered the identification of therapeutic targets to prevent these two debilitating conditions.

To this end, we hypothesized that PIH and PHH share a common pathophysiological mechanism characterized by host immune response to infection or hemorrhage, activation of the metalloprotease ADAM10 and cleavage of the cell-junctional protein, N-cadherin, which results in impaired VZ/SVZ differentiation and PVWM damage. The studies presented in this dissertation collectively explore novel overarching lines of scientific inquiry - the concept that there are unique host immune responses to PIH and PHH, as well as ones that are common to both conditions that underlie their observed clinical similarities.

To test the hypothesis, we leveraged the cerebrospinal fluid (CSF) of human PIH, PHH and matched non-infectious non-hemorrhagic hydrocephalic control infants. First, we defined the differentially expressed and transcriptome of PIH using high throughput and

RNA-seq, respectively. The integration of proteogenomic techniques defined critical

ii networks and pathway level mechanisms of PIH pathophysiology. Second, our validated proteomics pipeline was used to identify the common and unique molecular pathways that underlie the pathophysiology of PIH and PHH. Third, our CSF findings were related to VZ/SVZ development and PVWM microstructural damage with diffusion MRI (dMRI) in PHH and control infants. Neurocytology of human postmortem brain tissues of PIH, PHH and controls was performed to correlate the dMRI findings. Fourth, we defined the mechanistic triggers underlying

PIH and PHH pathogenesis utilizing a mouse in vitro cell culture model of periventricular VZ cells. Finally, we developed in vivo animal models of PIH (mice) and PHH (ferrets), to recapitulate our findings of VZ/SVZ disruption, neuroinflammation and PVWM injury in both PIH and PHH.

Through our comprehensive experiments, we determined the following: 1) human CSF profiles of

PIH and PHH reflect similar alterations in gene-activated pathways related to neuroinflammation and cell-cell junction structure; 2) neuroinflammation-associated cell junction (VZ/SVZ) disruption and PVWM injury is a pathophysiological mechanism that is common to both PIH and

PHH; 3) dMRI can non-invasively assess the VZ/SVZ region as well as differentiate and quantify tract-specific patterns of PVWM injury. Therefore, it can distinguish direct effects on axons/myelin from changes in the extracellular milieu to reflect neuroinflammation, axonal fiber loss, and dysmyelination; 4) VZ/SVZ disruption in PIH and PHH is mediated by metalloprotease

(e.g., ADAM10) cleavage of cell junctional proteins (e.g., N-cadherin); and 5) pharmacologic inhibition of ADAM10-mediated N-cadherin cleavage represents a viable therapeutic approach to prevent PIH and PHH.

iii These novel insights into the pathophysiology of PIH and PHH may enable investigation of

ADAM10 inhibitors and other therapeutic strategies to minimize the developmental disability in

PIH and PHH patients. The biomarkers we identified can be further investigated as diagnostic measures for monitoring and providing therapy to infants who develop neonatal sepsis or IVH to prevent PHH/PIH. Finally, the in vitro and in vivo experimental models we generated are poised for future preclinical and translational studies into the pathogenesis of these previously inaccessible and debilitating conditions.

iv Preface

This dissertation is original and independent work performed by the author, Albert M. Isaacs.

Some of the work has already been published, while others are in preparation for future publications. At the beginning of each chapter, the papers adapted for the chapter are cited. With permission from my supervisory committee, the majority of the text were verbatim from studies I played major roles in, including idea conception, research methodology, data analysis, synthesis and interpretation and writing of manuscripts. Permission from applicable sources, supervisors and co-investigators have been sought for all figures and tables presented in this dissertation. In each chapter, the institution or committee that provided ethics oversight for the specific studies are provided.

In Chapter 2, the majority of experiments were part of a broader work by the P3H and CONSHA consortiums led by Dr. Steven Schiff (Penn State University), investigating the neonatal septisomes of Sub-Saharan Africa. While I was responsible for all the CSF proteomics work performed at Washington University School of Medicine, I spent 6 weeks at the Center for

Personalized Medicine (Penn State University), where I learned the RNA-Seq wet lab techniques presented in this dissertation. I also spent 6 weeks in Mbale, Uganda at the CURE Children’s

Hospital where our Sub-Saharan African CSF samples were obtained. While in Uganda, I learned the workflow and observed surgical procedures performed on hydrocephalic infants, under the supervision of Drs. John Mugamba, Peter Ssenyonga and Justin Onen. I was involved in the logistical management of all proteomics samples. I designed the CSF proteomics workflows, including allocation of ten-plexes and bookending with large volume samples for each plex. At

v the proteomics core (Washington University), I observed all the wet lab processes, both manual and automated, and worked with the core’s statistician on post processing data quality assurance.

Upon receiving the protein raw counts, I performed all the proteomics data analyses presented here, and wrote the first draft of all manuscripts included in this chapter. Dr. Steven Schiff and Dr.

David Limbrick oversaw and guided all my work. Dr. Joseph Paulson (Genentech Inc.) taught me and supervised all and statistical analyses. I received incredible support and guidance on proteomics wet lab and processing methodology from Dr. Reid Townsend and all members of the Washington University proteomics core (Dr. Qiang Zhang, Ms. Petra Gilmore,

Mr. Jim Malone, Mr. Robert Sprung, and Ms. Yiling). Drs. Sarah Morton and Mercedeh

Movassagh (Harvard University) helped me with the transcriptomics and proteogenomics integrative methodologies. Ms. Christine Henhly (Penn State University) trained me on the RNA-

Seq wet lab, and Mr. Diego Morales (Washington University) helped coordinate the logistics of the CSF samples.

In Chapter 3, except for patient recruitment and human dMRI acquisition, I personally performed, or was heavily involved with all day to day experiments presented. Nevertheless, I received incredible support from our collaborators and colleagues. Unless otherwise specified, all the work and supervision were performed at Washington University. Indeed, all I learned in diffusion imaging are a reflection of the invaluable teaching and continuous guidance I received from Drs.

Jeff Neil, Christopher Smyser, Victor Song, Josh Shimony, and David Limbrick. Drs. Joseph

Paulson (Genetech Inc.) and Yan Yan supervised my statistical analyses. Drs. Shimony and Roland

Han helped me with the design of our novel lateral ventricular perimeter segmentation algorithm.

Ms Tara Smyser, Ms Jeanette Kenley, Mr. Dimitrios Alexopolous and Ms. Rachel Lean from the

vi Smyser lab helped with many aspects of dMRI data processing, logistics and troubleshooting. Drs.

Sun Peng, Harri Merisaari and Ajit George at the Song lab (Washington University) trained me on

DBSI fitting and methodology. Drs. Leandro Castaneyra-Ruiz, Pat McAllister and Sonika Dahiya helped tremendously to perform, analyze, interpret and correlate our neurocytology findings with dMRI.

In Chapter 4, I was involved in all and performed the majority of experiments presented.

Nevertheless, I had tremendous help from Dr. Leandro Castaneyra-Ruiz, who had developed the cell culture model a year prior to my arrival at Washington University. Dr. Pat McAllister and Dr.

David Limbrick mentored me through those projects. Dr. Steven Brody provided us with the mice and Dr. Juliane Bubeck-Wardenburg and her staff scientist, Dr. Kelly Tomaszewski, provided us with the 훂-hemolysin and gave guidance on the experiments.

In chapter 5, developing the animal models required a large infrastructure at Washington

University and the University of Calgary. I was heavily involved in all experiments. I took part in the surgeries, animal monitoring and immunohistochemistry. I performed the DBSI acquisition for all the ferrets and performed all data analyses presented in this chapter. Dr. Pat McAllister coordinated the day-to-day management of the ferret model. However, the ferret model was a collective effort by the entire Limbrick-McAllister lab, including Diego Morales and our many visiting students - Dr. Travis Crevecoeur, Alexis Hartman, Joel Brown, Sarah Zwick, and Brandon

Baksh. Members of the Garbow and Song labs, both at the Mallinckrodt Institute helped with the neuroimaging, especially Dr. Xia Ge, John Engelbach, Dr. Harri Merisaari, Dr. Tsen-Hun Lin, Dr.

Victor Song and Dr. Joel Garbow. Dr. Philip Bayly provided us with the control ferret scans for

vii comparisons. The mouse PIH model work was an extension of our animal model efforts at

Washington University, which was carried out at the University of Calgary. Drs. Jeff Dunn,

Castaneyra-Ruiz, McAllister and Limbrick were instrumental in the idea conception and provided guidance throughout the experiments. Dr. Castaneyra-Ruiz helped me immensely with the immunohistochemistry, especially during the Covid-19 pandemic. In Calgary, Dr. Jeff Dunn provided me with all the necessary resources at the Experimental Imaging Center and mentored me on this project. I had invaluable help from Dr. Ying Wu (who performed the mice intraventricular injections), as well as Max Hamilton, Runze Yang, Kevin Lee, David Rushforth and Tadeusz Foniok who helped with the neuroimaging and animal sacrifice. Dr. Bas Surewaard provided us with all the alpha toxin and guided us on dosing.

viii Acknowledgements

As I ponder on how this journey began, I cannot imagine how any of this would have been possible if not for the overflow of kindness, support, provisions and more importantly the mentorship I have received from Dr. David Limbrick, Dr. Pat McAllister and Dr. Mark Hamilton over the past several years. I honestly do not know what good I have done to deserve your devotion to my academic refinement both as a scientist and neurosurgeon and for always opening doors to ensure I continue to succeed in all aspects of life. Through these years you have shared in my successes, offered much needed encouragement in times of failures and have been the solid rock I have leaned on when the goings got tough, at work and in my personal life. I have always looked up to all three of you and you have not failed me once. I cannot thank you enough!

Pursuing a dual university degree at Washington University and the University of Calgary was not without challenges. I thank Dr. Jeff Dunn for his help navigating the complexities of the program.

As my PhD supervisor at the University of Calgary, Dr. Dunn helped develop me scientifically and provided me with resources, for which I will always be grateful. I will be remiss to not express my gratitude to Dr. Tara Beattie, Dr. David Limbrick, Dr. Pat McAllister, Dr. Jay Riva-Cambrin,

Dr. Mark Hamilton, Dr. Deborah Kurrasch, Dr. Richard Wilson, Dr. John Russell, Dr. Erik

Herzog, Dr. Larry Schneider, Dr. Tamara Hershey, Dr. Timothy Holy, Dr. Jalel Azaiez Ms. Sally

Vogt, Ms. Lesley Towill and Mr. Calvin Lac for working tirelessly to make the Cotutelle

Agreement a success.

ix Many generous, supportive and insightful people have helped develop me as a scientist over the past three years and were indispensable in the experiments presented here. Dr. Steve Schiff not only welcomed me to his massive P3H project, he entrusted a major part of his groundbreaking work to me and guided me through it every step of the way, ensuring my success. It still amazes me how far I have come and the amount of statistical and bioinformatics knowledge I have accumulated over these few years, as Dr. Joseph Paulson patiently tutored me days in and days out. I could also fill many pages listing the benchwork procedures Dr. Leandro Castaneyra-Ruiz taught me and how much he stimulated my knowledge in neurodevelopmental biology. Leandro helped me tremendously towards the finish line in the midst of the Covid-19 pandemic. Leandro has been a great friend, both at and outside of work, and we have shared many memories together.

Every member of my advisory committee at Washington University and the University of Calgary,

Drs. David Limbrick, Pat McAllister, Gregory Zipfel, Christopher Smyser, Joshua Shimony, Mark

Hamilton, Eric Smith and Billie Au, were fully invested in ensuring the productivity of my work and development as a scientist. I would like to specially thank Drs Reid Townsend, Jeffrey Neil, and Victor Song for training and guiding me through some of very complex scientific methodologies I used during this PhD. I also owe special thanks to Mr. Diego Morales, without his logistical help, advices, and experimental assistance, many of my work would not have been possible. Frankly, his occasional frivolity and our Limbrick-McAllister lab musings kept me sane.

I would also like to thank Dr. Ying Wu, Runze Yang and Max Hamilton for their help with all my experiments at the University of Calgary.

x A huge and special thank you to my wonderful wife, Gloria. You are hands down the tour-de-force of all I have accomplished and will ever be proud of, including this PhD. To my son Azariah - you have given meaning to this work, and everything I do! I dedicated my time to this PhD focused on finding ways to improve the health and life of preterm babies. Little did I know that your mom and

I will be having our very own lovely preterm baby born at 22 weeks, who will still be fighting in the NICU as I defend this thesis. We are blessed because of you. This work is dedicated to you because you have brought us unimaginable joy and I cannot wait to work with you to shatter the ceilings in neonatal health! May God continue to bless and keep you; May God’s face shine upon you and be gracious to you; May God lift up His countenance upon you, and give you joy and peace. We love you Azariah!

I thank my parents (Elizabeth and Isaac), parents-in law (Dora and Leo), siblings (Dorcas, Abigail, and Gideon), and sisters- and bother-in-law (Freda, Verona, and Andrew) for supporting me in unimaginable ways. I also thank my closest friends, Dr. Michael Opoku-Darko, Dr. Magalie

Cadieux, Mr. Felix Anin and Mr. Ryan Manse for always been there for me through this journey.

I thank members of the CONSHA consortium and all my colleagues at the Limbrick, Schiff,

Townsend, Dunn, Smyser and Song labs. Thank you, Dr. Praveen Ballabh, for facilitating my PhD thesis examination especially under these very unusual circumstances.

These studies would not have been possible if not for the children and their families who participated - thank you. I thank Vanier Canada, Killam Foundation, Hydrocephalus Association, and the University of Calgary Division of Neurosurgery for their financial support. I also

xi appreciate the support of the funding agencies of my supervisors including the National Institutes of Health, Child Neurology Foundation, Cerebral Palsy International Research Foundation, Dana

Foundation, March of Dimes Prematurity Research Center at Washington University, Doris Duke

Charitable Foundation, Eunice Kennedy Shriver National Institute of Child Health & Human

Development of the National Institutes of Health.

xii

Dedicated to my son, Azariah and my wife, Gloria

xiii TABLE OF CONTENTS

ABSTRACT ...... II PREFACE ...... V ACKNOWLEDGEMENTS ...... IX TABLE OF CONTENTS ...... XIV LIST OF TABLES ...... XVIII LIST OF ILLUSTRATIONS, FIGURES AND GRAPHICS ...... XIX LIST OF ABBREVIATIONS ...... XXI 1 CHAPTER 1: INTRODUCTION TO THE DISSERTATION ...... 1

1.1 HYDROCEPHALUS ...... 3 1.1.1 Classification of hydrocephalus ...... 3 1.1.2 Clinical presentation and diagnosis ...... 4 1.1.3 Global burden of hydrocephalus ...... 5 1.2 POST-INFECTIOUS HYDROCEPHALUS (PIH) ...... 8 1.3 POST-HEMORRHAGIC HYDROCEPHALUS (PHH) ...... 10 1.4 CURRENT TREATMENTS OF PIH AND PHH ...... 11 1.5 CURRENT UNDERSTANDING OF THE PATHOPHYSIOLOGY OF PIH/PHH ...... 13 1.5.1 Embryological overview of the cerebral ventricular system...... 14 1.5.1.1 Ventricular development ...... 14 1.5.1.2 Ependymal layer development ...... 18 1.5.1.3 Choroid plexus development ...... 19 1.5.2 Hydrodynamic and pulsatility concepts of hydrocephalus ...... 20 1.5.3 Phenotypic similarities between PIH and PHH ...... 22 1.5.4 Role of neuroinflammation in PIH/PHH pathogenesis ...... 22 1.5.5 Effect of PIH and PHH on neurodevelopment ...... 24 1.5.6 Periventricular white matter (PVWM) injury in PIH/PHH...... 26 1.6 PROPOSED STRATEGIES ...... 26 1.6.1 CSF and proteomics ...... 27 1.6.2 Proteogenomics ...... 30 1.6.3 Diffusion magnetic resonance imaging (dMRI) ...... 31 1.6.3.1 Diffusion tensor imaging (DTI) ...... 32 1.6.3.2 Diffusion basis spectrum imaging (DBSI) ...... 34 1.6.4 Neurocytology ...... 35 1.6.5 Experimental models of hydrocephalus ...... 36 1.7 THESIS OBJECTIVES AND AIMS ...... 38 1.7.1 Objectives ...... 38 1.7.2 Central hypothesis ...... 39 1.7.3 Specific Aims ...... 39 1.7.3.1 Specific Aim 1 ...... 39 1.7.3.2 Specific Aim 2 ...... 40 1.7.3.3 Specific Aim 3 ...... 40 1.7.3.4 Specific Aim 4 ...... 41 2 CHAPTER 2: PROTEOGENOMICS AND THE PIH-PHH CEREBROSPINAL FLUID EXPRESSOMES .. 42

2.1 INTRODUCTION ...... 43

xiv 2.2 RESULTS ...... 44 2.2.1 Concordant proteome and transcriptome of host immune response in PIH...... 44 2.2.1.1 Pathogen identification ...... 45 2.2.1.2 Patient characteristics ...... 46 2.2.1.3 Proteomics demonstrate neuroinflammation and cell junction disruption in PIH...... 47 2.2.1.4 RNA-Seq demonstrates neuroinflammation and cell junction disruption in PIH ...... 50 2.2.1.5 Clinical traits correlate with CSF gene expression in PIH ...... 52 2.2.1.6 Proteogenomic expressome of host immune response in PIH ...... 53 2.2.2 Activated gene pathways in PIH and PHH ...... 56 2.2.2.1 Patient characteristics ...... 56 2.2.2.2 Protein expression patterns in PIH and PHH...... 57 2.2.2.3 The independent of PIH and PHH ...... 60 2.2.2.4 Neuroinflammation is a common process in PIH and PHH pathophysiology ...... 62 2.3 DISCUSSION ...... 65 2.3.1 Summary of findings ...... 65 2.3.2 Proteogenomics as an approach for defining PIH and PHH pathophysiology ...... 66 2.3.3 Host-immune responses in PIH and PHH ...... 66 2.3.4 Inflammation and barrier integrity in PIH and PHH ...... 68 2.3.5 Limitations ...... 69 2.4 MATERIALS AND METHODS ...... 70 2.4.1 Patients and recruitment ...... 70 2.4.2 CSF Samples ...... 71 2.4.3 Transcriptomics ...... 72 2.4.3.1 RNA Extraction ...... 72 2.4.3.2 RNA Sequencing ...... 73 2.4.3.3 Data Analysis ...... 74 2.4.4 Proteomics ...... 74 2.4.4.1 Sample preparation and ...... 74 2.4.4.2 Protein identification ...... 75 2.4.4.3 Protein quantification ...... 76 2.4.4.4 Data analysis ...... 77 2.4.5 Proteogenomics ...... 80 3 CHAPTER 3: DIFFUSION MRI CHARACTERIZATION OF VZ/SVZ AND PVWM INJURY...... 81

3.1 INTRODUCTION ...... 81 3.2 RESULTS ...... 84 3.2.1 Segmentation of the lateral ventricular perimeter (LVP) on diffusion MRI ...... 84 3.2.1.1 Segmentation requires minimal user input ...... 84 3.2.1.2 The frontal and occipital horns of the lateral ventricle can be independently assessed ...... 85 3.2.1.3 Semi-automated algorithm strongly corelates with manually segmented LVP ...... 85 3.2.2 Region-specific dMRI-derived LVP disruption underlies PHH pathophysiology ...... 86 3.2.2.1 Patient characteristics ...... 86 3.2.2.2 PHH is associated with segmental aberration of dMRI measures in the LVP ...... 89 3.2.2.3 The FOHP region is more severely injured than the rest of the LVP ...... 92 3.2.2.4 Ventricular size correlates with dMRI measures in the LVP and FOHP ...... 94 3.2.3 DBSI differentiates PVWM pathology in PHH ...... 96 3.2.3.1 Subject characteristics ...... 96 3.2.3.2 Neuroinflammation and reduced fiber density in the corpus callosum ...... 98 3.2.3.3 Neuroinflammation, reduced fiber density, and demyelination in corticospinal tracts ...... 101 3.2.3.4 Neuroinflammation, reduced fiber density, and edema in optic radiations ...... 103 3.2.3.5 Ventricular size correlates with magnitude of PVWM disruption ...... 104 3.2.3.6 Postmortem tissue neurocytology relates DBSI metrics to VZ disruption ...... 106

xv 3.3 DISCUSSION ...... 109 3.3.1 The LVP is a critical region in PHH pathophysiology ...... 109 3.3.2 Delineating the LVP for dMRI analyses ...... 110 3.3.3 Cellular basis of dMRI-measured LVP injury in PHH ...... 111 3.3.4 PHH-associated myelin injury ...... 112 3.3.5 FOHP sustains a greater magnitude of injury than the rest of the LVP ...... 113 3.3.6 DBSI extends dMRI capabilities to delineate white matter cellular pathology ...... 115 3.3.7 dMRI variability of white matter structure and development ...... 116 3.3.8 Ventricular size correlates with magnitude LVP and large bundle PVWM injury...... 119 3.3.9 Limitations ...... 120 3.4 MATERIALS AND METHODS ...... 122 3.4.1 Participants ...... 122 3.4.2 Perinatal Clinical Variables...... 123 3.4.3 Imaging acquisition and preprocessing ...... 123 3.4.4 Diffusion tensor imaging ...... 124 3.4.5 Diffusion basis spectrum imaging ...... 124 3.4.6 Ventricular size Measurements ...... 126 3.4.7 Human Postmortem Brains Immunohistochemistry ...... 127 3.4.8 Statistical analysis ...... 127 4 CHAPTER 4: N-CADHERIN CLEAVAGE MEDIATES VZ DISRUPTION IN PHH AND PIH ...... 129

4.1 INTRODUCTION ...... 129 4.2 RESULTS ...... 130 4.2.1 Characterization of in vitro VZ differentiation to mature ependymal cells ...... 130 4.2.2 N-cadherin mediates cell-cell adhesion mediates in VZ development ...... 133 4.2.3 Effective dose and time of blood administration required to elicit VZ disruption ...... 134 4.2.4 Blood disrupts VZ cytoarchitecture and differentiation in a time-dependent manner ...... 136 4.2.5 VZ disruption occurs in the setting of cell-cell adherens junction damage ...... 138 4.2.6 Reactive astrocytosis (neuroinflammation) occur with VZ disruption ...... 140 4.2.7 ADAM10 inhibition abrogates N-cadherin cleavage and VZ disruption ...... 142 4.2.8 Bacterial toxin causes VZ disruption that can be modulated by ADAM10 ...... 143 4.3 DISCUSSION ...... 144 4.3.1 Summary of findings ...... 144 4.3.2 The in vitro model mimics normal human VZ differentiation ...... 145 4.3.3 N-cadherin cleavage underlies the cell-cell junction pathology in VZ disruption...... 146 4.3.4 ADAM10 Inhibition abrogates N-cadherin cleavage and VZ disruption ...... 147 4.3.5 Reactive astrocytosis (neuroinflammation) occurs at the site of VZ disruption ...... 148 4.4 MATERIALS AND METHODS ...... 149 4.4.1 Animals and surgery ...... 149 4.4.2 Ventricular zone cell cultures...... 150 4.4.3 Alpha hemolysin and whole blood induction...... 151 4.4.4 Immunohistochemistry and cellular imaging ...... 151 4.4.5 Western Blot...... 153 4.4.6 Statistical analysis ...... 154 5 CHAPTER 5: DEVELOPMENT OF IN VIVO MODELS OF PIH AND PHH ...... 155

5.1 INTRODUCTION ...... 155 5.2 RESULTS ...... 156 5.2.1 Ferret model of PHH ...... 156

xvi 5.2.1.1 Subjects ...... 156 5.2.1.2 Neurological Status and Health ...... 157 5.2.1.3 Ventriculomegaly ...... 158 5.2.1.4 VZ disruption and glial activation (neuroinflammation) ...... 159 5.2.1.5 Diffusion Basis Spectrum Imaging of ferrets ...... 161 5.2.2 Mouse model of PIH ...... 163 5.2.2.1 Neurological status and health ...... 163 5.2.2.2 Ventriculomegaly ...... 163 5.2.2.3 VZ disruption and reactive gliosis (neuroinflammation) ...... 164 5.3 DISCUSSION ...... 166 5.3.1 Prevalence and time course of ventriculomegaly in PIH and PHH models ...... 166 5.3.2 Cytopathology ...... 166 5.3.3 Surgical and Methodological Considerations ...... 167 5.3.4 Limitations ...... 169 5.4 METHODS ...... 170 5.4.1 Ferret model of IVH and PHH ...... 170 5.4.1.1 Subjects ...... 170 5.4.1.2 Induction of IVH and PHH ...... 171 5.4.1.3 Ferret Neuroimaging...... 173 5.4.1.4 Sacrifice and Neurocytology ...... 177 5.4.1.5 Statistical analysis...... 178 5.4.2 Mouse Model of Ventriculitis and PIH ...... 178 5.4.2.1 Subjects and Experimental Design ...... 178 5.4.2.2 Induction ...... 179 5.4.2.3 Imaging ...... 180 5.4.2.4 Euthanasia ...... 181 6 CHAPTER 6: DISCUSSION OF DISSERTATION ...... 182 6.1.1 Leveraging a translational neuroscience approach to PIH and PHH ...... 182 6.1.2 A unifying pathophysiological mechanism for PIH and PHH ...... 183 6.1.3 Neuroinflammation is central to PIH and PHH pathophysiology ...... 186 6.1.4 Role of modulating neuroinflammation pathways to mitigate PIH and PHH ...... 188 6.1.5 Ependymal barrier damage is associated with PVWM injury ...... 191 6.1.6 VZ/SVZ differentiation modulation and ependymal barrier and PVWM repair ...... 194 7 REFERENCES ...... 197 8 APPENDIX ...... 237

xvii List of Tables

Table 1. Demographics and clinical characteristics of PIH patients and controls ...... 46 Table 2: Characteristics of 158 infants in a prospective study to assess the effects of IVH/PHH on dMRI measures of the lateral ventricular layer...... 88 Table 3: Median dMRI measures among 157 full- and pre-term infants with/or without IVH/PHH ...... 90 Table 4: Groupwise comparisons between 157 full- and pre-term infants with/or without IVH/PHH ...... 91 Table 5: Region-specific dMRI measures examining between-group differences among 158 infants ...... 93 Table 6: Characteristics of infants in a prospective study to assess DBSI indices in PVWM ..... 97 Table 7: dMRI measures of PVWM in full term and preterm infants with/or without IVH/PHH and their frontal-occipital horn ratio-dMRI correlations ...... 99 Table 8: Levels of significance for one-way and groupwise comparisons of dMRI measures in full term and preterm infants with/or without IVH/PHH ...... 100 Table 9. Summary of ferrets used in the development of an animal model of PHH ...... 157

xviii List of Illustrations, Figures and Graphics

Figure 1: Global prevalence of pediatric hydrocephalus...... 6 Figure 2: Incidence of pediatric hydrocephalus...... 7 Figure 3: Representative CT scans of post-infectious hydrocephalus...... 9 Figure 4: Representative cranial ultrasounds of intraventricular hemorrhage of prematurity...... 11 Figure 5: Embryological origins of ventricular development...... 15 Figure 6: Schematic representation of ependymal layer and choroid plexus...... 19 Figure 7. Schematic of proteogenomics experimental and data analysis workflow...... 44 Figure 8. Proteomic profile of PIH and NPIH infants ...... 48 Figure 9. Gene ontology analyses based on cerebrospinal fluid assay...... 49 Figure 10. Transcriptomic profile of PIH and NPIH infants ...... 50 Figure 11. Weighted correlation network analysis (WGCNA) of gene modules ...... 52 Figure 12. Prominent Gene Ontological (GO) functions and interactions...... 53 Figure 13. Pathway analysis of differentially expressed markers in RNA-seq and proteomics ... 54 Figure 14. Interactome analysis of cross-omic differentially expressed markers...... 55 Figure 15. Proteomic profiles of infants with PIH, PHH and non-PHH non-PIH controls ...... 57 Figure 16. Differentially expressed proteins between PIH, PHH and control infants ...... 59 Figure 17. Proteins unique to PHH and controls ...... 61 Figure 18. Proteins unique to PIH and controls ...... 62 Figure 19. Gene ontology interactome of proteins common to PIH and PHH ...... 63 Figure 20. Reactome pathway analyses of proteins common to PIH and PHH ...... 64 Figure 21: Segmentation of Regions of Interest (ROIs) ...... 86 Figure 22: Flowchart of subjects screening and enrolment for study ...... 87 Figure 23: Assessment of ventricular size ...... 89 Figure 24: dMRI measures of the lateral ventricular perimeter (LVP) ...... 91 Figure 25: dMRI measures of the frontal occipital horn perimeter (FOHP) ...... 92 Figure 26: Effects of region of segmentation on dMRI measures ...... 93 Figure 27: Representative principal eigenvector maps of white matter orientation ...... 94 Figure 28: dMRI-ventricular size correlations in the LVP ...... 95 Figure 29: dMRI-ventricular size correlations in the FOHP ...... 95 Figure 30: Flowchart of patient enrolment and selection for study inclusion ...... 96 Figure 31: Ventricular size measurement and regions of interest segmentation ...... 98 Figure 32: dMRI measures of the corpus callosum (CC) ...... 101 Figure 33: dMRI measures of the corticospinal tracts (CST) ...... 102 Figure 34: dMRI measures of the optic radiations (OPRA) ...... 104 Figure 35: Ventricular size-dMRI correlations ...... 105 Figure 36. Representative neurocytology of post-mortem PVWM tissue ...... 106 Figure 37. Representative immunohistochemistry of postmortem brain tissue ...... 108 Figure 38: Schematic rendering of postulated pathology underlying different PVWM bundles 118 Figure 39: Differentiation of VZ cells in vitro...... 132

xix Figure 40: Cell-cell junction and astrocytic expression of differentiating of VZ cells in vitro. . 133 Figure 41: Time- and dose-dependent response of blood exposure to VZ cells in vitro...... 135 Figure 42: Time-dependent effect of blood in VZ cell cultures...... 137 Figure 43: Time-dependent effect of blood in N-cadherin expression in the VZ...... 139 Figure 44: Glial activation in the VZ...... 141 Figure 45: Effect of ADAM10 modulation on blood treated VZ cells...... 142 Figure 46: Effect of ADAM10 modulation on toxin treated VZ cells...... 144 Figure 47: Model summarizing the effects of the blood on the VZ...... 145 Figure 48. Ventricular enlargement in ferret a model of PHH ...... 158 Figure 49: Immunohistochemistry in a ferret model of PHH ...... 160 Figure 50. Reactive astrocytosis in a ferret mode of PHH ...... 161 Figure 51. Periventricular white matter dMRI characteristics in a ferret model of PHH ...... 162 Figure 52. Ventricular enlargement in a mouse model of PIH ...... 164 Figure 53: Immunohistochemistry in a mouse model of PIH...... 165 Figure 54: A proposed unifying pathophysiological mechanism for PIH and PHH ...... 185

Supplementary Figure 1. Patient baseline characteristics ...... 47 Supplementary Figure 2: Transcriptome profile of patients with CMV infection...... 51 Supplementary Figure 3. Differentially expressed proteins between PIH and control infants ..... 59 Supplementary Figure 4. Differentially expressed proteins between PHH and control infants ... 60 Supplementary Figure 5. Proteomics and RNA-Seq data preprocessing and quality control ...... 79 Supplementary Figure 6. ImageJ quantification of VZ cells of interest ...... 152

xx List of Abbreviations

AD Axial diffusivity ADAM10 A Disintegrin and metalloproteinase domain-containing protein 10 ALIC Anterior limb of internal capsule ANCOVA Analysis of covariance ANOVA Analysis of variance APP Amyloid precursor proteins AQP Aquaporin BBB Blood brain barrier BSA Bovine serum albumin CC Corpus callosum CCHU CURE Children's Hospital of Uganda CCL Chemokine C-C motif ligand CMV Cytomegalovirus CNS Central nervous system CPC Choroid plexus cauterization CSF Cerebrospinal fluid CST Corticospinal tracts CT Computed tomography CXCL Chemokine C-X-C motif ligand DAPI 4’6-diamidino-2-phenylindole DAVID Database for Annotation, Visualization and Integrated Discovery DBSI Diffusion basis spectrum imaging DMEM Dulbecco's modified eagle medium dMRI Diffusion magnetic resonance imaging DMSO Dimethyl sulfoxide DNA Deoxyribonucleic acid DTI Diffusion tensor imaging DWI Diffusion-weighted image EC Ependymal cells EDTA Ethylenediaminetetraacetic acid EGA Estimated gestational age ELISAs Enzyme-linked immunosorbent assays ETV Endoscopic third ventriculostomy FA Fractional anisotropy FAD Fiber axial diffusivity FBS Fetal bovine serum FF Fiber fraction

xxi FFA Fiber fractional anisotropy FGF Fibroblast growth factor FOHP Frontal and occipital horn perimeters FRD Fiber radial diffusivity FT Full-term GFAP Glial fibrillary acidic protein GO Gene ontology GSEA Gene set enrichment analysis H & E Hematoxylin and eosin HBSS Hank’s balanced salt solution HC Hydrocephalus HCD High-energy collisional dissociation HCRN Hydrocephalus clinical research network HG-IVH High-grade intraventricular hemorrhage IACUC Institutional Animal Care and Use Committee ICBDSR International Clearinghouse for Birth Defects Surveillance and Research ICP Intracranial pressure IHC Immunohistochemistry IL Interleukins IVH Intraventricular hemorrhage JAK Janus kinase Jhy Junctional cadherin complex regulator KEGG Kyoto Encyclopedia of and Genomes L1-CAM L1 cell adhesion molecule LC-MS Liquid -mass spectrometry LDS Lithium dodecyl sulfate LPA Lysophosphatidic acid LV Lateral ventricles LVP Lateral ventricular perimeter LVP Lateral ventricular perimeter MD Mean diffusivity MDS Multi-dimensional scaling MRI Magnetic resonance imaging N-cadherin Neural cadherin NCAM-1 Neural cell adhesion molecule 1 NF-κB Nuclear factor-κb NIH National Institute of Health NK Natural killer NPHH Non-posthemorrhagic hydrocephalus NPIH Non-postinfectious hydrocephalus

xxii NRF Non-restricted fraction NSC Neural stem cells OCT Optimal cutting temperature OPRA Optic radiations P/S Penicillin/streptomycin Paeni Paenibacillus spp. PBS Phosphate buffered saline PCA Principal component analysis PCR Polymerase chain reaction PHH Post-hemorrhagic hydrocephalus PIH Post-infectious hydrocephalus PLIC Posterior limb of internal capsule PSM Peptide spectrum matches PSU Pennsylvania state university PVWM Periventricular white matter RD Radial diffusivity RF Restricted fraction RIPA Radioimmunoprecipitation assay RNA Riboneucleic acid RNA-seq Ribonucleic acid sequencing ROI Region of interest ROS Reactive oxidative species SB Spina bifida SLCH St. Louis children’s hospital SPIA Signaling pathway impact analysis STAT Signal transducers and activators of transcription STRING Search tool for the retrieval of interacting genes SVZ Subventricular zone TBS Tris buffered saline TE Echo time TGF-β Transforming growth factor beta TH Temporal horn of lateral ventricle TI Inversion time TLR Toll-like receptor TMT Tandem mass tagging TNF Tumor necrosis factor TR Repetition time TREM-1 Triggering receptor expressed on myeloid cells 1 U of C University of Calgary USFDA United States Food and Drug Administration

xxiii VA Ventriculoatrial VAD Ventricular access device VP Ventriculoperitoneal VPT Very preterm VZ Ventricular zone WBC White blood cell WGCNA Weighted correlation network analysis WUSTL Washington University in St. Louis YAP Yes-associated protein βIV Beta-IV tubulin

xxiv 1 Chapter 1: Introduction to the Dissertation

Adapted from:

1. Isaacs AM, Dahiya S, McEvoy SD, Rich KM, Limbrick DD: Developmental Origins of

the Cerebral Ventricular System and Colloid Cysts. In Colloid Cysts of the Third Ventricle:

Natural History, Decision Making, and Operative Management. Edited by Dacey R, Nakaji

P, Beaumont TL: Thieme; 2020. (Under review).

2. Dunn JF, Isaacs AM: The impact of hypoxia on barriers that protect the brain—the blood

brain barrier and beyond. Journal of Applied Physiology 2020. (Under review).

3. Patel B, Castaneyra-Ruiz L, Isaacs AM, Morales DM, Eskandari R, Limbrick DD,

McAllister JP: Experimental Hydrocephalus. In Youmans & Winn Neurological Surgery.

Volume 2. Edited by Youmans JR, Winn HR: Elsevier; 2020. (Under Review).

4. Paulson JN, Williams BL, Hehnly C, Mishra N, Sinnar S, Zhang L, Ssentongo P, Mbabazi-

Kabachelor E, Wijetunge DSS, Bredow B, Mulondo R, Kiwanuka J, Bajunirwe F, Bazira

J, Bebell LM, Burgoine K, Couto-Rodriguez M, Ericson JE, Erickson T, Ferrari M,

Gladstone M, Guo C, Haran M, Hornig M, Isaacs AM, Kaaya BN, Kangere SM, Kulkarni

AV, Kumbakumba E, Li X, Limbrick DD, Magombe J, Morton SU, Mugamba J, Ng J,

Olupot-Olupot P, Onen J, Peterson MR, Roy F, Sheldon K, Townsend RR, Weeks AD,

Whalen AJ, Quackenbush J, Ssenyonga P, Galperin MY, Almeida M, Atkins H, Warf B

C, Lipkin WI, Broach JR, Schiff SJ: The Bacterial and Viral Complexity of Post-infectious

Hydrocephalus in Uganda. Science Translational Medicine 2020. (Accepted).

5. Baksh B, Botteron H, Morales DM, Isaacs AM, Akbari SH, Han RH, McAllister JPI,

Wallendorf M, Mathur AM, Smyser CD, Limbrick DD: Challenging common views

1 regarding cerebrospinal fluid parameters: infection, hemorrhage, and post-hemorrhagic

hydrocephalus. Journal of Neurosurgery Pediatrics 2020. (In preparation).

6. Isaacs AM, Limbrick DD: Cerebrospinal Fluid Biomarkers of Hydrocephalus. In

Cerebrospinal Fluid Disorders. Edited by Limbrick DD, Leonard JR. Cham: Springer

International Publishing; 2019: 47-70.

7. Isaacs AM, Shimony JS, Morales DM, Castaneyra-Ruiz L, Hartman A, Cook M, Smyser

CD, Strahle J, Smyth MD, Yan Y, McAllister JP, McKinstry RC, Limbrick DD: Feasibility

of fast brain diffusion MRI to quantify white matter injury in pediatric hydrocephalus. J

Neurosurg Pediatr 2019:1-8.

8. Isaacs AM, Riva-Cambrin J, Yavin D, Hockley A, Pringsheim TM, Jette N, Lethebe BC,

Lowerison M, Dronyk J, Hamilton MG: Age-specific global epidemiology of

hydrocephalus: Systematic review, metanalysis and global birth surveillance. PLoS One

2018, 13: e0204926.

2 1.1 Hydrocephalus

Hydrocephalus encompasses a heterogeneous group of complex neurological disorders characterized by brain injury in the context of cerebrospinal fluid (CSF) accumulation and abnormal dilatation of the cerebral ventricles. This can occur with or without an increase in intracranial pressure (ICP) that arises from an imbalance between CSF production and clearance1,2.

1.1.1 Classification of hydrocephalus

There are several approaches to classify the “syndrome” of hydrocephalus3-7. The classic, most common system dichotomizes hydrocephalus into communicating vs. non-communicating (or obstructive) subtypes based on the absence or presence of an identifiable obstruction in the CSF pathways, respectively3-8. An age-based classification stratifies hydrocephalus into neonatal, infantile, pediatric, juvenile, transitional, adult and elderly forms2,9. From an etiological standpoint, hydrocephalus may be classified as primary vs. secondary (or acquired), based on whether the cause is unknown or a result of an identifiable antecedent neurologic event3-7. Examples of secondary hydrocephalus include post-hemorrhagic hydrocephalus (PHH; occurs following intraventricular hemorrhage (IVH) in neonates)2,10, post-infectious hydrocephalus (PIH; due to perinatal sepsis)11,12, congenital hydrocephalus (caused by genetic aberrations)7,13-15, and spina- bifida-associated hydrocephalus (seen in patients with myelomeningocele)16-18.

3 1.1.2 Clinical presentation and diagnosis

The presenting signs and symptoms of hydrocephalus are often non-specific and may include any combination of the following: tense fontanels, splaying of cranial sutures and irritability in infants and headaches, nausea, vomiting, papilledema, cranial nerve abnormalities, seizures, depressed level of consciousness, urinary incontinence and gait difficulties in all age groups2,19. The clinical diagnosis of hydrocephalus requires both the presence of related clinical signs/symptoms and confirmation of abnormal ventriculomegaly on neuroimaging defined by metrics such as the frontal-occipital horn ratio and Evan’s Index20.

In preterm neonates, head ultrasounds are the most commonly used neuroimaging modality since they can be performed at the bedside and require minimal handling of the infants. Computed tomography (CT) scans may be utilized when feasible but their use in infants and children is limited, especially in well-resourced countries due to the risk of exposure to ionizing radiation, associated with developmental impairments and radiation-induced cancers later in life21. Magnetic resonance imaging (MRI) is the preferred neuroimaging modality in infants because it does not expose patients to this ionizing radiation. MRI is also ideal for identifying sites of obstruction to

CSF flow and hemorrhage that could contribute to hydrocephalus symptoms28,55. However, standard MRIs have lengthy acquisition times and often require sedation or general anesthesia, especially in neonates and uncooperative children, which subjects them to risks of anesthesia- related complications22,23. Recently, the United States Food and Drug Administration recommended against repeated elective exposure to anesthesia in young children24. Consequently, fast brain MRI protocols have been implemented since 2002 to circumvent the need for sedation in pediatric hydrocephalus patients. Fast brain MRIs utilize short pulse sequences to acquire MRI

4 images in 2-3 seconds per slice, which are adequate for making prompt clinical decisions in most pediatric hydrocephalus patients25,26. However, due to concerns of poor image quality and susceptibility to motion artifacts, indications for fast brain MRIs in hydrocephalus are typically limited to gross anatomical/structural and ventricular catheter placement assessments.

In addition to initial diagnosis, hydrocephalus patients undergo serial neuroimaging for a wide range of indications, including assessment of response to treatment and workup for future treatment failures27. However, the conventional symptomatologic-radiologic approach to hydrocephalus often poses a diagnostic dilemma. The current metrics are neither sensitive nor specific, and they do not provide any information on the pathophysiology or natural history of the disease28.

1.1.3 Global burden of hydrocephalus

There is heterogeneity in the reported prevalence and incidence of hydrocephalus, often without reference to age or etiology29. The global prevalence of hydrocephalus is often estimated as 1 in

500-1000 individuals2,30,31 with variations based on age, etiology, geography and economic strata1,7. We performed the first systematic review and metanalysis of population-based studies reporting prevalence of hydrocephalus in both pediatric and adult populations32. From a population of approximately 172 million extracted from 52 studies, we found that the overall global prevalence of hydrocephalus was 85 per 100 thousand [95% CI, 62-116]. The pooled prevalence of hydrocephalus in the pediatric population was 72 per 100 thousand [95% CI, 58-89] without, and 88 per 100 thousand [95% CI, 72-107] with spina bifida-associated hydrocephalus. The prevalence of pediatric hydrocephalus between continents was almost two-fold higher in Africa

5 (104 per 100 thousand [95% CI, 33-325]), compared to North America (56 per 100 thousand [95%

CI, 41-75]) (Figure 1).

Figure 1: Global prevalence of pediatric hydrocephalus. Assessment of the prevalence of pediatric hydrocephalus (HC) with and without spina bifida (SB) stratified by continent shows majority of cases occur in continents with highest number of low- income countries.

To assess the incidence of pediatric hydrocephalus, we analyzed annual reports from the

International Clearinghouse for Birth Defects Surveillance and Research (ICBDSR) database spanning the most recent 11 year period33. ICBDSR, a World Health Organization affiliate, collects data on birth defects including hydrocephalus and spina-bifida from 42 surveillance programs, spanning 36 countries. We identified that the mean annual incidence of pediatric hydrocephalus was 50 per 100 thousand [95% CI, 41-60] without and 81 per 100 thousand [95%

CI, 69-96] with spina-bifida associated hydrocephalus. In addition, countries with a higher level of income were associated with a significantly lower mean incidence of hydrocephalus, 78 per 100 thousand [95% CI 65-92], when compared to low- and middle-income countries combined, 106 per100 thousand [95% CI, 76-148], p < 001 (Figure 2).

6

Figure 2: Incidence of pediatric hydrocephalus. The mean annual incidence of pediatric hydrocephalus (A) with or without spina bifida (SB) remained steady from 2003 to 2014. (B) Countries with low- to medium-income status had relatively higher annual incidences than those in the high-income stratum.

Postnatally acquired hydrocephalus, including PIH and PHH, were not included in our hydrocephalus incidence analyses as those data are not reported to the ICBDSR. However, Dewan et al. performed a similar but limited systematic review on the incidence of pediatric hydrocephalus that included PIH and PHH populations34. Together, PIH and PHH represent the largest cause of infantile hydrocephalus worldwide with a majority of PIH cases in low- and middle-income countries34-37. As the largest single cause of childhood hydrocephalus and the need for neurological surgery in children worldwide, PIH afflicts 60%37 of the estimated 300,000 infants who develop hydrocephalus each year in low- to middle-income countries34. In sub-Saharan Africa alone, it is estimated that 1 in every 200 infants will develop PIH, resulting in >100,000 affected infants in that region each year38. On the other hand, PHH represents the most common cause of pediatric hydrocephalus in Europe and North America, affecting 1 in every 1000 newborns39,40. PHH occurs

7 in up to one half of infants who develop IVH39-41, the most frequent and severe neurological complication of preterm birth, occurring in nearly 20% of preterm infants42-44. PHH causes significant neurologic injury in patients. It is associated with some of the worst neurological outcomes in newborn medicine, with persisting cognitive deficits in >85% and cerebral palsy in

70% of the affected infants41,42,44-46. Unfortunately, these alarmingly high reported global incidences of PIH and PHH are likely an underestimation because most affected infants, particularly in developing countries, die in early childhood without presenting to healthcare facilities; therefore they are omitted from neonatal mortality surveillance47.

Hydrocephalus care results in a high financial burden for patients, their families, and healthcare infrastructure because of the diagnostic process, treatment, follow-up and long-term management.

Inpatient care of pediatric hydrocephalus patients (predominantly PHH) is reported to cost approximately $2 billion per year in the United States alone48. While already substantial, this does not account for the considerable costs associated with out-of-hospital care48.

1.2 Post-infectious hydrocephalus (PIH)

PIH is a serious sequelae of neonatal sepsis and a major cause of morbidity with one million annual deaths worldwide36,47,49,50. The PIH phenotype is complex with brain calcifications, seizures, neurological injury and developmental impairments36, and a typical clinical picture of ventriculitis without prior meningitis (Figure 3).

8

Figure 3: Representative CT scans of post-infectious hydrocephalus. Axial images showing (A) the normal lateral ventricles (LV) in a healthy infant in comparison to (B & C) infants with post-infectious hydrocephalus who have severe ventriculomegaly with loculated abscesses (as indicated by the stars).

PIH continues to be a major global health problem in spite of the recent clinical efforts to optimize treatment51. Strategies to prevent PIH have been thwarted for several reasons. First, there is a lack of identification of the key pathogens responsible for the underlying septic episodes that proceed to PIH. Moreover, the environmental factors that drive the incidences of PIH vary from one region to another12,52,53. For example, it is known that seasonal Neisseria meningitidis epidemics within the African meningitis belt can produce PIH54. However, there have been reports of a tendency toward gram negative coliform bacteria in infants in other Southern55 and Eastern12 African locations where Neisseria meningitidis is uncommon. Another major systemic problem impeding progress in the fight against PIH is the lack of diagnostic approaches. Whether lumbar puncture in infants, especially those without meningeal infection, could be a diagnostic tool during their neonatal sepsis evaluation is presently unknown. In addition, CSF cultures are difficult to employ due to limited armamentarium in low- and middle-income countries where PIH is most common38.

Even in well-resourced regions where CSF analyses are readily available, the commonly tested

9 indices (e.g., glucose, protein, cell count and differential, and gram stain) are often not reliable in identifying an infection and cultures underappreciate the polymicrobial nature of PIH56.

Alternatively, high throughput and unbiased pan-microbial approaches may be required to detect microbes in these complex PIH CSF samples, and to identify pathways toward more optimal treatment and prevention of the proximate neonatal infections.

1.3 Post-hemorrhagic hydrocephalus (PHH)

Infants who are born preterm (< 37 weeks gestational age) often sustain spontaneous intracranial hemorrhage within the germinal matrix44 which may extend into the cerebral ventricles to cause

IVH 57,58. IVH, in premature infants, is classified into four grades of severity (grades I to IV)57,59

(Figure 4). In grade I, the hemorrhage is restricted to the germinal matrix. The hemorrhage is considered to be grade II or III when it extends beyond the germinal matrix into the lateral ventricles and covers less than 50% or greater than 50% of the ventricular volume with associated ventriculomegaly, respectively60,61. The severest form, grade IV, is defined as having the features of grade III in addition to intraparenchymal hemorrhage60,61. Grades III and IV are grouped into a

“high grade IVH” category because they are associated with the most severe neuromotor and cognitive impairments of preterm birth62. In approximately half of the infants who develop high grade IVH, ventricular CSF dynamics are severely disrupted, which leads to the accumulation of

CSF, raised intracranial pressures, and PHH2,42.

10

Figure 4: Representative cranial ultrasounds of intraventricular hemorrhage of prematurity. The coronal sections show (A) the appearance of normal lateral ventricles (LV) and temporal horn (TH) in a healthy infant in comparison to (B-D) preterm infants with IVH. (B) Grade I, (C) grade II and (D) grade IV. Hemorrhages are outlined or identified by stars. Scale bars = 1 cm.

1.4 Current treatments of PIH and PHH

Timely treatment of hydrocephalus, particularly for acute cases with raised ICP is critical because it is associated with progressive neurologic injury and high mortality rates ranging from 20 to

87%30,31,45,63,64. With a primary goal of decreasing ventricular size, resolving raised ICP, mitigating patient symptoms and preventing further neurologic injury, the only effective treatment for hydrocephalus is surgical CSF diversion2. CSF diversion may be performed as a temporary or permanent intervention depending on the clinical indication51,65-68. Temporary procedures include

11 insertion of ventricular reservoirs or external ventricular drains69, which allow serial external CSF removal. However, the timing and volume of CSF removal are highly variable with little evidence- based practice or guidelines. Patients who demonstrate a need for long term treatment typically undergo ventriculoperitoneal (VP)70,71 or ventriculoatrial (VA)72-74 shunt insertion, which divert

CSF into the peritoneal cavity or right atrium of the heart, respectively74. Other extracranial compartments such as the lungs75 and gallbladder76, capable of absorbing CSF into systemic circulation may be used.

Shunts have been the mainstay of hydrocephalus treatment for over 60 years77. However, their associated risks and complications remain unacceptably high, with 50% failing within the first two years of implantation71, subjecting patients to multiple revision surgeries and poor quality of life.

An alternative approach to shunting is endoscopic third ventriculostomy (ETV), which establishes a CSF pathway between the ventricles and the subarachnoid space through a fenestration made in the floor of the third ventricle66,78,79. ETVs may be coupled with choroid plexus cauterization

(CPC) to gain added benefits of decreasing CSF production and dampening the negative effects of choroid plexus pulsatility51,80-82. However, ETV with or without CPC have not been shown to be more efficacious than shunting51,83.

Attempts at non-surgical treatment for hydrocephalus in humans have been unsuccessful, and there is no effective medical therapy yet84-88. Decorin, a transforming growth factor beta (TGF-β) inhibitor has been shown to prevent ventriculomegaly in rats with infantile communicating hydrocephalus89,90 and PHH91,92. Magnesium sulfate has been shown to improve gait performance and lower reactive astrocytosis in hydrocephalic rats93. In rat models of congenital hydrocephalus,

12 intraperitoneal administration of minocycline reduced the markers of neuroinflammation and increased cortical mantle thickness94-96. Intraventricular infusion of nimodipine, a calcium channel antagonist has been shown to reduce white matter damage and improve sensorimotor function and spatial learning in juvenile hydrocephalic rats 97,98. However, all the pharmacologic agents tested to date have shown to be variable with limited short-term success, even in the animal models84-98, and none have translated into an efficacious human hydrocephalus treatment88.

1.5 Current understanding of the pathophysiology of PIH/PHH

Although significant positive strides have been made in the context of advances in infection control71,99, shunt hardware100,101, and surgical technique51,66,78-82,87,102-107, the outcomes of hydrocephalus remain dismal71. Unfortunately, hydrocephalus surgery early in life is largely palliative and at the time of diagnosis over 80% of the deleterious effects of hydrocephalus are already established and are irremediable. Therefore, the most significant global health impact on

PIH and PHH will require directing efforts to defining disease pathophysiology to target both the underlying causes and mechanisms of neurological injury12,52,56. Characterizing the pathophysiology of PIH and PHH may also direct the discovery of biomarkers that will promote early identification and diagnosis, predict treatment efficacy, alert treatment failure in a timely fashion and facilitate the comparison of prevention strategies and treatment modalities in clinical trials. Furthermore, if PIH/PHH pathophysiology is better characterized and understood, emphasis could shift from palliation of CSF accumulation51 to prevention84,85,87.

13 1.5.1 Embryological overview of the cerebral ventricular system

As mentioned above, characterization of the pathophysiology of PIH and PHH renders in the development of diagnosis, treatment and prevention strategies. Knowledge of the ventricular system, ependymal barrier, and choroid plexus, particularly in the context of the development of the central nervous system (CNS) is essential to understand the PIH and PHH pathophysiology.

1.5.1.1 Ventricular development

Human development originates from the three-layer primitive embryonic structure: ectoderm, mesoderm and endoderm. These are formed through purposeful cell movement and organization following fertilization108. During neurulation, around the second week of embryonic development, the neural plate thickens and invaginates into a neural groove along its midline which then fuses into a rostro-caudal neural tube108,109. The neural tube momentarily remains in communication with the surrounding environment via its rostral (anterior) and caudal (posterior) neuropores, until they both eventually close off to form the ventricular system108 (Figure 5).

14

Figure 5: Embryological origins of ventricular development. During neurulation, the anterior and posterior neuropores close to isolate the ventricular system from its embryonic environment. Formation of the complex five-vesicle structure and differential expansion of regions around the neural tube flexures and the neural canal create the cerebral ventricles.

Around the third week of embryonic development, the rostral end of the neural tube undergoes rapid morphogenetic movement and expansion to develop three symmetric primary swellings: the prosencephalon (forebrain), the mesencephalon (midbrain), and the rhombencephalon (hindbrain).

This three-vesicle stage is facilitated by a symmetric folding of the neural tube rostrally along its lateral axis to form two flexures: the cephalic flexure which develops between the prosencephalon and mesencephalon, and the cervical flexure, which divides the rhombencephalon and the spinal cord108. Due to significantly restricted morphogenesis at the cephalic flexure in relation to the rest of the prosencephalon, two distinct structures develop: the rostral telencephalon and the caudal diencephalon. At approximately the sixth week, the pontine flexure develops to demarcate the rhombencephalon into the myelencephalon and the metencephalon. Thus, by the seventh week,

15 the three primary vesicles have developed into five secondary vesicles108, which are (from rostral to caudal): telencephalon (future cerebral hemispheres), diencephalon (future thalami, hypothalamic and optic cups) mesencephalon (future midbrain), metencephalon (future pons and cerebellum) and myelencephalon (future medulla oblongata)108 (Figure 5).

Development of the ventricular system is a fascinating and clinically important topic in its own right and is integral to understanding the complexities of ventricular-associated pathologies, such as hydrocephalus. In the developed human brain, the ventricular system comprises of two lateral

(telencephalic) ventricles that connect through the foramen of Monro to the third (diencephalic) ventricle, which in turn connects to the fourth (rhombencephalic) ventricle via the cerebral aqueduct. Although the ventricular system is continuous with the central canal of the spinal cord in development, and potentially thereafter, it communicates with the subarachnoid space through the foramen of Magendie and foramina of Lushka at the level of the pons.

Embryonic ventricular development is heralded by the closure of the anterior and posterior neuropores, which are connected at the midline by a CSF-filled lumen lined by ependymal cells along the neural canal. Osmotic absorption of water by the cells lining the primitive ventricles and secretion of fluid into the ventricular cavity from the choroid plexus lead to physical forces during neural tube expansion. These forces and interactions mediate the development of the three- dimensional ventricular structure from the primitive neural tube configuration. The segment of the neural canal that spans the telencephalon develops into the lateral ventricles. The lateral ventricles maintain a large inflated shape due to massive CSF production by the choroid plexus in those regions. The C-shaped configuration of the lateral ventricles occurs as a result of bilateral

16 ventromedial expansion of the ganglionic eminence bilaterally108. During the sixth week of gestation, the initial slit-like communication between the diencephalon and bilateral telencephalon vesicles enlarges forming the foramen of Monro. While the cavity in the diencephalon forms the third ventricle, the cerebral aqueduct forms from the remnant of the neural canal at the level of the mesencephalon. The fourth ventricle develops from the splaying of the pontine flexure. Unlike the fourth ventricle, which enlarges during formation of the rostral neural tube structures, the cerebral aqueduct is relatively large during early embryological stages but later narrows. The remainder of the neural canal stretches inferiorly as the central canal of the spinal cord. Around weeks 10-12 the lateral ventricles and the fornices are separated into the left and right by a thin membranous bridge; the septum pellucidum110. The septum pellucidum develops inextricably with the corpus callosum and the anterior and hippocampal commissures from the basal medial portion of the lamina terminalis known as the commissural plate110-112 (Figure 5).

From a bulk flow perspective, CSF moves rostro-caudally from the lateral ventricles, propelled by the ependymal cilia, through the foramen of Monro into the third ventricle. It then moves through the cerebral aqueduct and enters the spinal subarachnoid space via the foramina of Luschka and

Magendie. From the subarachnoid space, it has been traditionally thought that CSF is absorbed into systemic circulation exclusively at the arachnoid granulations113-116. However, other pathways including some cranial and dural routes113-116, and a glymphatic pathway117-121, have been recently described. The glymphatic pathway is a glia-based paravascular route for the clearance of soluble proteins and metabolic waste products from the brain to the lymphatic networks117-121.

17 1.5.1.2 Ependymal layer development

The inner perimeter of the cerebral ventricles is lined by the ependymal layer, a monolayer of multiciliated cuboidal to columnar epithelial cells122 held together near the apical surface by cell- cell junctional proteins. Embryonically, the ependymal layer is derived from the ventricular zone

(VZ), a single layer of monociliated neural stem cells (NSC) that populates the germinal matrix.

The VZ and the subjacent sub-ventricular zone (SVZ) are also the primary sites of neurogenesis for the brain123. During fetal development, NSCs in the VZ migrate via radial glial cells123-126 to differentiate either into neurons of the cerebral cortex or glia127,128 (e.g., ependymal cells)127. The

VZ spans the entire neural tube during neurulation. However, it is replaced by the ependymal layer as the brain matures, and stem cells are largely limited to specific areas of the SVZ. These stem cells retain restorative and repair capabilities; for example, they can regenerate neurons of the olfactory bulb and dentate gyrus via the rostral migratory stream129,130. One major role of the ependymal layer is to facilitate CSF flow through a highly coordinated beating of its apical cilia to move debris. Another major role is to serve as a protective barrier that shields the juxtaposed basal subependymal gray and white matter tissues from CSF toxins131. The predominant cell junction proteins that connect ependymal cells to maintain their structural integrity and function are adherens junctions (e.g., N-cadherin) and cell adhesion molecules (e.g., L1-CAM and NCAM-

1) (Figure 6). However, some gap junctional proteins, such as connexins132, and transitory expression of tight junctions during fetal development133 have been identified. However, the persistence of tight junctions in mature ependyma remains debatable.

18

Figure 6: Schematic representation of ependymal layer and choroid plexus. Mature multiciliated epithelial cells are structurally organized in a monolayer with intervening cell-cell adherens junctional proteins to form a partial CSF-brain barrier. The choroid plexus on the other hand comprises a monolayer of choroid plexus epithelial cells connected by tight junctions.

1.5.1.3 Choroid plexus development

As development of the neural tube progresses, a shallow sulcus limitans at the caudal end demarcates the roof and alar plates from the floor and basal plates. Further differentiation of non- neural tissue at the roof plate forms the highly vascularized choroid plexus that is found within each cerebral ventricle134. After neural tube closure, the choroid plexus appears first in the fourth ventricle, followed by the lateral ventricles synchronously, and lastly by the third ventricle135.

Despite their distinct appearance, the choroid plexuses in the lateral ventricles merge at the exit

19 levels of foramina of Monro and fuse with the third ventricle plexus to form a continuous choroid plexus structure135-137. The many functions of the choroid plexus, such as CSF secretion135,138, solute transport139, and immune system regulation140, have been extensively described in the literature. Histologically, the choroid plexus comprises a network of connective tissues and fenestrated capillaries, surrounded by cuboidal epithelial cells that are held together by tight and adherens junctions to form the blood-CSF barrier. A wide range of transporters, including aquaporins, Na+-K+-ATPase, Cl--HCO3, Na+-K+-Cl- and glucose transporter 1 have been identified as facilitators of the choroid plexus’ CSF production mechanism141.

1.5.2 Hydrodynamic and pulsatility concepts of hydrocephalus

The traditional pathophysiological perspective on hydrocephalus considers impairment of CSF flow dynamics, i.e. CSF overproduction and/or impaired uptake as the primary mechanisms1,142,143.

CSF overproduction has been mainly attributed to hypersecretion due to alterations in choroid plexus physiology144-147. In an acute septic ventriculitis mouse model, Cardia et al. reported ultrastructural changes of the choroid plexus epithelium that led to CSF overproduction and PIH144. In an experimental obstructive hydrocephalus model, Karimy et al. demonstrated that hydrocephalus was propagated by neuroinflammation-induced choroid plexus hypersecretion145.

In terms of CSF absorption impairment, the main theory is that CSF egress pathways, such as the cerebral aqueduct and arachnoid granulations undergo gliosis and scarring that impair CSF flow leading to CSF accumulation within the ventricles121.

20 Once hydrocephalus is established, there are progressive effects on the biomechanical properties of the brain. Di Rocco et al. demonstrated that ventricular dilation is likely propagated by abnormal intracranial pulsatility148, which may be attributed to an increased cerebral capillary pulsatility and/or the loss of an endogenous vascular dampening mechanism149-152. Increased capillary pulsatility in hydrocephalus may have several pathophysiologic consequences, including the loss of parenchymal microvessels153-156 and compromise of the blood-brain barrier157-159. Both of these effects might explain the marked loss of cerebral perfusion that has been well documented in clinical160-162 and experimental163-165 hydrocephalus. Importantly, it has been shown that excessive pulsatile stress can impair hemodynamics through changes in endothelial cell homeostasis166 mediated by nitric oxide167,168. These examples all provide a compelling case for the importance of CSF pulsatility in the pathogenesis of hydrocephalus.

As additional details of the biological mechanisms of CSF hydrodynamics and pulsatility emerge, pharmacological targeting of these systems may be utilized to modulate CSF physiology in PIH and PHH. Nevertheless, imbalances in CSF flow are only a part of a much more complex pathophysiology of hydrocephalus. Indeed, recent experimental169-178 and clinical studies169,179-183 have confirmed the pathophysiology of hydrocephalus is multifactorial, encompassing a complex interplay between neuroinflammation, edema, reactive gliosis, perturbations of the neurogenic niche and other neurodevelopmental processes, distortion of brain parenchyma, cerebral hypoxia and ischemia, axonal degeneration and demyelination, and neuroglial apoptosis87,102,184-190.

21 1.5.3 Phenotypic similarities between PIH and PHH

For the purposes of this work, it is critical to note that PIH and PHH have very similar features suggesting they may share common pathophysiological mechanisms. Clinically, the signs and symptoms associated with PIH and PHH after their respective antecedent event are indistinguishable. Radiologically, PIH and PHH are both classified as communicating hydrocephalus with features of diffuse grey and white matter injury, such as T2-weighted hyperintensities indicating tissue disruption191,192, susceptibility weighted imaging evidence of petechial hemorrhages193-195, restricted diffusion representing tissue ischemia193-195, and disrupted white matter water diffusivities on diffusion MRI (dMRI) suggesting PVWM damage. On gross pathology and histology, both PIH and PHH are characterized by many features of inflammation and basal cisterns fibrosis36,85-87,190,196,197. Interestingly, based on the many phenotypic similarities between PIH and PHH, a recent review by Karimy et al. classified them collectively as an

“inflammatory hydrocephalus” subtype198. This is critical because the commonalities suggest that in response to the antecedent neonatal sepsis or hemorrhage, neuroinflammation pathways are activated to mediate similar patterns of brain injury seen in both PIH and PHH86,87,190,197.

Furthermore, it is also possible that host-mediated neuroinflammation causes diffuse brain scarring and obstruction of CSF pathways, which results in hydrocephalus.

1.5.4 Role of neuroinflammation in PIH/PHH pathogenesis

Neuroinflammation, the host immune response to cues that threaten nervous tissue integrity, is a consistent finding in many forms of hydrocephalus and likely underlies the chronic problems that plague hydrocephalic patients due to glial scar formation199.

22 In PIH, the role of neuroinflammation may be well-defined in the case of a systemic infection.

Typically, once a pathogen breaches a host’s epithelial barriers, innate immune mechanisms eliminate the microbe. Sepsis, a systemic inflammatory response, occurs when the pathogen is not effectively eliminated. With a persistent pathogenic material, multiple pathways including the

Toll-like receptor (TLR)200, Janus kinase/signal transducers and activators of transcription

(JAK/STAT)201,202 and NF-κB/TREM-1 signaling203 pathways are activated elevating inflammation. Systemic inflammation typically makes the blood-brain barrier permeable to microbes and other systemic inflammatory mediators that traverse into the subarachnoid space.

This leads to the activation of neuroinflammatory cascades, production of nitric oxide metabolites, hypoxia, and decreased glucose metabolism in the brain and CSF204-206 culminating in brain injury and subsequent PIH.

In PHH, the presence of blood and blood products in the CSF initiates TLR2 and complement activation resulting in a release of cytokines (e.g., interleukins (IL) and tumor necrosis factors

(TNF)) and chemokines (e.g., Chemokines C-C (CCL) and C-X-C (CXCL)) that culminate in ependymal gliosis, arachnoiditis, and fibrotic blockage of CSF pathways to cause hydrocephalus85,196. Our group recently showed that the CSF of PHH infants contains significantly higher levels of IL-1α, IL-4, IL-6, IL-12, TNF-α, CCL-3, CCL-19, and CXCL-10 compared to healthy control infants207. Several other investigators have utilized targeted assays to demonstrate strong associations between PHH and CSF inflammatory markers199,208-210.

Therefore, it is important to characterize the nature of the host-immune response in PIH and PHH to understand the pathophysiology. This will allow us not only to address PIH and PHH as the

23 most prevalent variants of hydrocephalus, but also to gain further insights into other forms of hydrocephalus.

1.5.5 Effect of PIH and PHH on neurodevelopment

In humans, VZ/SVZ differentiation to form the ependymal layer continues into the third trimester and newborn period211. During these critical periods, any perturbations in the VZ/SVZ, particularly by IVH or infection, can have deleterious effects on brain development and CSF physiology.

McAllister et al. reported that VZ/SVZ disruption is a key feature of IVH in preterm human infants197 and is characterized by alterations in NSCs, VZ/SVZ cell loss, neuroepithelial injury, and gliosis in the affected areas. Blood and blood products, including iron, have also been shown to elicit similar effects on neurogenesis in experimental models85,212-214. Yung and colleagues reported that lysophosphatidic acid (LPA), a blood component that also plays a major role in neurogenesis, neuronal differentiation, and apoptosis of neural progenitor cells, consistently causes hydrocephalus when injected into the lateral ventricles of fetal mice214. LPA induction of hydrocephalus depends strongly on expression of the LPA1 receptor by neural progenitor cells in the VZ. Increased levels of LPA promotes the transfer of Yes-associated protein (Yap), a transcriptional regulator involved in the maturation of ependymal cells, from the cytoplasm to the nucleus in VZ cells. Nuclear Yap is associated with agenesis of ependymal cells leading to VZ disruption and hydrocephalus215. VZ disruption in the pathogenesis of other forms of hydrocephalus has been demonstrated by a series of studies on mice with genetic defects171,175-

177,216-220.

Accumulating evidence suggests that the VZ/SVZ disruption and loss of NSCs in many forms of hydrocephalus likely results from disruption of cell junctional proteins. For example, in congenital

24 hydrocephalus, in genes encoding proteins involved in intracellular trafficking and cell junctional complexes have been identified169,172,176,215,221-224. In addition, many forms of human congenital hydrocephalus result from genetic alterations of cellular adhesion proteins such as N- cadherin, connexin, and L1CAM169. A series of studies on hydrocephalus with hop gait (hyh) mice175,177,216-219, which develop perinatal aqueductal stenosis also support the presence of a genetic defect in cell junction complexes as an underlying cause of hydrocephalus. Furthermore, impairment of cell-cell junctional complexes has been implicated in the subcommissural organ alterations that lead to aqueductal stenosis170,225,226.

ADAM10, a metalloprotease, cleaves a wide range of cell-cell junctional proteins and regulates cell-cell interaction227. For example, hyperactivity of ADAM10 induces disruption of lung epithelial cells through the loss of cadherin-dependent junctions228. Preliminary results from in vitro approaches (described in Chapter 4) provide strong support for a potential mechanistic role of ADAM10 in the VZ/SVZ disruption observed consistently in PIH and PHH. It is quite possible that cleavage of adherens junction proteins such as N-cadherin by ADAM10 could create downstream effects on the neurogenic niche, such as ependymal denudation, eruption of neural precursors into the CSF, and formation of heterotopia. However, whether this disruption stimulates further inflammatory responses from astrocytes and microglia is another key question to be answered. Overall, there is a critical association between genetic susceptibility, cell-cell junction regulatory molecules, and hydrocephalus; therefore, setting the stage for exploring therapies that can mitigate these processes.

25 1.5.6 Periventricular white matter (PVWM) injury in PIH/PHH

One of the major effects of hydrocephalus is PVWM disruption. This disruption often presents as

T2 hyperintensities on MRI, and is histologically characterized by rarefactions, edema, astrogliosis, dysmyelination, hemosiderin-laded macrophages, and decreased neurofilament proteins175,178,197,229,230. Anatomically, the VZ/SVZ layer is juxtaposed by a thin layer of white matter fibers medial to large bundles of PVWM tracts such as the corpus callosum, internal capsule and optic radiations. When the VZ/SVZ and immediate PVWM fibers, a region we have collectively termed as the lateral ventricular perimeter (LVP)231, are disrupted by hydrocephalus232,233, the larger bundle of PVWM tracts, which carries ascending and descending motor and sensory fibers, is at risk of injury by CSF toxins and neuroinflammation234.

Hydrocephalus-related PVWM injury has been associated with neuromotor and cognitive developmental deficits in children131,187,235-239 and have been implicated in most of the debilitating neurological symptoms in affected infants240-242. Because the LVP can also be damaged by the expanding ventricles, PVWM tracts remain chronically vulnerable throughout the entire period of progressive hydrocephalus. Therefore, characterization of the LVP and large bundle PVWM tracts is indispensable for understanding PIH and PHH pathophysiology in addition to providing insights on the relationships between LVP injury and PVWM-related neurological function.

1.6 Proposed strategies

While important strides have been made in defining the pathophysiology of hydrocephalus, there remain unanswered questions in the field of PIH and PHH pathophysiology. Expert panelists at hydrocephalus research workshops coordinated by the National Institute of Health (NIH) have

26 outlined some of the significant gaps in hydrocephalus studies applicable to PIH and PHH research87,102. For example, how does ventriculitis or IVH cause CSF dysregulation? And what genetic mechanisms or pathology-induced epigenetic changes occur in the subset of patients who progress to develop PIH or PHH? Secondly, what are the reversible and irreversible cellular responses to inflammation and mechanical distortion by ventriculomegaly? Thirdly, which biomarkers are most useful for the diagnosis, prognosis, and monitoring of PIH and PHH infants?

And can those biomarkers be used to predict hydrocephalus before it develops? Lastly, perhaps more importantly, what pharmacologic strategies might be effective in reducing the myriad of pathophysiologic changes or preventing the development of PIH and PHH?

To address some of these questions, this dissertation utilizes genomics, proteomics, proteogenomics, dMRI, neurocytology and animal models as complementary approaches to characterize the pathophysiology of PIH and PHH and to examine potential therapeutic targets.

1.6.1 CSF genomics and proteomics

The molecular composition of CSF reflects the physiological status of the neural structures it bathes making it an indispensable medium for assessing changes that occur in the brain and for identifying genetic and proteomic footprints of disease pathogenesis. CSF also harbors molecular components necessary for neurodevelopment, neuromodulation and neurotransmission which may also be altered by pathological processes. In hydrocephalus, the initial injury, raised ICP, and ventricular stretch are all capable of initiating injurious cascades that can disrupt the CSF micro- and macro-environment39,87,102,185,187,189. Thus, the importance of CSF analyses in hydrocephalus cannot be overstated142,143,207,243-249.

27 There are several approaches to CSF analysis, including those done routinely to measure levels of clinical metabolites250,251 and targeted assays238,245-247,249,252,253 such as enzyme-linked immunosorbent assays (ELISAs) and western blots. While routine biochemical analyses and targeted assays are informative, they both have inherent limitations, such as lack of sensitivity and specificity, that preclude their utility for characterizing the multifactorial pathophysiology of and differentiating between PIH and PHH. We recently performed a retrospective review of the routine clinical lab CSF results of a cohort of 176 infants with CNS infections, IVH/PHH infants without infection, and controls with no known neurological injury. We found that the CSF profile of non- infected PHH infants is indistinguishable from that of infants with bacterial CNS infections. Both conditions presented with higher leukocyte counts, higher protein but lower glucose concentrations relative to healthy controls. Recently, the use of targeted assays to analyze CSF in hydrocephalus has gained momentum. Morales et. al. utilized ELISAs to analyze CSF samples obtained from preterm IVH infants and control infants246. They reported an increase in amyloid precursor proteins

(APP) and APP-related peptides in PHH246,249 and a correlation between ventricular size reduction and reduced levels of those markers245. Mounting evidence suggests that APP and its isoforms are associated with neurodevelopment and synaptogenesis254-256, and their disruption invariably leads to axonal injury257-259. As such, the elevated levels of APP and its isoforms in PHH may be attributed to a cascade of axonal disruption caused by the inciting hemorrhagic insult and the ventricular distension238,252. Presumably the reduction of APP levels following resolution of ventriculomegaly245 signals a reversal of the pathologic process. CSF targeted assays have also shown that infants with PHH have alterations in cell junction molecules including L1CAM,

NCAM-1 and N-cadherin247,253. Despite their utility, targeted assays require a priori knowledge of which molecules to investigate, thus only permitting the isolation of a limited number of proteins

28 instead of exploring the large CSF proteome of hydrocephalus207,245,246. These limitations necessitate the need for using high throughput discovery-based strategies such as genomics and proteomics to comprehensively define the molecular hallmarks of PIH and PHH pathophysiology.

A fundamental question in understanding any disease is the relationship between gene expression and biological function. Large scale genomic studies have already provided some insights into the role of genetics, especially those related to neurodevelopment in congenital hydrocephalus7,15.

Harnessing this genomic approach, Schiff and colleagues’ recent work involving Sub-Saharan

African infants with neonatal sepsis12,56 has offered evidence that genomics can be utilized to define PIH and PHH pathophysiology. However, the literature search did not reveal any studies to date that have used genomics to characterize the effect of hemorrhage, infection or PIH/PHH on the CSF genomic profile of infants.

On the other hand, CSF proteomics has been utilized, mostly by our group, to explore candidate protein biomarkers of hydrocephalus247. These biomarkers were subsequently examined with targeted assays to outline their clinical implications as they pertain to ventricular size and intracranial pressure245. Using tandem mass tagging (TMT)-10 proteomics, Morales et al. identified over 400 human CSF proteins involved with various neurodevelopmental processes including cell adhesion molecules, synaptogenesis, extracellular matrix and oxidative metabolism247. However, these experiments were performed in a small cross-sectional cohort of

PHH infants and such level of detailed analysis has not yet been applied to PIH.

29 1.6.2 Proteogenomics

Proteomics and genomics are important strategies for defining the pathophysiology of disease and identifying biomarkers, individual genes, sets of genes and biologically functional proteins.

However, these approaches in isolation cannot comprehensively explain the clinical phenotype of a disease260. Rather, disease pathogenesis involves the combinatorial interaction of regulatory elements within the proteome, transcriptome, and other environmental factors261. Proteogenomics is an emerging field that integrates deep-scale proteomics with next-generation genomic and transcriptomic data to define network and pathway level linkages to probe molecular mechanisms of disease262. Proteogenomic findings inform functional studies that lead to detailed insights into the biology of disease and, ultimately, new therapeutic approaches and novel drugs. The nucleotide sequencing data obtained from a proteogenomic study can be used to improve protein identification through comprehensive protein sequence database construction. Proteomic data can also be used to demonstrate the validity and functional relevance of novel findings based on large scale RNA-seq and novel coding transcripts262.

Following proteogenomic integration, gene set enrichment analysis (GSEA) can be utilized to perform signaling pathway analyses on proteogenomic data to identify dynamic interactions among genes263-265. In GSEA, differentially expressed proteogenomes are layered atop knowledge- based physical or biochemical interaction scaffold networks265. In PIH and PHH, these functional analyses can be carried out by ranking differentially expressed genes to calculate a score that reflects the degree to which a given pathway is represented at the extremes of the entire ranked list265. The significance of the gene set scores can be determined by using permutations of the samples266. It is important to note that like other existing pathways analyses, GSEA focuses on the

30 number of differentially expressed genes observed in a given pathway, but it is unable to either address topology or assess the impact of perturbations on gene-gene interactions. To address these important limitations, signaling pathway impact analysis (SPIA)267 may be used. SPIAs permit the calculation of a global pathway significance p value, which factors a perturbation p-value into the

GSEA p-values to limit data noise in signaling pathways and increase their sensitivity and specificity to the pathology of interest. To identify interactions beyond direct protein-protein binding, differentially expressed genes in PIH and PHH can also be entered into the search tool for the retrieval of interacting genes/proteins (STRING) database to capture functional links, such as substrates that connect PIH/PHH-related proteins in a metabolic pathway or proteins that are involved in large multi-protein assemblies268.

1.6.3 Diffusion magnetic resonance imaging (dMRI)

CSF perturbations due to an infection or hemorrhage invariably culminate in brain injury. This injury can be seen on conventional MRI as global changes in grey and white matter, including loculations, calcifications, necrosis and infarcts. However, conventional MRI is unable to examine pathophysiologic, microstructural changes of brain tissue. In contrast, dMRI is capable of delineating white matter integrity by assessing the magnitude and directionality of water molecules within tissues269-271 to inform microstructure and detect aberrations in white matter tissues that underlie PVWM injury272.

31 1.6.3.1 Diffusion tensor imaging (DTI)

Diffusion tensor imaging (DTI) is a commonly used dMRI technique, with many clinical and experimental applications. Standard DTI measures include: fractional anisotropy (FA), which quantifies water diffusion anisotropy; axial diffusivity (AD), which assesses the rate of water diffusion parallel to the direction of axons; radial diffusivity (RD), which assesses the rate of water diffusion orthogonal to the direction of axons; and mean diffusivity (MD), which is an average of

AD and RD270,271,273. With FA, AD, RD and MD measurements, DTI can determine PVWM integrity 271,273,274 and structural changes associated with hydrocephalus through its imaging of the orientation-independent magnitude of water diffusion. DTI has been widely used to assess PVWM integrity in hydrocephalus238,239,270,271,273,275-287 which has been shown to correlate with neurodevelopmental outcomes in hydrocephalic children238,276,284,287,288. For example, Mangano et al. showed that motor scores negatively correlated with MD scores in the internal capsule of hydrocephalic patients, who also had significantly lower conceptual, practical, and social ABAS-

II composite scores than healthy controls284. Higher diffusivity values in the corpus callosum have also been associated with poor psychomotor development index scores at approximately two years following hydrocephalus diagnosis289.

DTI measures have been shown to improve or normalize following shunt surgery284,287,290,291. For example, we recently reported a reduction in internal capsule and corpus callosum MD indices in

PHH and congenital hydrocephalus infants to levels similar to controls following 15 months of shunt surgery for hydrocephalus treatment292. Tan et al. demonstrated that higher FA measures correlated with better clinical outcomes as assessed by the Headache Disability Inventory and the

Hydrocephalus Outcome Questionnaire among 21 adolescent and young adult patients who had

32 been previously treated for hydrocephalus293. While these pieces of evidence suggest that serial

DTI may have both diagnostic and prognostic value294, there is an ongoing debate regarding the utility of DTI as a clinical tool for managing hydrocephalus in patients. This is because of the complete reliance on conventional high-resolution multi-sequence MRIs, which require long acquisition times up to 60 minutes per scan. However, we recently developed a fast brain MRI protocol that has an average entire brain acquisition time of approximately six minutes, from which

MD can be calculated. With our protocol, the diffusion tensor is computed by averaging the diffusion measurement in three axial directions derived from the conservation of the trace of the diffusion tensor under rotational transforms. We tested the feasibility of this protocol in 40 infants with PHH or congenital hydrocephalus alongside healthy controls. The results demonstrated that fast brain MRIs can be extended beyond anatomical assessments to evaluate the integrity of

PVWM in pediatric hydrocephalus patients, limiting the need for sedation for dMRI analyses292.

Despite its utility and many advantages, DTI has inherent limitations that make it challenging to delineate white matter pathology with adequate sensitivity and specificity295-297. For example, DTI is not capable of resolving crossing fibers296,298,299 or quantifying axonal density299,300.This is in part because DTI utilizes a single tensor model in which the white matter fibers in a voxel are represented as a single population, despite the fact that 60%-90% of white matter voxels contain a more complex configuration of white matter fibers, including crossing fibers. Secondly, in multi- faceted white matter pathologies such as PIH and PHH, each voxel may contain water populations representing myelinated, dysmyelinated or unmyelinated axons. Moreover, DTI has been shown to lack the sensitivity to quantify myelination under similar circumstances299,300. Furthermore, neuroinflammation, which is a major pathophysiology in PIH and PHH, leads to multiple water

33 compartments, including varying cell densities and morphologies due to reactive astrogliosis, edema, and axonal loss207,208,230. There are alternative dMRI approaches296,297,301-308, such as diffusion basis spectrum imaging (DBSI)307-312, that can address some of DTI’s limitations; however, they have yet to be utilized for PIH, PHH or other forms of hydrocephalus.

1.6.3.2 Diffusion basis spectrum imaging (DBSI)

Diffusion basis spectrum imaging (DBSI) is a relatively new dMRI technique best suited for examining complex multicompartmental cellular processes in the white matter. Unlike DTI, water displacements in DBSI are represented as multiple diffusion tensors each representing different water compartments. This allows separation of crossing fibers and estimation of crossing angles, directional diffusivity of individual crossing fibers, and axonal loss307-313. In addition to anisotropic diffusion compartments, DBSI includes isotropic tensors, which reflect water compartments in inflammatory cells and edema water307,311,313. DBSI has been validated in clinical308-311 and preclinical/phantom studies307,309,314-316 of multiple sclerosis307-309,311,314, epilepsy315, spinal cord injury317, and cervical spondylotic myelopathy310. The extended capabilities of DBSI suggest that it may provide more informative biomarkers than DTI for the differentiation and quantification of

PVWM integrity in hydrocephalus. There are several benefits of utilizing DBSI for the assessment of hydrocephalus-related PVWM injury. First, DBSI may provide parameters that more directly reflect underlying tissue microstructural changes. Second, DBSI may provide an opportunity to comprehensively investigate complex aspects of structural brain development in hydrocephalus and track their association with neurodevelopmental outcome. Third, DBSI may be used to delineate the relationship between ventricular volume, which is readily modified with current standard-of-care neurosurgical hydrocephalus treatments, and therapeutic response following

34 surgical treatment of hydrocephalus. Lastly, DBSI may be used to inform clinical trials that assess modalities focused on the clinical management or prevention of PVWM injury propagation.

1.6.4 Neurocytology

Immunohistochemistry is perhaps the most definitive modality for assessing the effects of hydrocephalus on brain microstructure. General cellular structure is often assessed with hematoxylin and eosin (H&E) stains. With a focus on neurodevelopment, neuroinflammation and

PVWM injury, single- and double-labeling immunohistochemistry with antibodies specific to cell adhesion molecules, neural stem cells, ependymal cells, and glial activity are valuable tools that can be leveraged to evaluate the effects of PIH/PHH on these processes. For example, anti-βIV tubulin can be used to evaluate multiciliated ependymal cells and undifferentiated monociliated cells, anti-N-cadherin for adherens junctions, anti-ADAM10 for metalloprotease activity, anti-

GFAP for immature ependymal cells and reactive astrocytes, and anti-Iba1 as a surrogate for microglia-related products. Following staining, epifluorescence or confocal microscopy are commonly used to visually examine cellular structure. VZ/SVZ damage are histologically characterized by disrupted adherens junctions, reduced number of multiciliated (mature) ependymal cells, radial glial cell disorganization, astrocytic proliferation, and ependymal rosette formation142,197. PVWM damage is typically observed as rarefactions, edema, astrocytosis, gliosis, dysmyelination, hemosiderin-laden macrophages, and decreased neurofilament proteins197,230,242.

Our group recently developed an in-house script that utilizes ImageJⓇ to quantify the numbers, length, area, volume, intensity, shape, and texture of cells318. Immunohistochemistry can also be

35 utilized to assess the brain regions of interests across species (i.e., humans vs. experimental models).

1.6.5 Experimental models of hydrocephalus

Animal models offer an environment that is amenable to manipulations of biological, cellular, and physiological processes to best understand the mechanisms of pathogenesis in human diseases and trial therapeutic strategies87,102. In vitro models, which involve the isolation and cultivation of cells or organ systems in specially formulated media provide micro-environments that can be precisely controlled to focus on mechanisms of action in disease pathogenesis. To date, only a few studies have utilized tissue cultures to investigate cellular processes involved in hydrocephalus. For example, Smith et al. utilized tissue culture chambers to examine the effects of increased pressure and to potentially slow stretch on ependymal and glial cells319. Other groups have examined explanted shunt catheters in tissue culture media to assess growth as well as specific responses to compression, stretch, and hypoxia. In several studies, Harris et al. have utilized a bioreactor to demonstrate the immediate deposition of protein biofilms that form a substrate for macrophage and glial cell attachment and provided evidence for cell surface modifications that reduce cell adhesion on silicone catheters320-324.

A major drawback of tissue culture systems is that they cannot mimic all the physiologic conditions of living organisms, necessitating the need for in vivo models. There is currently a plethora of in vivo models of several forms of hydrocephalus. Mouse knockout models such as the hydrocephalus with hop gait (hyh)176,216,217,219, hydrocephalus-3 (hy3)325-327, those with ciliopathies328-336 and mice with mutations in SUMS/NP 337,338, TGF-β1339-341, FGF-2342,343, L1-CAM344,345, Rnd-3346, and

36 AQP-4 347,348 have provided valuable insights into the genetic basis of congenital hydrocephalus15,338,349-351. In rats, the H-Tx352-356 and LEW/Jms357-360 models, which develop aqueductal stenosis, have been widely used to investigate the pathophysiology of obstructive hydrocephalus and, recently, to explore VZ/SVZ disruption, and the potential to transplant neurospheres for replacement therapy169,170,361,362. Acquired hydrocephalus models on the other hand, have mostly been achieved by an intraventricular injection of the substances depending on the variant of hydrocephalus of interest. The most popular injected substance is kaolin. The inert silicate is injected into the cisterna magna or the basal cisterns, to induce obstructive and communicating hydrocephalus, respectively151,363-369.

The vast majority of the existing animal models of hydrocephalus are mice370,371 and rats84,372,373, likely because of their reduced costs, ease of handling and availability of transgenic models133,170,220,359,374,375. However, these small animals are phylogenetically less advanced, usually develop severe ventriculomegaly rapidly and have a limited life span. Additionally, their lissencephalic cerebral cortex often presents challenges when translating research results to humans376. Recently, there has been an increasing focus on animals with gyrencephalic brains such as ferrets, dogs, sheep and piglets377-380 since they are more similar to human brains.

In PHH models, IVH has historically been induced with intraventricular injections of blood (whole or lysed) and blood products such as hemoglobin, iron and LPA85,212,213,223,242,372,381,382.

Ventriculomegaly in IVH models is believed to be related to mechanisms of neuroinflammation and damage to the VZ/SVZ. However, it is worth noting that up to 50% of control animals, typically injected with PBS intraventricularly, develop ventriculomegaly suggesting that the

37 intraventricular injection itself can produce collateral tissue damage91,383. Other studies have induced IVH and PHH via intraperitoneal injection of glycerol384, hemodynamic dysregulation and asphyxia378,385 and endovascular perforation of subarachnoid vessels386 with limited success.

In terms of CNS infections, there is extensive knowledge and documentation on several animal models387-391. However, the majority of these experiments were designed to study the virulence of pathogens, passive immunization and the efficacy of antimicrobial therapy392-395; and none have focused on PIH. A PIH model may be induced through inoculation of live bacteria or bacterial toxins via several routes including intraperitoneal, intranasal, oral and intraventricular387-389,391-

393,396-398. However, achieving meningitis/ventriculitis from bacteremia in animals is extremely challenging and is associated with unacceptably high risks of mortality from septic shock and multi-organ failure. Alternatively, direct intraventricular injection of toxins to cause ventriculitis may be more feasible, with the caveat that it bypasses the primary routes of human neonatal sepsis397,399-401.

1.7 Thesis objectives and aims

1.7.1 Objectives

With the long-term goal to improve patient outcomes by identifying targetable pathways to prevent the development of PIH and PHH, this dissertation leverages a complementary, yet comprehensive scientific approach that encompasses: 1) High throughput discovery-based analyses of human infant CSF to identify differentially expressed genes and proteins in PIH and PHH; 2)

Proteogenomic integration of proteomic and transcriptomic data to define network and pathway

38 level linkages to probe molecular mechanisms underlying PIH and PHH; 3) Characterization of the effects of hydrocephalus on the VZ, SVZ and PVWM using dMRI modalities that complement human hydrocephalus neurocytology; 4) Generation of experimental models of hydrocephalus to mimic the human condition and characterize hydrocephalus-associated neurobiology; and 5) examine the role of metalloprotease inhibition as a therapeutic strategy for abrogating PIH and

PHH pathology.

1.7.2 Central hypothesis

We hypothesized that VZ/SVZ disruption in PIH and PHH is caused by ADAM10-mediated N- cadherin cleavage. CSF protein profiling from PIH and PHH infants will demonstrate alterations in gene-activated pathways in immune-response, cell-cell junction adhesion, and neurodevelopment-neural injury. Pharmacologic inhibition of ADAM10 will represent a viable therapeutic approach to preventing these disabling diseases.

1.7.3 Specific Aims

1.7.3.1 Specific Aim 1

Determine the gene-pathway alterations that are common and unique to PIH and PHH.

Working hypothesis: Expressomes and specific pathways define the immune responses and dysregulation of neurodevelopmental mechanisms in PIH and PHH

Sub Aim 1: Quantify the PIH and PHH CSF expressome using high throughput mass spectrometry

(MS)/MS proteomics and RNA-seq transcriptomics

39 Sub Aim 2: Identify differentially expressed genes and proteins in the CSF of human PIH and

PHH infants using deep scale proteomics and define activated pathways relative to controls

1.7.3.2 Specific Aim 2

Define PIH and PHH related microstructural VZ and PVWM injury in human infants using advanced dMRI and immunohistochemistry.

Working hypothesis: VZ histopathology and abnormal diffusion metrics are associated with neuroinflammation in the PVWM of PIH/PHH human infants

Sub Aim 1: Quantify VZ and PVWM diffusion properties in PHH infants and age-matched control infants with dMRI.

Sub Aim 2: Determine the relationships between immune-associated PVWM microstructural histopathology and VZ disruption in human PIH/PHH postmortem tissues using immunohistochemistry

1.7.3.3 Specific Aim 3

Evaluate N-cadherin expression as a marker of ADAM10 activity and VZ disruption in vitro and examine the effect of ADAM-10 modulation on N-cadherin expression.

Working hypothesis: In vitro ADAM10-mediated cleavage of N-cadherin is associated with VZ disruption, and pharmacological inhibition of ADAM10 will ameliorate VZ disruption

Approach: Quantify N-cadherin disruption in vitro by exposing VZ cells to alpha-toxin (PIH) or whole blood (PHH). Then determine if treatment with the ADAM10 inhibitor G1254023X will preserve N-cadherin expression

40

1.7.3.4 Specific Aim 4

Evaluate hydrocephalus associated VZ disruption and PVWM injury in vivo using PIH mouse and

PHH ferret models.

Working hypothesis: N-cadherin is associated with VZ disruption and PVWM damage in the pathogenesis of PIH and PHH

Approach: Develop PIH mouse and PHH ferret hydrocephalus models and utilize MRI and immunohistochemistry to determine the relationships between N-cadherin expression, neuroinflammation and VZ and PVWM microstructure.

41 2 Chapter 2: Proteogenomics and the PIH-PHH Cerebrospinal Fluid Expressomes

Adapted from:

1. Paulson JN, Williams BL, Hehnly C, Mishra N, Sinnar S, Zhang L, Ssentongo P, Mbabazi-

Kabachelor E, Wijetunge DSS, Bredow B, Mulondo R, Kiwanuka J, Bajunirwe F, Bazira

J, Bebell LM, Burgoine K, Couto-Rodriguez M, Ericson JE, Erickson T, Ferrari M,

Gladstone M, Guo C, Haran M, Hornig M, Isaacs AM, Kaaya BN, Kangere SM, Kulkarni

AV, Kumbakumba E, Li X, Limbrick DD, Magombe J, Morton SU, Mugamba J, Ng J,

Olupot-Olupot P, Onen J, Peterson MR, Roy F, Sheldon K, Townsend RR, Weeks AD,

Whalen AJ, Quackenbush J, Ssenyonga P, Galperin MY, Almeida M, Atkins H, Warf B

C, Lipkin WI, Broach JR, Schiff SJ: The Bacterial and Viral Complexity of Post-infectious

Hydrocephalus in Uganda. Science Translational Medicine 2020. (Accepted).

2. Isaacs AM, Morton SU, Movassagh MJ, Zhang Q, Hehnly C, Zhang L, Morales DM,

Sinnar S, Ericson JE, Mbabazi-Kabachelor E, Ssenyonga P, Onen J, Mulondo R, Hornig

M, Warf BC, Broach JR, Townsend RR, Limbrick DD, Paulson JN, Schiff SJ: Central

nervous system immune response in postinfectious hydrocephalus. Nature Medicine 2020.

(Submitted).

3. Morton SU, Isaacs AM, Movassagh MJ, Zhang Q, Hehnly C, Zhang L, Morales DM,

Sinnar S, Ericson JE, Mbabazi-Kabachelor E, Ssenyonga P, Onen J, Mulondo R, Hornig

M, Warf BC, Broach JR, Townsend RR, Paulson JN, Schiff SJ, Limbrick DD:

Pathophysiological mechanisms common to post-hemorrhagic and post-infectious

Hydrocephalus. Manuscript in preparation.

42 2.1 Introduction

PIH and PHH are both likely propagated through analogous, yet poorly characterized host-genome interactions. While there is anecdotal understanding of their pathophysiology, there has been no systematic exploration of the regulatory pathways that drive the disease phenotype87. This has limited previous efforts in PIH and PHH research to develop biomarkers for the clinical management and comparative effectiveness studies, in turn hindering progress in diagnosis, monitoring of therapeutic efficacy, and providing information regarding long-term neurological outcomes. With a working hypothesis that expressomes and specific pathways define the host responses in PIH and PHH, the primary objectives of the experiments presented in this chapter were to identify key causative activated gene regulatory pathways and related functional protein products involved in the pathophysiology of PIH and PHH. To achieve this goal, we carried out two major studies examining the CSF of a cross-sectional cohort of infants with PIH, PHH and non-infectious non-hemorrhagic hydrocephalic controls. The first study examined the CSF proteogenomic expressomes of PIH, utilizing concurrent high throughput deep-scale proteomics and next-generation transcriptomics. Our proteogenomics pipeline (Figure 7), which leverages message-passing strategies to integrate gene expression and protein-protein interactions was utilized. The second study utilized isobaric labelling proteomics to define the proteome of PIH,

PHH and matched control infants to determine pathway alterations that are common to both conditions and those that are specific to each.

43

Figure 7. Schematic of proteogenomics experimental and data analysis workflow. Output data from concurrent proteomics and RNA-Seq on the same samples were processed, normalized, batch-controlled and explored independently. Differentially expressed transcriptomic and proteomic data were integrated following dimension reduction and feature selection. Gene ontology enrichment was evaluated and common reactome pathways were visualized to identify prominent molecular pathways implicated in the pathophysiology of post-infectious hydrocephalus.

2.2 Results

2.2.1 Concordant proteome and transcriptome of host immune response in PIH

In this study, ventricular CSF samples of 100 infants ≤ 3 months of age with PIH (n=64) and non- post-infectious hydrocephalus, NPIH (n=36) were concurrently analyzed with TMT-10 proteomics and Illumina-based RNA-Seq transcriptomics. Gene expression and protein abundance data were

44 analyzed for functional enrichment. Downstream analysis of proteomics and RNA-Seq data included dimension reduction, feature selection, gene ontology enrichment and reactome over- representation pathway analyses of the differentially expressed proteins and genes.

2.2.1.1 Pathogen identification

Prior to this study, we had identified a strain of Paenibacillus spp. in the CSF of our PIH infants in a background of cytomegalovirus infection (CMV)402, utilizing an extensive pathogen detection approach including Sanger sequencing, next generation sequencing, quantitative polymerase chain reaction (PCR), and VirCapSeq® 403. The Paenibacillus spp. strain we identified (which has been since named the “Mbale strain”) had a 99% identity and a whole-genome average nucleotide identity value of 97% to a known Paenibacillus thiaminolyticus strain NRRL B-4156T (=JCM

8360T, GenBank accession CP041405 – “Reference strain”)404. In our cohort, presence of the

Mbale strain in CSF inversely correlated with age and was associated with poorer clinical signs including seizures febrile episodes, and worse neuroimaging metrics assessing the presence of brain abscesses, calcifications loculations and debris on cranial CT scans. In 21-28-day old

C57BL/6J mice, intraperitoneal inoculation of the Mbale strain lead to a 93% mortality rate, and was associated with acute tubular necrosis in the kidneys, bone marrow myeloid hyperplasia, and lymphocyte apoptosis in the splenic periarteriolar sheaths. There were no significant lesions in the brains or hydrocephalus402. Similar inoculation with the Reference strain was associated with no adverse effects402. We are presently working on characterizing the pathogenicity of the Mbale strain, utilizing RNA-Seq and label-free data-dependent acquisition proteomics.

45 2.2.1.2 Patient characteristics

Informed by our findings of Paenibacillus spp. and CMV in the CSF of our cohort402, patients were sub-categorized as “Paeni-positive” or “Paeni-negative”, and “CMV-positive” or “CMV- negative”, respectively. Comparison of baseline characteristics between Paeni-positive and Paeni- negative patients: Paeni-positive patients had higher peripheral (p=0.002), and CSF (p<.001), white blood cell (WBC) counts, and lower hemoglobin (p=0.039) compared with Paeni-negative patients. There were no significant differences in age and gender between the groups. (Table 1).

Table 1. Demographics and clinical characteristics of PIH patients and controls

All Paeni-negative; n=62 Paeni- patients; positive; n=100 n=38 NPIH; n=36 PIH; n=26

Age in days, mean (SD) 57 (24) 43 (27) 75 (11) 59 (17) Sex Male (%) 51 (51) 16 (44) 14 (54) 21 (55) Female (%) 49 (49) 20 (56) 12 (46) 17 (45) CSF CMV Status, + (-) 8 (92) 0 (36) 2 (24) 6 (32) CSF WBC [1.0 x 103]/µL, mean (SD) 30 (62) 5 (0.5) 5 (0.0) 72 (86.0) Peripheral blood 10.3 (3.6) 8.8 (2.6) 9.8 (3.0) 11.9 (4.1) WBC [1.0 x 103]/µL, mean (SD) Hemoglobin g/dl, mean (SD) 11.5 (2.2) 13.0 (2.8) 10.5 (1.3) 10.8 (1.3) Hematocrit %, mean (SD) 36.8 (7.4) 41.6 (9.3) 33.6 (4.2) 34.4 (4.1) SD: Standard deviation, CSF: Cerebrospinal fluid, CMV: Cytomegalovirus; PIH: post-infectious hydrocephalus; NPIH: non-post-infectious hydrocephalus; Paeni-positive: Cerebrospinal fluid (CSF) Paenibacillus spp. positive; Paeni-negative: CSF Paenibacillus spp. negative.

46

Supplementary Figure 1. Patient baseline characteristics Composite of CT scans from all patients grouped by post-infectious hydrocephalus (PIH, upper groupings) and non-post-infectious (NPIH, lower group) hydrocephalus. The PIH group is further clustered by Paenibacillus spp. negative (left) and positive (right). Yellow and red stars indicate if a patient was PCR positive for cytomegalovirus (CMV) in the CSF and blood respectively. Two other patients met criteria for a bacterial identification for a putative pathogen, indicated as group B Streptococcus (GBS) in a case that was also Paenibacillus spp. positive, and one case with Bacillus sp. in the NPIH group. Two patients in the NPIH group were positive for Paenibacillus spp. (Paeni) using V1-V2 analysis. Note that no patient in the NPIH group that was CMV positive in the blood had CMV detected in the CSF.

2.2.1.3 Proteomics demonstrate neuroinflammation and cell junction disruption in PIH

Multi-dimensional scaling of proteomic data demonstrated clustering of PIH and NPIH infants and distinguished Paeni-positive from Paeni-negative patients. Of 616 proteins identified, 292 were differentially expressed based on Paenibacillus spp. status, with 144 upregulated and 148 proteins downregulated in the Paeni-positive group, respectively. Adjusting for CMV status did not change the differentially abundant proteins. (Figure 8).

47

Figure 8. Proteomic profile of PIH and NPIH infants The proteomics profiles of infants with (PIH) and without (NPIH) post-infectious hydrocephalus are based on 16s rRNA-determined Paenibacillus spp. status. (A) Multi-dimensional scaling (MDS) of normalized protein abundances demonstrating clustering of Paenibacillus spp. positive (Paeni-positive) infants from the remaining groups. The axes of the scatter plot of individual participants represent the first and second components. Each oval (not drawn to scale) encircles majority of patients belonging to the color-matched group, with orange as infants in the NPIH group, green as Paeni-negative PIH infants (Non-Paeni PIH), and purple as Paeni-positive infants (Paeni PIH). (B & C) Volcano plots demonstrating differential expression of genes between Paeni- positive and Paeni-negative infants. The vertical dashed lines demarcate fold changes of 1 and -1 respectively, and the horizontal dashed line represents an alpha significance level of 0.05. Each point represents a differentially expressed protein and those with an absolute fold change greater than 1 that met the significance level (red points) were selected for gene set enrichment analyses. (B) Differential expression of proteins based on Paenibacillus spp. status was similar with or without adjusting for CMV status (B). (C) There were no differentially expressed proteins based on CMV status alone that met fold change (blue points) and statistical significance (grey points) criteria.

48 Gene set enrichment analysis of the differentially expressed proteins demonstrated the most predominantly enriched functional gene sets identified were neuroinflammation related, particularly neutrophil-mediated inflammation, and negative regulation of and peptidase activity, and modulation of extracellular matrix and structure (Figure 9).

Figure 9. Gene ontology analyses based on cerebrospinal fluid assay. Differentially expressed (A) proteins and (B) RNA transcripts between infants with post-infectious hydrocephalus (PIH) and those without (NPIH) is based on 16s rRNA-determined Paenibacillus spp. status. In both proteomics and RNA-Seq, there was enrichment for functions associated with neuroinflammation, extracellular matrix structure and cell-cell adhesion. The abscissae of the dot plots correspond to the number of proteins/genes per total number of genes (gene ratio), and the size of each circle reflects the relative number of proteins/genes expressed that are enriched for the corresponding function (ordinates).

49 2.2.1.4 RNA-Seq demonstrates neuroinflammation and cell junction disruption in PIH

Principal component analysis (PCA) of genes identified with RNA-Seq demonstrated clustering of PIH and NPIH infants. Of the 11,114 genes expressed in the cohort, 2,161 were differentially expressed, and hierarchical clustering of those genes distinguished a subset of Paeni-positive from

Paeni-negative patients (Figure 10). Gene ontology analysis of the differentially expressed genes based on Paenibacillus spp. status demonstrated an enrichment for genes related to response to bacteria, host immune regulation, and cell motility, migration and adhesion (Figure 9).

Figure 10. Transcriptomic profile of PIH and NPIH infants The transcriptomic profiles of infants with post-infectious hydrocephalus (PIH) and those without (NPIH) based on 16s rRNA-determined Paenibacillus spp. status. (A) Principal component analysis of normalized protein abundances demonstrating clustering of PIH from NPIH. The axes of the scatter plot of individual participants represent the first and second components. Each oval (not drawn to scale) encircles majority of patients belonging to the color-matched group, with green as infants in the NPIH group, and blue PIH infants. (B) Heatmap of the 500 most differentially expressed genes, with the dendrogram demonstrating hierarchical clustering on Euclidean distance of gene identified in infants based on Paenibacillus spp. status.

50 Adjusting for CMV status did not significantly alter the gene ontology enrichment based on

Paenibacillus spp. status. Sixty-four genes were differentially expressed based on CMV status, and those genes were enriched for functions related to host response to virus. PCA using RNA abundance of all genes was not able to separate samples based on CMV status. (Supplementary

Figure 2)

Supplementary Figure 2: Transcriptome profile of patients with CMV infection.

Role of CMV infection on the genomic profile of infants with post-infectious hydrocephalus (PIH) and those without (NPIH), based on 16s rRNA-determined Paenibacillus spp. status. There were 64 differentially expressed genes between Paeni-positive and Paeni-negative infants based on CMV status, and (A) those genes were enriched for functions related to host response to virus demonstrated on the interactome. Genes are represented by grey circles and interactions are noted by lines between genes, clustered by their respective labelled gene ontology biological processes (cantaloupe circles). (B) However, there was no clustering of patients based on CMV status as shown on the PCA plot, where the axes of the scatter plot of individual participants represent the first and second components.

51 2.2.1.5 Clinical traits correlate with CSF gene expression in PIH

Using weighted correlation network analysis (WGCNA), 2 gene sets (Modules 1-2) were identified within a subset of 33 patients clustered by expression of differentially expressed genes between the Paeni-positive and Paeni-negative cohorts. These modules were assessed for correlation with

5 clinical variables: WBC counts and Paenibacillus spp. status in both CSF and peripheral blood, and, CSF CMV status in CSF (Figure 11). Module 1 contained all but 11 of the genes differentially expressed with Paenibacillus spp. status and was positively correlated with CSF cell count, and negatively correlated with Paenibacillus spp. status. Module 2 was positively correlated with

Paenibacillus spp. status, but those 11 genes were not enriched for any gene ontologies, indicating no specific functional enrichment for the small number of genes within this module.

Figure 11. Weighted correlation network analysis (WGCNA) of gene modules WGNCA gene modules identified within RNA-Seq data for post-infectious (PIH) and non-post- infectious (NPIH) hydrocephalus cohorts demonstrate Module 2 (A) was positively correlated with CSF cell count and negatively correlated with age and (B) was enriched for host immune responses, including leukocyte and neutrophil functions.

52 2.2.1.6 Proteogenomic expressome of host immune response in PIH

Unsupervised gene ontology analyses of concatenated differentially expressed proteins and genes assayed from proteomics and RNA-Seq respectively, recapitulated enrichment for the immune system, metabolism, response to oxidative stress, cell-cell junction interactions and extracellular matrix structure in association with peptidase activity. (Figure 12).

Figure 12. Prominent Gene Ontological (GO) functions and interactions. The Alluvial plot demonstrate GO functions enriched for the pathophysiology of post-infectious hydrocephalus (PIH) between Paenibacillus spp. positive and Paenibacillus spp. negative infants. Each ontological clustering occupies a column in the diagram and is horizontally connected to preceding and succeeding significance clustering by stream fields, representing similar gene involvement. Each stacked bar is color-coded based on the assay being assessed, with blue representing RNA-Seq data, orange for proteomics and green for genes common to both RNA-Seq and proteomics. The ordinate shows the number of genes represented in each cluster.

53 There were 33 genes that were common to both proteomics and RNA-Seq. Pathway enrichment analysis of those genes demonstrated an enrichment for functions predominantly associated with the immune system, particularly interleukin (IL)-4, IL-12, IL-13, interferon signalling, and genes involved with neutrophil activity and the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway. In addition, there was enrichment for processes involved with response to platelet activating factors and host recognition of microbes including antigen presentation and Class I MHC antigen processing (Figure 13).

Figure 13. Pathway analysis of differentially expressed markers in RNA-seq and proteomics Post-infectious and non-post-infectious hydrocephalus infants are stratified by cerebrospinal fluid 16s rRNA Paenibacillus spp. status. Corresponding pairs of upregulated (red boxes) and downregulated (green boxes) proteins (blue columns) and genes (purple columns) in the Paenibacillus spp. positive group that were identified with proteomics and RNA-Seq respectively are listed on the x-axis. The 33 common genes demonstrated predominant involvement of the immune system, particularly the innate system and those associated with neutrophil-mediated activity, interleukins, interferon and the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway (y-axis). Differential expression was defined as log2 fold change of >1 or <-1 at an alpha significance level of 0.05.

54 Interactome analysis405 demonstrated these 33 proteins had high physical protein-protein interactions with regards to their chromosome neighborhood, gene fusion, phylogenetic co- occurrence, homology, co-expression, experimentally determined interactions, database annotation and automated text mining405 (Figure 14).

Figure 14. Interactome analysis of cross-omic differentially expressed markers. Comparing the CSF profiles of Paenibacillus spp. positive post-infectious hydrocephalus infants compared to Paenibacillus spp. negative infants, the network nodes represent proteins produced by a single, protein-coding gene locus and first shell of interactors. Filled and empty nodes imply the 3D structure of the protein is known or unknown, respectively. Edges represent protein-protein associations determined from known interactions (curated and experimentally determined), predicted interactions (related to gene neighborhood, fusions and co-occurrence) and other interactions including text mining, co-expression and protein homology. The colors of edges, as demonstrated on the key, reflects the interactions. Shaded regions depict groupings into their predominant gene ontology function with grey, purple and orange regions representing those involved with the immune system, cytokine-mediated immune response and factors associated with platelet activity.

55 2.2.2 Activated gene pathways in PIH and PHH

Integration of RNA-Seq and proteomics was a useful approach to characterize PIH pathophysiologic mechanisms. Neuroinflammation and extracellular organization are critical processes in the pathophysiology of PIH and may also play a role in development of PHH. This study focused on parsing activated gene networks that are common to PHH and PIH, and just as importantly, those that are unique to each condition. We utilized our existing proteomics pipeline to assess ventricular CSF samples from PIH, PHH and non-infectious, non-hemorrhagic hydrocephalic infants (Controls).

2.2.2.1 Patient characteristics

There were 125 participants, which included 64 PIH, 17 PHH and 44 control infants. Parents of all participants provided informed consent. Among the PIH group, 38 were Paenibacillus spp. positive (Paeni-pos PIH) and 26 were Paenibacillus spp. negative (Paeni-neg PIH). The PHH and

8 control samples were obtained from infants from North America, whereas the PIH and 36 control samples were from Sub-Saharan Africa; regions where the conditions are most prevalent, respectively. All infants were less than 3 months of age and there was no difference in sex between groups.

56 2.2.2.2 Protein expression patterns in PIH and PHH

A z-log protein expression score was calculated as a ratio of the measured expression level to that of the common reference pool. These scores were then normalized, including batch correction and quantile normalization of z-log protein expression scores for each of 2,841 proteins identified.

Multi-dimensional scaling of proteomic data demonstrated clustering of patients based on groups.

As we identified in the previous experiments, controls clustered with Paeni-neg PIH, while Paeni- pos PIH clustered with PHH. To assess for differential expression of proteins between groups, we performed a linear modelling that included three sets of comparisons: PIH vs. controls; PHH vs. controls; and PIH vs. PHH. 1,832 proteins (64%) were differentially expressed between groups.

(Figure 15).

Figure 15. Proteomic profiles of infants with PIH, PHH and non-PHH non-PIH controls The proteomic profiles of post-infectious (PIH) and post-hemorrhagic (PHH) hydrocephalus and those with non-infectious non-hemorrhagic hydrocephalus (Controls). Panel A is a multi- dimensional Scaling of normalized protein abundances demonstrating clustering of Controls and Paenibacillus spp. negative (Paeni-neg PIH) from Paenibacillus spp. positive (Paeni-pos PIH) infants. The abscissa and ordinate of the scatter plot of individual participants represent the first and second components, respectively. Each oval (not drawn to scale) encircles majority of patients 57 belonging to the color-matched group. Panel B is a Euler diagram demonstrating counts of differentially expressed proteins among infants with PIH, PHH and Controls. Each branch (circle) is proportional to the number of proteins differentially expressed between the labelled pair-wise comparisons, with each intersection isolating proteins exclusive to the respective subgroup comparisons. Upregulated (↑) and downregulated (↓) counts of proteins are color-coded with red and blue, respectively.

Directed by our objective to identify activated pathways that are common or unique to PIH and

PHH, the following specific protein clusters from the linear model were selected for downstream analyses: 1) the proteins that were differentially expressed between PIH and PHH but not differentially expressed independently with controls (PIH-PHH group; n=84); 2) the proteins that were differentially expressed between PHH and controls (PHH-Control group; n=143) and those between PIH and controls (PIH-Control; n=59), respectively, but not differentially expressed between PIH and PHH. The former evaluated common pathways between PIH and PHH while the latter evaluated pathways that are uniquely activated in each condition. The nested F-test approach406 was utilized to achieve consistency between all related contrasts while ensuring proteins that were expressed in equal proportions between pairs of samples (log2 fold change of 0) were not excluded. (Figure 16). At all levels of the model, unsupervised hierarchical clustering of the differentially expressed proteins resulted in groups with PHH generally clustering separately from other groups. Two separate PIH populations also emerged, one of which is distinct from other groups, while the second shows some overlap control participants (Figure 16; Supplementary

Figures 3 and 4).

58

Figure 16. Differentially expressed proteins between PIH, PHH and control infants Protein expression profiles between post-infectious hydrocephalus (PIH), and post-hemorrhagic (PHH) hydrocephalus and control infants. (A) Heatmap demonstrating hierarchical clustering on Euclidean distance of proteins identified in the respective conditions. (B) Volcano plot demonstrating differential expression of proteins uniquely expressed in PIH and PHH infants, and not differentially expressed between either and controls. The vertical lines demarcate fold changes of 1 and -1, and the horizontal line represents an alpha significance level of 0.05.

Supplementary Figure 3. Differentially expressed proteins between PIH and control infants Proteomic profile of infants with post-infectious hydrocephalus (PIH) and control infants with non-infectious non-hemorrhagic hydrocephalus (control). (A) Heatmap demonstrating hierarchical

59 clustering on Euclidean distance of proteins identified in the respective conditions. (B) Volcano plot demonstrating differential expression of proteins uniquely expressed in PIH and Control, and not differentially expressed between PHH and post-hemorrhagic hydrocephalus. The vertical lines demarcate fold changes of 1 and -1, and the horizontal line represents an alpha significance level of 0.05.

Supplementary Figure 4. Differentially expressed proteins between PHH and control infants Proteomic profile of infants with post-hemorrhagic hydrocephalus (PHH) and control infants with non-infectious non-hemorrhagic hydrocephalus (Control). Panel A is a heatmap demonstrating hierarchical clustering on Euclidean distance of proteins identified in the respective conditions. Panel B is a volcano plot demonstrating differential expression of proteins uniquely expressed in PHH and Control, and not differentially expressed between PHH and post-infectious hydrocephalus. The vertical lines crossing the positive and negative abscissae demarcate fold changes of 1 and -1, and the horizontal line represents an alpha significance level of 0.05.

2.2.2.3 The independent proteomes of PIH and PHH

Gene set enrichment analysis of the differentially expressed proteins in the PIH-control and PHH- control groups independently demonstrated that inflammation and processes associated with extracellular structure, in association with protein metabolism, were predominantly enriched with minor variations. In PHH, expression markers of enrichment for inflammation was limited to acute

60 immune responses including neutrophil activity and platelet degranulation. Extracellular organization gene enrichment was associated with regulation of peptidases and hydrolases. As expected, there was also enrichment for factors related to blood coagulation and hemostasis.

(Figure 17).

Figure 17. Proteins unique to PHH and controls

Proteins uniquely expressed between post-hemorrhagic hydrocephalus (PHH) and controls. (A) Volcano plot demonstrating the differentially expressed proteins between infants with PHH and controls, that are not differentially expressed in post-infectious hydrocephalus. The vertical lines demarcate fold changes of 1 and -1, and the horizontal line represent an alpha significance level of 0.05. (B) Interactome of the differentially expressed proteins that met criteria (Panel A, red dots) demonstrating enrichment for processes predominantly involved with inflammation, extracellular organization, coagulation and wound healing. The size of each circle reflects the relative number of proteins expressed that are enriched for the corresponding function.

Similar to PHH, inflammatory responses enriched in PIH included those related to neutrophil activity and platelet degranulation. However, complement activation was also enriched.

Enrichment for extracellular organization processes occurred in the context of proteolysis, as well as in response to wound and wound healing (Figure 18).

61

Figure 18. Proteins unique to PIH and controls Analysis of proteins that are differentially expressed between infants with post-hemorrhagic (PIH) and controls, that are not differentially expressed in post-hemorrhagic hydrocephalus. (A) Volcano plot demonstrating differential expression of proteins. The vertical lines demarcate fold changes of 1 and -1, and the horizontal line represent an alpha significance level of 0.05. (B) Dot plot from gene set enrichment analysis of the differentially expressed proteins that met criteria (Panel A, red dots) demonstrating enrichment for processes predominantly involved with inflammation, extracellular organization, and wound healing. Each dot corresponds to the number of proteins per total number of proteins (gene ratio), and its size reflects the relative number of proteins expressed that are enriched for the corresponding function.

2.2.2.4 Neuroinflammation is a common process in PIH and PHH pathophysiology

Proteins that were uniquely differentially expressed in PIH or PHH were subjected to downstream analyses. Gene set enrichment analysis demonstrated that those proteins were also predominantly enriched for inflammation, but included both acute and humoral immune responses, complement activation and platelet degranulation. Neuroinflammation occurred in association with extracellular structure organization processes, which occurred in the context of protein activation cascades. (Figure 19).

62

Figure 19. Gene ontology interactome of proteins common to PIH and PHH Differentially expressed proteins that are common to infants with post-infectious (PIH) and post- hemorrhagic (PHH) hydrocephalus, but not differentially expressed in non-hemorrhagic non- infectious hydrocephalic infants were analyzed. Each point on the interactome represents a differentially expressed protein with an absolute fold change greater than 1. There is enrichment for functions associated with neuroinflammation and extracellular organization, occurring in the context of protein activation cascades. The size of each circle reflects the relative number of proteins expressed that are enriched for the corresponding function.

Having established neuroinflammation and extracellular organization as processes that are central to the pathophysiology of PIH and PHH, reactome analysis407 was performed to identify potentially targetable markers. Markers common to both the innate and adaptive systems that had a concomitant effect on cellular organization were of particular interest. Of 32 inflammation-

63 related proteins, five (CDC42, PSMB10, LGMN, NCF2 and TOM1) were common to both immune systems. CDC42 and PSMB10 are downstream markers involved with cytokine signaling

(Figure 20). KEGG pathway analyses408 (not shown) were performed independently on CDC42,

PSMB10, LGMN, NFC2, and TOM1 to identify their interactors. All but TOM1 were involved in various protein cascades reflecting their potential effects on extracellular organization.

Figure 20. Reactome pathway analyses of proteins common to PIH and PHH Differentially expressed inflammation related proteins unique to post-infectious (PIH) and post- hemorrhagic (PHH) hydrocephalus when compared to non-infectious non-hemorrhagic hydrocephalic controls were subjected to reactome analyses. CDC42, PSMB10, LGMN, NCF2 and TOM1 were found to common to the innate and adaptive systems. CDC42 and PSMB10, DDX58 and PRPS1 are involved with cytokine signalling in the immune system.

64 2.3 Discussion

2.3.1 Summary of findings

PIH and PHH are the most debilitating neurological sequelae of neonatal sepsis and intraventricular hemorrhage, respectively.35,36,42,409 The overarching goal of the many experiments presented in this chapter was to identify critical genes involved in the pathophysiology of PIH and

PHH and map those genes onto their activation pathways to define the common and unique pathophysiological mechanisms underlying both conditions. To circumvent some of the major independent limitations of proteomics and RNA-Seq, proteogenomics was utilized to integrate protein and gene data. Hierarchical clustering identified that PIH and PHH patients had differential expression of genes or proteins involved in neuroinflammation and extracellular matrix organization. Differential expression of host immune response genes and proteins in PIH (those with Paenibacillus spp. in CSF) was predominantly related to the innate immune system (i.e., neutrophil activity, IL-4, IL-12, IL-13 and interferon signaling, the JAK-STAT pathway, and platelet response/activating factors) when compared to controls. In addition, there was enrichment for processes involved with host recognition of microbes including Class I MHC antigen presenting complex in PIH. Similarly, host immune response in PHH was predominantly associated with enrichment of processes involving neutrophil activity and platelet degranulation.

However, gene sets involved in complement activation were enriched in PHH, as well as those involved in blood coagulation and hemostasis. In both PIH and PHH, protein cascades and processes such as proteolysis and hydrolysis were enriched, all within the context of extracellular matrix and cellular structural organization.

65 2.3.2 Proteogenomics as an approach for defining PIH and PHH pathophysiology

Large scale proteomics and genomics studies have been instrumental in the discovery of important biomarkers and have provided insights to the pathophysiology of central nervous system disease.

CSF proteomics have previously identified clinically useful protein biomarkers such as amyloid precursor protein and cell adhesion molecules in PHH.245,247 RNA-Seq of CSF has been widely used to study infectious diseases.410,411 Disease pathogenesis involves the combinatorial interaction of elements within the proteome, transcriptome, and environmental factors.260,412

Therefore, integrative methods that combine proteomic and genomic data into a network model are essential for understanding human disease.262 Our group is the first to utilize proteogenomics to characterize gene regulatory networks involved in PIH and PHH. Here we utilized clustering, reactome and interactome pathways, and gene ontology enrichment methodologies at the RNA and protein levels to unravel the underlying mechanisms involved in host response to PIH and

PHH. Through this effort we have identified the putative genes and pathway responses in the host involved in the pathophysiology of PIH due to Paenibacillus spp. Important future work will test the generalizability of host response observed in this study to other pathogens and other variants of hydrocephalus.

2.3.3 Host-immune responses in PIH and PHH

In addition to ventriculomegaly, both the PIH and PHH phenotype is characterized by granular ependymitis, ependymal gliosis, arachnoiditis, calcifications, and multi-loculated infiltrates36 suggesting that severe inflammation occurs locally during the antecedent neonatal sepsis or hemorrhage. Indeed, infants with PIH due to Paenibacillus spp. had ten-fold more calcifications,

66 abscesses, loculations and debris on head CT402. Collectively, these findings underscore the importance of characterizing the nature of the host-immune response in PIH and PHH.

Typically, once a pathogen breaches a host’s epithelial barriers, immune mechanisms are activated.

Although there is rapid progression of immunologic competence after birth, the innate immune system is the primary active defense against pathogens in infants less than 3 months of age since they lack the antigenic exposure required to develop acquired immunity. Consequently, the host immune response to microbial pathogens, mounted by the infants involved in this study, involved antigen-independent immune components such as neutrophils, phagocytes, natural killer and antigen presenting cells.201,413-416 However, complement activation, which has crossover with the adaptive immune system is a major mediator of host response to infection. Thus, it was not surprising to find that our PIH cohort had differential expression of proteins involved with the complement system. Immune response to non-infectious triggers such as hemorrhage in PHH would also involve activation of the innate immune system in response to endogenously released markers of cellular damage.

Our results support the role of the immune response in PIH and PHH pathophysiology. Persistent inflammation in the CSF of infants in our study may be pathologic rather than physiologic as they were remote from the original infection or hemorrhage and were already clinically well for surgery.

While host-immune response is necessary for clearing pathogens or non-infectious toxins, prolonged or overstimulated immune activation may be detrimental to the host.417 In terms of PIH and PHH, it is likely that the same host immune activity that clears pathogens also causes

67 hydrocephalus by damaging local tissues. Of the three cytokines we identified in PIH, IL-12 is the most diverse and is typically involved in augmentation of CD8+ T cell cytotoxicity. Upregulation of MHC Class I in our PIH cohort could be an indication of presentation to cytotoxic CD8+ cells and IL-12 activity.418 Further, IL-4 and IL-13 typically work synergistically to decrease inflammation and counteract the IL-12 activity, and thus may be upregulated in response to IL-12 upregulation. IL-4 has also been associated with Staphylococcus aureus infections, which like

Paenibacillus spp. is gram-positive. One hypothesis is that the balance of IL-12 and IL-4/IL-13 within the CSF contributes to risk of developing PIH. In Chapters 4 and 5, we present further evidence that inoculation of a gram-positive bacterial toxin in vitro and in vivo in experimental models is associated with neuroinflammation and disruption of the ependymal CSF-brain barrier, likely as an antecedent process to developing hydrocephalus.

2.3.4 Inflammation and barrier integrity in PIH and PHH

The activated immune pathways found in PIH and PHH support the perspective that neonatal immune response to toxins such as infection or hemorrhage inadvertently causes hydrocephalus through ependymal denudation, scarring of CSF conduits and disruption of CSF physiology to cause hydrocephalus. Our finding of differential abundance of RNA and protein involved in cell- cell integrity in PIH and PHH infants is also consistent with recent evidence from both experimental170,172,175-177,419 and clinical181-183,419 studies. In those studies, immune response is associated with ependymal cell-cell junction protein disruption as a critical pathogenetic mechanism of hydrocephalus. In a human postmortem histological study, McAllister et al. attributed the diffuse ependymal damage they identified in PHH infants to disruption of cell-cell

68 junctional proteins, which had occurred in the context of GFAP over-expression, a non-specific surrogate marker of neuroinflammation and glial scarring.197 Together, our findings and these evidences underscore the critical interplay between neuroinflammation and ependymal cellular structure in the pathogenesis of PIH and PHH.

2.3.5 Limitations

Limitations to this study include small cohort size for subset analyses such as in the case of the

CMV positive participants, low RNA abundance in the control samples, and reduced availability of clinical covariates to assess for correlation with gene abundance patterns. Identifying CMV in some of the infants in our cohort402 raises the important question of whether the host-immune response we observed was at least in part attributable to CMV. While there were differentially expressed transcripts at the genome level in response to viruses, we did not detect any functional proteins associated with CMV, nor was there any effect of CMV status on the differential expression of host-immune markers at the gene or protein level. It is possible that our inability to detect a CMV-related host-immune response was limited by having only a few (8/100) CMV- positive infants. Another possibility is that CMV tend to cycle through dormant and active state in affected individuals420, thus point sampling of CSF may not have captured an active state.

Therefore, it remains unknown whether CMV was a risk factor for developing Paenibacillus spp. infection or PIH in the first place, whether CMV can be implicated in the persistent host-immune response we observed, and whether it will have any long-term effects in these infants.

69 2.4 Materials and Methods

2.4.1 Patients and recruitment

PIH and NPIH infants were recruited at the CURE Children's Hospital of Uganda (CCHU), a pediatric neurosurgery hospital that serves as the largest referral center for patients with hydrocephalus in Uganda. Ethics oversight for those patients was provided by the CCHU

Institutional Review Board (IRB), Mbarara University of Science and Technology Research Ethics

Committee, the Ugandan National Council on Science and Technology, and the Pennsylvania State

University (PSU) IRB. A United States Centers for Disease Control permit for importation of infectious materials covered the transfer of specimens from CCHU to PSU. PHH and NPHH infants were recruited from the St. Louis Children’s Hospital (SLCH), a large tertiary pediatric hospital in St. Louis, Missouri. Ethics oversight for the PHH and NPHH infants was provided by the Washington University Human Research Protection Office (HRPO #s 201101887 and

201203126). Verbal informed consent was obtained from the parent(s) of each subject.

Each PIH infant had a history of febrile illness and/or seizures preceding the onset of hydrocephalus and/or ventriculitis, as detected by physical examination, endoscopy, or neuroimaging. No PIH infants had hydrocephalus at birth. NPIH infants were selected based on need for neurosurgical intervention of congenital hydrocephalus due to an obstructive aqueduct lesion, Dandy-Walker cyst, or other congenital malformation of the nervous system.

70 The PHH group included preterm infants (age < 35 weeks estimated gestational age) admitted to the SLCH Neonatal Intensive Care Unit who sustained IVH which was diagnosed with head ultrasound within the first month of life. Patients underwent weekly head ultrasounds for monitoring of ventricular size and were eventually deemed to require implantation of a ventricular access device (VAD). Well-established Hydrocephalus Clinical Research Network criteria were applied for the diagnosis of PHH and need for and timing of hydrocephalus interventions421. All

PHH infants included in this study underwent permanent CSF diversion surgery (ETV +/- CPC or

VP shunt). The NPHH group included age and gender matched infants who needed neurosurgical intervention congenital or aqueductal stenosis hydrocephalus who had no evidence of intraparenchymal or intraventricular hemorrhage on their head ultrasound.

Inclusion criteria for all patients were age less than 3 months and weight greater than 2.5 kg at time of CSF sampling. Exclusion criteria for all subjects were prior surgery on the nervous system or evidence of communication of nervous system with skin. Additionally, PHH patients with a history of systemic infection were excluded. Clinical patient information including demographics, computed tomography (CT) neuroimaging, CSF cell counts, and CSF culture results were obtained from review of the medical records.

2.4.2 CSF Samples

Sterile CSF samples were acquired directly from the cerebral ventricles of all patients at the time of surgery for treatment of hydrocephalus. After collection, samples were RNA shielded (Zymo

71 Research, Irvine CA) to preserve nucleic acid and protein contents when possible, then rapidly frozen and maintained in liquid nitrogen prior to shipment for final -80℃ storage until proteomics and/or RNA-Seq was performed. In all cases, the CSF microbiological cultures sent at the time of collection remained sterile. Any NPIH, PHH or NPHH that had gram stain or microbial cultures suggestive of a CNS infection was excluded.

2.4.3 Transcriptomics

2.4.3.1 RNA Extraction

All reagents and consumables required for RNA extraction were UV-treated in a polymerase chain reaction (PCR) workstation for 15 minutes. A TRIzol™ LS Reagent and Direct-zol™ RNA

MiniPrep Plus kit was used for all extractions. Each batch of extraction included a water control in which 500 μL of elution buffer was put through all extraction steps. CSF samples thawed at room temperature were processed with two aliquots of 250 μL mixed with 750 μL of TRIzol™ LS

Reagent. After bead lysis, 200 μL of chloroform was added, incubated at room temperature for 15 minutes then centrifuged at 12,000g for 15 minutes at 4oC. Once separated the aqueous layers from the two aliquots were put into the same tube. Then each organic layer was re-suspended in 400 μL of water and centrifuged again for 5 minutes. The 1.6 mL of aqueous phases were combined with

1.6 mL of 100% ethanol and put through the Zymo-Spin™ IC Column twice and proceeded per manufacturer’s protocol.

72 2.4.3.2 RNA Sequencing

Following RNA extraction, Illumina® TruSeq® Stranded RNA Sequencing kit was used for rRNA depletion and fragmentation, first strand cDNA synthesis, second strand cDNA synthesis, adenylate 3’ ends, adapters ligation, and DNA fragment enrichment following the manufacturer’s protocol. It was expected that due to the nature of low cell counts in CSF, varying concentrations of RNA would be extracted. Indeed, in our samples, majority of the RNA concentrations fell below the recommended input of 100 ng so a standard 10 ul of total RNA was used for input ranging from 10-150 ng of RNA. Using biotinylated, target-specific oligos combined with Ribo-Zero ribosomal RNA (rRNA) removal beads of divalent cations and high temperatures (60 – 100oC).

Ribosomal RNA was then depleted from the total RNA and the remaining RNA was purified, fragmented, and primed for cDNA synthesis. The cleaved RNA fragments were then copied into first strand cDNA using reverse transcriptase (Part # 18064-014) and random hexamers. A second strand cDNA synthesis was obtained by removing the RNA template, synthesizing a replacement strand, and incorporating dUTP in place of dTTP to generate the ds (double stranded) cDNA, using

DNA Polymerase I and RNaseH. A single 'A' nucleotide was then added to the 3' ends of the blunt fragments and a corresponding single 'T' nucleotide was added on the 3' end of the adapter.

Multiple indexing adapters were then ligated to the ends of the ds cDNA fragments to prepare them for hybridization. Polymerase chain reaction (PCR) was used to enrich the DNA fragments which was done without dUTP to ensure only the first strand was enriched. The libraries were quantified by fluorometry and size and quality checked on an Agilent Technologies 2100

Bioanalyzer. Libraries were then pooled to 4 nM, diluted and sequenced on the NovaSeq 6000 using the S2 flowcell with 2x100 reads aiming for 25 million reads per sample.

73 2.4.3.3 Data Analysis

All RNA-Seq data analyses were performed with R v.3.5TM (http://www.R-project.org) utilizing the following packages: clusterProfiler422, edgeR423, RSEM424, STAR425, YARN426. Pair-end RNA-

Seq were first mapped to the human reference genome hg38 using STAR425. Then RSEM424 was used for estimating gene and isoform expression levels from mapped RNA-Seq data. Gene read counts from all samples were combined into matrix for further analysis. YARN426 was used to create normalized gene expression sets from the RNA-Seq matrix data. edgeR423 was used to calculate differential expression between groups using a Genewise Negative Binomial Generalized

Linear Model. Differentially expressed genes were selected based on Benjamini-Hochberg adjusted p-value cutoff of 0.05 and absolute log fold change of 1 and were mapped to Gene ontology terms and assessed for enrichment using clusterProfiler422. Pathway enrichment analysis was performed utilizing Reactome Pathway Enrichment analysis and Database for Annotation,

Visualization and Integrated Discovery (DAVID v6.8)427.

2.4.4 Proteomics

2.4.4.1 Sample preparation and mass spectrometry

A standard validated proteomics workflow was utilized428. Briefly, the top six most abundant proteins in CSF (albumin, IgG, antitrypsin, IgA, transferrin and haptoglobin) were removed using a reproducible affinity method247. Samples were then enzymatically digested with trypsin, and peptides were isobarically labeled using a ten-channel peptide mass tagging (TMT-10) robotic

74 solid phase extraction system429,430. Labeled peptides were analyzed by high-resolution liquid chromatography-mass spectrometry (LC-MS) using a Q-Exactive Plus instrument coupled to an

EASY-nanoLC 1000 system (Thermo-Fisher Scientific). The pooled samples in batches of ten were loaded onto a 75 µm i.d. × 25 cm Acclaim® PepMap 100 RP column (Thermo-Fisher

Scientific). Peptide separations were started with 95% mobile phase A (0.1% FA) for 5 min and increased to 30% B (100% ACN, 0.1% FA) over 180 min, followed by a 25-min gradient to 45%

B, a 5-min gradient to 95% B and washed at 90% B for 7 min, with a flow rate of 300 nL/min.

Full-scan mass spectra were then acquired using the Orbitrap mass analyzer in the mass-to-charge ratio (m/z) of 375 to 1400 and with a mass resolving power set to 70,000. Fifteen data-dependent high-energy collisional dissociation (HCD) were performed with a mass resolving power set to

35,000, a fixed first m/z 100, an isolation width of 0.7 m/z, and the normalized collision energy setting of 32. The maximum injection time was 50 ms for parent-ion analysis and 105 ms for product-ion analysis. Target ions already selected for mass spectrometry (MS)/MS were dynamically excluded for 30 s. An automatic gain control target value of 3 x 106 ions was used for full MS scans and 1 x105 ions for MS/MS scans. Peptide ions with charge states of one or greater than six were excluded from MS/MS interrogation.

2.4.4.2 Protein identification

MS raw data were converted to peak lists using Proteome Discoverer (version 2.1.0.81, Thermo-

Fischer Scientific) with the integration of reporter-ion intensities of TMT-10-plex at a mass tolerance of ±3.15 mDa431. MS/MS spectra with charges +2, +3 and +4 were analyzed using

Mascot search engine432 (Matrix Science, London, UK; version 2.6.2). Mascot was set up to search

75 against a SwissProt database of human (version June 2016, 20,237 entries) and common contaminant proteins (cRAP, version 1.0, 116 entries). With trypsin as the digestion enzyme, four was the maximum allowed missed cleavages. The searches were performed with a fragment ion mass tolerance of 0.02 Da and a parent ion tolerance of 20 ppm. Carbamidomethylation of cysteine was specified in Mascot as a fixed modification. Deamidation of asparagine, formation of pyro- glutamic acid from N-terminal glutamine, acetylation of protein N-terminus, oxidation of , and pyro-carbamidomethylation of N-terminal cysteine were specified as variable modifications. Peptide spectrum matches (PSM) were filtered at 1% false-discovery rate (FDR) by searching against a reversed database and the ascribed peptide identities were accepted. The uniqueness of peptide sequences among the database entries was determined using the principal of parsimony. Protein identities were inferred using a greedy set cover algorithm from Mascot and the identities containing  2 Occam’s razor peptides were accepted433.

2.4.4.3 Protein quantification

All protein quantification and data analyses were performed with R v.3.5TM (http://www.R- project.org) utilizing the following packages: clusterProfiler422, Limma434, mixtools435. The processing, quality assurance and analysis of TMT data were performed with our tidyverse- based436 in-house pipeline, ProteoQ (version 1.0.0.0). Briefly, reporter-ion intensities under 10- plex TMT-10 channels were first obtained from Mascot®, followed by the removal of PSM entries from shared peptides or with intensity values lower than 1E3. Intensity of PSMs were converted to logarithmic ratios at base two, relative to the average intensity of reference samples within a 10- plex TMT. Under each TMT channel, Dixon’s437 outlier removals were carried out recursively for

76 peptides with greater than two identifying PSMs. The median of the ratios of PSM that can be assigned to the same peptide was first taken to represent the ratios of the incumbent peptide. The median of the ratios of peptides were then taken to represent the ratios of the incumbent protein.

To align protein ratios under different TMT channels, likelihood functions were first estimated for the log-ratios of proteins using finite mixture modelling, assuming two-component Gaussian mixtures using the mixtools435. The ratio distributions were then aligned in that the maximum likelihood of the log-ratios are centered at zero for each sample. Scaling normalization was performed to standardize the log-ratios of proteins across samples. To discount the influence of outliers from either log-ratios or reporter-ion intensities, the values between the 5th and 95th percentile of log-ratios and 5th and 95th percentile of intensity were used in the calculations of the standard deviations.

2.4.4.4 Data analysis

All proteomics data analyses were performed with R v.3.5TM utilizing the following packages: clusterProfiler422, edgeR423, and YARN426. Batch differences were visualized with MDS and

Principal Component Analyses (PCA) of the protein z-log ratios. YARN426 was then used to filter genes and correct for batch effects, while preserving group specificity. The data was then quantile normalized in a group-aware manner, accounting for global differences in protein distribution.

(Supplementary Figure 5). Next, empirical Bayes moderation were carried out to ensure variances were no longer dependent on mean protein expression levels438 Log fold change values of the protein abundances between groups were calculated by comparing mean expression levels using the Limma. The number of differentially expressed proteins were examined with significance set

77 at a Benjamini-Hochberg adjusted p-value cutoff of 0.05. Proteins that were differentially expressed in multiple comparisons were extracted and examined from smallest to largest adjusted p-value with their associated protein log fold changes. For assessing differentially expressed proteins common and unique to PIH and PHH, the nested F-test approach was used, and log fold change cut-off was set to zero in order to achieve better consistency between related contrasts while preserving proteins that were expressed at similar levels between groups. ClusterProfiler422 was then used to map differentially expressed proteins to their respective Gene ontology terms to assess for enrichment. Proteins enriched for similar and closely overlapping biological processes were further subjected to interactome405, reactome overrepresentation407 and KEGG408 pathway analyses.

78

Supplementary Figure 5. Proteomics and RNA-Seq data preprocessing and quality control Representative (A and C) multi-dimensional scaling and (B and D) protein density plots grouped by tandem mass tagging (TMT)-10 batches of proteomics experiments. The z-log normalized data had batch effects demonstrated as sparse patient samples in (A) first and second components and heterogeneity (“noise”) on the (B) density plot. After filtration of poor-quality samples, quantile normalization and correction for batch effects, (C) sparsity and (D) noise are reduced, while preserving group specificity. Each circle and spike represent a patient sample color-coded by the TMT-10 batch. Batch pos and Batch neg were CSF samples obtained from infants not in this study, whose CSF was used to bookmark all TMT-10 batches to act as batch controls, and to which each TMT-plex was normalized to obtain the z-log scores. The Batch pos samples were combined from a PIH and a PHH patient and Batch neg was from a non-infectious non-hemorrhagic control infant. Similar approach was used to assess RNA-Seq data prior to data analyses.

79 2.4.5 Proteogenomics

Integration of proteomics and transcriptomics data was performed following dimension reduction and feature selection of differentially expressed proteins and genes. Gene ontology enrichment using DAVID427 and reactome over-representation pathway analysis were performed by concatenating differentially expressed proteins and genes with a set Log fold change of < -1 and >

1 and a false discovery rate < 1 at an alpha of 0.05. In-house parsing scripts for data visualization and gene/pathway/GO enrichment commonality analyses were performed on the integrated data.

Physical and functional protein-protein interactions of the genes common to both proteomics and

RNA-Seq were assessed using the STRING405 online tools. Gene-gene relationships between differentially expressed genes were quantified by assigning a composite score to each pairwise comparison. Between each pair of genes, the composite score reflected their chromosome neighborhood, gene fusion, phylogenetic co-occurrence, homology, co-expression, experimentally determined interactions, database annotation and automated text mining405. The higher the composite score (i.e. closer to 1), the stronger the predicted gene-gene relationship405.

80 3 Chapter 3: Diffusion MRI Characterization of VZ/SVZ and PVWM Injury

3.1 Introduction

Adapted from

1. Isaacs AM, Han RH, Smyser CD, Limbrick DD, and Shimony JS. Semi-automated

segmentation of the lateral periventricular regions using diffusion magnetic resonance

imaging. MethodsX 2020. MEX-S-19-01180. (Under Review)

2. Isaacs AM, Neil JJ, McAllister JP, Dahiya S, Castaneyra-Ruiz L, Merisaari H, Botteron

HE, Alexopoulos D, George A, Peng S, Song SK, Limbrick DD, Smyser CD. Evaluating

white matter injury in posthemorrhagic hydrocephalus via diffusion basis spectrum

imaging. Brain 2020. (Submitted).

3. Isaacs AM, Smyser CD, Lean RE, Alexopoulos D, Han RH, Neil JJ, Zimbalist SA, Rogers

CE, Yan Y, Shimony JS, and Limbrick DD, Jr. MR diffusion changes in the perimeter of

the lateral ventricles demonstrate periventricular injury in post-hemorrhagic hydrocephalus

of prematurity. NeuroImage Clinical 2019. 24

In the previous chapter, we leveraged human CSF to establish that the pathophysiology of post- infectious (PIH) and post-hemorrhagic (PHH) hydrocephalus are similar, in that they are both propagated by inflammation-related disruption of the ependymal barrier. However, it is important to note that the infection and hemorrhage that herald PIH and PHH typically occur at a neonatal age when neural stem cells in the VZ and SVZ are actively differentiating to form the ependymal barrier. Therefore, to gain further insights into the pathophysiology of PIH and PHH, it is

81 imperative to characterize the ventricular zone (VZ) and subventricular zone (SVZ) in PIH/PHH infants. There is considerable histological evidence from human post-mortem tissues and animal models to support our CSF findings that the VZ and SVZ are critical regions in hydrocephalus pathophysiology133,175,197,318. However, there are presently no human studies that have been able to examine those regions in vivo. The ability to examine the VZ/SVZ non-invasively, would advance the diagnosis, care, monitoring, and prognostication of infants who develop perinatal infections and IVH. Additionally, non-invasive characterization of the VZ/SVZ will afford a foundation for future studies of therapeutic interventions which address the effects of hydrocephalus on those regions.

While diffusion tensor imaging (DTI) offers a unique opportunity to assess the VZ and SVZ regions in granular detail, it has not been utilized to date for characterizing those regions. Perhaps the biggest hurdle to utilizing any neuroimaging modality for these purposes has been the seemingly insurmountable challenge of overcoming volume averaging effects at the margin of the ventricle where the VZ and SVZ reside.

In this chapter, we present two major studies. The first study presents a novel semi-automated dMRI segmentation algorithm, which we developed, that can delineate the VZ, SVZ, and thin layer of PVWM to a high precision of 1.2 mm3 voxels (a region we have since named the Lateral

Ventricular Perimeter, LVP). We established the feasibility of our algorithm in human preterm infants who developed PHH or non-hydrocephalic IVH in comparison to preterm and full-term

82 infants with no brain injury. DTI characterization of the LVP helped gain further insights into the pathophysiology and deleterious effects of hydrocephalus on ependymal CSF-brain barrier.

There is evidence that disruption of the LVP exposes the subjacent large bundle PVWM to injury by toxin-laden CSF. PVWM injury, especially in tracts such as corpus callosum (CC), corticospinal tracts (CST), and optic radiations (OPRA), has been implicated in the majority of the debilitating neurological impairments seen in hydrocephalic infants. Therefore, our second study focused on assessing the effect of PHH on the CC, CST, and OPRA of our patient cohort. In that study, we used diffusion basis spectrum imaging (DBSI) instead of DTI for three principal reasons.

First, DTI has been extensively used by our group and others to study PVWM in various forms of hydrocephalus. Second, unlike DTI, DBSI can separate anisotropic tensors to model and resolve crossing fibers, assess injured axons and demyelination, and parse the isotropic tensor spectrum into components to reflect cellularity and edema. Third, DBSI related dMRI findings to histology, which not only helped to co-validate the dMRI findings but also provided further evidence for the associations between LVP disruption and PVWM injury.

83 3.2 Results

3.2.1 Segmentation of the lateral ventricular perimeter (LVP) on diffusion MRI

3.2.1.1 Segmentation requires minimal user input

Here we detail the semi-automated delineation of the LVP in infants whose DTI data were extracted from LVP to elucidate the role of VZ/SVZ disruption in the pathophysiology of PHH.

In MATLAB, the algorithm was set up to first read standard processed DTI files. Images showing mean diffusivity (MD) were then sequentially displayed to allowed us to select slices centered on the foramen of Monro. Once the central slice was determined, the algorithm automatically selected three total axial slices (one rostral and one caudal to the center slice) for further analysis. The MD image was used since it provides a sharp demarcation between the brain parenchyma and the CSF.

The script then displayed a bimodal histogram of the MD values with the lower peak representing

MD values in the brain parenchyma, and the higher peak representing CSF spaces. Based on the histogram, we determined a threshold value that best separated the parenchyma and CSF MD values. Following this, the script produced an image showing voxels with MD values above the threshold. Through automated sequential erode and dilate steps, the selected regions were smoothed, and small extraneous spaces were removed.

The algorithm was capable of automatically calculating the number of objects in each slice and the area of each object (blob analysis), then displaying the information. A predetermined minimum size (we used 10 voxels) was applied to display only areas likely to be involved in the ventricular system. The script also determined the center of mass of each object, with the assumption that true 84 ventricular objects will be close to the center of the brain. At this stage, the number of objects to be further processed was selected, including all objects that represent ventricular spaces. An image of all objects greater than the minimum size was then produced by the script, allowing us to click near the center of mass of each object to be included in the final selection. Once the relevant objects were selected, they were automatically saved, and extraneous objects were removed from further analysis. This process was repeated for each of the three contiguous slices automatically.

3.2.1.2 The frontal and occipital horns of the lateral ventricle can be independently assessed

In our cohort, we independently segmented the anterior (frontal) and posterior (occipital) horns of the lateral ventricle. To select only the frontal and occipital horns (FOHP), the previously segmented LVP objects were modified as follows: 1) The center point of the LVP in the anterior- posterior (A-P) direction was calculated, dividing the brain into an anterior and posterior segment;

2) The algorithm then located the most anterior and most posterior voxels that were already included in the LVP, to select all voxels that were within a predetermined A-P distance from the most extreme points for inclusion in the frontal and occipital horn perimeters.

3.2.1.3 Semi-automated algorithm strongly corelates with manually segmented LVP

With minimal user input as previously described, the algorithm automatically isolates periventricular voxels by performing two sequential dilation steps, incorporating voxels that are one and two spaces away from the ventricles and then subtracting the original ventricle objects from the dilated objects (Figure 21). Extraction of DTI data from the LVP was performed by applying numerical thresholds to MD maps to isolate the lateral ventricles as being central

85 structures with high MD values. To assess for potential differences between the algorithm and manually placed ROIs, dMRI of 20 neonates of varying ventricular morphologies were selected and manual ROIs were placed by an independent expert rater. Pearson correlation analyses between dMRI measures from both approaches indicated a strong positive association between the automated and manual segmentations fractional anisotropy (r (20) = .91, p <.001) and MD (r(20)

= .89, p <.001) measures.

Figure 21: Segmentation of Regions of Interest (ROIs)

Representative contiguous axial mean diffusivity maps of infants with variable ventricular morphologies demonstrating segmentation of the lateral ventricular perimeter (top row) and frontal and occipital horn perimeter (bottom row).

3.2.2 Region-specific dMRI-derived LVP disruption underlies PHH pathophysiology

3.2.2.1 Patient characteristics

This study leveraged a large cohort of prospectively recruited very preterm infants with/without

IVH/PHH and full-term healthy control infants in order to assess the effects of PHH on LVP. The first group included infants born ≤30 weeks gestational age with no or low-grade IVH (very

86 preterm, VPT group). The second group (high-grade intraventricular hemorrhage, HG-IVH group), was comprised of age-matched VPT infants who developed grade III/IV IVH identified on cranial ultrasound within the first month of life. The third group (post-hemorrhagic hydrocephalus,

PHH group), was comprised of VPT infants who sustained grade III/IV IVH identified on cranial ultrasound within the first month of life and required neurosurgical treatment for hydrocephalus.

The fourth group (full-term, FT group) were healthy infants born at 38–42 weeks gestational age, who were identified from a larger cohort undergoing neonatal neuroimaging scans as part of a contemporaneous fetal monitoring study. All patients underwent dMRI at term equivalent age.

A total of 206 infants (FT=76, VPT=91, HG-IVH=20, PHH=19) with DTI data were screened for possible inclusion in the study. Reasons for study exclusion across the groups included: acidosis

(FT=8), positive maternal urine drug screen (FT=5, VPT=1, HG-IVH=1), atypical pattern of brain injury/abnormal MRI finding (FT=2, VPT=2). (Figure 22).

Figure 22: Flowchart of subjects screening and enrolment for study

87 Of the 158 infants (77 males and 81 females) included in the final study cohort, there were 56 FT,

72 VPT, 17 HG-IVH, and 13 PHH infants. Median frontal-occipital horn ratios were higher in

PHH (0.50 ± 0.06) > HG-IVH (0.41 ± 0.04) = VPT (0.41 ± 0.02) > FT (0.37 ± 0.02) (Table 2;

Figure 23). All 13 PHH infants had undergone reservoir placement prior to their study MRI, with

6 of these infants also undergoing shunt placement prior to their MRI scan. There were no significant differences in MD or FA values either in the FOHP or LVP between PHH infants who underwent their shunt surgery before vs. after their MRI scan. Of the 13 PHH infants, 12 had reservoirs and 2 had subgaleal shunts placed prior to VP shunt or ETV surgery. For permanent hydrocephalus treatment, 10 infants had VP shunts placed and 2 underwent ETV surgery. As expected, the median ventricular size of the ETV infants was larger (0.60±0.01) than that of the

VP shunt infants (0.49±0.05) (p=0.008). Nevertheless, in this limited cohort, there was no difference in dMRI measures between infants who underwent ETV vs. VP shunt in both the LVP and FOHP regions. One infant underwent treatments for hydrocephalus with a reservoir but did not require permanent CSF diversion.

Table 2: Characteristics of 158 infants in a prospective study to assess the effects of IVH/PHH on dMRI measures of the lateral ventricular layer

FT VPT HG-IVH PHH # of patients 56 (35.4%) 72 (45.6%) 17 (10.8%) 13 (8.2%) Sex (# of Male/Female) 27 /29 30 /42 7/9 13/0 Birth gestational age (weeks) 39.0±1.2 27.0±1.7 25.0±2.0 24.0±2.1 Term age equivalent (weeks) 39.0±1.2 37.0±1.5 38.0±2.1 39.0±1.1 Birth weight (g) 3229.50±461.14 940±257.67 735±266.50 780±216.27 Clinical risk index score 0.00±0.00 1.00±1.87 3.00±1.69 3.00±2.33 Frontal-occipital horn ratio 0.37±0.02 0.41±0.02 0.41±0.04 0.50±0.06 All measures within the respective groups are medians ± standard deviations where necessary

88

Figure 23: Assessment of ventricular size

Ventricular size was determined by the frontal and occipital horn ratio. Axial T2W image of a normal neonate (left) and an infant with hydrocephalus (right). Ventricular size is determined by the frontal and occipital horn ratio which was calculated as (훼 + β)/2훿 or by the Evan’s Index calculated as (α/ 훿). Box and whisker diagram demonstrating the PHH group had the largest frontal-occipital horn ratio among 158 infants (p <.001, ***). There was no difference in ventricular size between the VPT and HG-IVH infants, although both groups had median ventricular sizes that were larger than that of the FT control infants.

3.2.2.2 PHH is associated with segmental aberration of dMRI measures in the LVP

In the LVP, family-wise comparisons with Benjamini-Hochberg correction revealed that PHH had consistently lower fractional anisotropy (FA) and higher mean (MD) and radial (RD) diffusivities than FT and VPT infants (p<.050). While PHH FA was lower, and PHH RD was higher than that of HG-IVH (p<.050) in the LVP, the MD and AD values did not differ (Tables 3 and 4; Figure

24). In the FOHP, Benjamini-Hochberg corrected family-wise comparisons revealed PHH infants had lower FA and higher RD than FT and VPT infants in the FOHP (p<.010) (Tables 3 and 4;

Figure 25). Similar to the LVP, PHH FA was lower than that of HG-IVH (p<.001) in the FOHP.

Except for MD and RD in the LVP region, there were no differences in dMRI measures between the HG-IVH and VPT infants. Among the PHH group, there was no difference in dMRI measures

89 between the ETV and VP shunt treated infants (p>.050) in both the LVP and FOHP regions. While the magnitude of PHH AD was consistently less than AD measures in the other groups, in both the

LVP and FOHP pairwise comparisons were not significant (p>.050). Comparison of LVP/FOHP dMRI measures to those of adjacent CST and OPRA revealed tract-specific associations between dMRI measures. Among the HG-IVH infants, dMRI metrics in both the LVP and FOHP were associated with OPRA microstructure, but no associations were found with the CST. In contrast, few associations were found for the PHH group.

Table 3: Median dMRI measures among 157 full- and pre-term infants with/or without IVH/PHH

FT VPT HG-IVH PHH Sig. Lateral ventricular perimeter FA 0.27±0.05 0.23±0.04 0.21±0.04 0.16±0.05 <0.001* MD (μm2/s) 1.46±0.09 1.47±0.08 1.55±0.11 1.58±0.15 <0.001* AD (μm2/s) 1.88±0.12 1.85±0.15 1.93±0.18 1.82±0.25 0.031* RD (μm2/s) 1.24±0.15 1.29±0.12 1.38±0.10 1.48±0.15 <0.001* Frontal-occipital horn perimeter FA 0.29±0.07 0.22±0.06 0.20±0.05 0.13±0.04 <0.001* MD (μm2/s) 1.59±0.11 1.66±0.19 1.73±0.14 1.77±0.24 <0.001* AD (μm2/s) 2.13±0.14 2.09±0.27 2.11±0.21 1.94±0.32 0.173 RD (μm2/s) 1.33±0.17 1.45±0.18 1.56±0.15 1.66±0.24 <0.001* All measures within the respective groups are medians ± standard deviations where necessary; One-way ANOVA was performed with Kruskall-Wallis test (Sig.). All p-values are Benjamini-Hochberg corrected with alpha level set at p <0.050 (*). FA = fractional anisotropy; MD = mean diffusivity; AD = axial diffusivity; RD = Radial diffusivity; PHH = post hemorrhagic hydrocephalus; HG-IVH = high grade intraventricular hemorrhage; VPT = very preterm; FT = full term

90 Table 4: Groupwise comparisons between 157 full- and pre-term infants with/or without IVH/PHH

FT:VPT FT:HG-IVH FT:PHH VPT:HG-IVH VPT:PHH HG-IVH:PHH Lateral ventricular perimeter FA <0.001* <0.001* <0.001* 0.100 <0.001* 0.004* MD (μm2/s) 1.000 0.011* 0.027* 0.008* 0.034* 1.000 AD (μm2/s) 0.299 1.000 1.000 0.055 1.000 0.869 RD (μm2/s) 0.057 0.003* <0.001* 0.014* <0.001* 0.048* Frontal-occipital horn perimeter FA <0.001* <0.001* <0.001* 0.349 <0.001* <0.001* MD (μm2/s) 0.022* 0.003* 0.102 0.213 0.700 1.000 AD (μm2/s) 1.000 1.000 0.281 1.000 1.000 0.567 RD (μm2/s) 0.002* <0.001* 0.007* 0.118 0.007* 0.869 All groupwise comparisons performed by Wilcoxon signed rank family-wise comparisons. All p-values are Benjamini-Hochberg corrected with alpha level set at p <0.050 (*). FA = fractional anisotropy; MD = mean diffusivity; AD = axial diffusivity; RD = Radial diffusivity; PHH = post hemorrhagic hydrocephalus; HG-IVH = high grade intraventricular hemorrhage; VPT = very preterm; FT = full term

Figure 24: dMRI measures of the lateral ventricular perimeter (LVP) The LVP demonstrated increasing severity of brain injury corresponds with decreasing fractional anisotropy (FA) and increasing mean (MD) and radial (RD) diffusivities. No significant changes were observed in axial diffusivity (AD) measures between groups.

91

Figure 25: dMRI measures of the frontal occipital horn perimeter (FOHP) The FOHP demonstrated increasing severity of brain injury corresponds with decreasing fractional anisotropy (FA) and increasing mean (MD) and radial (RD) diffusivities. No significant differences were observed in axial diffusivity (AD) measures between groups. 3.2.2.3 The FOHP region is more severely injured than the rest of the LVP

A linear mixed effects model, adjusted for term equivalent age at scan, revealed regional FA and

RD differences between the FOHP and LVP [F (3,153) = (FA (19.46), MD (0.98), AD (1.88), RD

(3.16); p<.001)]. In the FT infants, FA, MD, AD and RD measures in the FOHP were higher than the LVP, which reflects baseline differences between the two regions among healthy infants in this cohort. However, in the PHH group, while MD, RD and AD measures in the FOHP were higher than the LVP, FOHP FA was less than the LVP, directly opposite to the observed findings in the healthy infants. (Table 5; Figure 26). Nevertheless, directionality of the principal eigenvectors in both LVP and FOHP generally did not differ between groups (Figure 27).

92 Table 5: Region-specific dMRI measures examining between-group differences among 158 infants

Lateral ventricular perimeter Frontal occipital horn perimeter Mean difference Estimate Lower Upper Estimate Lower Upper (LVP-FOHP) Fractional anisotropy FT 0.26 0.24 0.27 0.29 0.27 0.30 -0.03 VPT 0.22 0.21 0.24 0.22 0.21 0.23 0.00 HG-IVH 0.20 0.18 0.23 0.20 0.17 0.23 0.00 PHH 0.15 0.12 0.18 0.12 0.09 0.15 0.03 Mean diffusivity(μm2/s) FT 1.46 1.42 1.50 1.60 1.56 1.63 -0.13 VPT 1.47 1.44 1.51 1.63 1.60 1.67 -0.16 HG-IVH 1.55 1.49 1.62 1.73 1.66 1.80 -0.17 PHH 1.58 1.51 1.66 1.73 1.65 1.81 -0.15 Axial diffusivity(μm2/s) FT 1.88 1.82 1.93 2.11 2.05 2.16 -0.23 VPT 1.82 1.77 1.86 2.02 1.97 2.07 -0.20 HG-IVH 1.90 1.80 2.00 2.10 2.00 2.20 -0.20 PHH 1.77 1.66 1.88 1.92 1.81 2.03 -0.15 Radial diffusivity(μm2/s) FT 1.26 1.22 1.31 1.35 1.31 1.39 -0.09 VPT 1.31 1.27 1.35 1.45 1.41 1.49 -0.14 HG-IVH 1.38 1.30 1.46 1.54 1.46 1.62 -0.16 PHH 1.51 1.42 1.60 1.65 1.56 1.74 -0.14 Summary of least square means from a linear mixed effects model. PHH = post hemorrhagic hydrocephalus; HG-IVH = high grade intraventricular hemorrhage; VPT = very preterm; FT = full term. Lower and upper values are confident intervals

Figure 26: Effects of region of segmentation on dMRI measures 93 Between groups the FOHP in PHH infants had higher MD, AD and RD as well as lower FA measures than the LVP, an indication the FOHP is more disrupted than the LVP. Error bars are standard error means.

Figure 27: Representative principal eigenvector maps of white matter orientation The eigen vector analyses of the periventricular white matter in healthy full-term (FT) and post hemorrhagic hydrocephalus (PHH) infants demonstrated orientation of the direction of maximum water displacements (arrows) along the perimeter of the ventricles (V) was generally preserved between groups in spite of the reduced anisotropy in the PHH group. 3.2.2.4 Ventricular size correlates with dMRI measures in the LVP and FOHP

Spearman’s rho revealed that frontal-occipital horn ratio correlated with dMRI measures in the

LVP. Ventricular size had negative correlation with FA (rs=-0.41 p<.001), with anticipated positive correlations with MD (rs=0.26, p=.005) and RD (rs=0.36, p<.001) in the LVP (Figure 28).

Relatively stronger relationships were found in the FOHP in that ventricular size was negatively correlated with FA (rs=-0.47, p<.001) and positively correlated with MD (rs=0.35, p<.001) and

RD (rs=0.42, p<.001) (Fig. 29). AD measures did not correlate with ventricular size in either the

LVP or FOHP segmentation analyses (rs=-0.05, p=1.000).

94

Figure 28: dMRI-ventricular size correlations in the LVP Spearman correlation analyses demonstrated that larger ventricular size was associated with higher MD and RD and lower FA, suggesting ventricular enlargement is associated with dMRI-measured LVP disruption.

Figure 29: dMRI-ventricular size correlations in the FOHP Spearman correlation analyses demonstrated larger ventricular size was associated with higher MD and RD and lower FA, suggesting ventricular enlargement is associated with dMRI-measured FOHP disruption.

95 3.2.3 DBSI differentiates PVWM pathology in PHH

3.2.3.1 Subject characteristics

Prospectively recruited infants assessed between May 2007 and August 2016 were categorized into four groups: full term (FT), very preterm (VPT), intraventricular hemorrhage (IVH) and post- hemorrhagic hydrocephalus (PHH). A total of 206 infants (FT=76, VPT=91, IVH=20, PHH=19) with dMRI data were assessed for possible inclusion. Sixty-six patients did not meet inclusion criteria for the reasons outlined in Figure 30, which included having a positive maternal urine drug screen, atypical pattern of brain injury/abnormal MRI finding that precluded accurate segmentation of ROIs, and dMRI data that could not be fitted with DBSI due to having less than

25 diffusion-encoding directions and/or b-values.

Figure 30: Flowchart of patient enrolment and selection for study inclusion

96 Among 140 infants (70 males, 70 females) who underwent dMRI, 45 FT, 68 VPT, 15 IVH and 12

PHH infants were included in the analyses. The median term age equivalent of dMRI acquisition was 38±2 weeks. By definition, the PHH group had larger ventricles than the remaining groups

(median frontal-occipital horn ratio of 0.52.10 vs. 0.40.04). The median clinical risk scores of

IVH and PHH infants were similar and significantly higher than that of the VPT and FT infants.

(Table 6; Figure 31). Prior to their dMRI scan, all PHH infants (n=12) had undergone a temporary ventricular access device placement for serial CSF removal, with 10 of these infants eventually undergoing permanent shunt placement and two undergoing ETV to treat hydrocephalus. There were no differences between the shunt- and ETV-treated infants on all measures.

Table 6: Characteristics of infants in a prospective study to assess DBSI indices in PVWM

Full term Very preterm IVH PHH # of patients 45 68 15 12 Sex # of male infants 22 29 7 12 # of female infants 23 39 8 0 GA at birth (weeks) 39.0±2.0 27.0±2.8 25.0±3.0 24.5±4.0 TEA at dMRI acquisition (weeks) 39.0±2.0 37.0±1.0 38.0±4.0 39.0±1.0 Birth weight (g) 3195±714.0 932.5±350.0 740.0±330.0 807.5±304.5 Frontal-occipital horn ratio 0.36±0.03 0.41±0.03 0.41±0.04 0.52±0.10 Clinical risk score 0±0 1±3 3±2 3±4 All continuous variables are reported as median ± interquartile range within the respective groups. IVH = intraventricular hemorrhage; PHH= post-hemorrhagic hydrocephalus; GA = gestational age; TEA = term equivalent age

97

Figure 31: Ventricular size measurement and regions of interest segmentation Representative axial T2W of a preterm infant with post-hemorrhagic hydrocephalus (PHH) (top row). Ventricular size was determined by the frontal-occipital horn ratio which was calculated as (a + c)/2b 20. The Box and whisker diagram demonstrate the PHH group had the largest ventricular size among 140 infants (p<.001, ***). There was no difference in ventricular size between the VPT and IVH infants, although both groups had median ventricular sizes that were larger than that of the FT control infants. Bottom row demonstrates representative contiguous axial fractional anisotropy maps of infants with demarcation of the segments of the white matter bundles of interest within the corpus callosum, bilateral corticospinal tracts and bilateral optic radiations.

3.2.3.2 Neuroinflammation and reduced fiber density in the corpus callosum

In the CC, PHH infants had the lowest FA and highest RD (p<.001), suggesting they had more severe white matter disruption than non-PHH infants. Quantification of fiber density demonstrated that the PHH group had 71%, 70% and 53% less white matter fiber density than the FT, VPT and

IVH groups, respectively (p<.001). However, the IVH group also had lower FA, higher RD and

32% and 38% lower fiber density in comparison to the FT and VPT infants, respectively (p<.01).

This indicates hemorrhage had an independent detrimental effect on white matter integrity, albeit exacerbated by hydrocephalus. Isotropic diffusion metrics revealed the PHH group had the highest restricted fraction, suggesting cellular infiltration when compared to the non-PHH groups

98 (p<.001). Hypercellularity would be expected to be associated with reduced FA and AD measures,

and, indeed, fiber level analyses confirmed the PHH group had the lowest FFA and FAD (p<.001).

FRD and the non-restricted fraction (a marker of edema) did not differ between groups (Figure

32). Across tracts, dMRI measures and p values for one-way and groupwise comparisons are

presented in Tables 7 and 8, respectively.

Table 7: dMRI measures of PVWM in full term and preterm infants with/or without IVH/PHH and their frontal-occipital horn ratio-dMRI correlations

Measures Ventricular size-dMRI PHH IVH VPT FT Rho P-value FA 0.11 ± 0.11 0.34 ± 0.40 0.42 ± 0.44 0.43 ± 0.57 -0.34 <0.001* AD 2.02 ± 1.48 2.25 ± 1.03 2.18 ± 0.94 2.15 ± 1.61 -0.06 0.474 RD 1.61 ± 1.39 1.20 ± 0.89 1.08 ± 1.03 1.01 ± 1.59 0.28 0.001* MD 1.73 ± 1.42 1.56 ± 0.77 1.43 ± 0.81 1.38 ± 1.29 0.22 0.011* FFA 0.46 ± 0.32 0.70 ± 0.48 0.71 ± 0.29 0.69 ± 0.50 -0.14 0.106 CC FAD 1.80 ± 1.30 2.42 ± 1.19 2.21 ± 1.19 2.18 ± 1.27 -0.20 0.018* FRD 0.71 ± 0.59 0.57 ± 0.38 0.56 ± 0.59 0.58 ± 0.61 -0.02 0.847 FF 0.17 ± 0.30 0.36 ± 0.47 0.53 ± 0.66 0.58 ± 0.77 -0.28 0.001* RF 0.53 ± 0.56 0.34 ± 0.74 0.28 ± 0.72 0.22 ± 0.71 0.32 <0.001* NRF 0.28 ± 0.80 0.29 ± 0.80 0.16 ± 0.58 0.18 ± 0.75 0.05 0.547 FA 0.39 ± 0.34 0.51 ± 0.36 0.51 ± 0.22 0.56 ± 0.40 -0.10 0.236 AD 1.58 ± 0.55 1.77 ± 0.62 1.75 ± 0.25 1.76 ± 0.52 0.16 0.054 RD 0.84 ± 1.02 0.69 ± 0.84 0.72 ± 0.35 0.66 ± 0.40 0.14 0.109 MD 1.13 ± 0.87 1.05 ± 0.68 1.03 ± 0.28 1.02 ± 0.17 0.14 0.109 FFA 0.65 ± 0.40 0.71 ± 0.57 0.71 ± 0.38 0.76 ± 0.31 -0.25 0.003* CST FAD 1.45 ± 0.86 1.92 ± 1.11 1.96 ± 0.42 1.87 ± 0.49 0.21 0.011* FRD 0.66 ± 0.22 0.42 ± 0.44 0.48 ± 0.61 0.42 ± 0.37 0.21 0.011* FF 0.49 ± 0.52 0.59 ± 0.65 0.62 ± 0.44 0.65 ± 0.56 -0.09 0.273 RF 0.39 ± 0.44 0.29 ± 0.42 0.25 ± 0.43 0.27 ± 0.57 0.00 0.973 NRF 0.05 ± 0.73 0.03 ± 0.23 0.02 ± 0.10 0.03 ± 0.19 0.06 0.493 FA 0.24 ± 0.68 0.36 ± 0.32 0.37 ± 0.22 0.38 ± 0.23 -0.18 0.032 AD 2.04 ± 1.03 1.91 ± 0.91 1.95 ± 0.35 1.81 ± 0.72 0.48 <0.001* RD 1.41 ± 1.41 1.21 ± 0.60 1.21 ± 0.54 1.20 ± 0.60 0.16 0.056 MD 1.65 ± 1.04 1.48 ± 0.55 1.45 ± 0.42 1.39 ± 0.61 0.29 0.001* FFA 0.63 ± 0.32 0.68 ± 0.28 0.67 ± 0.25 0.72 ± 0.26 -0.18 0.031* OPRA FAD 2.53 ± 0.62 2.18 ± 0.92 2.22 ± 0.54 2.01 ± 0.54 0.47 <0.001* FRD 0.74 ± 0.72 0.76 ± 0.45 0.74 ± 0.47 0.65 ± 0.33 0.28 0.001* FF 0.36 ± 0.41 0.51 ± 0.33 0.53 ± 0.33 0.45 ± 0.38 -0.02 0.798 RF 0.53 ± 0.66 0.40 ± 0.44 0.41 ± 0.34 0.41 ± 0.44 -0.03 0.700 NRF 0.10 ± 0.28 0.04 ± 0.34 0.03 ± 0.24 0.05 ± 0.24 0.13 0.135 All continuous variables are reported as median ± standard deviation within the respective groups. IVH = intraventricular hemorrhage; PHH= post-hemorrhagic hydrocephalus; Groupwise comparisons were done with the Wilcoxon signed rank test. Ventricular size- diffusion MRI (dMRI) correlations were assessed

99 with Spearman Rho correlations. All p-values were Bonferroni corrected and significant p-values (p<.050) are asterisked (*).

Table 8: Levels of significance for one-way and groupwise comparisons of dMRI measures in full term and preterm infants with/or without IVH/PHH

Groupwise comparisons One- PHH vs PHH- PHH- PHH- IVH- IVH- VPT-

way Non-PHH FT VPT IVH FT VPT FT FA <.001 <.001 <0.001* <0.001* <0.001* 0.004* 0.002* 0.462 AD 0.489 0.278 0.394 0.314 0.196 0.379 0.235 0.854 RD <.001 <.001 <0.001* <0.001* 0.026* 0.003* 0.001* 0.313 MD 0.002 0.023 0.024* 0.018* 0.428 0.005* 0.005* 0.192 FFA <.001 <.001 <0.001* <0.001* <0.001* 0.612 0.867 0.221 CC FAD <.001 <.001 <0.001* <0.001* <0.001* 0.158 0.240 0.377 FRD 0.480 0.160 0.213 0.152 0.367 0.973 0.606 0.522 FF <.001 <.001 <0.001* <0.001* 0.002* 0.003* 0.001* 0.585 RF <.001 <.001 <0.001* <0.001* 0.016* 0.002* 0.035* 0.010* NRF 0.127 0.145 0.305 0.070 0.683 0.446 0.086 0.208 FA <.001 <.001 0.002* <0.001* <0.001* 0.102 0.438 0.114 AD <.001 <.001 0.001* <0.001* <0.001* 0.919 0.745 1.000 RD <.001 <.001 <0.001* <0.001* <0.001* 0.543 0.069 0.022* MD <.001 <.001 <0.001* <0.001* 0.003* 0.117 0.504 0.114 FFA <.001 <.001 <0.001* <0.001* 0.025* 0.163 0.864 <0.001* CST FAD <.001 <.001 <0.001* <0.001* <0.001* 0.139 0.329 <0.001* FRD <.001 <.001 <0.001* <0.001* <0.001* 0.407 0.372 <0.001* FF <.001 <.001 0.004* <0.001* 0.002* 0.488 0.623 0.483 RF 0.001 <.001 0.038* <0.001* 0.012* 0.906 0.179 0.344 NRF <.001 0.005 0.080 <0.001* 0.152 0.879 0.047* 0.001* FA 0.004 0.001 0.001* 0.001* 0.015* 0.397 0.692 0.092 AD <.001 0.037 0.009 0.135 0.069 0.242 0.042* <0.001* RD 0.139 0.029 0.066 0.026* 0.109 0.477 0.489 0.498 MD 0.005 0.016 0.018* 0.019* 0.134 0.060 0.582 0.007* FFA <.001 0.012 0.005* 0.025* 0.121 0.028* 0.701 <0.001* OPRA FAD <.001 <.001 <0.001* <0.001* 0.001* 0.005* 0.674 <0.001* FRD <.001 0.307 0.031* 0.729 0.959 <0.001* 0.854 <0.001* FF <.001 <.001 0.022* <0.001* 0.003* 0.417 0.295 0.009* RF 0.025 0.004 0.018* 0.003* 0.024* 0.467 0.799 0.338 NRF <.001 0.001 0.042* <0.001* 0.027* 0.761 0.154 0.008* All continuous variables are reported as median ± standard deviation within the respective groups. IVH = intraventricular hemorrhage; PHH= post-hemorrhagic hydrocephalus; Groupwise comparisons were done with the Wilcoxon signed rank test. All p-values were Bonferroni corrected and significant p-values (p<.050) are asterisked (*).

100

Figure 32: dMRI measures of the corpus callosum (CC) The CC dMRI metrics demonstrated PHH infants had the most severe white matter disruption, which was marked by having the lowest fiber level FA and AD indices when compared to IVH, VPT and FT infants. PHH was also associated with a reduced white matter fiber density (Fiber Fraction), and the relatively highest measures of cellular infiltration (Restricted Fraction) in the CC. No differences were observed in fiber RD and markers of edema (Non-restricted Fraction) between groups. ***represents p-value<.001; FT=full term; VPT= very preterm; IVH=intraventricular hemorrhage; PHH=post-hemorrhagic hydrocephalus; FA= fractional anisotropy; AD=axial diffusivity; RD=radial Diffusivity.

3.2.3.3 Neuroinflammation, reduced fiber density, and demyelination in corticospinal tracts

In the CST, the PHH group again had the lowest FA, lowest AD and highest RD, implying more severe white matter disruption than in PHH compared to non-PHH infants (p<.001). Fiber-specific metrics demonstrated PHH had the lowest FFA and FAD, and the highest FRD when compared to

101 the non-PHH groups (p<.001). White matter fiber density quantification showed the PHH group had 25%, 21% and 17% less fibers than the FT (p=.004), VPT (p<.001) and IVH (p=.002) groups, respectively. The PHH group also had the highest measures of restricted fraction, suggesting a higher cellular infiltration than in the non-PHH groups (p<.001). Non-restriction fraction, a marker of edema, only differed between the PHH and VPT groups (p<.001). (Figure 33).

Figure 33: dMRI measures of the corticospinal tracts (CST) The CST dMRI metrics demonstrated PHH infants had the most severe white matter disruption, which was marked by having the lowest fiber level FA and AD indices, as well as highest measures of RD when compared to IVH, VPT and FT infants. PHH was also associated with reduced white matter fiber density and the highest measures of cellular infiltration in the. No between group differences were observed in non-restricted fraction, a marker of vasogenic edema. ***, ** and * represents p-value <.001, <.01 and <.05, respectively; FT=full term; VPT= very preterm; IVH=intraventricular hemorrhage; PHH=post-hemorrhagic hydrocephalus; FA= fractional anisotropy; AD=axial diffusivity; RD=radial Diffusivity.

102 3.2.3.4 Neuroinflammation, reduced fiber density, and edema in optic radiations

The OPRA in the PHH group also demonstrated disruption when compared to the non-PHH groups. However, the pattern differed from that of the CC and CST. Specifically, while the PHH group had the lowest FA values (p=.001), their AD and RD measures did not differ from the non-

PHH groups. Fiber level analysis showed that FFA in the PHH group was lower than that of the

FT (p=.005) and VPT (p=.025) but did not differ from the IVH group (p=.12). FFA of the IVH and VPT groups were lower than that of the FT group (p=.028 and p<.001, respectively), but did not differ from each other (p=.70). Fiber level RD was lower in FT than very preterm infants: PHH

(p=.03), IVH (p<.001), and VPT (p<.001). Together, these findings suggest hemorrhage and preterm birth were independent contributing factors to impaired white matter disruption in the

OPRA. White matter fiber density quantification demonstrated the PHH infants had 20%, 32% and 29% less fibers than the FT (p=.02), VPT (p<.001) and IVH (p=.003) infants, and the PHH group had the highest measures of fiber level AD (p<.001). Isotropic tensor analyses demonstrated evidence of inflammation in the OPRA of PHH infants, given their increased measures of white matter edema (NRF; p=.001) and cellularity (RF; p=.004) compared to non-PHH groups (Figure

34).

103

Figure 34: dMRI measures of the optic radiations (OPRA) The OPRA dMRI metrics demonstrated PHH infants had the most severe white matter disruption, which was marked by having the lowest fiber level FA when compared to IVH, VPT and FT infants. PHH was also associated with reduced white matter fiber density (Fiber Fraction) and the highest measures of fiber level AD. In addition, the PHH group demonstrated evidence of severe inflammation marked by the highest measures of vasogenic edema (Non-restricted Fraction) and cellular infiltration (Restricted Fraction). ***, ** and *represents p-value <.001, <.01 and <.05, respectively; FT=full term; VPT= very preterm; IVH=intraventricular hemorrhage; PHH=post- hemorrhagic hydrocephalus; FA= fractional anisotropy; AD=axial diffusivity; RD=radial diffusivity.

3.2.3.5 Ventricular size correlates with magnitude of PVWM disruption

Across all three white matter bundles, greater ventricular size demonstrated varying correlation strength with poorer dMRI measures with variable patterns. In the CC, ventricular size related

104 negatively with FA (p<.001) and fiber density (p=.001) and positively with RD (p<.001) and cellular infiltration (p<.001). In the CC, FAD negatively correlated with ventricular size (p=.018), while in the CST (p=.01) and OPRA (p<.001) positive correlations were observed. This likely reflects differences between the CC and CST/OPRA at this developmental stage including myelination and compactness of the respective white matter fiber bundles. (Figure 35).

Figure 35: Ventricular size-dMRI correlations Spearman correlation analyses demonstrated increase in ventricular size was correlated with poorer dMRI measures, but in variable patterns across the corpus callosum (CC), corticospinal tracts (CST) and optic radiations (OPRA). In the CC, as ventricular size increases, a decrease in fiber AD and white matter fiber density and an increase in cellular infiltration are observed. However, in both the CST and OPRA, increases in ventricular size were associated with decreases in fiber FA and AD and increases in fiber RD. Ventricular size was obtained with frontal occipital 105 horn ratio approach20. Significant correlations are represented by bold image borders where ***, ** and * represents p-value <.001, <.010 and <.050 respectively; FT=full term; VPT= very preterm; IVH=intraventricular hemorrhage; PHH=post-hemorrhagic hydrocephalus.

3.2.3.6 Postmortem tissue neurocytology relates DBSI metrics to VZ disruption

Immunohistochemical analyses of the CC from postmortem tissues validated the dMRI findings.

In comparison to controls, PHH infants demonstrated white matter tracts with relatively higher cellularity, reflecting their high restricted fractions seen on dMRI. In addition, the PHH infants had decreased synaptophysin staining suggesting decreased neuropil and synaptic processes, which reflects their low fiber fractions on dMRI. While variable amount of cytoplasmic vacuolation suggesting edema was seen on histology, the dMRI correlate, non-restricted fractions did not differ between PHH and non-PHH groups, presumably the non-restricted fractions reflect interstitial or extracellular edema (Figure 36).

Figure 36. Representative neurocytology of post-mortem PVWM tissue

106 Representative histological staining of post-mortem tissue from corpus callosum (CC) of preterm infants. (A & B) H&E and (C & D) synaptophysin of preterm infants (A & C) without or (B & D) with PHH. PHH infants had relatively increased cellular infiltration (B & D) and vacuolation of their cytoplasm to suggest edema (B). Decreased axonal staining in PHH (D) suggests a relatively decreased fiber density in comparison to controls (C). The CC is not myelinated at term equivalent.

Analyses of the VZ/SVZ and adjoining PVWM demonstrated that IVH, and to a greater extent

PHH, were associated with hemorrhage as well as VZ/SVZ disorganization, as characterized by variable granular ependymitis on H&E staining. IVH/PHH was associated with a 20-30% relative increase in VZ disruption when compared with (p=.016) and FT infants (p=.011). In addition,

IVH/PHH was associated with disruption of radial glial cells in the VZ, characterized by loss of their basal projections and VZ lattice. Furthermore, there was relatively increased expression of

GFAP and IBA1, representing reactive astrocytosis and microglia/macrophage-associated inflammation in the VZ/SVZ regions of IVH/PHH when compared to FT infants. (Figure 37).

107

Figure 37. Representative immunohistochemistry of postmortem brain tissue In the full term, FT (A, E, H, K), very preterm, VPT (B, F and I) and infants with intraventricular hemorrhage or post-hemorrhagic hydrocephalus, IVH/PHH (C, G, J, and L), the H&E images (A- C) demonstrate an intact ventricular zone (VZ) with normal multiciliated ependymal cells (A), a region undergoing early VZ disruption with protrusion of the VZ toward the ventricular space with preserved cilia (B) and an established VZ disruption with denuded ependyma that has no cilia (C). (D) Percent VZ disruption, quantified as the cumulative length of the disrupted ependymal regions as a factor of the entire length of VZ available on the same slide, demonstrate IVH/PHH is associated with the severest VZ damage than VPT and FT infants (p<.05). Adjacent slides of the same infant’s double-labeled with anti-GFAP and anti-dapi demonstrates the characteristic shape of radial glial cells (E) with basal projections undergoing progressive changes in the pattern of GFAP expression, (F) initially showing a lack of basal projections and subsequently a proliferation of GFAP-positive

108 3.3 Discussion

3.3.1 The LVP is a critical region in PHH pathophysiology

Many of the debilitating symptoms of IVH and PHH have been attributed to PVWM injury

241,242,439,440. As such, the anatomical, physiological and molecular relationships between the LVP and PVWM have several critical research and clinical implications133,175,197,318. First, IVH/PHH in preterm infants typically occurs during a critical developmental period of VZ/SVZ differentiation, interfering with neural stem cell maturation and ependymal layer formation. Impairment in ependymogenesis leads to denuded regions that expose PVWM to injury from CSF contents, ciliopathy that impairs CSF circulation and overall dysregulation of cerebral homeostasis, immunologic response to injury and other normal cellular processes 232,233. In addition, the irritation effects of blood, ferritin and reactive oxidative species likely propagates injurious mechanisms of neuroinflammation, hypoxia and ischemia 131,197 that further compromises the LVP barrier and damages PVWM 122,133,184. Thus, examining LVP with dMRI in PHH patients may help focus future research on strategies that enhance LVP barrier properties in preterm infants to avert

PVWM injury and its neurological sequalae. Second, in PHH, the ventricular dilatation exerts mechanical pressure on both the LVP and PVWM 39,185, which may exacerbate ependymal denudation and axonal dysfunction due to direct axonal stretch and/or compression. Further, from a hydrocephalus treatment standpoint, CSF diversion surgery is typically performed to relieve the mechanical pressure, thereby preventing further damage and permitting recovery 284,287,291.

Therefore, using dMRI metrics to assess the relationships between ventricular size and LVP

109 integrity in IVH patients with or without PHH may help define the compounding effect of PHH on PVWM on top of IVH, and the role of therapeutic ventricular decompression.

3.3.2 Delineating the LVP for dMRI analyses

There are numerous technical challenges to using dMRI in neonates and high-risk clinical populations. From an acquisition perspective, specialized medical facilities, equipment, protocols and staff who are experienced in handling neonates and performing population-specific procedures implemented for collection of high-quality dMRI data are imperative 441,442. Moreover, since the majority of neonatal MRIs are acquired without sedation, there can be excessive motion leading to severe signal dropouts that compromise dMRI anisotropy. Furthermore, the relative differences in infant head sizes, cortical gyrification, and anatomical heterogeneity resulting from different forms/severities of brain injury also pose significant challenges to image acquisition and analyses

441. Thus, standard dMRI analysis pipelines (usually for adults) will typically fail when applied to neonatal data, compelling development of tailored pipelines that are able to account for those factors.

The LVP has been challenging to study with dMRI because of partial volume effects that may occur when adjacent or surrounding structures are included in the segmentation. This is because, in addition to being in close proximity to ventricular CSF and other tissues of varying signal intensities, the thickness of the LVP is only a few cell layers. Indeed, the ependymal layer is a single layer of epithelial cells (~5μm thickness), and the VZ/SVZ comprise a mix of radial glial

110 and neuroblast cells,123,133 which makes it technically challenging to segment a homogeneous ROI for the LVP. In this study, we developed a novel MATLAB-based algorithm capable of segmenting the innermost layer of the LVP and its adjoining PVWM with a spatial resolution of

1.2 mm3. This voxel size is larger than the SVZ. As a result, the ROIs likely contain a mixture of

SVZ and subjacent white matter. The ROIs had nonzero diffusion anisotropy, and evaluation of

Eigen vector maps (Fig. 26) showed fiber orientation parallel to the walls of the lateral ventricles, consistent with the orientation of white matter in these areas. In contrast, the SVZ would be expected to have isotropic diffusion or a primary orientation of maximum water displacements orthogonal to the walls of the lateral ventricles. Thus, the measured diffusion parameters likely reflect those of white matter, and injury to the SVZ was inferred through evidence of injury to the adjacent white matter. Further, the ROIs were generated with an automated algorithm, which helped eliminate manual and inter-/intra-rater related errors. In addition, the ROIs were averaged from three contiguous axial slices to increase signal to noise and to reduce through-plane partial volume averaging effects.

3.3.3 Cellular basis of dMRI-measured LVP injury in PHH

During typical white matter development, PVWM FA increases, while MD, RD and AD decrease in a time-dependent fashion 443,444. Our finding of lower FA and higher MD and RD in VPT compared to FT infants is consistent with previous studies showing that preterm birth is associated with dMRI-measured tissue abnormalities 445-447 and poor neurological outcomes, which have been attributed to disrupted neuronal proliferation, migration and synaptogenesis 448. In HG-IVH and

PHH the premature brain’s sensitivity to perturbations in normal cellular processes by hemorrhage

111 and hydrocephalus contributes significantly to their poorer neurological outcomes when compared to non-IVH preterm infants and healthy full term controls 439,440,449. Our finding of more prominent abnormalities in FA, MD and RD in HG-IVH and PHH infants recapitulates the patterns of

IVH/PHH-associated tissue injury that have been previously reported by dMRI studies of large bundle PVWM tracts such as the corpus callosum and internal capsules 272,284,450-452. In Chapter 4 we propose a molecular mechanism for LVP injury in the setting of IVH. Using cell cultures of

VZ cells from the LVP to demonstrate VZ disruption occurs as a result of cleavage of adherens junctions proteins 318, thereby exposing the PVWM to severe inflammation and gliosis 197,318.

Several other histopathological studies have shown that IVH is associated with LVP tissue damage, which is characterized by ependymal denudation, heterotopia and rosette formation, radial glial cell disorganization, and astrocytic proliferation in the VZ and SVZ 175,178,197.

3.3.4 PHH-associated myelin injury

Diffusion anisotropy data in white matter are typically interpreted in terms of changes to myelin, with an increase in RD thought to represent injury to myelin with relative sparing of axons. In the present context, this interpretation is complicated by the fact that most of PVWM is not myelinated at term equivalent 453. While white matter tracts associated with primary motor and sensory areas, particularly CST and OPRA, are myelinated at term equivalent and do show greater values for diffusion anisotropy than unmyelinated areas 443, the majority of PVWM is unmyelinated at this stage of development 453. Thus, for this study, the relative increase in RD compared with AD suggests greater spacing of axons, possibly related to transependymal migration of CSF, and/or an increase in axonal membrane permeability, perhaps related to stretching and injury 454-456. The

112 influence of these unmyelinated axons on the diffusion measurement is supported by the finding that the orientation of the direction of maximum water displacements (which indicates the orientation of white matter fiber tracts) was generally preserved in spite of the reduced anisotropy, suggesting that the axons were still present (Figure 26). Nevertheless, disruption of the LVP may be associated with subsequent abnormalities of myelination through injury to oligodendrocyte progenitor cells that are present in this region at term equivalent. Indeed studies of the CC of older hydrocephalic children after myelination 238,450 show MD changes driven by changes to RD rather than to AD, indicating myelin disruption. Other studies have identified morphological defects in oligodendrocytes, which occur as a result of hemorrhage and other multifactorial mechanisms including neuroinflammation and ischemia-induced excitotoxity 457-462. In addition, PHH likely compounds oligodendrocyte disruption and PVWM dysmyelination through its compressive effects on oligodendrocyte progenitor cells in the LVP 457,458,461.

3.3.5 FOHP sustains a greater magnitude of injury than the rest of the LVP

Across the LVP, there are regional differences in the cytoarchitecture of the VZ and organization of neural stem cells within the subjacent SVZ 133,197,463. While radial glial cells are sequentially replaced by ependymal cells along the LVP during fetal development, the lateral wall of the frontal horn is the only region that retains these stem cells into infancy 463, which may contribute to adverse neurodevelopmental outcomes when those regions are damaged by PHH. Indeed, in our cohort, the FOHP demonstrated more prominent evidence of injury than the LVP. These findings are consistent with the more prominent patterns of FOHP than LVP injury seen on conventional

MRI scans of hydrocephalus patients, which is characterized by T2W periventricular

113 hyperintensities 449,464, petechial hemorrhages on susceptibility weighted imaging 193-195 and restricted diffusion on diffusion weighted imaging 465. There is also a disproportionate distribution of stress and strain on the stem cell rich FOHP in patients with hydrocephalus 466-468. For example, relative to the LVP, the FOHP region is subjected to higher stress and strain in the setting of hydrocephalus because of its concave geometry and acute anterolateral angles, which in turn expands the extracellular spaces in the FOHP and facilitates water extravasation and tissue edema

466-468. It is important to note however, that in this study, all PHH infants had undergone intervention for hydrocephalus, and their intracranial pressure was expected to have normalized at the time of dMRI acquisition. Nevertheless, we suspect the dMRI disruptions seen in the PHH group may reflect complex pathology from the initial IVH, injury from raised intracranial pressure sustained prior to their shunt/ETV treatment, and the subsequent associated secondary injury.

Several clinical and experimental studies have shown that PHH is strongly associated with significant neurological disabilities, which are mediated in part by impaired cerebral white matter289,469-475. Conventional dMRI has been widely employed to characterize alterations in cerebral development45,476 and to assess white matter disruptions in PHH, but the microstructural basis underlying the reported diffusion changes are not resolved307,309,311. DBSI, an innovative analysis technique uniquely capable of simultaneously modeling anisotropic and isotropic tensors307,309,311 was utilized in this study to quantify and differentiate white matter pathologies in prospectively collected dMRI data obtained at term equivalent age of preterm infants with PHH, those who sustained IVH but did not develop PHH and those who had no identifiable neurologic events (VPT), and full term healthy infants (FT). DBSI anisotropic diffusion tensors resolved fibers to assess axonal and myelin injury as well as white matter fiber density, while isotropic

114 tensor analyses reflected cellularity and edema. Three clinically relevant white matter tracts, the

CC, OPRA and CST were assessed. Correlations between ventricular volume and DBSI measures were assessed. It is important to emphasize that the dMRI images in this cohort were acquired at term equivalent age, when some but not all white matter tracts are expected to have been myelinated, therefore, the findings are interpreted in the context of whether the assessed white matter is myelinated (CST and OPRA) or unmyelinated (CC).

3.3.6 DBSI extends dMRI capabilities to delineate white matter cellular pathology

Several clinical and experimental studies have shown that PHH exhibits multifactorial white matter disruption that encompasses a mix of axonal and myelin disruption, as well as multi- compartmental microstructural processes such as inflammation, hypercellularity and edema. These nuances are beyond the capabilities of DTI to accurately parse out. Advanced post-processing computational methods such as DBSI address some of DTI limitations without need for dramatically changing data acquisition307-312. DBSI utilizes standard high-resolution dMRI data

(with 25 diffusion encodings or more) and extracts multiple diffusion tensors across microstructural compartments307-313. These diffusion tensors then enable the separation of crossing fibers, estimation of crossing angles, recovery of directional diffusivity of individual crossing fibers, assessment of axonal loss, distinction of myelinated, dysmyelinated, or unmyelinated axons, and modelling crossing fibers, extra-axonal inflammation, cellularity and edema307,308,311,313,314. DBSI has been validated experimentally in preclinical mouse models of white matter injury307,309,314,315,477 and used to investigate white matter pathology across clinical populations in adults308-311. It lends itself as a uniquely powerful, non-invasive neuroimaging tool

115 with high sensitivity for delineating white matter alterations in the developing brain, both normal and in PHH pathology.

Beyond the LVP, the white matter aberrations we identified in the CC, CST and OPRA with DBSI in PHH infants are all factors that have been associated with impaired neuromotor and cognitive development. This is likely because PHH disturbs processes that underlie early white matter development, engendering local and widespread effects through direct cellular mechanisms and disruption of subsequent cerebral development240,478-482. Collectively, LVP injury, characterized by the loss of progenitor cells and impaired neuroblast migration215,223,318,325,483, further impacted by pressure-related structural deformation, ischemia, oxidative stress, inflammation, and other phenomena86,185, has brain-wide effects, impacting both cortical and subcortical regions and the development of critical white matter tracts123-126.

3.3.7 dMRI variability of white matter structure and development

Across all fibers examined with DBSI, both conventional FA and fiber level FA were lower in the

PHH group, indicating white matter injury. However, when assessing white matter integrity in infants, especially in PHH, variabilities in the stages of white matter development, as well as their response to injury must be taken into consideration. This is particularly important for this study because in conventional dMRI, RD is typically regarded as a marker of myelin integrity and the higher the RD, the worse the myelin injury. Yet, in this study, there was significant variability in

116 RD indices, which were subject to different interpretations in the presence or absence of myelin in the fiber tracts of interest.

In large bundle tracts, myelination begins and ends at variable ages among different white matter bundles. In general, myelination in the CST and OPRA starts at 38- and 40-weeks’ gestation, respectively484, whereas myelination in the CC does not begin until after 2 months postnatally484,485. Kinney et. al.485 examined human postmortem tissues obtained less than 24 hours after expiration using immunohistochemistry and biochemical measurements of different lipids and observed that sphingomyelin, cerebroside, and sulfatide were not detectable until early infancy and myelin basic protein staining was not detectable until the 2nd postnatal month485 in the CC, similar to Sarnat et al.’s observations484. DTI RD in the unmyelinated CC of PHH infants in this study was significantly higher than non-PHH infants, but fiber level RD, while still highest in

PHH, was not statistically significantly different. This suggests that the higher RD detected with

DTI was related to increased fiber spacing rather than direct axonal injury, which is consistent with the roughly 70% reduction in fiber density found with DBSI and supported by histology. In contrast, the myelinated CST consistently demonstrated higher RD in the PHH than non-PHH group, suggesting myelin disruption/injury. For the myelinated OPRA, on the other hand, RD findings were not consistently different between groups. This may be attributed to intrinsic variabilities in myelination of segments within the OPRA that were not detected with dMRI. To complicate matters further, there is variability in the effects of PHH on different segments of the

PVWM, even along the uniform lateral ventricular perimeter. Using a segmentation algorithm that quantified dMRI measures in the 1 mm layer subjacent to the ventricular wall along the lateral ventricular perimeter, Isaacs et al. demonstrated that the frontal and occipital horns of the lateral

117 ventricle had the worst dMRI-measured PVWM injury231. Anatomically, the OPRAs are in very close proximity to the occipital horns, which are subjected to the highest mechanical pressures and transependymal flow injury in PHH, leading to significant water extravasation and tissue edema in those regions.466-468 (Figure 38)

Figure 38: Schematic rendering of postulated pathology underlying different PVWM bundles Diffusion MRI voxels from healthy myelinated and unmyelinated white matter are shown in the top row. Bottom row shows voxels from the corpus callosum (CC), corticospinal tracts (CST) and optic radiations (OPRA) of PHH infants, drawn to same scale as the healthy voxels, to demonstrate relative fiber density, inflammatory cells, and estimated inter-axonal spacing. The CC is not myelinated at this developmental stage76,77, depicted by the lack of myelin sheath. Relative to healthy white matter, there was significantly reduced fiber fraction and increased cellularity in the CC, CST and OPRA of PHH infants in comparison to healthy white matter. In the CST, increased radial diffusivity suggests likely myelin injury, which is represented as reddish sleeves around the axons.

118 3.3.8 Ventricular size correlates with magnitude LVP and large bundle PVWM injury

Although ventricular size appears to be a central risk factor for neurodevelopmental impairment in PHH, and early surgical decompression may improve outcomes, little is known regarding the link between ventricular size and white matter integrity. Collectively, the tensile and compressive forces of enlarged ventricles in PHH on the LVP and distant PVWM such as the CC, CST and

OPRA leads to impairment of arterial blood flow and venous stasis, which may cause axonal disruption and dysmyelination that sets off a cascade of ischemia-induced white matter injury

458,486,487. These findings beg the question whether ventricular size can be used as a surrogate marker for disease severity in PHH patients. In assessing the LVP/FOHP, ventricular size correlated negatively with FA and positively with MD and RD, suggesting that expanded ventricular size likely has a negative effect on dMRI-measured LVP/FOHP tissue integrity.

In the CC, CST and OPRA, DBSI parsed the relationships between ventricular size and white matter at the cellular level. In the CC, increases in ventricular size correlated with increases in hypercellularity and reduction fiber density, which may have been secondary to reactive cellular infiltration extracellularly and axonal loss, respectively. In PHH, the mechanical pressure from the large ventricles distorts PVWM fiber organization, decreases cerebral perfusion, and initiates an inflammatory cascade, which likely mediates axonal injury and loss185,210,242. These findings have critical clinical implications as several studies have shown that progressive ventricular distension in PHH is associated with higher rates of neurological disability. In a prospective study of 118 children, including 54 with hydrocephalus, Mangano et al. showed that changes in FA in the CC and CST correlated with conceptual, general adaptive, and motor skills284. Strahle et al. found that larger ventricular size was associated with impaired hippocampal development and worse

119 cognitive scores at age 2 years in infants with PHH488. The findings of this study and the existing literature demonstrate the deleterious impact of PHH and ventricular enlargement on white matter integrity, brain development and its persistent neurodevelopmental effects. We also provide strong support for utilizing DBSI to investigating the role of ventricular size – a readily modifiable factor

– in the neurodevelopmental disability of PHH.

3.3.9 Limitations

A primary limitation of this study is the small sample size in the PHH and, to a lesser extent, IVH cohorts which may have limited our ability to observe subtle differences in dMRI parameters.

Notably, all PHH patients were male, which may have introduced a sex bias in the PHH group, although there is a male preponderance in IVH reported in the literature 489,490. In addition, while we were able to establish similarities between PHH and PIH in terms of VZ, SVZ and PVWM disruption on postmortem neurocytology, we did not have any PIH subjects with dMRI data to correlate to cytology. To minimize the effect of age on brain development, all scans were acquired cross-sectionally at term-equivalent age, which precludes assessment of the relationships between dMRI parameters and brain development over time. Almost all PHH infants had shunts in situ at the time of dMRI acquisition, which may have caused through plane distortions and susceptibility artifacts due to signal and geometric distortions. Critical steps and modifications to the imaging pipeline such as aligning the relative frequency encoding directions to the shunt catheters, using wider receiver bandwidths and reducing echo times may be helpful for decreasing geometric distortions and minimize signal loss441,491. In the OPRA AD and RD measures did not consistently differ between the PHH and non-PHH groups. While it is possible that the inflammation, edema,

120 decreased fiber density and increased axonal spacing could have affected AD and RD measures, the OPRA has relatively small tracts that in close proximity to many structures of varying signal intensities. These signals may have inadvertently been included in OPRA segmentation, thereby affecting the AD and RD measures due to partial volume artifacts. Due to the relative novelty of the analysis techniques employed in this dissertation and the lack of concomitant histological specimens in the same infants who underwent dMRI scans, our ability to directly link dMRI and immunochemistry results limits the interpretability of some key findings (i.e., as neurobiological or technical), such as the diffusivity measures in the OPRA. Another limitation is that our study contained only term-equivalent scans with a cross-sectional design. Thus, we were unable to make direct comparisons of measures in subjects with hydrocephalus studied during both the days before and after neurosurgical intervention; information critical for evaluating what these approaches add to our understanding of the reversible effects of neonatal hydrocephalus and optimal timing of neurosurgical interventions. A longitudinal study including multiple scans at predetermined time- points for each subject would allow more detailed evaluation of relationships between dMRI parameters, frontal occipital horn ratio, and neurobehavioral correlates over time. In addition, the neuroimaging measures from the LVP and DBSI analyses have not been related to neurodevelopmental outcomes, limiting the interpretation of the clinical significance of our key findings. Successfully linking the brain and behavioral measures in this high-risk population would enhance the clinical relevance of our work and contribute to their validation as a biomarker. We are planning future studies to investigate prospective associations between dMRI measures (DTI and DBSI) in the LVP, FOHP and PVWM with subsequent neurodevelopmental outcomes to elucidate the neurological mechanisms underlying impairment in high-risk VPT infants, as well as

121 to potentially identify VPT infants in need of early referral to intervention services and longer- term follow-up care.

3.4 Materials and Methods

3.4.1 Participants

All patients were recruited after written informed consent was obtained from at least one parent of every infant. Institutional Review Board approval for this study was obtained from the Washington

University Human Research Protection office. The FT infants were identified from a larger cohort of infants undergoing neonatal neuroimaging scans as part of an existing contemporaneous fetal monitoring study conducted at an adjoining maternity hospital. All VPT, HG-IVH/IVH and PHH patients were recruited from a Level III Neonatal Intensive Care Unit. IVH diagnosis was made within the first month of life by cranial ultrasound and graded using the Papile classification system59. Well-established Hydrocephalus Clinical Research Network criteria were applied for the diagnosis of PHH and the timing of neurosurgical treatment421, which included a reservoir or sub- galeal shunt placement for temporary ventricular access, or permanent treatment with endoscopic third ventriculostomy (ETV) or ventriculoperitoneal (VP) shunt. To ensure the FT group had no confounding health issues at birth, additional exclusion criteria included acidosis (pH < 7.20) on cord blood gas or blood gas obtained during the first hour of life or evidence of in utero illicit substance exposure. Exclusion criteria for all four groups included inability to obtain informed consent from an infant’s parent, presence of chromosomal abnormalities and/or proven congenital infections (e.g., cytomegalovirus, toxoplasma, rubella).

122 3.4.2 Perinatal Clinical Variables

To account for differences in medical status between groups, perinatal clinical information obtained for each infant was used to calculate their clinical risk index score 470. The presence or absence of each of 10 factors were scored as 1 or 0 respectively. There were 9 patient-related factors, which included: 1) small for gestational age at birth or intrauterine growth restriction; 2) received oxygen therapy at 36 weeks; 3) received postnatal steroids; 4) developed necrotizing enterocolitis; 5) had a patent ductus arteriosus requiring indomethacin or ibuprofen therapy; 6) had retinopathy of prematurity; 7) developed culture-positive neonatal sepsis; 8) weight-for- height/length ratio at the time of term equivalent MRI acquisition >3 standard deviations below the measured ratio at birth; and 9) duration receiving total parenteral nutrition above the 75th percentile. A point was also scored if the mother did not receive antenatal steroids. All scores were collated to obtain a composite risk index score on a Likert scale of 10 - the higher the score, the poorer the presumed neonatal health 470.

3.4.3 Imaging acquisition and preprocessing

Human infant diffusion MRI (dMRI) was acquired without sedation and during natural sleep or when the subject was resting quietly following institutional neonatal guidelines 492. Full-term infants were scanned within the first four days of birth. To match the expected developmental stage of the full-term infants, preterm infants were scanned at or near-term equivalent age (35-43 weeks postmenstrual age). All scans were acquired using a 3-T Siemens TIM Trio System (Erlangen,

Germany), using an infant-specific, quadrature head coil (Advanced Imaging Research, Cleveland,

123 OH, USA). Anatomical T1W with repetition time (TR) 1550 ms, inversion time (TI) 1100 ms, echo time (TE) 3.05 ms, flip angle 15°, 1 × 1 × 1.25 mm3 voxels; and T2W sequences, sagittal, magnetization-prepared rapid gradient echo sequence [TR 8210 ms, TE 161 ms, 1 × 1 × 1 mm3] voxels were obtained. Then, a diffusion-weighted image (DWI) [TR 13300 ms, TE 112 ms,

1266 Hz/Px bandwidth, 128 mm field of view, 1.2 × 1.2 × 1.2 mm3 voxels], with 27-48 b-directions and amplitudes ranging 0-1200 s/mm2 was obtained.

3.4.4 Diffusion tensor imaging

Diffusion weighted image (DWI) data was processed using FSL tools493,494 and corrected for eddy current distortions using Eddy495 susceptibility induced distortions using ApplyTopup495 and motion corrected using FLIRT496,497. The diffusion signal attenuation curve was modeled as a mono-exponential function plus a constant, and diffusion parameters were estimated using

Bayesian Probability theory498. FA, AD, RD and MD and Principal Eigen Vector maps were generated with DSI studio(http://dsi-studio.labsolver.org).

3.4.5 Diffusion basis spectrum imaging

DWI data were processed using FSL tools 493,494 and corrected for eddy current distortions using Eddy 495, susceptibility induced distortions using ApplyTopup 495, and motion corrected using FLIRT 496,497. The CC, bilateral posterior limbs of the internal capsules, designated as CST, and bilateral OPRA were selected a priori as our regions of interest (ROIs) as they have been

124 shown to demonstrate improvement in dMRI indices following hydrocephalus treatment and relate to PHH-related neuropsychological outcomes 272,284,452,499. For each subject, ROIs were manually segmented on native diffusion parametric maps by three experienced reviewers (Fig. 1), with high mean inter-rater reliabilities (range .70-.99). Each ROI was segmented on three contiguous axial slices to minimize through-slice partial volume averaging using Analyze 10.0 software

(AnalyzeDirect, Inc., Overland Park, KS, USA).

The preprocessed 27-48 directional DWI images were analyzed via the DBSI pipeline 307,310 in MatLab® (MathWorks, Natick, MA, USA). As demonstrated in Equation 1, the DBSI model is a sum of the multiple anisotropic diffusion tensors (first term) and a spectrum of isotropic diffusion tensors (second term):

푁 퐴푛푖푠표 → → → 2 푏 푆푘 −|푏푘|휆⊥푖 −|푏푘|(휆∥푖−휆⊥푖) 푐표푠 휙푖푘 −|푏푘|퐷 = ∑ 푓푖 푒 푒 + ∫ 푓(퐷)푒 푑퐷 (푘 = 1,2,3, … ). [1] 푆0 푖=1 푎

Here bk is the kth diffusion gradient; Sk/S0 is the acquired diffusion weighted signal at direction of bk normalized to non-diffusion-weighted signal; NAniso is the number of anisotropic tensors to be determined; ik is the angle between the diffusion gradient bk and the principal direction of the ith

→ anisotropic tensor; |푏푘| is b-value of the kth diffusion gradient; ||i and ⊥i are the axial and radial diffusivity of the ith anisotropic tensor under the assumption of cylindrical symmetry; fi is the signal-intensity-fraction (non-diffusion-weighted) of the ith anisotropic tensor; a, b are low and

125 high diffusivity limits of the isotropic diffusion spectrum; f(D) is the spectrum of signal-intensity- fraction (non-diffusion-weighted) at all possible isotropic diffusivity D.

A priori division of isotropic components was established using an isotropic apparent diffusion coefficient (ADC) cutoff of ≤0.3 µm2/ms representing restricted diffusion; ADC values between

0.3-2.5 µm2/ms representing hindered diffusion; and ADC ≥2.5 µm2/ms representing free diffusion. Following DBSI analysis, ROI-specific metrics were obtained via voxel-based computations. DBSI metrics included: fiber specific FA (fiber FA, FFA), AD (fiber AD, FAD), and RD (fiber RD, FRD); axonal density (fiber fraction, FF); and non-fiber related restricted fraction (RF), and non-restricted fraction (NRF).

3.4.6 Ventricular size Measurements

All ventricular size measurements were taken by two independent expert raters on axial T2W image using the frontal-occipital horn ratio approach20, which sums the distance between the widest lateral walls of the frontal and occipital horns, divided by twice the widest biparietal diameter at the level of the foramen of Monro. Reproducibility of FRONTAL-OCCIPITAL HORN RATIO measures was established across groups using measurements made by two independent raters, with inter-rater comparisons demonstrating a high degree of agreement (intraclass correlation coefficient=.91; p<.001).

126 3.4.7 Human Postmortem Brains Immunohistochemistry

Autopsy specimens from preterm neonates were obtained from the St. Louis Children’s Hospital

(SLCH) at the Washington University School of Medicine, St. Louis, Missouri, USA. The relative age of these patients was estimated from the time of expected fertilization and recorded as

Estimated Gestational Age (EGA). Brain specimens were dissected from regions of the frontal cortex containing the corpus callosum. Formalin-fixed tissues were paraffin-embedded, and 15 µm sections were immunostained with H&E to assess cellular structure, edema and cellularity, synaptophysin to assess axonal integrity and GFAP (1:300, catalog #7260, Abcam) to assess astrocytes. To examine associations between PVWM and the subjacent ventricular (VZ) and subventricular (SVZ) zones, the VZ/SVZ regions of adjacent slides were stained with H&E to assess cytoarchitecture, double-label immunostained with 4’6-diamidino-2-phenylindole (DAPI)

(#D1306 Thermo Fisher Scientific) and GFAP to assess immature ependymal cells and reactive astrocytes, and with DAPI and Iba1 (1:100, #PA5-18039, Invitrogen) as a surrogate for microglia- and macrophage-associated products. All slides were examined with fluorescence microscopy using Nanozoomer-XR (Hamamatsu Photonics). Qualitative comparisons were confirmed by with a neuropathologist. Percent VZ disruption was quantified as the cumulative length of the disrupted ependymal regions as a factor of the entire length of VZ available on the same slide.

3.4.8 Statistical analysis

Statistical analyses were performed with R version 3.5.0 (The R Foundation, Vienna, Austria)500 and SAS version 9.4 (The SAS institute, Cary, NC). Normality of data was assessed with the

127 Shapiro-Wilk test, which determined the use of non-parametric methods for all data analyses.

Descriptive statistics of clinical factors including gestational age at birth, postmenstrual age at

MRI scan, birth weight and clinical risk index scores 470, as well as measured factors, including frontal-occipital horn ratio and dMRI measures in the LVP, FOHP, CC, CST and OPRA were obtained. Kruskal-Wallis one-way ANOVA was used to determine between groups differences.

Wilcoxon signed rank test was used to perform family-wise comparisons, and all resulting p-values were corrected with Benjamini-Hochberg adjustment. Correlations between ventricular size and dMRI measures were examined using Spearman’s rank-order correlations because the patient groups correspond with disease severity (ordinal ranking). To examine group differences in dMRI measures between the LVP and FOHP, a linear mixed effect model with a random patient intercept was fit to each measure. Specifications of the model included a main effect of region (LVP and

FOHP), group (FT, VPT, HG-IVH and PHH) and region-by-group interactions with Bonferroni correction. Given the reported effect of age on dMRI measures 238, postmenstrual age at scan was set as a covariate in the linear model. Where Mauchly’s test of sphericity was violated,

Greenhouse-Geisser correction was used to adjust degrees of freedom for the averaged tests of significance. Similarly, FT, VPT, IVH, PHH groups were compared for all DBSI measures with

Kruskal-Wallis test, followed by Wilcoxon Rank-Sum tests when Kruskal-Wallis was statistically significant. frontal-occipital horn ratio was compared to DBSI scalars for non-linear association with Spearman’s rank correlation test. Bonferroni-corrected p value <.050 was considered statistically significant. Statistically significant results were further subjected to an ANCOVA where number of voxels, number of diffusion directions, and laterality were set as covariates to limit any potential confounding effects of those factors on the results.

128 4 Chapter 4: N-cadherin cleavage mediates VZ disruption in PHH and PIH

Adapted from:

1. Castaneyra-Ruiz L, Morales DM, McAllister JP, Brody SL, Isaacs AM, Strahle JM, Dahiya

SM, and Limbrick DD. Blood Exposure Causes Ventricular Zone Disruption and Glial

Activation In Vitro. J Neuropathol Exp Neurol 2018. 77: 803-813

2. Castaneyra-Ruiz, L., Morales, D. M., McAllister, J. P., Brody, S. L., Isaacs, A. M. and

Limbrick DD. Preterm Intraventricular Hemorrhage In Vitro: Modeling the Cytopathology

of the Ventricular Zone. Fluids and Barriers of CNS 2020. FBCN-D-20-00029R2.

(Accepted)

4.1 Introduction

In the previous chapters, we identified key biological processes central to the pathophysiology of

PIH and PHH in humans. In CSF, we observed transcriptomic and proteomic evidence of neuroinflammation and ependymal barrier damage in the context of VZ and SVZ disruption. We confirmed those findings in human brain parenchyma in vivo and ex vivo utilizing dMRI and immunohistochemistry, respectively. We also related VZ/SVZ/ependymal disruptions and neuroinflammation to PVWM injury. While our findings to date provide insights on relating neuroinflammation and brain injury, we have not yet probed the pathogenesis of PIH and PHH.

Strong evidence demonstrates that proteins of adherens junctions help to form and preserve the ependymal barrier. Of the four classical cadherins, neural cadherin (N-cadherin) is the most highly expressed in the ependymal layer. In addition to mediating cell-cell adhesion, N-cadherins

129 stimulate CDC42, which mediates apical-basal polarity of neuroepithelial cells and ciliogenesis during VZ/SVZ differentiation and ependymal barrier formation. Like other cadherins, N-cadherin is susceptible to disruption by metalloproteases. Specifically, our collaborators228,501,502 and several others503-507 have shown that the metalloprotease ADAM10 mediates inflammation and cleaves cadherins to regulate several epithelial cell pathologies including the lung and liver injury in S. aureus sepsis, and in cancer. Therefore, we hypothesized that ADAM10-mediated cleavage of N- cadherin is associated with VZ disruption, and pharmacological inhibition of ADAM10 will ameliorate this disruption and preserve ependymal integrity. We developed a novel in vitro ependymal cell culture model of IVH and ventriculitis to characterize the effect of blood and pathogens on N-cadherin during VZ differentiation while modulating ADAM10 activity.

4.2 Results

4.2.1 Characterization of in vitro VZ differentiation to mature ependymal cells

I describe details of our cell culture model in the Methods section. Briefly, in P0/P4 mice, we harvested VZ cells from the perimeter of the lateral ventricles and modulated their proliferation and differentiation in media. First, characterized normal ependymogenesis in our in vitro model to establish a baseline for future comparisons and to determine the precise timing for inoculating the cells with blood or pathogens. To evaluate differentiation, we immunolabeled cells against the cilia marker βIV-tubulin, which allowed us to discriminate between multiciliated, monociliated, and nonciliated VZ cells. To determine the stage of ependymal cells maturation, we used the glial marker GFAP. Ependymal cells exhibit GFAP-positive attributes in early stages and become

130 GFAP-negative with maturation.197,508-510 This approach permitted identification of six different cell types: 1) cells not expressing GFAP or βIV-tubulin, 2) cells expressing GFAP but not βIV- tubulin, 3) monociliated βIV-tubulin cells not expressing GFAP, 4) monociliated βIV-tubulin cells expressing GFAP, 5) multiciliated βIV-tubulin cells expressing GFAP, and 6) multiciliated βIV- tubulin cells not expressing GFAP. At this stage, we measured successful VZ differentiation by the number and quality of multiciliated ependymal cells (mature ependyma) (Figure 41).

Multiciliated ependymal cells developed from monociliated cells in a time-dependent fashion, comprising 3.73 ± 4.29%, 23.01 ± 7.56%, and 52.29 ± 11.07% (p < 0.001) of all viable cells at 3,

5, and 7 days after addition of differentiation media. Overall, the total number of cells changed very little at the same time points (day 3: 42.80 ± 7.64, day 5: 42.78 ± 8.61, day 7: 44.31 ± 6.87) after the initiation of differentiation. This observation indicates that cells were changing from progenitor-like VZ cells to those that are typically found in the well-differentiated ependyma.

Double-labeling was performed against βIV-tubulin and GFAP took place at days 5 and 7, when a significant number of VZ cells had differentiated to ependymal cells. We found no significant increase in the percentage of GFAP-negative cells from among all multiciliated cells (day 5; 29.45

± 18.84, day 7; 35.37 ± 33.77). At both time points, over 60% of the ependymal cells remained in a premature stage (still expressing GFAP). In addition, the total number of GFAP-positive cells did not change over time (day 5: 72.78 ± 9.42, day 7: 73.61 ± 12.47) (Figure 39).

131

Figure 39: Differentiation of VZ cells in vitro. Differentiation of VZ cells in vitro. Representative images of VZ cells at different stages of differentiation and maturation. (A) VZ cells undergo differentiation into multiciliated ependymal cells from day 3 to day 7, as identified by cilia marker βIV tubulin. (B) Quantification of percentage of multiciliated ependymal cells during differentiation. Data represent means of the percentage of ciliated cells from two experiments with at least n=3 wells at days 3, 5 and 7, respectively. (C) Expression of GFAP (green) in cultures of multiciliated ependymal cells identified by βIV tubulin (red) at days 5 and 7. (D) Detail of double-labeling of GFAP and βIV tubulin showing a mixed population of cells, including those not expressing GFAP or βIV tubulin (1), expressing GFAP only (2), GFAP-negative monociliated cells (3), GFAP positive

132 monociliated cells (4), GFAP-positive multiciliated cells (5) and GFAP-negative multiciliated cells (6). Scale bars = 50 μm in A and C; 10 μm in D.

4.2.2 N-cadherin mediates cell-cell adhesion mediates in VZ development

The plasma membranes of all VZ cells contained N-cadherin-based adherens junctions. This phenomenon probably contributed to the characteristic polygonal shape that occurred regardless of whether the ependymal cells were less (day 5) or more (day 7) differentiated. Interestingly, the ratio of N-cadherin to total number of cells (N-cadherin expression) initially declined from day 5 to day 6 (2.93 ± 2.41 vs. 2.11 ± 2.03, respectively; p>0.05). However, levels stabilized by day 7

(2.22 ± 1.87). This observation suggests a possible heightened requirement for this cell junction protein during early stages of normal differentiation. Double-labeling with GFAP and N-cadherin showed that the polygonal shape existed independent of GFAP expression, indicating that GFAP may not affect N-cadherin expression. The glial projections of the GFAP-positive cells did not express N-cadherin. (Figure 40)

Figure 40: Cell-cell junction and astrocytic expression of differentiating of VZ cells in vitro. Representative images of VZ cells at different stages of differentiation and maturation. (A) N- cadherin-based cell-cell junctions, with characteristic membrane marker localization and polygonal morphology at day 5 and 7. (B) Expression of N-cadherin (red) and GFAP (green)

133 showing that the glial projections do not colocalize with cell-cell junctions (arrowheads) (B). Scale bars = 50 μm.

4.2.3 Effective dose and time of blood administration required to elicit VZ disruption

VZ cultures were used to assess whether blood exposure elicits neuroepithelial and ependymal cell loss and to determine the effective timing and dosage of blood needed to elicit the pathology. At day 5 of differentiation, we exposed VZ cultures to syngeneic blood, and evaluated treatment effects over time. Pilot experiments using 25 (2.5%), 30 (3%) and 40 µL (4%) determined the amount of blood needed for this model. After three hours of 40-µL treatment, the number of total cells and N-cadherin expression obviously decreased, preventing observations on the cytopathology of IVH beyond this time. Treatment with 25 or 30 µL of blood permitted long-term experiments because the model offered a sufficient and reliable number of VZ cells even after 48 h, similar to the human condition. (Figure 41)

134

Figure 41: Time- and dose-dependent response of blood exposure to VZ cells in vitro. (A) Three hours of 40 µL of syngeneic blood treatment dramatically decreased the number of total cells and N-cadherin expression, precluding observations of IVH cytopathology beyond this time. (B) Treatment with 25 or 30 µL of blood permitted long-term experiments because the model offered a sufficient and reliable number of VZ cells even after 48 h, similar to the human condition.

135 4.2.4 Blood disrupts VZ cytoarchitecture and differentiation in a time-dependent manner

Based on findings from our dose- and time-response experiments, we used 30 µL of blood treatment for the remainder of the experiments. To characterize VZ disruption, we evaluated the total number of cells, the percentage of ependymal cells and programmed cell death over time.

After 3 hours of phosphate buffered saline (PBS) control or blood treatment, we found no differences in the total number of VZ DAPI-positive cells per 0.0225 mm2 (38.62± 11.47 vs.

42.78± 8.614). However, we observed a significant reduction of total VZ cells after 24 hours

(41.45± 5.62 vs. 32.08± 10.68 p<0.05) and after 48 hours (44.31± 6.87 vs. 26.94± 7.50 p<0.001) of blood exposure. A similar effect occurred in multiciliated cells, as shown by changes in βIV- tubulin signal. We observed was no change in multiciliated cells after three hours of treatment, but we noted a decrease after 24 hours (control 31.07 ± 7.78 vs. IVH 11.84 ± 7.86 p<0.001) and after

48 hours (control: 52.29 ± 11.07 vs. IVH: 14.65 ± 8.49 p<0.001). We then calculated the percentage of total cell loss (loss of DAPI-positive cells) after blood exposure and compared this value with the percentage of ependymal cell diminution (diminution of multiciliated βIV-tubulin- positive cells) at three, 24 and 48 hours of blood treatment. After three hours, we observed essentially no discrepancy between total cell loss and ependymal cell diminution (-2.92 ± 28.72 vs

-8.60 ± 24.84, respectively). However, after 24 hours, the ependymal cell diminution was twice as high as total cell loss (-59.68% ± 25.10 vs. -27.12% ± 21.94, respectively; p<0.05) and even greater after 48 hours (-85.16% ± 9.15 vs. -36.90% ± 23.98, respectively; p<0.001). These results demonstrated that the ependymal cell loss exceeded total cell loss, suggesting that cell loss does not solely control the decrease of the multiciliated ependymal cells after blood treatment. Indeed, other factors, such as alterations in neuroepithelial cell/ependymal cell differentiation may contribute to the diminution in ependymal cells following blood exposure (Figure 42).

136 Interestingly, immunohistochemistry against active caspase 3, a marker of apoptosis, showed no change in caspase 3-positive cells at the same time points (control vs. blood at 3 h: 2.93 ± 0.70 vs.

3.39 ± 2.63; at 24 h: 3.72 ± 0.42 vs. 2.50 ± 2.80; at 48 h: 3.19 ± 1.00 vs. 1.823 ± 1.16). These results suggest that programmed cell death did not affect cell loss following blood exposure

(Figures 41 and Figure 42).

Figure 42: Time-dependent effect of blood in VZ cell cultures. After 5 days of differentiation, samples were analyzed at 3, 24, and 48 h of control (Cont) or blood exposure. (A & C) Controls exhibit normal differentiation from NSC to ependymal cells with significant increases in the percentage of βIV-tubulin multiciliated cells (C). (B) Blood exposure significantly reduces both the total number of cells (DAPI-positive cells/0.0225 mm2) and (C) the percentage of multiciliated cells at 24 h and 48 h. * p<0.05; **p<0.01; *** p<0.001. (D) Double labeling with active caspase 3 (green) and βIV-tubulin (red) showed no difference in the number

137 of apoptotic cells. (E) Blood exposure-dependent proportional change of total cells vs. ependymal cells (EC). EC decrease was twice as high after 24 h and even greater after 48 h than the cell loss, showing a significant discrepancy between total cell loss and the lack of EC. * p<0.05; *** p<0.001. Data represent means of the percentage of ciliated cells from two experiments with at least n=6 wells at 3, 24, and 48 h, respectively. Scale bars = 50 μm in all panels.

4.2.5 VZ disruption occurs in the setting of cell-cell adherens junction damage

To gain insights into the contributing factors in VZ disruption following blood exposure, the integrity of the cell-cell junctions was examined with N-cadherin expression as a surrogate measure. N-cadherin expression was quantified as a ratio of the number of N-cadherin positive cells to total number of cells. After three hours of blood exposure, we found no changes in N- cadherin expression (control: 2.29 ± 2.40 vs. blood: 2.76 ± 2.37). After 24 hours, the N-cadherin signal profile became fragmented, with loss of the typical lattice-like cell membrane expression and appearance of abnormal subcellular cytosolic depositions. However, the total amount of N- cadherin was not significantly different when immunodensity ratios were quantified (control: 2.11

± 2.03 vs. blood: 2.06 ± 2.10). After 48 hours, the abnormal deposition and fragmentation persisted along with significant loss of N-cadherin signal (control: 2.21 ± 1.87, blood: 1.12 ± 1.07; p<0.05;

Figure 42 A to C). Western blots confirmed significant reduction of N-cadherin signal by measuring the N-cadherin/GAPDH ratio (median of control: 0.47 ± 0.17 vs. blood: 0.22 ± 0.03; p<0.05) after 48 hours of blood treatment. (Figure 43)

138

Figure 43: Time-dependent effect of blood in N-cadherin expression in the VZ. (A) Representative VZ cultures after 48 hours of control (left) or blood exposure (right). Compared to the prominent expression of N-cadherin (red) at the cell membrane in controls (arrows), N- cadherin expression was weaker and cytoplasmically dislocated after blood exposure (asterisk). (B) High magnification of VZ N-cadherin labeling in which 3 hours after exposure to blood, no changes were found in the expression of N-cadherin (red). However, after 24 h of treatment, the expression of N-cadherin fragmented, with loss of the lattice-like configuration. After 48 hours, there was fragmentation, dislocation from the cell membrane, and loss of N-cadherin expression. (C) Quantification and comparison of N-cadherin expression between control and blood exposure, normalized by the total number of cells. There was a significant decrease in N-cadherin expression after 48 h of blood treatment (p<0.05). Data represent means of the immunodensity/cells from two experiments with at least n=6 wells at 3, 24, and 48 h, respectively (C) and protein levels (N- cadherin/GAPDH) by western blot (* p<0.05). (D) Data were analyzed from two experiments with n=4 per condition. Scale bars = 50 μm in all A panels and 10 μm in all B panels.

139 4.2.6 Reactive astrocytosis (neuroinflammation) occur with VZ disruption

We also used GFAP immunolabeling to investigate astroglial differentiation following blood exposure. The percentage of non-ciliated GFAP-positive cells did not change after 3 hours

(control: 49.44 ± 12.96 vs. blood: 45.94 ± 16.33) or 24 hours (control: 47.16 ± 17.33 vs. blood:

59.94 ± 13.53) but increased significantly after 48 hours of blood treatment (control: 47.10 ± 13.21 vs. blood: 79.63 ± 11.81; p<0.05). GFAP-expressing cells also demonstrated morphological changes from 3 to 48 hours of blood exposure. In particular, GFAP-positive cells exhibited branch- like projections and a larger GFAP-positive cytoskeleton; at 48 hours, the GFAP-positive processes appeared elongated, consistent with reactive astrocyte morphology. Treating cells with blood for 48 hours increased the GFAP/GAPDH ratio as measured by western blot (median of control: 1.99 ± 1.01 vs. blood: 4.75 ± 4.1; p<0.05), further supporting the findings above (Figure

44).

140

Figure 44: Glial activation in the VZ. (A) Representative VZ culture immunostained for GFAP (arrowheads) after 48 h of blood treatment. (B) High-resolution images of cells double-labeled with βIV-tubulin and GFAP antibodies after 3, 24, and 48 h of blood treatment vs. control. After 48 h treatment, most GFAP- positive cells exhibited numerous long, thin projections (arrowhead) typical of reactive astrocytes. (C) Quantification of non-ciliated GFAP-positive cells. After 48 h treatment, the mean number of cells expressing GFAP increased significantly (***, p<0.001). Data represent means from two experiments with at least n=6 wells at 3, 24, and 48 h, respectively (D) GFAP protein levels quantified by western blot and normalized to GAPDH showing increased levels of GFAP after 48 h of blood treatment (*, p<0.05; the filled circle is an outlier). Data were analyzed from three experiments with n=4 per condition. Scale bars = 50 μm in all A panels and 10 μm in all B panels.

141 4.2.7 ADAM10 inhibition abrogates N-cadherin cleavage and VZ disruption

Under physiological conditions, ADAM10 cleaves N-cadherin, yielding a soluble isoform of N- cadherin.511 In multiple organ systems, regulation of ADAM10 activity512-514 causes pathological shedding of cadherins, resulting in loss of adherens junctions, damage to epithelium, or disruption of endothelium.228,505,515. Importantly, the ADAM10 inhibitor GI254023X protects against epithelial disruption.516 To examine the role of ADAM10 modulation in IVH, we treated VZ cell cultures with either blood alone or blood+GI254023X (20 μM, Catalog #260264-93-5, Sigma-

Aldrich, St. Louis, MO). Treatment of blood elicited similar findings as before, including significantly decreased N-cadherin expression compared to DMSO vehicle-treated cells. However, the addition of GI254023X partially preserved N-cadherin expression. Accounting for number of cells, N-cadherin expression did not change between inhibitor-treated and vehicle-treated cells, both of which showed higher expression than the blood-treated cells (Figure 45)

Figure 45: Effect of ADAM10 modulation on blood treated VZ cells.

142 A significant decrease in N-cadherin expression is observed in blood-treated VZ cells after 48 hours when compared to DMSO (vehicle)-treated cells. Addition of the ADAM10 inhibitor GI254023X preserved N-cadherin expression similarly to DMSO-treated cells.

4.2.8 Bacterial toxin causes VZ disruption that can be modulated by ADAM10

To determine if N-cadherin-mediated VZ disruption and astrogliosis contribute to PIH pathogenesis and to examine the effects of ADAM10 modulation on VZ cytopathology in ventriculitis, we treated VZ cell cultures with ⍺-hemolysin. ⍺-hemolysin, the most cytotoxic pathogen secreted by Staphylococcous aureus, commonly causes hydrocephalus-related infections. In addition, mounting evidence suggests that ADAM10 carries a selective epitope activated by ⍺-hemolysin during systemic S. aureus infection. This epitope activation leads to cadherin cleavage and epithelial barrier disruption.512-514,516 Therefore, following differentiation, we separated equivalent numbers of wells containing VZ cell-mounted coverslips into three groups. Treatments included DMSO, 15 µg/mL of α-hemolysin, or 15 µg/mL of α- hemolysin+GI254023X. We cultured the cells for a total of 3.5 hours following suspension.

Treatment with α-hemolysin severely reduced N-cadherin expression. In addition, GI254023X treatment preserved N-cadherin structure, similar to DMSO. (Figure 46)

143

Figure 46: Effect of ADAM10 modulation on toxin treated VZ cells. Compared to the VZ cells of controls (A & D) treated with DMSO, those treated with (B & E) α- hemolysin was associated with severely reduced N-cadherin expression and higher GFAP signal. However, addition of (C & F) GI254023X to α-hemolysin-treated cells was associated with preserved N-cadherin structure. (Right panel) The box and whisker plot demonstrate the relative intensity of N-cadherin as a ratio of the total number of cells per well was lowest with the addition of α-hemolysin when compared to control DMSO-treated cells. Addition of GI254023X in the setting of α-hemolysin preserved the relative N-cadherin intensity, to levels similar to that of controls. *** p<0.001.

4.3 Discussion

4.3.1 Summary of findings

In summary, we showed that exposing VZ cells to blood or a bacterial toxin triggers a time- dependent chain of events starting with the dislocation of N-cadherin. This dislocation impairs differentiation and induces loss of ependymal cells, culminating in reactive astrocytosis (Figure

49). The findings provide a mechanistic basis for PIH and PHH pathogenesis in support of our

144 findings from previous chapters. Furthermore, results from existing literature on human and animal models of hydrocephalus support our findings.171,172,175,176,178,183,197,215,216,223,517,518

Figure 47: Model summarizing the effects of the blood on the VZ. During maturation, neural stem cells (NSC) with adherens junctions transform into multiciliated ependymal cells (EC). Blood exposure produces a time-dependent chain of events starting with the dislocation of cell junctions (N-cadherin) from the cell membrane into the cytoplasm and loss of multiciliated cells. Subsequently, glial activation occurs, as evidenced by astrocytes with numerous processes. In vivo observations suggest that glial activation occurs at the site of VZ disruption.

4.3.2 The in vitro model mimics normal human VZ differentiation

Our murine in vitro model recapitulated the normal VZ cell differentiation that occurs across species. In rats, differentiation of the dorsolateral wall of the lateral ventricle takes place postnatally. At postnatal day one, the VZ comprises approximately 70% neural stem cells, 23% non-ciliated cells and 7% multiciliated ependymal cells. Neural stem cells then differentiate into multiciliated ependymal cells. However, the percentage of non-ciliated cells remains constant.518

A similar pattern of postnatal ependymal maturation occurs in mice.519 In humans, ependymal cell

145 maturation occurs earlier in intrauterine life, and multiciliated ependymal cells appear in the dorsolateral wall from 23-40 gestational weeks.197,518,520 To model the clinical timeline of VZ development as closely as possible, we performed our experiments during the stage of differentiation between in vitro days five and seven, when the cultured cells progressed from approximately 20% to 55% multiciliated ependymal cells. During this time, the VZ also exhibited preterm human-like features such as the presence of undifferentiated monociliated cells with or without expression of GFAP, immature multiciliated ependymal cells expressing GFAP (~65% of all ependymal cells), and mature ependymal cells not expressing GFAP (~35%). The presence of monocilia characterizes neural stem cells123, and GFAP marks astrocytes and immature ependymal cells. Thus, monociliated cells failing to express GFAP may belong to another lineage of stem cells not committed to microglia.

4.3.3 N-cadherin cleavage underlies the cell-cell junction pathology in VZ disruption

Cell junction pathobiology may mediate VZ disruption in clinical and experimental hydrocephalus.171,175,176,216-220,521 In areas of VZ disruption, N-cadherin localizes to the cytoplasm rather than binding to the cell membrane.518 Furthermore, many genetic alterations directly or indirectly affecting cell junction stability associate with VZ disruption and hydrocephalus.174,183,216,522,523 The blood component lysophosphatidic acid (LPA) also alters cell junctions, disrupts VZ cytoarchitecture, and produces hydrocephalus.215,223 These authors noted

VZ disruptions in both the lateral ventricles and the cerebral aqueduct and therefore suggested that aqueductal stenosis produced ventriculomegaly in their rodent model. Our in vitro IVH model corroborates these well-known cell junction alterations; we observed dislocation of N-cadherin

146 after 24 hours of blood treatment and significantly decreased expression of N-cadherin after 48 hours of treatment. Adherens junctions mediate VZ cell attachment. Therefore, impairment of these junctions results in detachment and loss of those cells. Because cell junctions appear fundamental to the genesis of cytopathology in many forms of hydrocephalus, modulators of those molecules such as metalloproteases should play a role in the etiology of PIH and PHH.

4.3.4 ADAM10 Inhibition abrogates N-cadherin cleavage and VZ disruption

ADAM10 cleaves more than 40 transmembrane proteins in the extracellular region,524 most of which are cell adhesion proteins including L1-CAM,525 E-cadherin, and N-cadherin (reviewed in

526). ADAM10 activation disrupts lung epithelial cells in mice,228 presenting a pattern of cadherin alteration similar to what we observed in our in vitro model. Interestingly, our group previously reported that many of the proteins cleaved by ADAM10 such as amyloid precursor protein and

L1-CAM show increased levels in the CSF of both congenital hydrocephalus and PHH.246,249,527,528

In our model, exposure of VZ cells to the ADAM10 agonist α-hemolysin enhanced the activity of

ADAM10 and triggered VZ disruption by cleaving N-cadherin. Perhaps more importantly, we prevented VZ disruption after both α-hemolysin and blood induction by using the ADAM10 inhibitor GI254023X. These observations support the potential for this inhibitor, or perhaps other metalloprotease inhibitors, to have important direct therapeutic implications. However, future studies need to rigorously examine pharmacological inhibition of ADAM10 as a potential therapeutic strategy for preventing the development of PIH and PHH.

147 4.3.5 Reactive astrocytosis (neuroinflammation) occurs at the site of VZ disruption

The in vitro model also resulted in fewer total VZ cells after 24 hours of treatment with blood or bacterial toxin, possibly due to loss of adherens junctions. Interestingly, we observed fewer multiciliated cells at 24 and 48 hours, suggesting that those cells may be more affected by this detachment. However, the percentage of VZ cells lost was not proportional to the percentage of ependymal cell diminution. We found that ependymal cells decrease twice as much as observed cell loss, indicating that factors other than cell loss contribute to ependymal cell absence. Also, impaired ependymal cell differentiation appears to take place, probably due to the aforementioned blood- or pathogen-dependent N-cadherin effects. This observation is consistent with the findings reported by Muniz-Talavera and Schmidt (2017)529 where mutant mice lacking the junctional cadherin complex regulator (Jhy) showed protracted ependymal cell differentiation and developed hydrocephalus. In both human and animal cases, VZ disruption relates to glial activation by possibly forming a new layer to specifically repair the disrupted area.133,171,517 Our experimental model corroborates this theory by displaying a significant increase in the number of GFAP-positive cells at 48 hours and by exhibiting multiple projections reminiscent of a reactive astroglial response. This glial response occurred 24 hours after disruption of N-cadherin, and we detected loss of multiciliated cells, indicating that this reaction may respond to cell junction impairment and VZ cell loss. These alterations seem to promote neural stem cell differentiation into reactive astrocytes, even though these stem cells originally committed to an ependymal cell fate. This altered program of VZ differentiation also explains the discrepancy between cell loss and ependymal cell differentiation. Glial activation, especially the addition of branched processes, influences the overall decrease of N-cadherin levels found after 48 hours of blood treatment because glial projections do not express this junction protein.

148 4.4 Materials and Methods

4.4.1 Animals and surgery

All animal procedures were approved by the Institutional Animal Care and Use Committee

(IACUC) of the Washington University School of Medicine. C57BL/6 mice at P0 to P4 were used.

Each pup was exposed to wet ice for approximately 2 minutes until circulatory arrest, then sacrificed by removing the head and gently stripping the scalp. The brain was removed through a combination of a lateral incision from the foramen magnum toward the external auditory canal (~3 mm) and a superficial sagittal incision from the foramen magnum all the way through the midline; incisions were made using Metzenbaum scissors. The frontal and parietal bones were then removed with curved surgical forceps. The brain was then deposited in a Petri dish with ice cold

Hank’s Balanced Salt Solution (HBSS), 1% PSA. Under a stereomicroscope, both hemispheres, the brainstem, and the olfactory bulbs were dissected by cutting in the midline using a single edge razor blade. The medial hemispheric side was positioned superiorly; using ultrafine forceps, one tip was introduced into the lateral ventricle in an anteroposterior direction all the way through the ventricle until reaching the parahippocampal gyrus. The opposite tip of the forceps was kept in a ventral position and closed completely leaving the hippocampus and the thalamus over the forceps and the lateral wall underneath. With a single twist of the wrist, the hippocampus and thalamus were removed from the lateral wall. The meninges were then stripped off.

149 4.4.2 Ventricular zone cell cultures

Cells were dissociated with an enzymatic digestion solution (17 µL of Papain #37A17241,

Worthington, Lakewood, NJ; 15 µL of DNAse, #EN0521, Thermo Fisher Scientific, Waltham,

MA; and 288 µg of L-Cysteine, #C7352, Sigma, Saint Louis, MO in 1 mL of DMEM (Dulbecco's

Modified Eagle Medium)/Glutamax) per brain for 1 h. Dissociated VZ cells were plated into 25 cm2 flasks and grown in proliferation media composed of DMEM/Glutamax supplemented with

10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (P/S) at 37 °C to select for ependymal cells as described by Delgehyr et al. (2015). Half of the media was replaced the following day. After the cultures were confluent (~4 days), the flasks were shaken at 250 rpm overnight at room temperature to remove weakly attached cells, which were mainly differentiated neurons. The remaining cells were rinsed with Ca2+/Mg2+-free PBS and incubated for 5 minutes at

37 °C with 1 mL of trypsin-EDTA (ethylenediaminetetraacetic acid) to detach them from the flask.

These cells were resuspended in proliferation media and re-plated onto 12 mm diameter round glass coverslips in 24-well plates. The cell suspension (20 µL of 2x106 cells/mL) was carefully placed onto the coverslip and incubated for 1 h, allowing the cells to adhere at high density, after which 1 mL of the proliferation media was added to each well. The following day,

DMEM/Glutamax supplemented with 1% P/S (differentiation media) was used for continued culture; cells progressively differentiated from neural stem cells to ependymal cells in this media.

519

150 4.4.3 Alpha hemolysin and whole blood induction

Five days after differentiation was initiated, the VZ cells were submerged in either non-heparinized whole blood (collected using orbital sinus puncture530 on animals within the same colony) or, alternatively, in α-hemolysin. All samples were held at 4 °C for less than 5 minutes. Cells on coverslips were treated with 25, 30, or 40 µL of blood or 15 µg/mL α-hemolysin with or without the ADAM10 inhibitor GI254023X (#260264-93-5, Sigma-Aldrich, St. Louis, MO) for a total of

3, 24, or 48 h in the pilot dose-response experiment. This experimental group was compared to control coverslips that were submerged in PBS under similar conditions. Subsequently, time- related experiments were performed comparing exposure of cells to 30 µL of PBS, blood, or α- hemolysin for 3, 24, or 48 h.

4.4.4 Immunohistochemistry and cellular imaging

At 3, 24, and 48 h after treatment, the coverslips were washed three times with PBS and fixed with

4% paraformaldehyde (PFA) in PBS for 7 minutes. After washing again in PBS, the cells were permeabilized with 5% bovine serum albumin (BSA) in PBS and 1% Triton X-100 for 1 h; subsequently the cells were incubated for 2 h with antibodies diluted in PBS supplemented with

0.1% Triton X-100. Antibodies and dilutions included: N-cadherin (#180224, Thermo Fisher

Scientific, Waltham, MA), 1:25; GFAP (#7260, Abcam, Cambridge, MA), 1:300; βIV tubulin

(#11315, Abcam, Cambridge, MA), 1:100; and active caspase-3 (#AF835, Novus Biologicals,

Littleton, CO), 1:100. After rinsing three times with PBS, the coverslips were incubated for 1 h with the appropriate secondary antibody and diluted in PBS supplemented with 0.1% Triton X-

151 100. Secondary antibodies included goat anti-mouse Alexa fluor 488 (#A11001, Thermo Fisher

Scientific, Waltham, MA), goat anti-rabbit Alexa fluor 488 (#A11034, Invitrogen, Carlsbad, CA) at 1:300 or goat anti-mouse Alexa fluor 555 (#AQ21422, Thermo Fisher Scientific, Waltham, MA) at 1:300. Lastly, the cells were stained with DAPI at 1:5000 in PBS (#D1306, Thermo Fisher

Scientific, Waltham, MA) for 5 minutes to label cell nuclei. Images of immunostained tissue were obtained using Pascal confocal microscopy at 20X (0.50 Zeiss, Oberkochen, Germany) and 63X

(1.4 Zeiss, Oberkochen, Germany). Leveraging the National Institute of Health’s ImageJ software, we created an in-house script that quantified cell variations (total cells, number of cells expressing specific proteins, and immunodensity) in areas of 0.0225mm2. (Supplementary Figure 6).

Supplementary Figure 6. ImageJ quantification of VZ cells of interest Representative image (screenshot) of in-house script (A) incorporated into ImageJ for the quantification of cells of interest. By selecting an area of 0.0225mm2 (A), the cells of interest, in this case DAPI-positive (blue) cells are recognized (B). Cells in focus within the same plane are automatically selected (C), demarcated (yellow circles), and quantified. Quantification measures include total number of cells expressing the proteins of interest and their mean immunodensity.

152 4.4.5 Western Blot

After 48 h of treatment, the cells were rinsed with PBS and then treated with 50 µL of radioimmunoprecipitation assay (RIPA) buffer (#R0278 Sigma, Saint Louis, MO) to homogenize the cultures. Proteins were denatured in lithium dodecyl sulfate (LDS) sample buffer (NP0007,

Invitrogen, Carlsbad, CA) and reduced using β-mercaptoethanol (#M7154, Sigma, Saint Louis,

MO). After heating at 95 °C for 5 minutes, each 30 μL sample was run on an SDS-PAGE gel (4-

12% polyacrylamide) (#NP0321, Invitrogen, Carlsbad, CA) for 1 h at 120 V in SDS running buffer

(#NP0001, Novex, Carlsbad, CA). The proteins were then blotted onto a nitrocellulose membrane

(#LC2006, Novex, Carlsbad, CA) at 170 mA for 2.5 h in 15% methanol transfer buffer (#NP0006,

Novex, Carlsbad, CA). After blocking for 1 h in Tris buffered saline (TBS) with 5% powdered milk, samples were incubated with primary antibodies in TBS and 0.1% Tween (TBST) at 4 °C overnight. Primary antibodies and dilutions were GFAP (#7260, Abcam, Cambridge, MA),

1:1000; N-cadherin (#180224, Thermo Fisher Scientific, Waltham) 1:50, and GAPDH (#2118S,

Cell Signaling Technology, Danvers), 1:500. Detection was performed by enhanced chemiluminescence (#7003, Cell Signaling Technology, Danvers, MA) after 1 h of incubation with horseradish peroxidase-conjugated anti-rabbit (#70742, Cell Signaling Technology, Danvers,

MA), 1:10,000 or anti-mouse (#SC-2005, Santa Cruz Biotechnology, Dallas, TX), 1:5,000.

Western blot images were taken with the molecular imager ChemidocTM XRS+ using Image LabTM

3.0 from BioRad (Hercules, CA).

153 4.4.6 Statistical analysis

One-way ANOVA followed by the Tukey post-hoc test or the Student’s t-test were used for parametric data and a Mann-Whitney U test was used for non-parametric data. Results were considered statistically significant if p < 0.05. Data are presented as group medians (non- parametric) or means ± SD (parametric) and were considered statistically significant at p < 0.05.

All analyses were conducted using Prism 5 (Graphpad software company, San Diego, CA).

154 5 Chapter 5: Development of in vivo models of PIH and PHH

Adapted from:

1. McAllister JP, Castaneyra Ruiz L, Morales D, Isaacs AM, Ge X, Hartman A, Crevecoeur

T, Brown J, Engelbach J, Talcott M, Baksh B, Garbow J, Bayly P, Limbrick DD.

Development of a Novel Model of Intraventricular Hemorrhage and Post-Hemorrhagic

Hydrocephalus in Infant Ferrets. (In preparation)

2. Isaacs AM, Castaneyra Ruiz L, McAllister JP, Wu Y, Hamilton M, Yang R, Morales D,

Dunn J, Limbrick DD. Intraventricular Injection of Gram-Positive Bacterial Toxin as a

Mouse Model of Post-infectious Hydrocephalus. (In preparation)

5.1 Introduction

While cell cultures are ideal for testing disease mechanisms and manipulating biological processes in a controlled fashion, they are not able to provide a complete physiological environment of an intact organism. Therefore, we anticipate our future work beyond this thesis will require the use of in vivo animal models, not only to establish the mechanisms we have identified in vivo but also as a foundation for preclinical drug studies and easier translation into FDA–approved clinical trials. Our primary objective was to create animal models of PHH and PIH that mimic the human disease characterized by VZ disruption, impairment of neural stem cells differentiation, ependymal denudation, and neuroinflammation occurring in the setting of IVH or infection.

155 For PIH, we developed a mouse model because their small size, relatively cheaper cost, ease of handling, and availability of transgenics make them more feasible for a disease with very limited information on previous experimental models. On the other hand, PHH has many existing experimental models including mice531,532, rats372,533-536, rabbits384,537-540, cats541, dogs378,542-548, sheep378,542-548, and piglets379,380,549,550. Nevertheless, we developed a novel ferret model to circumvent some of the major limitations of the existing models. Unlike the lissencephalic cerebral cortex of rodents, ferrets have a gyrencephalic brain that is similar to a human brain, providing investigators with access to very early stages of neural development551. Particularly important for the investigation of preterm IVH, ferret brain maturation at birth is equivalent to the human brain during the early second trimester552. Their smaller size in comparison to other USDA-covered species makes them relatively easier to handle and cost-effective.

5.2 Results

5.2.1 Ferret model of PHH

5.2.1.1 Subjects

Thirty-two ferret kits (ages 14-21 days old) were included in this study. Five intact controls did not undergo any surgery. Stereotactic intraventricular injection was performed in 4 sham PBS induction controls, 13 autologous blood inductions, and 10 syngeneic lysed blood inductions.

Variations in age of induction, which occurred due to logistical challenges, were factored into future analyses. (Table 9)

156 Table 9. Summary of ferrets used in the development of an animal model of PHH

Intact Control Sham Control Autologous Blood Syngeneic Blood

- - - -

ID ID ID ID

induction induction induction

induction*

DaysPost DaysPost DaysPost DaysPost

InjectionSite InjectionSite InjectionSite InjectionSite F3 29 NA F22 48 Bilateral F19 44 Bilateral F38 28 Bilateral F4 121 NA F23 48 Bilateral F20 44 Bilateral F39 28 Bilateral F8 7 NA F24 48 Bilateral F21 44 Bilateral F40 28 Bilateral F17 44 NA F25 48 Bilateral F26 6 Unilateral F42 28 Bilateral F35 54 NA F27 23 Bilateral F43 28 Bilateral F28 52 Unilateral F44 28 Bilateral F29 52 Unilateral F45 28 Bilateral F30 52 Unilateral F63 22 Bilateral F31 35 Unilateral F64 22 Bilateral F32 35 Bilateral F66 22 Bilateral F33 35 Unilateral F34 35 Bilateral F36 28 Unilateral * Calculated post-induction age based on induction at 20 days of age.

5.2.1.2 Neurological Status and Health

None of the ferret kits developed post-operative neurological deficits or signs of discomfort. Jills consistently provided normal maternal care and nourishment. Early in the study, one jill removed the skin sutures within an hour after surgery; this experience prompted us to carefully place single, interrupted subcuticular sutures and to cover the incision with skin glue. Subsequently, no incisions were opened by the jill in the remaining cases. Except for the expiration of one kit at the end of an

MRI scan, probably from a thermoregulatory impairment, all kits recovered well after neuroimaging. All groups exhibited body weight increases along a similar growth curve (Figure

48C).

157 5.2.1.3 Ventriculomegaly

The extent of ventriculomegaly was variable within and among induction groups, with a clear trend toward more frequent and more severe ventricular enlargement occurring in kits that received syngeneic lysed blood compared to those receiving autologous blood. The volume of syngeneic lysed blood injected into each lateral ventricle (150-250 μL) was considerably higher than the amount of autologous blood delivered (10-50 μL). The pattern of ventriculomegaly was quite consistent, with much greater enlargement of the occipital and temporal horns of the lateral ventricles compared to the frontal horns. Often ventriculomegaly was asymmetric even though equal volumes of blood were usually injected into both frontal horns. Unilateral injections rarely produced bilateral ventriculomegaly (Figure 48).

Figure 48. Ventricular enlargement in ferret a model of PHH (A) Representative T2-weighted coronal MRI images illustrating ventricular size in an intact control (animal F035) and (B) following bilateral intraventricular injection of syngeneic blood (animal F050). Selected images are viewed with the animals’ left corresponding to the reader’s 158 left, arranged from anterior (left) to posterior (right) in each row and aligned at approximately the same coronal plane. Note the extremely small size of the frontal (FH) and temporal (TH) horns of the lateral ventricles in the control ferret (A). Following blood injections (in this case, 9 days), the occipital (OH) and temporal (TH) horns of the lateral ventricles expanded dramatically, and a CSF flow void (hypointense signal) was apparent in the cerebral aqueduct (CA). The needle trajectory for injecting blood is drawn with a dashed line in (B). (C) Line plot depicting the progression of mean body weights for each experimental group up to 25 days post-induction. All groups followed similar growth curves, although the PBS-injected sham controls were lighter (non-significantly), probably because they were selected for less traumatic procedures due to their lower initial weights. (D) Bar plot depicting ventricular volumes for the controls (intact and sham) and blood- induced (autologous and syngeneic) groups. While the ventricular volume of the blood injected group was much greater than the control group, the difference was not statistically significant due to large within-group variation.

5.2.1.4 VZ disruption and glial activation (neuroinflammation)

Histological and immunohistochemical methods confirmed the patterns of ventriculomegaly detected with MRI and, importantly, revealed VZ disruption neuroinflammation. The pattern of

VZ disruption was quite similar to our observations in preterm humans with IVH and PHH197 and with in vitro studies of maturing ependymal cells318, namely loss of ciliated ependyma, eruption of VZ cells into the lateral ventricle, loss or spatial rearrangement of N-cadherin adherens junctions, and the appearance of reactive astrocytes in the areas of VZ disruption. These impairments, found as early as nine days following blood induction, were found throughout the lateral ventricle, but seemed more frequent and prominent in the occipital and temporal horns where the ventricular enlargement was greatest. It is important to note that the pattern of cytopathology we observed occurred in all animals that had been injected with blood regardless of the extent of ventriculomegaly (Figure 49).

159

Figure 49: Immunohistochemistry in a ferret model of PHH Representative photomicrographs comparing astrocyte (GFAP), ependymal cell cilia (βIV tubulin), adherens junction (N-cadherin) proteins, and a known modulator of N-cadherin (ADAM10) in the temporal horn (LVt) of PBS-injected control (A,C,C’,E,E’) and blood injected IVH (B,D,D’,F,F’) ferrets. (A & B) In H&E-stained sections, the monolayer of ependymal cells (Ep) is much thinner in IVH and contains a prominent area of disruption (*). (C/C’&D/D’) Viewed at low (C & D) and high (C’ & D’) magnification, IVH is associated with a conspicuous reduction in βIV tubulin-positive cilia, and the disrupted area contains numerous reactive GFAP-positive astrocytes. (E/E’ & F/F’) Likewise, in IVH there is a reduction in N-cadherin-positive (Ncad) adherens junctions and a conspicuous increase in ADAM10 immunostaining. Taken together, these results demonstrate that ventricular zone disruption and reactive astrocytosis are associated with IVH, even in mild forms of ventriculomegaly. (G) Box and whisker plots of the mean immunodensity of N-cadherin and ADAM10 in the ventricular wall of control and IVH/PHH ferrets. The decrease in N-cadherin expression and the increase in ADAM10 expression are both statistically significant, suggesting a causative role for ADAM10 in the ependymal disruption that occurs in PHH. Additional abbreviations: CP – choroid plexus; GFAP – glial fibrillary acidic protein.

Lateral to the VZ/SVZ, the PVWM contained many more GFAP-positive astrocytes compared to controls, and most of these cells had the classical appearance of reactive astrocytes, suggesting neuroinflammation in the setting of VZ/SVZ disruption in the blood-treated group (Figure 50).

160

Figure 50. Reactive astrocytosis in a ferret mode of PHH Representative histological and immunohistochemical images (of increasing magnification from A-C) comparing VZ and periventricular white matter region cytology in control and blood-injected ferrets. GFAP-positive reactive astrocytes cluster at the areas of ependymal denudation. LV- lateral ventricle

5.2.1.5 Diffusion Basis Spectrum Imaging of ferrets

The brains of 13 age- and weight-matched female ferrets, including 7 blood-injected (IVH/PHH group) and 6 PBS-injected (Control group), underwent ex-vivo DBSI scans following sacrifice at

~50 days old. The corpus callosum (CC) and bilateral anterior limbs (ALIC) and posterior limbs

(PLIC) of internal capsules were demarcated as regions of interest (ROIs). DBSI measures were extracted from the ROIs. The IVH/PHH group exhibited a 50% proportional increase in non- restricted fractions within the PVWM compared to controls (p=0.001) reflecting PVWM edema.

However, 68% of this change was due to an increase in the hindered fraction (p<0.0004), implying that there was a component of increased cellularity in the PVWM, which would suggest inflammation. The proportional increases in non-restricted and hindered fractions in the CC were

96% (p=0.002) and 120% (p=0.0006), respectively, and 49% (p=0.0006) and 51% (p=0.0005) in the ALIC, respectively. There was no significant change in the PLIC. Assessing intrinsic PVWM

161 injury, the CC and ALIC appear to sustain the most impact from PHH. This is evident as their respective fiber density proportionally decreased by 7% (p=0.007) and 10% (p=0.03). In addition to axonal fiber loss, the ALIC also demonstrated evidence of axonal injury by a decrease in axial diffusivity of 8% (p=0.008) when compared to controls. While similar trends were observed in the

PLIC, none were statistically significant. (Figure 51)

Figure 51. Periventricular white matter dMRI characteristics in a ferret model of PHH Proportional change in mean Diffusion Basis Spectrum Imaging (DBSI) of intraventricular hemorrhage and posthemorrhagic hydrocephalic (IVH/PHH) from control ferrets in three white matter tracts: corpus callosum (CC) and bilateral anterior limbs (ALIC) and posterior limbs (PLIC) of the internal capsules. In the CC and ALIC, there was an approximately 50-90% increase in the non-restricted fraction, reflecting PVWM edema in the IVH/PHH group. At the same time, there was over a 100% increase in the hindered fraction, reflecting increased cellularity, typically as a result of inflammation, contributed to the edema. No significant changes were observed in the PLIC. ★ p< 0.05. Abbreviations: MA – mean diffusivity, FA – fractional anisotropy, AD – axial diffusivity, FF – fiber fraction, RF – restricted fraction, NRF – non-restricted fraction, HF – hindered fraction.

162 5.2.2 Mouse model of PIH

5.2.2.1 Neurological status and health

Of 23 mice, 14 underwent bilateral intraventricular injection of ⍺-hemolysin (⍺-hemolysin group) and 9 received PBS (Control group). There was no difference in pre-induction weights between groups (p=0.66). There was a steep learning curve during the development of the model, as there was no existing literature to guide the concentration and volume of ⍺-hemolysin that would yield the desired endpoint (hydrocephalus). Injection of 25 µL/ventricle in the first cohort (5 controls and 5 ⍺-hemolysin) had unacceptably high mortality (80-90%), with the ⍺-hemolysin-injected mice dying within hours of surgery. The majority of control mice in that cohort died within 24-48 hours of injection, suggesting there was also a volume effect, perhaps causing delayed intracranial hypertension, hydrocephalus, and death. Therefore, the volume was reduced for subsequent injections. Further volume response trials showed that injections of 2-2.5 µL/ventricle was ideal, as they did not cause any issues in the control mice. Additionally, ⍺-hemolysin injection at that volume yielded hydrocephalus in 50% of the animals without acute issues.

5.2.2.2 Ventriculomegaly

For dose response trials, three concentrations of 휶-hemolysin were tested: low (27.4 µg/mL), medium (54.8 µg/mL), and high (91.3µg/ml) doses. Likely due to technical reasons, early (<48h) mortality rates in the low and high dose were similar (~15%), whereas mice in the medium dose had no change in mortality. Neuroimaging was obtained 7 days after surgery. Approximately 50% of mice developed hydrocephalus to varying severity of ventriculomegaly. However, there were

163 no significant differences in ventricle size between the animals receiving different doses of ⍺- hemolysin (p=0.403). The mice receiving medium and high doses mice did demonstrate significant white matter T2W hyperintensity suggestive of inflammation/edema (Figure 52).

Figure 52. Ventricular enlargement in a mouse model of PIH Representative T2W coronal images of mice undergoing bilateral intraventricular injection of PBS (control) and low (27.4 µg/mL), medium (54.8 µg/mL), or high (91.3µg/ml) doses of α-hemolysin at 2.5 µL/ventricle. Note the Hamilton syringe needle tract on the left (red dotted arrow). All α - hemolysin-injected mice developed hydrocephalus as compared to the ventricular size of the controls. However, there was significant white matter hyperintensity observed in the medium and high dose mice in comparison to the hypointense signal seen in the low dose and control mice (blue solid arrow).

5.2.2.3 VZ disruption and reactive gliosis (neuroinflammation)

Immunohistochemistry confirmed the pathology detected with MRI, revealing ependymal denudation and astrogliosis (neuroinflammation). The pattern of ependymal disruption recapitulated our observations in preterm humans, in in vitro studies of maturing ependymal cells in both the blood and ⍺-hemolysin-treated cells, and in our ferret model of IVH/PHH. These

164 findings provide further evidence that ependymal disruption and neuroinflammation are an underlying pathophysiology that is common to PIH and PHH (Figure 53).

Figure 53: Immunohistochemistry in a mouse model of PIH Representative photomicrographs comparing astrocyte (GFAP) and ependymal cell cilia (βIV tubulin) in (A-C) non-disrupted and (D-F) disrupted regions of mice injected with intraventricular α -hemolysin. Cell nuclei are stained with DAPI. Viewed at low (A & D), medium (B & E), and high (C & E) magnification, the disrupted regions showed a conspicuous reduction in βIV tubulin- positive cilia and contained numerous reactive GFAP-positive astrocytes. These results demonstrate that ventricular zone disruption and reactive astrocytosis are associated with PIH. GFAP – glial fibrillary acidic protein.

165 5.3 Discussion

5.3.1 Prevalence and time course of ventriculomegaly in PIH and PHH models

Our observations indicate that the progression of PHH and PIH begin with periventricular edema followed by expansion of the lateral ventricles. In the mouse PIH model, animals that were scanned within 24 hours of surgery demonstrated diffuse white matter hyperintense T2W signal suggestive of edema. However, those that made it to 7 days to be scanned had developed much larger ventricles compared to their initial perioperative scans. In the PHH ferret model, ventriculomegaly typically occurred 10-15 days post-induction. Occasionally, ventriculomegaly occurred abruptly and severely within 7-10 days post-induction. The frequency of ventriculomegaly in both the PIH and PHH models was approximately 30% to 50%, which is similar to those reported in other species 379,553,554 and quite consistent with the incidence of PIH and PHH in human neonates who develop sepsis or IVH11,38,44,240,555. Thus, both the mice and ferret models appear to mimic the clinical occurrence of PIH and PHH. This feature also raises the pertinent and unanswered question of why some humans and animals do not develop ventriculomegaly after neonatal sepsis or IVH.

5.3.2 Cytopathology

Ependymal disruption in both models were characterized by ependymal cell denudation, loss of motile cilia, and accumulation of reactive astrocytes, possibly in the same location as ependymal alterations. In the ferret model, we also observed lost or spatial translocation of N-cadherin from the cell membrane to the cytoplasm in addition to lost or misdirected radial glial cells. These

166 features are consistent with those observed in human preterm infants182,197,556,557 and rabbits558,559 with IVH/PHH as well as rodents with congenital hydrocephalus182,183,560. Furthermore, a glial response similar to reactive astrocytosis occurred in the ependyma and the PVWM. This finding matches a growing line of evidence from in vitro and in vivo models of IVH/PHH, as well as the observations from ferret kits receiving intracisternal injections of kaolin to induce obstructive hydrocephalus561.

5.3.3 Surgical and Methodological Considerations

Several important lessons were learned during the development of the animal models. For the mouse model, the main issues we encountered, which were later overcome, were related to finding the optimal dose and volume to achieve ventriculomegaly without acute death. There remains the limitation of bypassing the normal routes of human infection that lead to PHH when the intraventricular route is used. Future work will include inducing PHH via systemic infection.

In the ferret model, a considerable volume of injected blood was required in order to promote ventriculomegaly. Autologous blood, which could only be obtained in limited quantities from the tail, seldom exceeded 25 μL/injection and the subsequent ventricular volumes in these preparations were lower than when larger volumes of blood were injected. In order to obtain a sufficient quantity of injectable blood, syngeneic cardiac samples had to be collected from a ferret littermate.

These preparations delivered 150-250 μL/injection and produced much larger ventricles. It should

167 be noted that Whitehall and colleagues ultimately employed a second intraventricular injection of blood with a raised hematocrit in order to increase their yield of rats with ventriculomegaly.562

The ferret model does not feature an expandable skull. Even though the sagittal and coronal sutures appeared flexible during the injection surgery at 20 days of age, none of the kits demonstrated an enlarged skull, even those with relatively severe ventriculomegaly. To our knowledge, there are no reports on when the cranial sutures of the ferret ossify, but it may be that the sutures close within a few days of our injections, prior to when intracranial pressure may increase. The lack of an expandable skull and the prevalence of mild-moderate ventriculomegaly in this model are important, especially because other models in infant-juvenile rats, cats, and rabbits do present with a remarkably expansive skull and open anterior fontanelle.

A complete stereotaxic atlas of the adult or developing ferret is not available. As a result, we had to rely on measurements and trajectories planned from MRI scans and gross brain dissections. In addition, the short snout and relatively flat face of the infant ferret required a nose cone for anesthesia, which interfered with the standard bite bar used in commercial stereotaxic frames.

Furthermore, the external auditory meatus of a 20-day-old ferret kit is still closed. We attempted to carefully secure the animal in the usual stereotaxic planes with the ear bars directed toward the openings of the external auditory meatuses and the dorsum of the skull perfectly horizontal, but positioning can still be variable. Further studies with short-term post-induction neuroimaging are still needed to determine the accuracy of intraventricular injections.

168 None of the ferret kits, regardless of the severity of ventriculomegaly or the amount of blood injected, developed detectable neurological deficits. This feature of the model may be advantageous for long-term studies, but it appears that functional outcome assessments will have to rely on more stringent behavioral, cognitive, and neurological tests. Such tests are not impossible; however, as di Curzio et al. have demonstrated, quantitative open field tests can be performed on ferret kits from 10 to 44 days old563. Depending on the outcome measures to be used

(e.g., advanced MRI), studies with the infant ferret IVH/PHH model can become quite expensive.

Because a ferret is a covered species by the USDA and Institutional Animal Care and Use

Committees, purchase from purpose-bred vendors, domestic and local transportation costs, and per diem charges can amount to about $1500 for a lactating jill and a litter of 4-5 kits.

5.3.4 Limitations

This study has several limitations. As stated previously, the modest yield of, and relatively low variability in ventriculomegaly in both models contributed to the low sample sizes, making it difficult to draw firm predictions on the severity of hydrocephalus and to accumulate cohort sizes amenable to statistics. The mice used in this model were older so we could assess the ventricular zone as their ependyma had already developed. However, we used older mice in order to be able to obtain neuroimaging as the small brains of immature rodents create difficulties for high- resolution neuroimaging86,564. Ongoing experiments are utilizing P0/P1 mice for intraventricular injections. We also recognize that an intraventricular injection approach bypasses the typical systemic route of infection; however, as first steps we had to ensure adequate ventriculitis was achieved, which can be further advanced to include systemic inoculation. The absence of an

169 expandable skull in the ferret may require inductions in younger kits; it may also indicate that intracranial pressure measurements need to be taken, which was not the case in the present study.

5.4 METHODS

5.4.1 Ferret model of IVH and PHH

5.4.1.1 Subjects

Ferrets were delivered to the Washington University School of Medicine animal facility as a litter of one jill and 4-6 kits when the kits were 12-14 days old. Following a 5-8-day quarantine period, kits underwent a pre-induction baseline MRI 2-3 days prior to receiving intraventricular injections to mimic IVH. Autologous blood or syngeneic blood obtained from an age-matched littermate was used to induce IVH. Controls received intraventricular injections of sterile saline or phosphate buffered saline (PBS) or no injections to serve as intact controls. All of the induction and control procedures were conducted when the kits were 14-21 days old; these times were chosen to match the age during which most premature humans present with IVH240,552,565. Post-induction MRI scans were performed to document the development of ventriculomegaly, indicative of PHH. To conclude each study, kits were euthanized with an overdose of Fatal Plus (sodium pentobarbital

95%, ketamine 5%; IM), and the brains were fixed by cardiac perfusion of 4% paraformaldehyde in PBS. All procedures were conducted in accordance with National Institutes of Health Guide for the Care and Use of Laboratory Animals and approved by the Washington University in St. Louis

Institutional Animal Care and Use Committee (IACUC protocol #2015005).

170 5.4.1.2 Induction of IVH and PHH

Initially, IVH/PHH was induced in infant ferrets by unilateral or bilateral intraventricular injections of heparinized autologous blood obtained from the tail vein of the animal. This method was modified from procedures described previously by Whitelaw et al379,562. Approximately 20-

150 μL of autologous blood, depending on each animal, was obtained from the tail using a 22- gauge hypodermic needle and drawn into sterile heparinized 70 μL glass capillary tubes. These blood samples were kept on ice for about 10 minutes to allow heparin to be mixed with the blood; this step proved to be critical to prevent blood clotting and obstruction in the microliter syringe.

About 20-50 μL of the sample was drawn into a sterile Hamilton 50 or 100 μL syringe, which was then mounted on a stereotactic carrier for intraventricular injection.

In order to mimic larger hemorrhages (i.e., IVH grades III and IV566, unilateral or bilateral intraventricular injections of syngeneic lysed blood were employed. Syngeneic blood was obtained from an anesthetized, age-matched littermate by cardiac puncture using routine methods. This approach yielded approximately 500-5,000 μL of blood, which was sufficient for induction of 4-5 littermates. The blood samples were placed immediately in 1.5 mL heparinized tubes and frozen in liquid nitrogen. As needed, samples were lysed by 3 subsequent thaw-freeze cycles before being drawn into a sterile 1.0 mL syringe for intraventricular injection into each animal.

For all intraventricular injections, the following procedures were performed using sterile technique. Kits were anesthetized with 1-4% isoflurane vapor in 100% oxygen. Heart rate and

171 oxygen saturation were monitored by pulse oximetry using a probe clipped to a rear paw.

Respiratory rate was monitored by manual observation, and body temperature was monitored with a rectal probe. Kits were placed on a heating pad in the prone position and the head stabilized in a stereotaxic frame. Blunt ear bars were placed against the external auditory meatus because the ear canal was not patent at this age. The mouth was secured to a standard bite bar onto which the anesthesia nose cone was taped. Because maintaining body temperature in ferret kits at this age is very important for survival, towels were placed over the animal to keep it warm. The skin on the dorsal neck and skull were alternately swabbed three times each with Betadine and 70% alcohol to obtain sterility. The surgical field was covered with sterile drapes and a small hole cut in the outermost drape for access to the dorsum of the skull. A small skin incision was made over the dorsum of the skull and the skin retracted to expose bregma and the coronal suture. Based on measurements taken from pre-induction MRIs and prior gross morphology dissections, stereotaxic coordinates were set at 1.0-1.5 mm lateral to bregma, 2.0-3.0 mm anterior to bregma, and 5.5-

6.5mm deep to the pial surface to target the frontal horn of the lateral ventricle. A small (~1.0 mm diameter) burr hole was drilled in the skull. After bone and dura mater bleeding had been controlled with saline flushes, bone wax and Gelfoam, the syringe was loaded with PBS, saline or blood and advanced into the brain to the approximate depth. Initially this depth was 5.0 mm from the skull surface, subsequently 10.0 mm proved to be more accurate. After a 30-second equilibration period, the induction agent was injected slowly into each lateral ventricle. In general, the complete injection process consisted of short (about 10 second) injections of about 5.0 μL of sample followed by equilibration intervals of about 20 seconds. After the final injection, the needle was left in place for about 30 seconds and then withdrawn. When bilateral injections were performed, the same needle was used, having been pre-loaded with a sufficient amount of induction agent,

172 and the same injection procedure repeated on the opposite side. The skin incision was closed with absorbable suture using subcutaneous suturing techniques after surrounding tissue had been infiltrated with a mixture of Lidocaine-Marcaine (2.0 mg/kg). The wound area was cleaned with sterile saline and minimal amounts of skin glue applied to the incision to prevent the jill from opening the incision. Post-operative analgesia and infection control were initiated with

Buprenorphine (0.01-0.03 mg/kg IM) and Cephalexin (10mg/kg SC). Isoflurane inhalation (but not oxygen) was discontinued and the animal allowed to recover; once sternal and reactive, the kit was returned to its litter.

Following surgery all kits were weighed daily and monitored for signs of neurological deficits

(ataxia, alertness, reflexes), general health (dehydration, lethargy), and discomfort (vocalization, stiff hair).

5.4.1.3 Ferret Neuroimaging

5.4.1.3.1 In vivo

Anatomic MRI techniques, performed sequentially during the same setting or individually during separate sessions, were used to determine gross morphological changes in the brain, especially volumetric increases in the cerebral ventricles and the location of blood deposits and needle tracks.

Imaging was performed pre-induction or post-induction (5-75 days, depending on the experiment) on an Agilent/Varian 4.7T small animal MR scanner equipped with a DirectDriveTM console

173 (Santa Clara, CA), built around an Oxford Instruments (Oxford, United Kingdom) horizontal magnet with a 40-cm clear bore diameter. MRI images were collected with a custom-built actively decoupled transmit (volume, 9-cm inner diameter) and receive (surface, 3.3, 3.8 or 4.5 cm outer diameter, depending on ferret head size) coil pair.

Ferrets were anesthetized with isoflurane/oxygen mixture (3-4% isoflurane) in an induction chamber. When all reflexes had been abolished, the head of the ferret was placed in the prone position and secured in a custom-built head-holder with a C-clamp on the head and a pallet bar. A custom-built nose cone was taped in place for delivery of 1-3% Isoflurane for maintenance of anesthesia during the scan. Isoflurane excavation was accomplished by a silicone suction tubing placed next to the animal's head. The eyes were moistened with ophthalmic ointment. The ferret body was placed prone onto a padded heating bed that circled warm water through a water pump

(Isotemp® 2100). A pulse oximeter (Nonin® 7500) secured to the ferret paw was used to monitor heart rate and oxygen saturation during MRI scan. Ferret respiratory rate (a pillow attached to animal abdomen) and body temperature (rectal probe) were monitored and heat-compensated with a MR-compatible small animal monitoring & gating system (SA Instruments, Inc.). Total preparation time was about 15-30 minutes. All monitored physiological parameters were recorded every 15 minutes until the animal had been recovered and had assumed a sternal posture. The in vivo MRI scans required 60-150 minutes, depending on the MRI sequences obtained. While recovering from anesthesia, the ferret kits were placed on a heating pad and body temperature monitored.

174 For anatomical sequences, depending on brain size, 24-30 slices of T2-weighted transaxial images were collected with a 2D fast spin-echo (fsems) sequence with an echo train length (ETL) of 8,

Kzero = 8, TR 3 S, effective TE 96-112 ms; matrix size 256 x 256 and field of view (FOV) 48 x

48 to 60 x 60 mm2 (depending on ferret brain size) with a slice thickness 1.0 mm. Total data collection time for four averages was 6 minutes 48 seconds. The T2-weighted images were loaded into MatLab (MathWorks®) and converted into NIfTI (.nii) format with zero-padding to a matrix size of 512 x 512 as anatomic images. These T2-weighted images were used for ventricular volume segmentation with ITK-SNAP.

5.4.1.3.2 Ex-vivo DBSI

To limit cost, the explanted brains of a subset of age and weight matched ferrets (n=13) sacrificed at approximately 50 days old underwent DBSI to assess their PVWM integrity. The brains were placed in a 35mL syringe tube in 4% PFA then scanned ex vivo in an Agilent® 4.7 Tesla magnet with a solenoid coil. T2-weighted images, along with a multi-echo spin-echo diffusion weighted sequence in 99 diffusion directions (TR 3000 ms, TE 60 ms, Matrix = 192 x 192, b-value max =

3000 s/mm2) was acquired for a total scan time of 14 hours per animal. The raw data files were converted to NIfTI and registered to a single subject space.

All datasets were processed with our in-house DBSI pipeline described above. Two anisotropic components were used to model primary and secondary direction of fibers within imaged voxels.

175 For division of the isotropic components, 2.0 µm2/ms and higher ADC was chosen to represent free diffusion, 0.6-2.0 µm2/ms hindered diffusion, and 0.6 µm2/ms and less for restricted diffusion.

ROIs of the white matter tracts we examined were drawn on de-noised T2W coronal images by two independent blinded operators. The ventricles on de-noised T2W coronal slices were marked as ROIs for ventricular volumetric assessment. PVWM ROIs were identified in coronal slices. The genu – the anterior end of the corpus callosum was demarcated medial to the frontal lobes and the anterior horns of the lateral ventricles, behind and below the cingulate gyrus. The internal capsule was demarcated as a white matter structure that begins at the level of the thalamus, resting between the caudate nucleus and the thalamus, connected to cortical white matter. Thus, in the same slice as the genu, the anterior limb of internal capsule was identified. The most posterior end of the corpus callosum the splenium was identified coming through the mid-region of the coronal slice.

The position was determined in the second to last slice in which the corpus callosum still existed, knowing that fibers continued all the way through the structure still. Positioned between the splenium and genu, the posterior limb of internal capsule was identified using the same guidelines as the anterior limb, being sure to exclude the crus cerebri (cerebral peduncle). Rostrally, the crus cerebri was observed as a bright, dense bundle of white nerve fibers emerging from the ventral cerebral cortex.

All PVWM and ventricular volume ROIs were delineated using ITK-SNAP (open source software, www.itk-snap.org). After delineation, the voxel intensity values of all ROIs were extracted and analyzed.

176 5.4.1.4 Sacrifice and Neurocytology

Routine histology and immunohistochemistry (IHC) of PFA-fixed brains collected at sacrifice were used to characterize cytology and cytoarchitecture, as described previously197. Each brain was blocked in the coronal plane and embedded in paraffin following routine methods. Coronal sections were cut serially at a thickness of 10 μm, collected into staining groups in which sections were mounted onto glass slides at intervals of approximately 100 μm and stained with Hematoxylin and Eosin (H&E). For routine immunohistochemistry, primary antibodies directed against cilia

(βIV tubulin 1:100; catalog #11315, Abcam), immature ependymal cells and astrocytes (glial fibrillary acidic protein, GFAP; 1:300; catalog #7260, Abcam), microglia (ionized calcium binding adaptor molecule 1, Iba1; 1:100, catalog #PA5-27436, Thermo Fisher Scientific), and adherens junctions (N-cadherin, Ncad; 1:25; catalog #180224, Thermo Fisher Scientific), were employed.

Secondary antibodies included goat anti-rabbit Alexa Fluor 488 (1:300; #A11034, Thermo Fisher

Scientific) and goat anti-mouse Alexa Fluor 555 (1:300; #AQ21422, Thermo Fisher Scientific).

Finally, the tissue was treated with 4’6-diamidino-2-phenylindole (DAPI 405, 1:5000; #D1306

Thermo Fisher Scientific) to label all cell nuclei. Sections were analyzed by light microscopy and multiple fluorescence confocal microscopy. Quantification of immunohistochemistry was performed with our in-house script that quantified cell variations (total cells, number of cells expressing specific proteins, and immunodensity) per slide (Supplementary Figure 6). Relative quantifications of the cells of interest were obtained as a ratio of the total number of DAPI-positive cells

177 5.4.1.5 Statistical analysis

All statistical analyses were performed using in-house Python v2.7 and R package v3.4.1 scripts.

Parameter map (PM) intensity values inside PVWM ROIs and ventricular volumes were analyzed with Shapiro-Wilk test for normality. Paired and non-paired t-tests and non-parametric measures

(Wilcoxon signed rank tests) were used for between group comparisons as determined by tests of normality. Correlations between PVWM and ventricular volumes were analyzed between and within groups.

5.4.2 Mouse Model of Ventriculitis and PIH

5.4.2.1 Subjects and Experimental Design

Mice were delivered to the University of Calgary animal facility as a litter of 12 pups. Following a 5-day quarantine period, pups underwent intraventricular injections to mimic ventriculitis and

PIH. ⍺-hemolysin (Medimmune, Mountain View, CA) provided by our collaborator was used.

Controls received intraventricular injections of saline. Post-induction MRI scans were performed to document the development of ventriculomegaly, indicative of PHH. All procedures were approved by the Calgary University Institutional Animal Care and Use Committee (IACUC protocol #AC17-0151). Throughout the study, mice were monitored daily for neurological and general health status until the time of euthanasia. In addition to routine assessments of alertness, locomotion, hydration, pain, anxiety, and infection, special attention was focused on signs and symptoms that typically accompany hydrocephalus and increased intracranial pressure.

178 5.4.2.2 Induction

Mice were anesthetized with isoflurane 2−4% via inhalation through a nose cone. Their head was secured in a stereotactic head frame in the prone position. A surgical plane of anesthesia was obtained, as determined by multiple clinical signs such as limb withdrawal following paw pressure and any response to painful stimuli. Buprenorphine hydrochloride (0.05 mg/kg) was administered subcutaneously with a 27-gauge needle to maximize animal comfort during recovery. Strict aseptic techniques were followed at all times during surgery. The skin on the dorsal neck and skull was alternately swabbed three times each with Betadine and 70% alcohol to obtain sterility. The surgical field was covered with sterile drapes, sterile instruments and materials were used, and sterile surgical gloves were worn at all times.

A small skin incision was made over the dorsum of the skull and the skin retracted to expose bregma. A small (~1mm diameter) burr hole was drilled in the skull 1.0 mm lateral to bregma and

0.5 mm posterior to the coronal suture. After bone bleeding had been controlled with saline flushes or bone wax, the stereotactic frame was positioned to its coordinates for the frontal horn of the lateral ventricle. A Hamilton 30-gauge needle connected to a 5-µL gas-tight glass syringe

(Hamilton, Reno, NV, USA) that was loaded with different concentrations and volumes of ⍺- hemolysin in saline (27.4 µg/ml, 2.5 µl each side; 54.8 µg/ml, 2.5 µl each side; 91.3 µg/ml, 2 µl each side; 68.5 µg/ml, 2 µl each side) was lowered to the predetermined depth so that the tip will be located in the frontal horn of the lateral ventricle. The drug was then dispensed into the ventricle.

After a 1-minute equilibration period, the needle was withdrawn slowly, and any leaking CSF stopped with a piece of bone wax. The similar process was performed on the opposite hemisphere.

179 The incision was closed with absorbable suture, the wound was cleaned, and the animal was allowed to recover prior to return to its cage. The induction procedure took ~30 minutes skin-to- skin per mouse.

5.4.2.3 Imaging

Imaging was undertaken 7 days post-injection or sooner if the mouse showed signs of humane endpoint. Imaging was performed on a Bruker BioSpin 9.4T MRI using a helium cooled cryocoil, which is a dual channeled surface coil. Anatomical scans were obtained as T2W RARE sequences

(Matrix = 256 x 256, FOV =1.92 x 1.92 cm, slice thickness = 0.5 mm, TE = 30 ms, TR= 4000 ms,) to determine morphological changes in the brain, especially volumetric increases in the cerebral ventricles. Animals were anesthetized with isoflurane (2-4%) while in an induction chamber and then through a nose cone while positioned in the MRI scanner. After the animal was sufficiently anesthetized, the animal’s eyes were moistened by the application of ophthalmic ointment to further reduce the amount of drying to the eyes. The animal was placed into the scanner headfirst for imaging in prone position while lying on a bed of pillows with a heart rate and respiration monitor. Core temperature (36°C) was controlled using regulated air flow and a feedback to a rectal probe. Respiration was monitored and maintained by adjustments in anesthesia.

The total time in the MRI lasted 30-45 minutes after which the animal was removed from the MRI and monitored until the anesthetic has worn off and normal movements had returned. While recovering from anesthetic, the mouse was placed on a heating pad to keep body temperature as

180 close to normal as possible. After normal movement has been restored, the animal was returned to its litter.

5.4.2.4 Euthanasia

To conclude each study, mice were euthanized with an overdose of Sodium Pentobarbital (200 mg/kg). After all reflexes had been abolished, the pleural cavity was opened surgically, and a transcardial perfusion was performed with PBS. The extracted brains were cryopreserved in

Optimal cutting temperature (OCT) compound prior to storage in -80 °C freezer.

Immunohistochemistry of coronal sections of the cryofixed brains were performed with primary antibodies directed against cilia (βIV tubulin 1:100; catalog #11315, Abcam), and immature ependymal cells and astrocytes (GFAP; 1:300; catalog #7260, Abcam). Secondary antibodies included goat anti-rabbit Alexa Fluor 488 (1:300; #A11034, Thermo Fisher Scientific) and goat anti-mouse Alexa Fluor 555 (1:300; #AQ21422, Thermo Fisher Scientific). The tissues were treated with DAPI (405, 1:5000; #D1306 Thermo Fisher Scientific) to label all cell nuclei. Sections were analyzed by light microscopy and multiple fluorescence confocal microscopy. However, immunodensitometry was not performed.

181 6 Chapter 6: Discussion of dissertation

6.1.1 Leveraging a translational neuroscience approach to PIH and PHH

In the past decade alone, over 11,000 hydrocephalus-related research articles have been published, accounting for one third of all papers published on the subject since 1860. In this extensive volume of data are many useful ideas that could potentially change the landscape of the field. However, there seems to be a dichotomy of the domains of hydrocephalus research, i.e. experimental versus clinical, and only a few studies have endeavored to merge the two approaches into a translational framework87,102. In addition, the majority of studies tend to focus on a single variant of hydrocephalus366,567-569, and those combining two or more forms of hydrocephalus are uncommon.

Since the 1990s, “Translational Neuroscience” has gained widespread recognition, facilitating critical advances in many neurological conditions570. In addition to bridging the gap between fundamental neurosciences (typically in experimental models) and human brain function, the scope of translational neuroscience has widened to include multi-disciplinary consortiums, the design and implementation of new technologies and the development of diagnostic, prognostic, treatment and monitoring strategies570,571.

The studies presented in this dissertation epitomizes the use of translational neuroscience principles to further hydrocephalus research. Rather than focusing on a single disease, two similar variants of hydrocephalus (PIH and PHH) were examined together as sharing a common pathophysiology. In addition, all experiments were performed within a cross-disciplinary, multi- institutional and international consortium including samples and expert representations from North

182 America and Sub-Saharan Africa. Furthermore, not only did we examine different anatomical regions of the brain including the VZ, SVZ and PVWM, as well as CSF physiology, we defined

PIH and PHH with in-depth granularity by characterizing their pathophysiology at the gene, protein and cellular levels. We also developed and/or implemented several novel experimental approaches, including dMRI segmentation of the VZ/SVZ, DBSI, proteogeonomics and in vitro cell cultures to understand the pathophysiology and underlying mechanisms of PIH and PHH.

Finally, we developed animal models that mimic the human disease and our findings, and are amenable to the modulation of physiological processes to further understand the pathophysiology of PIH and PHH, and perhaps more importantly, for utilization by animal trials that can be transitioned into human clinical trials for PIH and PHH therapy.

6.1.2 A unifying pathophysiological mechanism for PIH and PHH

With a long-term goal of improving the care of infants affected by PIH and PHH, and identifying strategies to prevent both conditions, the objective of this dissertation was to define the common and unique pathways underlying the pathophysiology of PIH and PHH. With proteogenomics, we established that both conditions are associated with the differential expression of genes involved in neuroinflammation and brain-CSF barrier cell-cell junction structure in CSF. While the neuroinflammation was predominantly innate, there was overlap with the adaptive immune system through complement activation.

183 Assessment of the ventricular perimeter of human infants with dMRI confirmed that, in the setting of neuroinflammation, PIH and PHH are associated with microstructural disruptions of the neural stem cell rich VZ/SVZ regions. The most aberrant dMRI measures were found in the FOHP regions and were attributed to compounding mechanical injury caused by pressures exerted on those regions by the expanded ventricles. The neuroinflammation and VZ/SVZ pathology were confirmed with neurocytology in humans, ferrets and mice with PIH or PHH.

Utilizing in vitro cell cultures, we identified that the VZ/SVZ disruption was mediated by cleavage of adherens junctional proteins (e.g. N-cadherin). N-cadherin cleavage was found to be facilitated by activation of metalloproteases (e.g. ADAM10), which led to a reduction in the number of

VZ/SVZ cells available for differentiation into ependymal cells and neuroglia. In addition, differentiation of the remaining cells was also impaired, culminating in a disrupted ependymal barrier. Disruption of the VZ/SVZ and ependymal barrier was associated with injury to the subjacent PVWM, in which axonal injury, reduced axonal fibers, increased cellularity and edema, neuroinflammation, and myelin disruption of myelinated axons were observed in human and ferret dMRI and immunohistochemistry.

Together, these findings confirm our central hypothesis and provide verifiable evidence to posit the following regarding PIH and PHH: 1) the antecedent events of hemorrhage and infection and the hydrocephalus activate neuroinflammation cascades that, while physiologically necessary, inadvertently cause ependymal injury; 2) these events occur in the setting of alterations in gene- activated pathways of host immune-responses and also during a stage of active differentiation of

184 VZ/SVZ neural stem cells, making neuroinflammation-related VZ/SVZ disruption central to the pathogenesis of PIH and PHH; 3) VZ/SVZ disruption is mediated by ADAM10 cleavage of N- cadherin-based cell-cell junctions, leading to denudation of the ependymal barrier, which compromises the brain-CSF barrier critical for CSF homeostasis; 4) PVWM injury occurs when the brain-CSF barrier is damaged, which has been previously related to neurodevelopmental deficits in affected infants, and even later in adult life; 5) pharmacologic modulation of ADAM10 represents a viable therapeutic approach to mitigating the deleterious pathogenesis of PIH and

PHH. (Figure 54)

Figure 54: A proposed unifying pathophysiological mechanism for PIH and PHH The ventricular zone (VZ) and subventricular (SVZ) zones are the main neurogenic niche for brain development. During fetal development, neuroepithelial cells in the VZ/SVZ migrate to differentiate into cortical neurons, astroglia and ependymal cells, utilizing radial glial cells. During this critical period, a systemic infection or intraventricular hemorrhage (1) may elicit a host- immune response and activation of metalloproteases (e.g., ADAM10) (2), which in turn cleave adherens junction molecules (e.g., N-cadherin) (3), leading to disruption of the brain-CSF ependymal barrier (4). Together, the VZ/SVZ and ependymal disruption lead to impairment of CSF dynamics (5), and the juxtaposed periventricular white matter to injury from persistent neuroinflammation and CSF-laden toxins. Inhibition of ADAM10 abrogates the N-cadherin mediated VZ/SVZ disruption in cell cultures. (Modified after Rodriguez et. al.172 and Gao et. al.572).

185 6.1.3 Neuroinflammation is central to PIH and PHH pathophysiology

Neuroinflammation has long been associated with the pathogenesis of infection-related hydrocephalus. Earlier reports described inflammation-mediated granular ependymitis and scarring of CSF pathways as the cause of infectious hydrocephalus573-576. Similar mechanisms of host immune responses have been implicated in the pathogenesis of PHH following IVH242,577 and congenital hydrodrocephalus.145,207,242 Indeed, reactive astrocytosis has been demonstrated repeatedly in both congenital and acquired models of hydrocephalus, and has been further shown to increase with the progression of hydrocephalus. Additionally, there is evidence to suggest that in many forms of hydrocephalus, neuroinflammation has widespread deleterious effects on brain function, including disturbances in oxidative metabolism, neurotransmission, periventricular cell- cell junctional interaction, brain-CSF barriers, iron transport mechanisms, and choroid plexus function.84,145,169,182,197,212,318,336,382,578-580 Bearing in mind the effects of neuroinflammation across many variants of hydrocephalus, we elected to use non-hemorrhagic non-infectious hydrocephalic infants as controls in order to detect molecular mechanisms that were more specific to PIH and

PHH.

Neuroinflammation can be mediated through many different pathways. The TLR system, especially the TLR4 ligand for lipopolysaccharides (LPS), is perhaps the best understood. It involves LPS binding to the lymphocyte antigen 96 which then triggers dimerization of TLR4 to enhance cytokine and chemokine production downstream. TLR4 can also be activated by blood products.581 In a rat model, Karimy et al. demonstrated that TLR4-mediated activation of SPAK-

NKCC1 led to hypersecretion of CSF by the choroid plexus epithelium, leading to

186 hydrocephalus.145 While the TLR immune system pathway is commonly implicated in hydrocephalus,145,198,336,582 the JAK-STAT pathway is considered the most efficient form of innate immunity, especially to intracellular pathogens.201,202 In fact, protein mutations in the JAK-STAT pathway have been implicated in poor inflammation mediation.201,202 For example, impaired JAK function in severe combined immunodeficiency is associated with susceptibility to infections due to the absence of natural killer (NK) cells, B cells or T cells.583,584 In our PIH and PHH human cohorts, JAK-STAT-dependent processes predominated, likely because all the infants were less than 3 months of age and had not had the antigenic exposure required to mount a detectable humoral response.

The proteogenomics experiments validated our previous findings from immunometric assays confirming that activation of the immune system in PIH and PHH increases cytokine and chemokine production207. In addition, there was differential expression of products involved in the innate immune system-related metabolism such as iron depletion and generation of reactive oxidative species (ROS). During an acute infection585 or IVH586,587, there is a high rate of oxidative metabolism and response to ROS such as hydrogen peroxide, superoxide radicals and nitric oxide.

ROS activation typically triggers cascades of cellular excitotoxicity targeted toward the blood- brain barrier (BBB), which secondarily results in brain injury that impairs brain oxygenation, leading to cerebral vascular dysfunction and ischemia.588 The role of BBB disruption in CNS infections and IVH have been broadly studied. In sepsis, the BBB acts as a nexus for integrating signals of the brain and periphery and permits extravasation of microbes and infection-related mediators into the CNS (reviewed in 589-591). In IVH, BBB abnormalities identified as decreased pericyte laminin, which is critical for maintaining their tight junctions 592,593 and type IV collagen

187 that provide structural support for the capillaries 594 have been reported. However, it is unclear whether the BBB abnormalities in hemorrhage or infections are triggers of hydrocephalus as there are many inconsistencies in the literature on the role of BBB in hydrocephalus pathophysiology

39,595,596. Nevertheless, our finding of abundant expression of neutrophil-generated ROS in PIH and PHH infants raises important questions as to whether BBB disruption contributes to the sustained (chronic) inflammatory milieu in PIH and PHH even months after the initial infection or hemorrhage, when the CSF samples were collected.

6.1.4 Role of modulating neuroinflammation pathways to mitigate PIH and PHH

From CSF to neural stem cell organization, ependymal barrier structure and PVWM, across human, mice and ferret species, and at both the gene and protein levels, our results consistently supported host-immune response as being central to PIH and PHH pathophysiology. However, it would be premature to suggest that our findings support the general use of anti-inflammatory or immunosuppressive agents during neonatal sepsis or IVH to mitigate the risk of developing hydrocephalus. For one, it remains unclear whether neuroinflammation drives the development of

PIH and PHH or vice versa. Moreover, the time course and permanence of neuroinflammation- mediated brain injury have yet to be elucidated187. In experimental models, non-specific modulation of immune response has yielded variable results. For example, minocycline, a tetracycline antibiotic that has many undefined effects but has been shown to inhibit microglial cells has been explored. In the H-Tx rat model of congenital hydrocephalus, intraperitoneal administration of minocycline significantly reduced the numbers of astrocytes and microglia and increased cortical mantle thickness 94,96. It is important to note that minocycline has many side

188 effects including tooth discoloration in children and long-term severe osteoporosis; which may limit its translation to clinical use88.

The use of steroids remains a controversial topic in perinatal medicine, and in particular, in neonatal sepsis and IVH597-600. There have been many attempts to use corticosteroids in treating infants with acute bacterial meningitis, and despite a few notable exceptions such as Hemophilus influenza and pneumococcal meningitis, there have been many failures, some with devastating outcomes601-609. In experimental models, corticosteroids and glucocorticoids have been used for decades with variable benefit in hydrocephalus88. For example, intravenous administration of dexamethasone in kaolin-induced hydrocephalic dogs yielded a 40 % reduction in CSF flow.

However, the effect, was short-lived, lasting for approximately six hours. 610,611.

Neuroinflammation tends to involve several biological pathways, each of which encompasses scores of mediators that are also involved in other related processes. As such, while it may not be advisable to broadly modulate upstream ligands of neuroinflammation such as TLRs or JAK receptors, specific mediators (e.g., TGF-β1) may be targeted to minimize inadvertent effects on the patient’s physiology. A few decades ago, it was reported that compared to controls, the CSF of preterm IVH infants expressed relatively higher levels of TGF-β1, which is released from platelets at sites of blood clotting to regulate fibroblasts proliferation612. This line of work prompted several experimental studies to explore specific targeting of TGF-β1 as a potential therapy for hydrocephalus. In experimental models, transgenic mice with TGF-β1 overexpression

613,614, and wild type mice who were injected with TGF-β1 into the subarachnoid compartment

189 both had hydrocephalus615,616. Perhaps more importantly, the induced hydrocephalus mice could be remediated with intraventricular infusion TGF-β1 antagonists617. In addition, Botfield et al. showed that in kaolin-induced hydrocephalic rats with, treatment with decorin, a TGF-β1 binding proteoglycan, at the time of Kaolin injection was associated with reduced inflammation and fibrosis in the subarachnoid compartment and prevention of hydrocephalus 89. Other studies have also shown that decorin has the therapeutic potential to decrease ventricle size and PVWM microstructural damage, as well as cognitive outcome in rats90,92. While the experimental work on

TGF-β1 modularion is exciting, none of these studies have translated to humans. Another good example of specific targeting is the recent work of Karimy et al145. In a rat model of PHH, Karimy et al showed that IVH causes a TLR4 and NF-κB dependent inflammatory response in choroid plexus epithelium, which was associated with an approximately three-fold increase in CSF secretion and hydrocephalus. It was found then that the SPAK gene, which binds, phosphorylates, and stimulates the NKCC1 co-transporter at the choroid plexus epithelium apical membrane was responsible for TLR4-dependent choroid plexus CSF hypersecretion. To that effect, genetic depletion of TLR4 or SPAK, as well as antagonism of TLR4-NF-κB signaling or the SPAK-

NKCC1 co-transporter complex all led to normalized CSF secretion rates145. Future studies are needed to explore these specific targets in humans.

Our human proteogenomic work does point to novel intervention pathways and supports that selective and targeted modulation of certain aspects of immune response pathways may be a more successful approach in the prevention of hydrocephalus.For example, in addition to pro- inflammatory immune factors, we found a differential expression of IL-4 and IL-13, which generally have anti-inflammatory effects. IL-4 and IL-13 can share a common receptor618-620 and

190 typically work synergistically to decrease inflammation by counteracting the activity of pro- inflammatory cytokines such as IL-12.621,622 IL-4 and IL-13 are also considered neuroprotective as they can induce death of microglial cells that mediate neuronal damage623-625. As a caution, Park and colleagues reported that, in experimental models, IL-4 and IL-13 potentiate oxidative stress- related injury to hippocampal neurons.626,627 Thus, one hypothesis is that the balance of IL-12 and

IL-4/IL-13 within the CSF contributes to the risk of PIH, suggesting that their targeted modulation may be a potential therapeutic target. Further studies are needed to examine the role of cytokine/chemokine modulators as potential adjunctive treatment targets to regulate the immune response during such infections and IVH in an effort to reduce the chance of developing hydrocephalus.

It is also critical emphasize that inflammation itself, at least in the acute phase of CNS injury, is a physiologically helpful process for appropriately responding to and clearing microbes and toxins.

Since there are no set thresholds, it is challenging to define an inappropriate inflammatory response and the point at which modulation would do more benefit than detriment. Time course and levels of inflammatory markers in biological samples could be helpful, but more research is needed to identify robust biomarkers and obtain normative data on appropriate levels of response.

6.1.5 Ependymal barrier damage is associated with PVWM injury

As we have consistently demonstrated in the setting of neuroinflammation, disruption of neural stem cells in the VZ/SVZ compromises their differentiation into ependymal cells. While this

191 dissertation focused on the ependyma due to its central role in hydrocephalus pathogenesis, we acknowledge that VZ/SVZ disruption also negatively impacts neurogliogenesis via both the radial and tangential migration streams572. With regards to ependymogenesis, the ependymal barrier, which is responsible for many functions including brain-CSF homeostasis and immune response232,233, is defective when the VZ/SVZ differentiation is disrupted, demonstrating regions of denudation and heterotopias. As a result, the protective barrier properties of the ependymal layer to shield the subjacent PVWM from injurious CSF toxins is compromised. With dMRI and immunohistochemistry, we demonstrated ependymal layer microstructural damage was intimately related to transependymal CSF migration, and PVWM dysmyelination and axonal loss across species.

While VZ/SVZ injury involves the majority of the LVP, we found segmental variations in the magnitude of injury. The FOHP regions tend to be more vulnerable than other regions, perhaps due to relatively higher pressures exerted on the FOHP by the expanded ventricles. Across the lateral ventricle there are regional differences in the cytoarchitecture of the VZ and in the organization of neural stem cells within the SVZ.133,197,463 While radial glial cells are sequentially replaced by ependymal cells along the LVP during fetal development, the lateral wall of the frontal horn is the only region that retains these stem cells into infancy463, which may be associated with adverse neurodevelopment when those specific regions are damaged by PHH. Our findings of more aberrant VZ/SVZ and ependyma in the FOHP than in the rest of the ventricle are consistent with the patterns of injury seen on conventional MRIs of hydrocephalus patients193-195,465, and reflect the disproportionate distribution of stress and strain on the FOHP region.466-468 Indeed, porohyperelastic finite element anatomical models have showed that, compared to the rest of the

192 lateral ventricle, the FOHP is subjected to increased stress and strain and a 3.5-times increased edema468. The disproportionate risk of injury at the FOHP has been attributed to the acute anterolateral angles at those regions, which subject them to high ICP- and hydrocephalus-related tensile forces466-468. In contrast to the convex shape of the body of the lateral ventricle, the concave geometry of the FOHP is associated with expanded extracellular spaces, which makes those regions more susceptible to free water accumulation and edema, leading to impairment of arterial blood flow, venous stasis, ischemia-induced PVWM injury, dysmyelination, axonal disruption.458,466-468,486,487 This segmental difference in VZ/SVZ injury is critical because it begs the question as to whether therapeutic strategies should be focused on localized regions at the highest risk of injury, and those most populated by neural stem cells that have the capacity to regenerate (the FOHP).

In our cohort, ventricular size negatively correlated with the magnitude of VZ/SVZ and PVWM injury, suggesting that the expansion of the ventricles likely has a negative effect on ependymal and PVWM tissue integrity, speaking to whether ventricular size can be used as a surrogate marker for disease severity in PIH and PHH. However, we must note that there is ongoing debate regarding ventricular size-PVWM injury correlations, as variable results have been reported in the literature.450,628,629 For example, Kulkarni et al. did not find any association between FRONTAL-

OCCIPITAL HORN RATIO and PVWM injury in 23 pediatric patients with treated, stable obstructive hydrocephalus629. Our findings are similar to those of Akbari et al.450 and Cauley et al.628, who showed significant positive correlations between FRONTAL-OCCIPITAL HORN

RATIO and dMRI measures. At the very least, we can conclude that our findings and some of the existing literature underscore the importance of evaluating the role of ventricular size – a readily

193 modifiable factor – on white matter integrity, brain development and neurodevelopmental disability associated with PIH and PHH.

6.1.6 VZ/SVZ differentiation modulation and ependymal barrier and PVWM repair

Given the many downsides of broadly interfering with acute neuroinflammation, the paradigm could probably be shifted towards mitigating chronic inflammation, which has an implicated role in the perpetuation of brain injury following IVH and CNS infections198. ADAM10 has been implicated in some chronic inflammatory diseases such as multiple sclerosis and osteoarthritis.630,631 With our cell culture model, we established that ADAM10 activation led to cleavage of N-cadherin, which impaired VZ/SVZ differentiation and ependyma cell-cell junction adhesion. During systemic infections, ADAM10 dysregulates BBB function through ectodomain shedding of barrier proteins such as LRP1, endothelial barrier damage and increased vascular permeability632,633, which likely perpetuates chronic inflammation. As such, there is a growing body of evidence involving ADAM10 therapy in the treatment of other conditions.526,633 For example, the -secretase activity of ADAM10 is presently being investigated as a potential therapeutic target for limiting Aβ production in Alzheimer’s disease633-635. Administration of

ADAM10 inhibitors have also been shown to be effective at protecting against lung and liver epithelia disruption during staphylococcal infection502,516. In our experiments, the inhibition of

ADAM10 enabled us to preserve VZ/SVZ cytoarchitecture in vitro in the setting of blood and bacterial toxins inoculation. Therefore, there is potential for ADAM10 as a modifiable, therapeutic target for preserving ependymal barrier cytoarchitecture, to limit progression to PIH and PHH following an infection or hemorrhage. From a patient management standpoint, ADAM10 cleavage

194 of N-cadherin yields soluble isoforms of N-cadherin,511 which can be detected in the CSF of human

PHH infants in addition to other ADAM10-related substrates such as L1CAM, NCAM-1, and

APP.245,247,636 Nevertheless, it must be noted that while the ADAM10/N-cadherin mechanisms we tested in this dissertation brings necessary focus, there may be other proteases or important pathways involved in the pathophysiology of PIH and PHH. In addition, the effect of hemorrhage or infection on choroid plexus physiology, as well as the secretory/absorptive capacity of the ependymal layer can be further explored.

In terms of PVWM, Del Bigio et al. demonstrated that in kaolin-induced hydrocephalic rats axonal damage was in part mediated by increased levels of proteolytic enzymes mediated by intracellular calcium637. This prompted modulation of calcium as therapy in experimental models of hydrocephalus. Although rats treated with the calcium channel blocker nimodipine for two weeks after hydrocephalus induction did not show a reduction in ventricular size, their corpus callosum was relatively thicker and they performed better on motor and cognitive behavior testing when compared to untreated hydrocephalic control rats97. In two-week-old ferrets, parenteral injections of magnesium sulfate, a calcium competitor, was associated with lethargy and there was no evidence for protection against the effects of hydrocephalus at the behavioral, structural, or biochemical level364. In addition, administration of magnesium sulfate, with or without nimodipine was associated with substantial side effects including dysregulation of systemic arterial blood pressure, cerebral perfusion and intracerebral pulse pressure459. Nevertheless, it is worth mentioning that there are several experimental therapies geared towards PVWM repair and recovery of neurological function. For example, in rat models of PHH, Ballabh and colleagues have explored therapies related to epidermal growth factor638, AMPA-Kainate Receptor

195 inhibitors540, hyaluronidase and hyaluronan oligosaccharides639, thyroxine640, bone morphogenetic protein641 and cox-2 inhibitors642. On the horizon, there are also other therapeutic strategies for hydrocephalus that are focused on reparative or regenerative processes of already disrupted ependyma. For example, Rodriguez and colleagues have explored neurosphere transplantation therapy for the regeneration of denuded ependyma. Unilateral intraventricular implantation of neurospheres obtained from normal HTx rats helped to repair VZ disruption in hydrocephalic littermates643. McAllister and colleagues have also utilized Decorin, a proteoglycan capable of inhibiting fibrosis in the subarachnoid space, to prevent the development of ventriculomegaly and reduce reactive astrogliosis and inflammation in congenital hydrocephalus models89,90. While the current non-surgical therapies for hydrocephalus are experimental, they provide a glimmer of hope that the progression to PIH and PHH following infection or hemorrhage can be mitigated, the outcomes of affected patients can be improved, and that PIH and PHH can eventually be prevented by pursuing robust scientific discovery.

196 7 References

1 Rekate, H. L. A contemporary definition and classification of hydrocephalus. Semin Pediatr Neurol 16, 9-15, doi:10.1016/j.spen.2009.01.002 (2009). 2 Kahle, K. T., Kulkarni, A. V., Limbrick, D. D., Jr. & Warf, B. C. Hydrocephalus in children. Lancet 387, 788-799, doi:10.1016/S0140-6736(15)60694-8 (2016). 3 Aronyk, K. E. The history and classification of hydrocephalus. Neurosurg Clin N Am 4, 599-609 (1993). 4 Mori, K. in Hydrocephalus: pathogenesis and treatment (eds S. Matsumoto & N. Tamaki) 362-368 (Springer Verlag, 1991). 5 Raimondi, A. J. A unifying theory for the definition and classification of hydrocephalus. Childs Nerv Syst 10, 2-12, doi:10.1007/bf00313578 (1994). 6 Rekate, H. L. A consensus on the classification of hydrocephalus: its utility in the assessment of abnormalities of cerebrospinal fluid dynamics. Childs Nerv Syst 27, 1535- 1541, doi:10.1007/s00381-011-1558-y (2011). 7 Tully, H. M. & Dobyns, W. B. Infantile hydrocephalus: a review of epidemiology, classification and causes. Eur J Med Genet 57, 359-368, doi:10.1016/j.ejmg.2014.06.002 (2014). 8 Dandy, W. E. & Blackfan, K. D. An experimental and clinical study of internal hydrocephalus. Journal of the American Medical Association 61, 2216-2217, doi:DOI 10.1001/jama.1913.04350260014006 (1913). 9 Williams, M. A. et al. The clinical spectrum of hydrocephalus in adults: report of the first 517 patients of the Adult Hydrocephalus Clinical Research Network registry. J Neurosurg, 1-12, doi:10.3171/2019.2.JNS183538 (2019). 10 Volpe, J. J. Intraventricular hemorrhage and brain injury in the premature infant. Diagnosis, prognosis, and prevention. Clin Perinatol 16, 387-411 (1989). 11 Chu, S. M. et al. Neurological complications after neonatal bacteremia: the clinical characteristics, risk factors, and outcomes. PLoS One 9, e105294, doi:10.1371/journal.pone.0105294 (2014). 12 Li, L. et al. Association of bacteria with hydrocephalus in Ugandan infants. J Neurosurg Pediatr 7, 73-87, doi:10.3171/2010.9.PEDS10162 (2011). 13 van der Knaap, M. S. & Valk, J. Classification of congenital abnormalities of the CNS. AJNR Am J Neuroradiol 9, 315-326 (1988). 14 Muniz-Talavera, H. & Schmidt, J. V. The mouse Jhy gene regulates ependymal cell differentiation and ciliogenesis. PLoS One 12, e0184957, doi:10.1371/journal.pone.0184957 (2017). 15 Zhang, J., Williams, M. A. & Rigamonti, D. Genetics of human hydrocephalus. J Neurol 253, 1255-1266, doi:10.1007/s00415-006-0245-5 (2006). 16 Bamforth, S. J. & Baird, P. A. Spina bifida and hydrocephalus: a population study over a 35-year period. Am J Hum Genet 44, 225-232 (1989). 17 Marchbanks, R. J. Spina Bifida and Congenital Hydrocephalus: The U.K. Incidence and Number of Shunt Insertions. Z Kinderchir 44, 13-15 (1989).

197 18 Williams, H. A unifying hypothesis for hydrocephalus, Chiari malformation, syringomyelia, anencephaly and spina bifida. Cerebrospinal Fluid Res 5, 7, doi:10.1186/1743-8454-5-7 (2008). 19 Wellons, J. C., 3rd et al. The assessment of bulging fontanel and splitting of sutures in premature infants: an interrater reliability study by the Hydrocephalus Clinical Research Network. J Neurosurg Pediatr 11, 12-14, doi:10.3171/2012.10.PEDS12329 (2013). 20 O'Hayon, B. B., Drake, J. M., Ossip, M. G., Tuli, S. & Clarke, M. Frontal and occipital horn ratio: A linear estimate of ventricular size for multiple imaging modalities in pediatric hydrocephalus. Pediatr Neurosurg 29, 245-249, doi:10.1159/000028730 (1998). 21 Brenner, D. J. & Hall, E. J. Computed tomography--an increasing source of radiation exposure. N Engl J Med 357, 2277-2284, doi:10.1056/NEJMra072149 (2007). 22 Malviya, S. et al. Sedation and general anaesthesia in children undergoing MRI and CT: adverse events and outcomes. Br J Anaesth 84, 743-748, doi:10.1093/oxfordjournals.bja.a013586 (2000). 23 DiMaggio, C., Sun, L. S. & Li, G. Early childhood exposure to anesthesia and risk of developmental and behavioral disorders in a sibling birth cohort. Anesth Analg 113, 1143-1151, doi:10.1213/ANE.0b013e3182147f42 (2011). 24 Administration, U. S. F. D. FDA approves label changes for use of general anesthetic and sedation drugs in young children, (2016). 25 Ashley, W. W., Jr. et al. Use of rapid-sequence magnetic resonance imaging for evaluation of hydrocephalus in children. J Neurosurg 103, 124-130, doi:10.3171/ped.2005.103.2.0124 (2005). 26 Christy, A., Murchison, C. & Wilson, J. L. Quick Brain Magnetic Resonance Imaging With Diffusion-Weighted Imaging as a First Imaging Modality in Pediatric Stroke. Pediatr Neurol 78, 55-60, doi:10.1016/j.pediatrneurol.2017.09.020 (2018). 27 Dincer, A. & Ozek, M. M. Radiologic evaluation of pediatric hydrocephalus. Childs Nerv Syst 27, 1543-1562, doi:10.1007/s00381-011-1559-x (2011). 28 Warf, B. et al. Neurocognitive outcome and ventricular volume in children with myelomeningocele treated for hydrocephalus in Uganda. J Neurosurg Pediatr 4, 564- 570, doi:10.3171/2009.7.PEDS09136 (2009). 29 Martin-Laez, R., Caballero-Arzapalo, H., Lopez-Menendez, L. A., Arango-Lasprilla, J. C. & Vazquez-Barquero, A. Epidemiology of Idiopathic Normal Pressure Hydrocephalus: A Systematic Review of the Literature. World Neurosurg 84, 2002-2009, doi:10.1016/j.wneu.2015.07.005 (2015). 30 Jaraj, D. et al. Prevalence of idiopathic normal-pressure hydrocephalus. Neurology 82, 1449-1454, doi:10.1212/WNL.0000000000000342 (2014). 31 Hamilton, M. G. Treatment of hydrocephalus in adults. Semin Pediatr Neurol 16, 34-41, doi:10.1016/j.spen.2009.02.001 (2009). 32 Isaacs, A. M. et al. Age-specific global epidemiology of hydrocephalus: Systematic review, metanalysis and global birth surveillance. PLoS One 13, e0204926, doi:10.1371/journal.pone.0204926 (2018). 33 Research, I. C. f. B. D. S. a. Annual Report, (2017). 34 Dewan, M. C. et al. Global hydrocephalus epidemiology and incidence: systematic review and meta-analysis. J Neurosurg, 1-15, doi:10.3171/2017.10.JNS17439 (2018).

198 35 Warf, B. C., Dagi, A. R., Kaaya, B. N. & Schiff, S. J. Five-year survival and outcome of treatment for postinfectious hydrocephalus in Ugandan infants. J Neurosurg Pediatr 8, 502-508, doi:10.3171/2011.8.PEDS11221 (2011). 36 Ciurea, A. V., Coman, T. C. & Mircea, D. in Pediatric Hydrocephalus Ch. Chapter 15, 201-218 (Springer, 2005). 37 Warf, B. C. & East African Neurosurgical Research, C. Pediatric hydrocephalus in East Africa: prevalence, causes, treatments, and strategies for the future. World Neurosurg 73, 296-300, doi:10.1016/j.wneu.2010.02.009 (2010). 38 Muir, R. T., Wang, S. & Warf, B. C. Global surgery for pediatric hydrocephalus in the developing world: a review of the history, challenges, and future directions. Neurosurg Focus 41, E11, doi:10.3171/2016.7.FOCUS16273 (2016). 39 McAllister, J. P., 2nd & Chovan, P. Neonatal hydrocephalus. Mechanisms and consequences. Neurosurg Clin N Am 9, 73-93 (1998). 40 Christian, E. A. et al. Trends in hospitalization of preterm infants with intraventricular hemorrhage and hydrocephalus in the United States, 2000-2010. J Neurosurg Pediatr 17, 260-269, doi:10.3171/2015.7.PEDS15140 (2016). 41 Limbrick, D. D., Jr. et al. Neurosurgical treatment of progressive posthemorrhagic ventricular dilation in preterm infants: a 10-year single-institution study. J Neurosurg Pediatr 6, 224-230, doi:10.3171/2010.5.PEDS1010 (2010). 42 Adams-Chapman, I., Hansen, N. I., Stoll, B. J., Higgins, R. & Network, N. R. Neurodevelopmental outcome of extremely low birth weight infants with posthemorrhagic hydrocephalus requiring shunt insertion. Pediatrics 121, e1167-1177, doi:10.1542/peds.2007-0423 (2008). 43 McCrea, H. J. & Ment, L. R. The diagnosis, management, and postnatal prevention of intraventricular hemorrhage in the preterm neonate. Clin Perinatol 35, 777-792, vii, doi:10.1016/j.clp.2008.07.014 (2008). 44 Stoll, B. J. et al. Trends in Care Practices, Morbidity, and Mortality of Extremely Preterm Neonates, 1993-2012. JAMA 314, 1039-1051, doi:10.1001/jama.2015.10244 (2015). 45 Murphy, B. P. et al. Posthaemorrhagic ventricular dilatation in the premature infant: natural history and predictors of outcome. Arch Dis Child Fetal Neonatal Ed 87, F37-41, doi:10.1136/fn.87.1.f37 (2002). 46 Vassilyadi, M., Tataryn, Z., Shamji, M. F. & Ventureyra, E. C. Functional outcomes among premature infants with intraventricular hemorrhage. Pediatr Neurosurg 45, 247- 255, doi:10.1159/000228982 (2009). 47 Warf, B. C. et al. Costs and benefits of neurosurgical intervention for infant hydrocephalus in sub-Saharan Africa. J Neurosurg Pediatr 8, 509-521, doi:10.3171/2011.8.PEDS11163 (2011). 48 Simon, T. D. et al. Hospital care for children with hydrocephalus in the United States: utilization, charges, comorbidities, and deaths. J Neurosurg Pediatr 1, 131-137, doi:10.3171/PED/2008/1/2/131 (2008). 49 John, C. C. et al. Global research priorities for infections that affect the nervous system. Nature 527, S178-186, doi:10.1038/nature16033 (2015). 50 Cortese, F. et al. Early and Late Infections in Newborns: Where Do We Stand? A Review. Pediatr Neonatol 57, 265-273, doi:10.1016/j.pedneo.2015.09.007 (2016). 51 Kulkarni, A. V. et al. Endoscopic Treatment versus Shunting for Infant Hydrocephalus in Uganda. N Engl J Med 377, 2456-2464, doi:10.1056/NEJMoa1707568 (2017).

199 52 Kiwanuka, J. et al. The microbial spectrum of neonatal sepsis in Uganda: recovery of culturable bacteria in mother-infant pairs. PLoS One 8, e72775, doi:10.1371/journal.pone.0072775 (2013). 53 Schiff, S. J., Ranjeva, S. L., Sauer, T. D. & Warf, B. C. Rainfall drives hydrocephalus in East Africa. J Neurosurg Pediatr 10, 161-167, doi:10.3171/2012.5.PEDS11557 (2012). 54 Aziz, I. A. Hydrocephalus in the Sudan. J R Coll Surg Edinb 21, 222-224 (1976). 55 Handler, L. C. & Wright, M. G. Postmeningitic hydrocephalus in infancy. Ventriculography with special reference to ventricular septa. Neuroradiology 16, 31-35, doi:10.1007/978-3-642-66959-0_11 (1978). 56 Schiff, S. J. et al. Separating Putative Pathogens from Background Contamination with Principal Orthogonal Decomposition: Evidence for Leptospira in the Ugandan Neonatal Septisome. Front Med (Lausanne) 3, 22, doi:10.3389/fmed.2016.00022 (2016). 57 Inder, T. E., Perlman, J. M. & Volpe, J. J. in Volpe's Neurology of the Newborn (ed Joseph J. Volpe) 637-698.e621 (Elsevier Inc., 2018). 58 Mukerji, A., Shah, V. & Shah, P. S. Periventricular/Intraventricular Hemorrhage and Neurodevelopmental Outcomes: A Meta-analysis. Pediatrics 136, 1132-1143, doi:10.1542/peds.2015-0944 (2015). 59 Papile, L. A., Burstein, J., Burstein, R. & Koffler, H. Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birth weights less than 1,500 gm. J Pediatr 92, 529-534, doi:10.1016/s0022-3476(78)80282-0 (1978). 60 Ancel, P. Y. et al. Cerebral palsy among very preterm children in relation to gestational age and neonatal ultrasound abnormalities: the EPIPAGE cohort study. Pediatrics 117, 828-835, doi:10.1542/peds.2005-0091 (2006). 61 Sherlock, R. L., Anderson, P. J., Doyle, L. W. & Victorian Infant Collaborative Study, G. Neurodevelopmental sequelae of intraventricular haemorrhage at 8 years of age in a regional cohort of ELBW/very preterm infants. Early Hum Dev 81, 909-916, doi:10.1016/j.earlhumdev.2005.07.007 (2005). 62 Klebermass-Schrehof, K. et al. Impact of low-grade intraventricular hemorrhage on long- term neurodevelopmental outcome in preterm infants. Childs Nerv Syst 28, 2085-2092, doi:10.1007/s00381-012-1897-3 (2012). 63 Smith, E. R., Butler, W. E. & Barker, F. G., 2nd. In-hospital mortality rates after ventriculoperitoneal shunt procedures in the United States, 1998 to 2000: relation to hospital and surgeon volume of care. J Neurosurg 100, 90-97, doi:10.3171/ped.2004.100.2.0090 (2004). 64 Jaraj, D. et al. Mortality and risk of dementia in normal-pressure hydrocephalus: A population study. Alzheimers Dement 13, 850-857, doi:10.1016/j.jalz.2017.01.013 (2017). 65 Drake, J. M. & Kulkarni, A. V. Cerebrospinal fluid shunt infections. Neurosurgery Quarterly 3, 283-294 (1993). 66 Kulkarni, A. V. et al. Endoscopic third ventriculostomy vs cerebrospinal fluid shunt in the treatment of hydrocephalus in children: a propensity score-adjusted analysis. Neurosurgery 67, 588-593, doi:10.1227/01.NEU.0000373199.79462.21 (2010). 67 Warf, B. C. & Campbell, J. W. Combined endoscopic third ventriculostomy and choroid plexus cauterization as primary treatment of hydrocephalus for infants with myelomeningocele: long-term results of a prospective intent-to-treat study in 115 East African infants. J Neurosurg Pediatr 2, 310-316, doi:10.3171/PED.2008.2.11.310 (2008).

200 68 Warf, B. C., Mugamba, J. & Kulkarni, A. V. Endoscopic third ventriculostomy in the treatment of childhood hydrocephalus in Uganda: report of a scoring system that predicts success. J Neurosurg Pediatr 5, 143-148, doi:10.3171/2009.9.PEDS09196 (2010). 69 Weninger, M. et al. External ventricular drainage for treatment of rapidly progressive posthemorrhagic hydrocephalus. Neurosurgery 31, 52-57; discussion 57-58, doi:10.1227/00006123-199207000-00008 (1992). 70 Kestle, J. Ventriculoperitoneal shunts in children: Indications, equipment and techniques. Pan Arab Journal of Neurosurgery 12, 26-32 (2008). 71 Kulkarni, A. V. et al. Outcomes of CSF shunting in children: comparison of Hydrocephalus Clinical Research Network cohort with historical controls: clinical article. J Neurosurg Pediatr 12, 334-338, doi:10.3171/2013.7.PEDS12637 (2013). 72 Gonzalez, L. F., Kim, L., Rekate, H. L., McDougall, C. G. & Albuquerque, F. C. Endovascular placement of a ventriculoatrial shunt. Technical note. J Neurosurg 106, 319-321, doi:10.3171/ped.2007.106.4.319 (2007). 73 Isaacs, A. M., Krahn, D., Walker, A. M., Hurdle, H. & Hamilton, M. G. Transesophageal Echocardiography-Guided Ventriculoatrial Shunt Insertion. Oper Neurosurg (Hagerstown), doi:10.1093/ons/opz353 (2019). 74 Vernet, O., Campiche, R. & de Tribolet, N. Long-term results after ventriculoatrial shunting in children. Childs Nerv Syst 9, 253-255, doi:10.1007/bf00306265 (1993). 75 Kunert, P. Pleural cavity as an alternative for cerebrospinal shunting in hydrocephalic treatment. Neurol Neurochir.Pol. 31, 1065-1067 (1997). 76 Aldana, P. R., James, H. E. & Postlethwait, R. A. Ventriculogallbladder shunts in pediatric patients. J Neurosurg Pediatr 1, 284-287, doi:10.3171/PED/2008/1/5/284 (2008). 77 Drake, J. M. & Sainte-Rose, C. The shunt book. (Blackwell Science, 1995). 78 Drake, J. M., Kulkarni, A. V. & Kestle, J. Endoscopic third ventriculostomy versus ventriculoperitoneal shunt in pediatric patients: a decision analysis. Childs Nerv Syst 25, 467-472, doi:10.1007/s00381-008-0761-y (2009). 79 Kulkarni, A. V. et al. Endoscopic third ventriculostomy in children: prospective, multicenter results from the Hydrocephalus Clinical Research Network. J Neurosurg Pediatr 18, 423-429, doi:10.3171/2016.4.PEDS163 (2016). 80 Kulkarni, A. V. et al. Endoscopic third ventriculostomy and choroid plexus cauterization in infant hydrocephalus: a prospective study by the Hydrocephalus Clinical Research Network. J Neurosurg Pediatr 21, 214-223, doi:10.3171/2017.8.PEDS17217 (2018). 81 Warf, B. C. Comparison of endoscopic third ventriculostomy alone and combined with choroid plexus cauterization in infants younger than 1 year of age: a prospective study in 550 African children. J Neurosurg 103, 475-481, doi:10.3171/ped.2005.103.6.0475 (2005). 82 Warf, B. C. Endoscopic third ventriculostomy and choroid plexus cauterization for pediatric hydrocephalus. Clin Neurosurg 54, 78-82 (2007). 83 Kestle, J. R. et al. Lack of benefit of endoscopic ventriculoperitoneal shunt insertion: a multicenter randomized trial. J Neurosurg 98, 284-290, doi:10.3171/jns.2003.98.2.0284 (2003). 84 Garton, T. et al. Intraventricular Hemorrhage: the Role of Blood Components in Secondary Injury and Hydrocephalus. Transl Stroke Res 7, 447-451, doi:10.1007/s12975- 016-0480-8 (2016).

201 85 Strahle, J. et al. Mechanisms of hydrocephalus after neonatal and adult intraventricular hemorrhage. Transl Stroke Res 3, 25-38, doi:10.1007/s12975-012-0182-9 (2012). 86 Koschnitzky, J. E. et al. Opportunities in posthemorrhagic hydrocephalus research: outcomes of the Hydrocephalus Association Posthemorrhagic Hydrocephalus Workshop. Fluids Barriers CNS 15, 11, doi:10.1186/s12987-018-0096-3 (2018). 87 McAllister, J. P., 2nd et al. An update on research priorities in hydrocephalus: overview of the third National Institutes of Health-sponsored symposium "Opportunities for Hydrocephalus Research: Pathways to Better Outcomes". J Neurosurg 123, 1427-1438, doi:10.3171/2014.12.JNS132352 (2015). 88 Del Bigio, M. R. & Di Curzio, D. L. Nonsurgical therapy for hydrocephalus: a comprehensive and critical review. Fluids Barriers CNS 13, 3, doi:10.1186/s12987-016- 0025-2 (2016). 89 Botfield, H. et al. Decorin prevents the development of juvenile communicating hydrocephalus. Brain 136, 2842-2858, doi:10.1093/brain/awt203 (2013). 90 Aojula, A. et al. Diffusion tensor imaging with direct cytopathological validation: characterisation of decorin treatment in experimental juvenile communicating hydrocephalus. Fluids Barriers CNS 13, 9, doi:10.1186/s12987-016-0033-2 (2016). 91 Hoque, N., Thoresen, M., Aquilina, K., Hogan, S. & Whitelaw, A. Decorin and colchicine as potential treatments for post-haemorrhagic ventricular dilatation in a neonatal rat model. Neonatology 100, 271-276, doi:10.1159/000327842 (2011). 92 Yan, H. et al. Decorin alleviated chronic hydrocephalus via inhibiting TGF- beta1/Smad/CTGF pathway after subarachnoid hemorrhage in rats. Brain research 1630, 241-253, doi:10.1016/j.brainres.2015.11.004 (2016). 93 Khan, O. H., Enno, T. & Del Bigio, M. R. Magnesium sulfate therapy is of mild benefit to young rats with kaolin-induced hydrocephalus. Pediatr Res 53, 970-976, doi:10.1203/01.PDR.0000061561.42921.5B (2003). 94 McAllister, J. P., 2nd & Miller, J. M. Minocycline inhibits glial proliferation in the H-Tx rat model of congenital hydrocephalus. Cerebrospinal Fluid Res 7, 7, doi:10.1186/1743- 8454-7-7 (2010). 95 Miller, J. M. & McAllister, J. P., II. in Hydrocephalus2006. 96 Xu, H. et al. Minocycline reduces reactive gliosis in the rat model of hydrocephalus. BMC neuroscience 13, 148, doi:10.1186/1471-2202-13-148 (2012). 97 Del Bigio, M. R. & Massicotte, E. M. Protective effect of nimodipine on behavior and white matter of rats with hydrocephalus. J Neurosurg 94, 788-794, doi:10.3171/jns.2001.94.5.0788 (2001). 98 Khan, O. H., McPhee, L. C., Moddemann, L. N. & Del Bigio, M. R. Calcium antagonism in neonatal rats with kaolin-induced hydrocephalus. J Child Neurol 22, 1161-1166, doi:10.1177/0883073807306259 (2007). 99 Kestle, J. R. et al. A new Hydrocephalus Clinical Research Network protocol to reduce cerebrospinal fluid shunt infection. J Neurosurg Pediatr 17, 391-396, doi:10.3171/2015.8.PEDS15253 (2016). 100 Drake, J. M., Kestle, J. R. & Tuli, S. Cerebrospinal fluid shunt technology. Clin Neurosurg 47, 336-345 (2000). 101 Drake, J. M., Kestle, J. R. & Tuli, S. CSF shunts 50 years on--past, present and future. Childs Nerv Syst 16, 800-804, doi:10.1007/s003810000351 (2000).

202 102 Williams, M. A. et al. Priorities for hydrocephalus research: report from a National Institutes of Health-sponsored workshop. J Neurosurg 107, 345-357, doi:10.3171/PED- 07/11/345 (2007). 103 He, M., Ouyang, L., Wang, S., Zheng, M. & Liu, A. Laparoscopy versus mini- laparotomy peritoneal catheter insertion of ventriculoperitoneal shunts: a systematic review and meta-analysis. Neurosurg Focus 41, E7, doi:10.3171/2016.5.FOCUS1637 (2016). 104 Sosin, M. et al. Laparoscopic-assisted Peritoneal Shunt Insertion for Ventriculoperitoneal and Lumboperitoneal Shunt Placement: An Institutional Experience of 53 Consecutive Cases. Surg Laparosc Endosc Percutan Tech 25, 235-237, doi:10.1097/SLE.0000000000000141 (2015). 105 Gilard, V. et al. Comparison of electromagnetic neuronavigation system and free-hand method for ventricular catheter placement in internal shunt. Clin Neurol Neurosurg 158, 93-97, doi:10.1016/j.clineuro.2017.05.007 (2017). 106 Xu, L. W., Sussman, E. S. & Li, G. Frameless, electromagnetic image-guided ventriculostomy for ventriculoperitoneal shunt and Ommaya reservoir placement. Clin Neurol Neurosurg 147, 46-52, doi:10.1016/j.clineuro.2016.05.024 (2016). 107 Zanaty, M. et al. The Wishbone: A Cranial Midline Localizing Device. World Neurosurg 128, 600-605 e601, doi:10.1016/j.wneu.2019.05.046 (2019). 108 Carpenter, E. M. in The Choroid Plexus and Cerebrospinal Fluid 15-27 (2016). 109 Smith, J. L. & Schoenwolf, G. C. Neurulation: coming to closure. Trends Neurosci 20, 510-517, doi:10.1016/s0166-2236(97)01121-1 (1997). 110 Loeser, J. D. & Alvord, E. C., Jr. Agenesis of the corpus callosum. Brain 91, 553-570, doi:10.1093/brain/91.3.553 (1968). 111 Sarwar, M. The septum pellucidum: normal and abnormal. AJNR Am J Neuroradiol 10, 989-1005 (1989). 112 Loeser, J. D. & Alvord, E. C., Jr. Clinicopathological correlations in agenesis of the corpus callosum. Neurology 18, 745-756, doi:10.1212/wnl.18.8.745 (1968). 113 Nagra, G. et al. Elevated CSF outflow resistance associated with impaired lymphatic CSF absorption in a rat model of kaolin-induced communicating hydrocephalus. Cerebrospinal Fluid Res 7, 4, doi:10.1186/1743-8454-7-4 (2010). 114 Boulton, M. et al. Raised intracranial pressure increases CSF drainage through arachnoid villi and extracranial lymphatics. Am J Physiol-Reg I 275, R889-R896 (1998). 115 Johnston, M. The importance of lymphatics in cerebrospinal fluid transport. Lymphatic research and biology 1, 41-44; discussion 45, doi:10.1089/15396850360495682 (2003). 116 Johnston, M., Zakharov, A., Papaiconomou, C., Salmasi, G. & Armstrong, D. Evidence of connections between cerebrospinal fluid and nasal lymphatic vessels in humans, non- human primates and other mammalian species. Cerebrospinal Fluid Research 1, 2 (2004). 117 Hladky, S. B. & Barrand, M. A. Mechanisms of fluid movement into, through and out of the brain: evaluation of the evidence. Fluids Barriers CNS 11, 26, doi:10.1186/2045- 8118-11-26 (2014). 118 Iliff, J. J. et al. Brain-wide pathway for waste clearance captured by contrast-enhanced MRI. The Journal of clinical investigation 123, 1299-1309, doi:10.1172/JCI67677 (2013).

203 119 Yang, L. et al. Evaluating glymphatic pathway function utilizing clinically relevant intrathecal infusion of CSF tracer. Journal of translational medicine 11, 107, doi:10.1186/1479-5876-11-107 (2013). 120 Iliff, J. J. et al. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci 33, 18190-18199, doi:10.1523/JNEUROSCI.1592-13.2013 (2013). 121 Benveniste, H., Heerdt, P. M., Fontes, M., Rothman, D. L. & Volkow, N. D. Glymphatic System Function in Relation to Anesthesia and Sleep States. Anesth Analg 128, 747-758, doi:10.1213/ANE.0000000000004069 (2019). 122 Del Bigio, M. R. The ependyma: a protective barrier between brain and cerebrospinal fluid. Glia 14, 1-13, doi:10.1002/glia.440140102 (1995). 123 Kriegstein, A. & Alvarez-Buylla, A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci 32, 149-184, doi:10.1146/annurev.neuro.051508.135600 (2009). 124 Bayatti, N. et al. A molecular neuroanatomical study of the developing human neocortex from 8 to 17 postconceptional weeks revealing the early differentiation of the subplate and subventricular zone. Cereb Cortex 18, 1536-1548, doi:10.1093/cercor/bhm184 (2008). 125 Bystron, I., Blakemore, C. & Rakic, P. Development of the human cerebral cortex: Boulder Committee revisited. Nat Rev Neurosci 9, 110-122, doi:10.1038/nrn2252 (2008). 126 De Juan Romero, C. & Borrell, V. Coevolution of radial glial cells and the cerebral cortex. Glia 63, 1303-1319, doi:10.1002/glia.22827 (2015). 127 Rowitch, D. H. & Kriegstein, A. R. Developmental genetics of vertebrate glial-cell specification. Nature 468, 214-222, doi:10.1038/nature09611 (2010). 128 Molofsky, A. V. et al. Astrocytes and disease: a neurodevelopmental perspective. Genes & development 26, 891-907, doi:10.1101/gad.188326.112 (2012). 129 Wang, C. et al. Identification and characterization of neuroblasts in the subventricular zone and rostral migratory stream of the adult human brain. Cell Res 21, 1534-1550, doi:10.1038/cr.2011.83 (2011). 130 Sanai, N. et al. Corridors of migrating neurons in the human brain and their decline during infancy. Nature 478, 382-+, doi:10.1038/nature10487 (2011). 131 Del Bigio, M. R., da Silva, M. C., Drake, J. M. & Tuor, U. I. Acute and chronic cerebral white matter damage in neonatal hydrocephalus. Can J Neurol Sci 21, 299-305, doi:10.1017/s0317167100040865 (1994). 132 Sohl, G., Odermatt, B., Maxeiner, S., Degen, J. & Willecke, K. New insights into the expression and function of neural connexins with transgenic mouse mutants. Brain research. Brain research reviews 47, 245-259, doi:10.1016/j.brainresrev.2004.05.006 (2004). 133 Jimenez, A. J., Dominguez-Pinos, M. D., Guerra, M. M., Fernandez-Llebrez, P. & Perez- Figares, J. M. Structure and function of the ependymal barrier and diseases associated with ependyma disruption. Tissue Barriers 2, e28426, doi:10.4161/tisb.28426 (2014). 134 Lun, M. P., Monuki, E. S. & Lehtinen, M. K. Development and functions of the choroid plexus-cerebrospinal fluid system. Nature Reviews Neuroscience 16, 445-457, doi:10.1038/nrn3921 (2015).

204 135 Lun, M. P. et al. Spatially heterogeneous choroid plexus transcriptomes encode positional identity and contribute to regional CSF production. J Neurosci 35, 4903-4916, doi:10.1523/JNEUROSCI.3081-14.2015 (2015). 136 Lehtinen, M. K. & Walsh, C. A. in Annual review of cell and developmental biology Vol. 27 653-679 (2011). 137 Shechter, R. et al. Recruitment of beneficial M2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus. Immunity 38, 555-569, doi:10.1016/j.immuni.2013.02.012 (2013). 138 Doetsch, F., Caille, I., Lim, D. A., Garcia-Verdugo, J. M. & Alvarez-Buylla, A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell 97, 703-716, doi:10.1016/s0092-8674(00)80783-7 (1999). 139 Redzic, Z. B., Preston, J. E., Duncan, J. A., Chodobski, A. & Szmydynger-Chodobska, J. in Current Topics in Developmental Biology Vol. 71 1-52 (2005). 140 Hébert, J. M., Mishina, Y. & McConnell, S. K. BMP Signaling Is Required Locally to Pattern the Dorsal Telencephalic Midline. Neuron 35, 1029-1041, doi:10.1016/s0896- 6273(02)00900-5 (2002). 141 Redzic, Z. Molecular biology of the blood-brain and the blood-cerebrospinal fluid barriers: similarities and differences. Fluids Barriers CNS 8, 3, doi:10.1186/2045-8118- 8-3 (2011). 142 Larroche, J. C. Post-haemorrhagic hydrocephalus in infancy. Anatomical study. Biol Neonate 20, 287-299, doi:10.1159/000240472 (1972). 143 Boop, F. A. in Pediatric Hydrocephalus (ed G. Cinalli) Ch. 8, 121-131 (Springer- Verlag, 2005). 144 Cardia, E. et al. Morphological modifications of the choroid plexus in a rodent model of acute ventriculitis induced by gram-negative liquoral sepsis. Possible implications in the pathophysiology of hypersecretory hydrocephalus. Childs Nerv Syst 11, 511-516, doi:10.1007/bf00822840 (1995). 145 Karimy, J. K. et al. Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalus. Nat Med 23, 997-1003, doi:10.1038/nm.4361 (2017). 146 Aziz, A. A., Coleman, L., Morokoff, A. & Maixner, W. Diffuse choroid plexus hyperplasia: an under-diagnosed cause of hydrocephalus in children? Pediatr Radiol 35, 815-818, doi:10.1007/s00247-005-1456-0 (2005). 147 Bettegowda, C. et al. Treatment of choroid plexus tumors: a 20-year single institutional experience. J Neurosurg Pediatr 10, 398-405, doi:10.3171/2012.8.PEDS12132 (2012). 148 Di, R. C., Di, T. G., Pettorossi, V. E. & Caldarelli, M. On the pathology of experimental hydrocephalus induced by artificial increase in endoventricular CSF pulse pressure. Childs Brain 5, 81-95 (1979). 149 Wagshul, M. E., Eide, P. K. & Madsen, J. R. The pulsating brain: A review of experimental and clinical studies of intracranial pulsatility. Fluids Barriers CNS 8, 5, doi:10.1186/2045-8118-8-5 (2011). 150 Rashid, S., Li, J., McAllister, J. P., II, Tian, G. W. & Wagshul, M. E. Microvascular pulsatility in the hydrocephalic rat neocortex by two-photon microscopy. Neuroscience Meeting Planner, Chicago, IL, Society for Neuroscience Program #406.5 (2009).

205 151 Wagshul, M. E. et al. Ventricular dilation and elevated aqueductal pulsations in a new experimental model of communicating hydrocephalus. Exp Neurol 218, 33-40, doi:10.1016/j.expneurol.2009.03.034 (2009). 152 Wagshul, M. E., Egnor, M. R., Kelly, E. J. & Yu, H. J. Resonance and intracranial pressure pulsations: An experimental study. Clin Neurol Neurosurg 110, S5 (2008). 153 Luciano, M. G. et al. Cerebrovascular adaptation in chronic hydrocephalus. J Cereb Blood Flow Metab 21, 285-294, doi:10.1097/00004647-200103000-00012 (2001). 154 Del Bigio, M. R. & Bruni, J. E. Changes in periventricular vasculature of rabbit brain following induction of hydrocephalus and after shunting. J Neurosurg 69, 115-120, doi:10.3171/jns.1988.69.1.0115 (1988). 155 Oka, N., Nakada, J., Endo, S. & Takaku, A. Angioarchitecture in experimental hydrocephalus. Pediatr Neurosci 12, 294-299 (1985). 156 Okuyama, T., Hashi, K., Sasaki, S., Sudo, K. & Kurokawa, Y. Changes in cerebral microvasculature in congenital hydrocephalus of the inbred rat LEW/Jms: light and electron microscopic examination. Surg Neurol 27, 338-342, doi:10.1016/0090- 3019(87)90008-5 (1987). 157 Castejon, O. J. Transmission electron microscope study of human hydrocephalic cerebral cortex. J Submicrosc Cytol Pathol 26, 29-39 (1994). 158 Nakagawa, Y., Cervos-Navarro, J. & Artigas, J. Tracer study on a paracellular route in experimental hydrocephalus. Acta Neuropathol 65, 247-254, doi:10.1007/bf00687004 (1985). 159 Sada, Y., Moriki, T., Kuwahara, S., Yamane, T. & Hara, H. Immunohistochemical study on blood-brain barrier in congenitally hydrocephalic HTX rat brain. Zentralblatt fur Pathologie 140, 289-298 (1994). 160 Momjian, S. et al. Pattern of white matter regional cerebral blood flow and autoregulation in normal pressure hydrocephalus. Brain 127, 965-972, doi:10.1093/brain/awh131 (2004). 161 Hanigan, W. C. & Bogner, D. Cerebrovascular physiology in perinates with congenital hydrocephalus. Childs Nerv Syst 26, 775-780, doi:10.1007/s00381-009-1075-4 (2010). 162 Shirane, R. et al. Cerebral blood flow and oxygen metabolism in infants with hydrocephalus. Childs Nerv Syst 8, 118-123, doi:10.1007/bf00298263 (1992). 163 Cosan, T. E. et al. Cerebral blood flow alterations in progressive communicating hydrocephalus: transcranial Doppler ultrasonography assessment in an experimental model. J Neurosurg 94, 265-269, doi:10.3171/jns.2001.94.2.0265 (2001). 164 Wozniak, M., McLone, D. G. & Raimondi, A. J. Micro- and macrovascular changes as the direct cause of parenchymal destruction in congenital murine hydrocephalus. Journal of Neurosurgery 43, 535-545 (1975). 165 Nakada, J., Oka, N., Nagahori, T., Endo, S. & Takaku, A. Changes in the cerebral vascular bed in experimental hydrocephalus: an angio-architectural and histological study. Acta Neurochir (Wien) 114, 43-50, doi:10.1007/bf01401113 (1992). 166 de la Torre, J. C. & Aliev, G. Inhibition of vascular nitric oxide after rat chronic brain hypoperfusion: spatial memory and immunocytochemical changes. J Cereb Blood Flow Metab 25, 663-672, doi:10.1038/sj.jcbfm.9600057 (2005). 167 Bilfinger, T. V. & Stefano, G. B. Human aortocoronary grafts and nitric oxide release: relationship to pulsatile pressure. Ann Thorac Surg 69, 480-485, doi:10.1016/s0003- 4975(99)01083-8 (2000).

206 168 Klinge, P. M. et al. Cerebral hypoperfusion and delayed hippocampal response after induction of adult kaolin hydrocephalus. Stroke 34, 193-199, doi:10.1161/01.str.0000048820.17198.15 (2003). 169 Guerra, M. et al. A cell junction pathology of neural stem cells Is associated to ventricular zone disruption, hydrocephalus and neurogenesis abnormalities. J Neuropath Exp Neur 74, 653-671 (2015). 170 Ortloff, A. R. et al. Role of the subcommissural organ in the pathogenesis of congenital hydrocephalus in the HTx rat. Cell and tissue research 352, 707-725, doi:10.1007/s00441-013-1615-9 (2013). 171 Roales-Bujan, R. et al. Astrocytes acquire morphological and functional characteristics of ependymal cells following disruption of ependyma in hydrocephalus. Acta Neuropathol 124, 531-546, doi:10.1007/s00401-012-0992-6 (2012). 172 Rodriguez, E. M. et al. A cell junction pathology of neural stem cells leads to abnormal neurogenesis and hydrocephalus. Biological research 45, 231-242, doi:10.4067/S0716- 97602012000300005 (2012). 173 Guerra, M. et al. in Cerebrospinal Fluid Res Vol. 7 S56 (2010). 174 Ferland, R. J. et al. Disruption of neural progenitors along the ventricular and subventricular zones in periventricular heterotopia. Hum Mol Genet 18, 497-516, doi:10.1093/hmg/ddn377 (2009). 175 Jimenez, A. J. et al. Disruption of the neurogenic niche in the subventricular zone of postnatal hydrocephalic hyh mice. J Neuropathol Exp Neurol 68, 1006-1020, doi:10.1097/NEN.0b013e3181b44a5a (2009). 176 Paez, P. et al. Patterned neuropathologic events occurring in hyh congenital hydrocephalic mutant mice. J Neuropathol Exp Neurol 66, 1082-1092, doi:10.1097/nen.0b013e31815c1952 (2007). 177 Batiz, F. et al. Clinical and neuropathological evolution of the hydrocephalus developed by the mutant mouse hyh. Cerebrospinal Fluid Research (2005). 178 Dominguez-Pinos, M. D. et al. Ependymal denudation and alterations of the subventricular zone occur in human fetuses with a moderate communicating hydrocephalus. J Neuropathol Exp Neurol 64, 595-604 (2005). 179 Acabchuk, R. L. et al. 3D Modeling of the Lateral Ventricles and Histological Characterization of Periventricular Tissue in Humans and Mouse. J Vis Exp, e52328, doi:10.3791/52328 (2015). 180 Shook, B. A. et al. Ventriculomegaly associated with ependymal gliosis and declines in barrier integrity in the aging human and mouse brain. Aging Cell 13, 340-350, doi:10.1111/acel.12184 (2014). 181 Halling, M. An Analysis of the Association Between Ependymal Integrity and Ventriculomegaly Masters thesis, University of Connecticut, (2013). 182 Ortega, E. et al. The value of early and comprehensive diagnoses in a human fetus with hydrocephalus and progressive obliteration of the aqueduct of Sylvius: Case Report. BMC Neurol 16, 45, doi:10.1186/s12883-016-0566-7 (2016). 183 Sival, D. A. et al. Neuroependymal denudation is in progress in full-term human foetal spina bifida aperta. Brain Pathol 21, 163-179, doi:10.1111/j.1750-3639.2010.00432.x (2011).

207 184 Di Curzio, D. L. Neuropathological Changes in Hydrocephalus—A Comprehensive Review. Open Journal of Modern Neurosurgery 08, 1-29, doi:10.4236/ojmn.2018.81001 (2018). 185 Del Bigio, M. R. Neuropathology and structural changes in hydrocephalus. Dev Disabil Res Rev 16, 16-22, doi:10.1002/ddrr.94 (2010). 186 Khan, O. H., Enno, T. L. & Del Bigio, M. R. Brain damage in neonatal rats following kaolin induction of hydrocephalus. Exp Neurol 200, 311-320, doi:10.1016/j.expneurol.2006.02.113 (2006). 187 Del Bigio, M. R. Cellular damage and prevention in childhood hydrocephalus. Brain Pathol 14, 317-324, doi:10.1111/j.1750-3639.2004.tb00071.x (2004). 188 Del Bigio, M. R., Wilson, M. J. & Enno, T. Chronic hydrocephalus in rats and humans: white matter loss and behavior changes. Ann Neurol 53, 337-346, doi:10.1002/ana.10453 (2003). 189 Del Bigio, M. R. & McAllister, J. P., II. in Pediatric Neurosurgery Vol. 4 (eds M. Choux, R. DiRocco, A. D. Hockley, & M. L. Walker) 217-236 (Churchill Livingstone, 1999). 190 McAllister, J. P., 2nd. Pathophysiology of congenital and neonatal hydrocephalus. Semin Fetal Neonatal Med 17, 285-294, doi:10.1016/j.siny.2012.06.004 (2012). 191 Ho, M. L., Rojas, R. & Eisenberg, R. L. Cerebral edema. AJR Am J Roentgenol 199, W258-273, doi:10.2214/AJR.11.8081 (2012). 192 Segev, Y. et al. Morphometric study of the midsagittal MR imaging plane in cases of hydrocephalus and atrophy and in normal brains. AJNR Am J Neuroradiol 22, 1674-1679 (2001). 193 Niwa, T. et al. Punctate white matter lesions in infants: new insights using susceptibility- weighted imaging. Neuroradiology 53, 669-679, doi:10.1007/s00234-011-0872-0 (2011). 194 Perlman, J. M. White matter injury in the preterm infant: an important determination of abnormal neurodevelopment outcome. Early Hum Dev 53, 99-120, doi:10.1016/s0378- 3782(98)00037-1 (1998). 195 Childs, A. M. et al. Cerebral maturation in premature infants: Quantitative assessment using MR imaging. American Journal of Neuroradiology 22, 1577-1582 (2001). 196 Hudgins, R. J. et al. Intrathecal urokinase as a treatment for intraventricular hemorrhage in the preterm infant. Pediatr Neurosurg 26, 281-287, doi:10.1159/000121207 (1997). 197 McAllister, J. P. et al. Ventricular Zone Disruption in Human Neonates With Intraventricular Hemorrhage. J Neuropathol Exp Neurol 76, 358-375, doi:10.1093/jnen/nlx017 (2017). 198 Karimy, J. K. et al. Inflammation in acquired hydrocephalus: pathogenic mechanisms and therapeutic targets. Nat Rev Neurol, doi:10.1038/s41582-020-0321-y (2020). 199 Sival, D. A. et al. Neonatal high pressure hydrocephalus is associated with elevation of pro-inflammatory cytokines IL-18 and IFNgamma in cerebrospinal fluid. Cerebrospinal Fluid Res 5, 21, doi:10.1186/1743-8454-5-21 (2008). 200 Stearns-Kurosawa, D. J., Osuchowski, M. F., Valentine, C., Kurosawa, S. & Remick, D. G. The pathogenesis of sepsis. Annu Rev Pathol 6, 19-48, doi:10.1146/annurev-pathol- 011110-130327 (2011). 201 Villarino, A. V., Kanno, Y. & O'Shea, J. J. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol 18, 374-384, doi:10.1038/ni.3691 (2017).

208 202 O'Shea, J. J. et al. The JAK-STAT Pathway: Impact on Human Disease and Therapeutic Intervention. Annual Review of Medicine, Vol 66 66, 311-328, doi:10.1146/annurev-med- 051113-024537 (2015). 203 Wong, H. R. et al. Identification of pediatric septic shock subclasses based on genome- wide expression profiling. BMC medicine 7, 34, doi:10.1186/1741-7015-7-34 (2009). 204 Srinivasan, L., Kilpatrick, L., Shah, S. S., Abbasi, S. & Harris, M. C. Cerebrospinal fluid cytokines in the diagnosis of bacterial meningitis in infants. Pediatr Res 80, 566-572, doi:10.1038/pr.2016.117 (2016). 205 Mustafa, M. M. et al. Cerebrospinal fluid prostaglandins, interleukin 1 beta, and tumor necrosis factor in bacterial meningitis. Clinical and laboratory correlations in placebo- treated and dexamethasone-treated patients. Am J Dis Child 144, 883-887, doi:10.1001/archpedi.1990.02150320047024 (1990). 206 van Furth, A. M., Roord, J. J. & van Furth, R. Roles of proinflammatory and anti- inflammatory cytokines in pathophysiology of bacterial meningitis and effect of adjunctive therapy. Infect Immun 64, 4883-4890 (1996). 207 Habiyaremye, G. et al. Chemokine and cytokine levels in the lumbar cerebrospinal fluid of preterm infants with post-hemorrhagic hydrocephalus. Fluids Barriers CNS 14, 35, doi:10.1186/s12987-017-0083-0 (2017). 208 Savman, K. et al. Cytokine response in cerebrospinal fluid from preterm infants with posthaemorrhagic ventricular dilatation. Acta Paediatr 91, 1357-1363, doi:10.1111/j.1651-2227.2002.tb02834.x (2002). 209 Baumeister, F. A., Pohl-Koppe, A., Hofer, M., Kim, J. O. & Weiss, M. IL-6 in CSF during ventriculitis in preterm infants with posthemorrhagic hydrocephalus. Infection 28, 234-236, doi:10.1007/s150100070043 (2000). 210 Schmitz, T. et al. Interleukin-1beta, interleukin-18, and interferon-gamma expression in the cerebrospinal fluid of premature infants with posthemorrhagic hydrocephalus-- markers of white matter damage? Pediatr Res 61, 722-726, doi:10.1203/pdr.0b013e31805341f1 (2007). 211 Malik, S. et al. Neurogenesis continues in the third trimester of pregnancy and is suppressed by premature birth. J Neurosci 33, 411-423, doi:10.1523/JNEUROSCI.4445- 12.2013 (2013). 212 Strahle, J. M. et al. Role of hemoglobin and iron in hydrocephalus after neonatal intraventricular hemorrhage. Neurosurgery 75, 696-705; discussion 706, doi:10.1227/NEU.0000000000000524 (2014). 213 Xi, G., Strahle, J., Hua, Y. & Keep, R. F. Progress in translational research on intracerebral hemorrhage: is there an end in sight? Prog Neurobiol 115, 45-63, doi:10.1016/j.pneurobio.2013.09.007 (2014). 214 Yung, Y. C., Stoddard, N. C., Mirendil, H. & Chun, J. Lysophosphatidic Acid signaling in the nervous system. Neuron 85, 669-682, doi:10.1016/j.neuron.2015.01.009 (2015). 215 Park, R. et al. Yap is required for ependymal integrity and is suppressed in LPA-induced hydrocephalus. Nat Commun 7, 10329, doi:10.1038/ncomms10329 (2016). 216 Batiz, L. F. et al. Heterogeneous expression of hydrocephalic phenotype in the hyh mice carrying a point in alpha-SNAP. Neurobiol Dis 23, 152-168, doi:10.1016/j.nbd.2006.02.009 (2006).

209 217 Perez-Figares, J. M. et al. Spontaneous congenital hydrocephalus in the mutant mouse hyh. Changes in the ventricular system and the subcommissural organ. J Neuropathol Exp Neurol 57, 188-202, doi:10.1097/00005072-199802000-00010 (1998). 218 Jimenez, A. J. et al. A programmed ependymal denudation precedes congenital hydrocephalus in the hyh mutant mouse. J Neuropathol Exp Neurol 60, 1105-1119, doi:10.1093/jnen/60.11.1105 (2001). 219 Wagner, C. et al. Cellular mechanisms involved in the stenosis and obliteration of the cerebral aqueduct of hyh mutant mice developing congenital hydrocephalus. J Neuropathol Exp Neurol 62, 1019-1040, doi:10.1093/jnen/62.10.1019 (2003). 220 Batiz, L. F. et al. New ependymal cells are born postnatally in two discrete regions of the mouse brain and support ventricular enlargement in hydrocephalus. Acta Neuropathol 121, 721-735, doi:10.1007/s00401-011-0799-x (2011). 221 Oliver, C. et al. Disruption of CDH2/N-cadherin-based adherens junctions leads to apoptosis of ependymal cells and denudation of brain ventricular walls. J Neuropathol Exp Neurol 72, 846-860, doi:10.1097/NEN.0b013e3182a2d5fe (2013). 222 Batiz, L. F. et al. Sperm from hyh mice carrying a point mutation in alphaSNAP have a defect in acrosome reaction. PLoS.One. 4, e4963 (2009). 223 Yung, Y. C. et al. Lysophosphatidic acid signaling may initiate fetal hydrocephalus. Sci Transl Med 3, 99ra87, doi:10.1126/scitranslmed.3002095 (2011). 224 Yamamoto, H. et al. Genetic deletion of afadin causes hydrocephalus by destruction of adherens junctions in radial glial and ependymal cells in the midbrain. PLoS One 8, e80356, doi:10.1371/journal.pone.0080356 (2013). 225 Vio, K., Rodriguez, S., Yulis, C. R., Oliver, C. & Rodriguez, E. M. The subcommissural organ of the rat secretes Reissner's fiber glycoproteins and CSF-soluble proteins reaching the internal and external CSF compartments. Cerebrospinal Fluid Res 5, 3, doi:10.1186/1743-8454-5-3 (2008). 226 Vio, K. et al. Hydrocephalus induced by immunological blockage of the subcommissural organ-Reissner's fiber (RF) complex by maternal transfer of anti-RF antibodies. Exp Brain Res 135, 41-52, doi:10.1007/s002210000474 (2000). 227 Yang, P., Baker, K. A. & Hagg, T. The ADAMs family: coordinators of nervous system development, plasticity and repair. Prog Neurobiol 79, 73-94, doi:10.1016/j.pneurobio.2006.05.001 (2006). 228 Inoshima, I. et al. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat Med 17, 1310-1314, doi:10.1038/nm.2451 (2011). 229 Cherian, S., Thoresen, M., Silver, I. A., Whitelaw, A. & Love, S. Transforming growth factor-betas in a rat model of neonatal posthaemorrhagic hydrocephalus. Neuropathol Appl Neurobiol 30, 585-600, doi:10.1111/j.1365-2990.2004.00588.x (2004). 230 Cherian, S. S. et al. Posthemorrhagic ventricular dilation in the neonate: development and characterization of a rat model. J Neuropathol Exp Neurol 62, 292-303, doi:10.1093/jnen/62.3.292 (2003). 231 Isaacs, A. M. et al. MR diffusion changes in the perimeter of the lateral ventricles demonstrate periventricular injury in post-hemorrhagic hydrocephalus of prematurity. NeuroImage. Clinical 24, 102031, doi:10.1016/j.nicl.2019.102031 (2019).

210 232 Mishra, P. K. & Teale, J. M. Transcriptome analysis of the ependymal barrier during murine neurocysticercosis. J Neuroinflammation 9, 141, doi:10.1186/1742-2094-9-141 (2012). 233 Alvarez, J. I. & Teale, J. M. Differential changes in junctional complex proteins suggest the ependymal lining as the main source of leukocyte infiltration into ventricles in murine neurocysticercosis. J Neuroimmunol 187, 102-113, doi:10.1016/j.jneuroim.2007.05.005 (2007). 234 Gotz, M. & Huttner, W. B. The cell biology of neurogenesis. Nat Rev Mol Cell Biol 6, 777-788, doi:10.1038/nrm1739 (2005). 235 Fletcher, J. M. et al. Cerebral white matter and cognition in hydrocephalic children. Arch Neurol 49, 818-824, doi:10.1001/archneur.1992.00530320042010 (1992). 236 Fletcher, J. M., Francis, D. J., Thompson, N. M., Davidson, K. C. & Miner, M. E. Verbal and nonverbal skill discrepancies in hydrocephalic children. J Clin Exp Neuropsychol 14, 593-609, doi:10.1080/01688639208402847 (1992). 237 Hannay, H. J. Functioning of the corpus callosum in children with early hydrocephalus. J Int Neuropsychol Soc 6, 351-361, doi:10.1017/s1355617700633106 (2000). 238 Yuan, W. et al. Diffusion tensor imaging properties and neurobehavioral outcomes in children with hydrocephalus. AJNR Am J Neuroradiol 34, 439-445, doi:10.3174/ajnr.A3218 (2013). 239 Yuan, W., McAllister, J. P. I. & Mangano, F. T. Neuroimaging of white matter abnormalities in pediatric hydrocephalus. Journal of Pediatric Neuroradiology 2, 119- 128, doi:10.3233 (2013). 240 Robinson, S. Neonatal posthemorrhagic hydrocephalus from prematurity: pathophysiology and current treatment concepts. J Neurosurg Pediatr 9, 242-258, doi:10.3171/2011.12.PEDS11136 (2012). 241 Shooman, D., Portess, H. & Sparrow, O. A review of the current treatment methods for posthaemorrhagic hydrocephalus of infants. Cerebrospinal Fluid Res 6, 1, doi:10.1186/1743-8454-6-1 (2009). 242 Cherian, S., Whitelaw, A., Thoresen, M. & Love, S. The pathogenesis of neonatal post- hemorrhagic hydrocephalus. Brain Pathol 14, 305-311, doi:10.1111/j.1750- 3639.2004.tb00069.x (2004). 243 Tarnaris, A. et al. Use of cerebrospinal fluid amyloid-beta and total tau protein to predict favorable surgical outcomes in patients with idiopathic normal pressure hydrocephalus. J Neurosurg 115, 145-150, doi:10.3171/2011.2.JNS101316 (2011). 244 Santangelo, R. et al. Cerebrospinal Fluid Amyloid-beta 42, Total Tau and Phosphorylated Tau are Low in Patients with Normal Pressure Hydrocephalus: Analogies and Differences with Alzheimer's Disease. J Alzheimers Dis 60, 183-200, doi:10.3233/JAD- 170186 (2017). 245 Morales, D. M. et al. Cerebrospinal fluid levels of amyloid precursor protein are associated with ventricular size in post-hemorrhagic hydrocephalus of prematurity. PLoS One 10, e0115045, doi:10.1371/journal.pone.0115045 (2015). 246 Morales, D. M. et al. Lumbar Cerebrospinal Fluid Biomarkers of Posthemorrhagic Hydrocephalus of Prematurity: Amyloid Precursor Protein, Soluble Amyloid Precursor Protein alpha, and L1 Cell Adhesion Molecule. Neurosurgery 80, 82-90, doi:10.1227/NEU.0000000000001415 (2017).

211 247 Morales, D. M. et al. Alterations in protein regulators of neurodevelopment in the cerebrospinal fluid of infants with posthemorrhagic hydrocephalus of prematurity. Mol Cell Proteomics 11, M111 011973, doi:10.1074/mcp.M111.011973 (2012). 248 Isaacs, A. M. & Limbrick, D. D. in Cerebrospinal Fluid Disorders (eds David D. Limbrick Jr & Jeffrey R. Leonard) Ch. Chapter 3, 47-70 (Springer International Publishing, 2019). 249 Limbrick, D. D., Jr. et al. Cerebrospinal Fluid Biomarkers of Pediatric Hydrocephalus. Pediatric neurosurgery 52, 426-435, doi:10.1159/000477175 (2017). 250 Bajaj, M., Lulic-Botica, M. & Natarajan, G. Evaluation of cerebrospinal fluid parameters in preterm infants with intraventricular reservoirs. J Perinatol 32, 786-790, doi:10.1038/jp.2011.190 (2012). 251 Lenfestey, R. W. et al. Predictive value of cerebrospinal fluid parameters in neonates with intraventricular drainage devices. J Neurosurg 107, 209-212, doi:10.3171/PED- 07/09/209 (2007). 252 Rajagopal, A. et al. White matter microstructural abnormality in children with hydrocephalus detected by probabilistic diffusion tractography. AJNR Am J Neuroradiol 34, 2379-2385, doi:10.3174/ajnr.A3737 (2013). 253 Morales, D. M. et al. Lumbar cerebrospinal fluid biomarkers of post-hemorrhagic hydrocephalus of prematurity - Amyloid Precursor Protein, soluble APPα, and L1 cell adhesion molecule. Neurosurgery in press (2016). 254 Priller, C. et al. Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci 26, 7212-7221, doi:10.1523/JNEUROSCI.1450-06.2006 (2006). 255 Kamenetz, F. et al. APP processing and synaptic function. Neuron 37, 925-937, doi:10.1016/s0896-6273(03)00124-7 (2003). 256 Dawkins, E. & Small, D. H. Insights into the physiological function of the beta-amyloid precursor protein: beyond Alzheimer's disease. J Neurochem 129, 756-769, doi:10.1111/jnc.12675 (2014). 257 Gentleman, S. M., Nash, M. J., Sweeting, C. J., Graham, D. I. & Roberts, G. W. β- Amyloid precursor protein (βAPP) as a marker for axonal injury after head injury. Neuroscience Letters 160, 139-144, doi:10.1016/0304-3940(93)90398-5 (1993). 258 Gorrie, C., Oakes, S., Duflou, J., Blumbergs, P. & Waite, P. M. Axonal injury in children after motor vehicle crashes: extent, distribution, and size of axonal swellings using beta- APP immunohistochemistry. J Neurotrauma 19, 1171-1182, doi:10.1089/08977150260337976 (2002). 259 Sherriff, F. E., Bridges, L. R., Gentleman, S. M., Sivaloganathan, S. & Wilson, S. Markers of axonal injury in post mortem human brain. Acta Neuropathol 88, 433-439, doi:10.1007/bf00389495 (1994). 260 Rédei, G. P. Encyclopedia of genetics, genomics, proteomics & informatics., 2201 (Springer, Dordrecht, 2008). 261 Heintzman, N. D. & Ren, B. The gateway to transcription: identifying, characterizing and understanding promoters in the eukaryotic genome. Cellular and molecular life sciences : CMLS 64, 386-400, doi:10.1007/s00018-006-6295-0 (2007). 262 Ruggles, K. V. et al. Methods, Tools and Current Perspectives in Proteogenomics. Mol Cell Proteomics 16, 959-981, doi:10.1074/mcp.MR117.000024 (2017).

212 263 Khatri, P., Sirota, M. & Butte, A. J. Ten years of pathway analysis: current approaches and outstanding challenges. PLoS computational biology 8, e1002375, doi:10.1371/journal.pcbi.1002375 (2012). 264 Craig, R., Cortens, J. P. & Beavis, R. C. Open source system for analyzing, validating, and storing protein identification data. J Proteome Res 3, 1234-1242, doi:10.1021/pr049882h (2004). 265 Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102, 15545- 15550, doi:10.1073/pnas.0506580102 (2005). 266 Fang, H. et al. Signaling pathway impact analysis by incorporating the importance and specificity of genes (SPIA-IS). Comput Biol Chem 71, 236-244, doi:10.1016/j.compbiolchem.2017.09.009 (2017). 267 Tarca, A. L. et al. A novel signaling pathway impact analysis. Bioinformatics 25, 75-82, doi:10.1093/bioinformatics/btn577 (2009). 268 Snel, B., Lehmann, G., Bork, P. & Huynen, M. A. STRING: a web-server to retrieve and display the repeatedly occurring neighbourhood of a gene. Nucleic Acids Res 28, 3442- 3444, doi:10.1093/nar/28.18.3442 (2000). 269 Beaulieu, C. & Allen, P. S. Determinants of anisotropic water diffusion in nerves. Magnetic Resonance in Medicine 31, 394-400, doi:10.1002/mrm.1910310408 (1994). 270 Pierpaoli, C., Jezzard, P., Basser, P. J., Barnett, A. & Di Chiro, G. Diffusion tensor MR imaging of the human brain. Radiology 201, 637-648, doi:10.1148/radiology.201.3.8939209 (1996). 271 Neil, J. J. et al. Normal brain in human newborns: apparent diffusion coefficient and diffusion anisotropy measured by using diffusion tensor MR imaging. Radiology 209, 57- 66, doi:10.1148/radiology.209.1.9769812 (1998). 272 Lean, R. E. et al. Altered neonatal white and gray matter microstructure is associated with neurodevelopmental impairments in very preterm infants with high-grade brain injury. Pediatr Res 86, 365-374, doi:10.1038/s41390-019-0461-1 (2019). 273 Beaulieu, C. & Allen, P. S. Determinants of anisotropic water diffusion in nerves. Magn Reson Med 31, 394-400, doi:10.1002/mrm.1910310408 (1994). 274 Pierpaoli, C. & Basser, P. J. Toward a quantitative assessment of diffusion anisotropy. Magn Reson Med 36, 893-906, doi:10.1002/mrm.1910360612 (1996). 275 Assaf, Y., Ben-Sira, L., Constantini, S., Chang, L. C. & Beni-Adani, L. Diffusion tensor imaging in hydrocephalus: initial experience. AJNR Am J Neuroradiol 27, 1717-1724 (2006). 276 Yuan, W. et al. Changes of White Matter Diffusion Anisotropy in Response to a 6-Week iPad Application-Based Occupational Therapy Intervention in Children with Surgically Treated Hydrocephalus: A Pilot Study. Neuropediatrics 47, 336-340, doi:10.1055/s- 0036-1584938 (2016). 277 Ben-Sira, L. et al. Clinical benefits of diffusion tensor imaging in hydrocephalus. J Neurosurg Pediatr 16, 195-202, doi:10.3171/2014.10.PEDS13668 (2015). 278 Yuan, W. et al. Diffusion tensor imaging correlates with cytopathology in a rat model of neonatal hydrocephalus. Cerebrospinal Fluid Res 7, 19, doi:10.1186/1743-8454-7-19 (2010). 279 Yuan, W. et al. Diffusion tensor imaging of white matter injury in a rat model of infantile hydrocephalus. Childs Nerv Syst 28, 47-54, doi:10.1007/s00381-011-1590-y (2012).

213 280 Basser, P. J. & Pierpaoli, C. A simplified method to measure the diffusion tensor from seven MR images. Magn Reson Med 39, 928-934, doi:10.1002/mrm.1910390610 (1998). 281 Pierpaoli, C. & Basser, P. J. Toward a quantitative assessment of diffusion anisotropy. Magnetic Resonance in Medicine 36, 893-906, doi:10.1002/mrm.1910360612 (1996). 282 Pajevic, S. & Pierpaoli, C. Color schemes to represent the orientation of anisotropic tissues from diffusion tensor data: application to white matter fiber tract mapping in the human brain. Magn Reson Med 43, 921, doi:10.1002/1522-2594(200006)43:6<921::aid- mrm23>3.0.co;2-i (2000). 283 Basser, P. J., Mattiello, J. & LeBihan, D. MR diffusion tensor spectroscopy and imaging. Biophys J 66, 259-267, doi:10.1016/S0006-3495(94)80775-1 (1994). 284 Mangano, F. T. et al. Diffusion tensor imaging study of pediatric patients with congenital hydrocephalus: 1-year postsurgical outcomes. J Neurosurg Pediatr 18, 306-319, doi:10.3171/2016.2.PEDS15628 (2016). 285 Ben-Sira, L. et al. Clinical benefits of diffusion tensor imaging in hydrocephalus. J Neurosurg-Pediatr 16, 195-202, doi:10.3171/2014.10.PEDS13668 (2015). 286 Keong, N. C. et al. Diffusion tensor imaging profiles reveal specific neural tract distortion in normal pressure hydrocephalus. PLoS One 12, e0181624, doi:10.1371/journal.pone.0181624 (2017). 287 Buckley, R. T. et al. Longitudinal comparison of diffusion tensor imaging parameters and neuropsychological measures following endoscopic third ventriculostomy for hydrocephalus. J Neurosurg Pediatr 9, 630-635, doi:10.3171/2012.2.PEDS11331 (2012). 288 Ambady, P. et al. Neuro/Medical Oncology. Neuro-Oncology 15, iii98-iii135, doi:10.1093/neuonc/not182 (2013). 289 Thompson, D. K. et al. Corpus callosum alterations in very preterm infants: perinatal correlates and 2 year neurodevelopmental outcomes. Neuroimage 59, 3571-3581, doi:10.1016/j.neuroimage.2011.11.057 (2012). 290 Air, E. L. et al. Longitudinal comparison of pre- and postoperative diffusion tensor imaging parameters in young children with hydrocephalus. J Neurosurg Pediatr 5, 385- 391, doi:10.3171/2009.11.PEDS09343 (2010). 291 Scheel, M., Diekhoff, T., Sprung, C. & Hoffmann, K. T. Diffusion tensor imaging in hydrocephalus--findings before and after shunt surgery. Acta Neurochir (Wien) 154, 1699-1706, doi:10.1007/s00701-012-1377-2 (2012). 292 Isaacs, A. M. et al. Feasibility of fast brain diffusion MRI to quantify white matter injury in pediatric hydrocephalus. J Neurosurg Pediatr, 1-8, doi:10.3171/2019.5.PEDS18596 (2019). 293 Tan, K. et al. Diffusion tensor imaging and ventricle volume quantification in patients with chronic shunt-treated hydrocephalus: a matched case-control study. J Neurosurg 129, 1611-1622, doi:10.3171/2017.6.JNS162784 (2018). 294 Sun, M. et al. Diffusion tensor imaging findings in young children with benign external hydrocephalus differ from the normal population. Childs Nerv Syst 28, 199-208, doi:10.1007/s00381-011-1651-2 (2012). 295 Tournier, J. D., Mori, S. & Leemans, A. Diffusion tensor imaging and beyond. Magn Reson Med 65, 1532-1556, doi:10.1002/mrm.22924 (2011). 296 Jeurissen, B., Tournier, J. D., Dhollander, T., Connelly, A. & Sijbers, J. Multi-tissue constrained spherical deconvolution for improved analysis of multi-shell diffusion MRI data. Neuroimage 103, 411-426, doi:10.1016/j.neuroimage.2014.07.061 (2014).

214 297 Jones, D. K. et al. Microstructural imaging of the human brain with a 'super-scanner': 10 key advantages of ultra-strong gradients for diffusion MRI. Neuroimage 182, 8-38, doi:10.1016/j.neuroimage.2018.05.047 (2018). 298 Jeurissen, B., Leemans, A., Tournier, J. D., Jones, D. K. & Sijbers, J. Investigating the prevalence of complex fiber configurations in white matter tissue with diffusion magnetic resonance imaging. Hum Brain Mapp 34, 2747-2766, doi:10.1002/hbm.22099 (2013). 299 Jones, D. K., Knosche, T. R. & Turner, R. White matter integrity, fiber count, and other fallacies: the do's and don'ts of diffusion MRI. Neuroimage 73, 239-254, doi:10.1016/j.neuroimage.2012.06.081 (2013). 300 Beaulieu, C. The basis of anisotropic water diffusion in the nervous system - a technical review. NMR Biomed 15, 435-455, doi:10.1002/nbm.782 (2002). 301 Wedeen, V. J., Hagmann, P., Tseng, W. Y., Reese, T. G. & Weisskoff, R. M. Mapping complex tissue architecture with diffusion spectrum magnetic resonance imaging. Magn Reson Med 54, 1377-1386, doi:10.1002/mrm.20642 (2005). 302 Tuch, D. S. et al. High angular resolution diffusion imaging reveals intravoxel white matter fiber heterogeneity. Magn Reson Med 48, 577-582, doi:10.1002/mrm.10268 (2002). 303 Assaf, Y. & Basser, P. J. Composite hindered and restricted model of diffusion (CHARMED) MR imaging of the human brain. Neuroimage 27, 48-58, doi:10.1016/j.neuroimage.2005.03.042 (2005). 304 Assaf, Y., Blumenfeld-Katzir, T., Yovel, Y. & Basser, P. J. AxCaliber: a method for measuring axon diameter distribution from diffusion MRI. Magn Reson Med 59, 1347- 1354, doi:10.1002/mrm.21577 (2008). 305 Zhang, H., Schneider, T., Wheeler-Kingshott, C. A. & Alexander, D. C. NODDI: practical in vivo neurite orientation dispersion and density imaging of the human brain. Neuroimage 61, 1000-1016, doi:10.1016/j.neuroimage.2012.03.072 (2012). 306 Sotiropoulos, S. N. et al. Advances in diffusion MRI acquisition and processing in the . Neuroimage 80, 125-143, doi:10.1016/j.neuroimage.2013.05.057 (2013). 307 Wang, X. et al. Diffusion basis spectrum imaging detects and distinguishes coexisting subclinical inflammation, demyelination and axonal injury in experimental autoimmune encephalomyelitis mice. NMR Biomed 27, 843-852, doi:10.1002/nbm.3129 (2014). 308 Wang, Y. et al. Differentiation and quantification of inflammation, demyelination and axon injury or loss in multiple sclerosis. Brain 138, 1223-1238, doi:10.1093/brain/awv046 (2015). 309 Chiang, C. W. et al. Quantifying white matter tract diffusion parameters in the presence of increased extra-fiber cellularity and vasogenic edema. Neuroimage 101, 310-319, doi:10.1016/j.neuroimage.2014.06.064 (2014). 310 Sun, P. et al. Diffusion Assessment of Cortical Changes, Induced by Traumatic Spinal Cord Injury. Brain Sci 7, doi:10.3390/brainsci7020021 (2017). 311 Wang, Y. et al. Quantification of increased cellularity during inflammatory demyelination. Brain 134, 3590-3601, doi:10.1093/brain/awr307 (2011). 312 Xu, J. et al. Magnetic resonance diffusion characteristics of histologically defined prostate cancer in humans. Magn Reson Med 61, 842-850, doi:10.1002/mrm.21896 (2009).

215 313 Cross, A. H. & Song, S. K. "A new imaging modality to non-invasively assess multiple sclerosis pathology". J Neuroimmunol 304, 81-85, doi:10.1016/j.jneuroim.2016.10.002 (2017). 314 Lin, T. H. et al. Diffusion MRI quantifies early axonal loss in the presence of nerve swelling. J Neuroinflammation 14, 78, doi:10.1186/s12974-017-0852-3 (2017). 315 Zhan, J. et al. Diffusion Basis Spectrum and Diffusion Tensor Imaging Detect Hippocampal Inflammation and Dendritic Injury in a Virus-Induced Mouse Model of Epilepsy. Front Neurosci 12, 77, doi:10.3389/fnins.2018.00077 (2018). 316 Lin, S. D. et al. The Use of Ultrasound-Measured Optic Nerve Sheath Diameter to Predict Ventriculoperitoneal Shunt Failure in Children. Pediatr Emerg Care 35, 268-272, doi:10.1097/PEC.0000000000001034 (2019). 317 Lin, T. H. et al. Noninvasive Quantification of Axonal Loss in the Presence of Tissue Swelling in Traumatic Spinal Cord Injury Mice. J Neurotrauma 36, 2308-2315, doi:10.1089/neu.2018.6016 (2019). 318 Castaneyra-Ruiz, L. et al. Blood Exposure Causes Ventricular Zone Disruption and Glial Activation In Vitro. J Neuropathol Exp Neurol 77, 803-813, doi:10.1093/jnen/nly058 (2018). 319 Smith, M. E. & Eskandari, R. A novel technology to model pressure-induced cellular injuries in the brain. Journal of neuroscience methods 293, 247-253, doi:10.1016/j.jneumeth.2017.10.004 (2018). 320 Harris, C. A. & McAllister, J. P., 2nd. What we should know about the cellular and tissue response causing catheter obstruction in the treatment of hydrocephalus. Neurosurgery 70, 1589-1601; discussion 1601-1582, doi:10.1227/NEU.0b013e318244695f (2012). 321 Harris, C. A. & McAllister, J. P., 2nd. Does drainage hole size influence adhesion on ventricular catheters? Childs Nerv Syst 27, 1221-1232, doi:10.1007/s00381-011-1430-0 (2011). 322 Harris, C. A. et al. Reduction of protein adsorption and macrophage and astrocyte adhesion on ventricular catheters by polyethylene glycol and N-acetyl-L-cysteine. J Biomed Mater Res A 98, 425-433, doi:10.1002/jbm.a.33130 (2011). 323 Harris, C. A. et al. Effects of surface wettability, flow, and protein concentration on macrophage and astrocyte adhesion in an in vitro model of central nervous system catheter obstruction. J Biomed Mater Res A 97, 433-440, doi:10.1002/jbm.a.33078 (2011). 324 Harris, C. A. et al. Mechanical contributions to astrocyte adhesion using a novel in vitro model of catheter obstruction. Exp Neurol 222, 204-210, doi:10.1016/j.expneurol.2009.12.027 (2010). 325 Wilson, G. R. et al. Deletion of the Parkin co-regulated gene causes defects in ependymal ciliary motility and hydrocephalus in the quakingviable mutant mouse. Hum Mol Genet 19, 1593-1602, doi:10.1093/hmg/ddq031 (2010). 326 Lechtreck, K. F., Delmotte, P., Robinson, M. L., Sanderson, M. J. & Witman, G. B. Mutations in Hydin impair ciliary motility in mice. The Journal of cell biology 180, 633- 643, doi:10.1083/jcb.200710162 (2008). 327 Davy, B. E. & Robinson, M. L. Congenital hydrocephalus in hy3 mice is caused by a frameshift mutation in Hydin, a large novel gene. Hum Mol Genet 12, 1163-1170, doi:10.1093/hmg/ddg122 (2003).

216 328 Vogel, T. W., Carter, C. S., Abode-Iyamah, K., Zhang, Q. & Robinson, S. The role of primary cilia in the pathophysiology of neural tube defects. Neurosurg Focus 33, E2, doi:10.3171/2012.6.FOCUS12222 (2012). 329 Swiderski, R. E. et al. Structural defects in cilia of the choroid plexus, subfornical organ and ventricular ependyma are associated with ventriculomegaly. Fluids Barriers CNS 9, 22, doi:10.1186/2045-8118-9-22 (2012). 330 Norris, D. P. & Grimes, D. T. Mouse models of ciliopathies: the state of the art. Dis Model Mech 5, 299-312, doi:10.1242/dmm.009340 (2012). 331 Carter, C. S. et al. Abnormal development of NG2+PDGFR-alpha+ neural progenitor cells leads to neonatal hydrocephalus in a ciliopathy mouse model. Nat Med 18, 1797- 1804, doi:10.1038/nm.2996 (2012). 332 Breunig, J. J., Arellano, J. I. & Rakic, P. Cilia in the brain: going with the flow. Nat Neurosci 13, 654-655, doi:10.1038/nn0610-654 (2010). 333 Agassandian, K. et al. Ciliopathy is differentially distributed in the brain of a Bardet- Biedl syndrome mouse model. PLoS One 9, e93484, doi:10.1371/journal.pone.0093484 (2014). 334 Bryan, J. H. The immotile cilia syndrome. Mice versus man. Virchows Arch A Pathol Anat Histopathol 399, 265-275, doi:10.1007/bf00612944 (1983). 335 Appelbe, O. K. et al. Disruption of the mouse Jhy gene causes abnormal ciliary microtubule patterning and juvenile hydrocephalus. Developmental biology 382, 172- 185, doi:10.1016/j.ydbio.2013.07.003 (2013). 336 Lattke, M., Magnutzki, A., Walther, P., Wirth, T. & Baumann, B. Nuclear factor kappaB activation impairs ependymal ciliogenesis and links neuroinflammation to hydrocephalus formation. J Neurosci 32, 11511-11523, doi:10.1523/JNEUROSCI.0182-12.2012 (2012). 337 Jones, H. C., Dack, S. & Ellis, C. Morphological aspects of the development of hydrocephalus in a mouse mutant (SUMS/NP). Acta Neuropathol 72, 268-276, doi:10.1007/bf00691100 (1987). 338 Bruni, J. E., del Bigio, M. R., Cardoso, E. R. & Persaud, T. V. Neuropathology of congenital hydrocephalus in the SUMS/NP mouse. Acta Neurochir (Wien) 92, 118-122, doi:10.1007/bf01401981 (1988). 339 Aliev, G. et al. Ultrastructural analysis of a murine model of congenital hydrocephalus produced by overexpression of transforming growth factor-beta1 in the central nervous system. J Submicrosc Cytol Pathol 38, 85-91 (2006). 340 Cohen, A. R. et al. Characterization of a model of hydrocephalus in transgenic mice. J Neurosurg 91, 978-988, doi:10.3171/jns.1999.91.6.0978 (1999). 341 Crews, L., Wyss-Coray, T. & Masliah, E. Insights into the pathogenesis of hydrocephalus from transgenic and experimental animal models. Brain Pathol 14, 312-316, doi:10.1111/j.1750-3639.2004.tb00070.x (2004). 342 Johanson, C. E. et al. Altered formation and bulk absorption of cerebrospinal fluid in FGF-2-induced hydrocephalus. The American journal of physiology 277, R263-271, doi:10.1152/ajpregu.1999.277.1.R263 (1999). 343 Ohmiya, M. et al. Administration of FGF-2 to embryonic mouse brain induces hydrocephalic brain morphology and aberrant differentiation of neurons in the postnatal cerebral cortex. J Neurosci Res 65, 228-235, doi:10.1002/jnr.1146 (2001). 344 Dahme, M. et al. Disruption of the mouse L1 gene leads to malformations of the nervous system. Nat Genet 17, 346-349, doi:10.1038/ng1197-346 (1997).

217 345 Fransen, E. et al. L1 knockout mice show dilated ventricles, vermis hypoplasia and impaired exploration patterns. Hum Mol Genet 7, 999-1009, doi:10.1093/hmg/7.6.999 (1998). 346 Lin, X. et al. Genetic deletion of Rnd3 results in aqueductal stenosis leading to hydrocephalus through up-regulation of Notch signaling. Proc Natl Acad Sci U S A 110, 8236-8241, doi:10.1073/pnas.1219995110 (2013). 347 Bloch, O., Auguste, K. I., Manley, G. T. & Verkman, A. S. Accelerated progression of kaolin-induced hydrocephalus in aquaporin-4-deficient mice. J Cereb Blood Flow Metab 26, 1527-1537, doi:10.1038/sj.jcbfm.9600306 (2006). 348 Papadopoulos, M. C. & Verkman, A. S. Aquaporin-4 and brain edema. Pediatr Nephrol 22, 778-784, doi:10.1007/s00467-006-0411-0 (2007). 349 Borit, A. & Sidman, R. L. New mutant mouse with communicating hydrocephalus and secondary aqueductal stenosis. Acta Neuropathol 21, 316-331, doi:10.1007/bf00685139 (1972). 350 Shu, T., Butz, K. G., Plachez, C., Gronostajski, R. M. & Richards, L. J. Abnormal development of forebrain midline glia and commissural projections in Nfia knock-out mice. J Neurosci 23, 203-212 (2003). 351 das Neves, L. et al. Disruption of the murine nuclear factor I-A gene (Nfia) results in perinatal lethality, hydrocephalus, and agenesis of the corpus callosum. Proc Natl Acad Sci U S A 96, 11946-11951, doi:10.1073/pnas.96.21.11946 (1999). 352 Kohn, D. F., Chinookoswong, N. & Chou, S. M. A new model of congenital hydrocephalus in the rat. Acta Neuropathol 54, 211-218, doi:10.1007/bf00687744 (1981). 353 Jones, H. C. Molecular genetics of congenital hydrocephalus. Experimental Neurology 190, 79-90 (2004). 354 Jones, H. C., Depelteau, J. S., Carter, B. J., Lopman, B. A. & Morel, L. Genome-wide linkage analysis of inherited hydrocephalus in the H-Tx rat. Mammalian genome : official journal of the International Mammalian Genome Society 12, 22-26, doi:10.1007/s003350010226 (2001). 355 Jones, H. C., Yehia, B., Chen, G. F. & Carter, B. J. Genetic analysis of inherited hydrocephalus in a rat model. Exp Neurol 190, 79-90, doi:10.1016/j.expneurol.2004.06.019 (2004). 356 Cai, X. et al. Hydrocephalus in the H-Tx rat: a monogenic disease? Exp Neurol 163, 131- 135, doi:10.1006/exnr.1999.7301 (2000). 357 Jones, H. C., Carter, B. J. & Morel, L. Characteristics of hydrocephalus expression in the LEW/Jms rat strain with inherited disease. Childs Nerv Syst 19, 11-18, doi:10.1007/s00381-002-0671-3 (2003). 358 Sasaki, S. et al. Congenital hydrocephalus revealed in the inbred rat, LEW/Jms. Neurosurgery 13, 548-554, doi:10.1227/00006123-198311000-00011 (1983). 359 Jones, H. C., Totten, C. F., Mayorga, D. A., Yue, M. & Carter, B. J. Genetic loci for ventricular dilatation in the LEW/Jms rat with fetal-onset hydrocephalus are influenced by gender and genetic background. Cerebrospinal Fluid Res 2, 2, doi:10.1186/1743- 8454-2-2 (2005). 360 Yamada, H., Oi, S., Tamaki, N., Matsumoto, S. & Sudo, K. Prenatal Aqueductal Stenosis as a Cause of Congenital Hydrocephalus in the Inbred Rat Lew Jms. Childs Nervous System 7, 218-222 (1991).

218 361 Guerra, M. Neural stem cells: are they the hope of a better life for patients with fetal- onset hydrocephalus? Fluids Barriers CNS 11, 7, doi:10.1186/2045-8118-11-7 (2014). 362 Garcia-Bonilla, M. et al. Neocortical tissue recovery in severe congenital obstructive hydrocephalus after intraventricular administration of bone marrow-derived mesenchymal stem cells. Stem cell research & therapy 11, 121, doi:10.1186/s13287-020- 01626-6 (2020). 363 Del Bigio, M. R., Khan, O. H., da Silva Lopes, L. & Juliet, P. A. Cerebral white matter oxidation and nitrosylation in young rodents with kaolin-induced hydrocephalus. J Neuropathol Exp Neurol 71, 274-288, doi:10.1097/NEN.0b013e31824c1b44 (2012). 364 Di Curzio, D. L., Turner-Brannen, E., Mao, X. & Del Bigio, M. R. Magnesium sulfate treatment for juvenile ferrets following induction of hydrocephalus with kaolin. Fluids Barriers CNS 13, 7, doi:10.1186/s12987-016-0031-4 (2016). 365 Eskandari, R. et al. Differential vulnerability of white matter structures to experimental infantile hydrocephalus detected by diffusion tensor imaging. Childs Nerv Syst 30, 1651- 1661, doi:10.1007/s00381-014-2500-x (2014). 366 McAllister, J. P., 2nd, Maugans, T. A., Shah, M. V. & Truex, R. C., Jr. Neuronal effects of experimentally induced hydrocephalus in newborn rats. J Neurosurg 63, 776-783, doi:10.3171/jns.1985.63.5.0776 (1985). 367 McAllister, J. P., II et al. in Soc.Neurosci.Abstr. 1316. 368 Wagshul, M. E. et al. 369 Yamada, H., Yokota, A., Haratake, J. & Horie, A. Morphological study of experimental syringomyelia with kaolin-induced hydrocephalus in a canine model. J Neurosurg 84, 999-1005, doi:10.3171/jns.1996.84.6.0999 (1996). 370 Xue, M. & Del Bigio, M. R. Comparison of brain cell death and inflammatory reaction in three models of intracerebral hemorrhage in adult rats. J Stroke Cerebrovasc Dis 12, 152- 159, doi:10.1016/S1052-3057(03)00036-3 (2003). 371 Zhu, X. et al. Plasmalemma permeability and necrotic cell death phenotypes after intracerebral hemorrhage in mice. Stroke 43, 524-531, doi:10.1161/STROKEAHA.111.635672 (2012). 372 Lodhia, K. R., Shakui, P. & Keep, R. F. Hydrocephalus in a rat model of intraventricular hemorrhage. Acta Neurochir Suppl 96, 207-211, doi:10.1007/3-211-30714-1_45 (2006). 373 Aquilina, K., Chakkarapani, E., Love, S. & Thoresen, M. Neonatal rat model of intraventricular haemorrhage and post-haemorrhagic ventricular dilatation with long-term survival into adulthood. Neuropathol Appl Neurobiol 37, 156-165, doi:10.1111/j.1365- 2990.2010.01118.x (2011). 374 Miller, J. M., Kumar, R., McAllister, J. P., 2nd & Krause, G. S. Gene expression analysis of the development of congenital hydrocephalus in the H-Tx rat. Brain research 1075, 36-47, doi:10.1016/j.brainres.2005.12.094 (2006). 375 Harris, N. G., McAllister, J. P., 2nd & Jones, H. C. The effect of untreated and shunt- treated hydrocephalus on cortical pyramidal neurone morphology in the H-Tx rat. Eur J Pediatr Surg 3 Suppl 1, 31-32 (1993). 376 Perlman, R. L. Mouse models of human disease: An evolutionary perspective. Evol Med Public Health 2016, 170-176, doi:10.1093/emph/eow014 (2016). 377 Ment, L. R., Stewart, W. B., Duncan, C. C., Scott, D. T. & Lambrecht, R. Beagle puppy model of intraventricular hemorrhage. Journal of Neurosurgery 60, 737-742 (1984).

219 378 Ment, L. R., Stewart, W. B., Duncan, C. C. & Pitt, B. R. Beagle puppy model of perinatal cerebral insults. Cerebral blood flow changes and intraventricular hemorrhage evoked by hypoxemia. J Neurosurg 65, 847-850, doi:10.3171/jns.1986.65.6.0847 (1986). 379 Aquilina, K. et al. A neonatal piglet model of intraventricular hemorrhage and posthemorrhagic ventricular dilation. J Neurosurg 107, 126-136, doi:10.3171/PED- 07/08/126 (2007). 380 Aquilina, K., Chakkarapani, E. & Thoresen, M. Early deterioration of cerebrospinal fluid dynamics in a neonatal piglet model of intraventricular hemorrhage and posthemorrhagic ventricular dilation. J Neurosurg Pediatr 10, 529-537, doi:10.3171/2012.8.PEDS11386 (2012). 381 Ahn, S. Y. et al. Mesenchymal stem cells prevent hydrocephalus after severe intraventricular hemorrhage. Stroke 44, 497-504, doi:10.1161/STROKEAHA.112.679092 (2013). 382 Gao, C. et al. Role of red blood cell lysis and iron in hydrocephalus after intraventricular hemorrhage. J Cereb Blood Flow Metab 34, 1070-1075, doi:10.1038/jcbfm.2014.56 (2014). 383 Aquilina, K., Hobbs, C., Tucker, A., Whitelaw, A. & Thoresen, M. Do drugs that block transforming growth factor beta reduce posthaemorrhagic ventricular dilatation in a neonatal rat model? Acta Paediatr 97, 1181-1186, doi:10.1111/j.1651-2227.2008.00903.x (2008). 384 Georgiadis, P. et al. Characterization of acute brain injuries and neurobehavioral profiles in a rabbit model of germinal matrix hemorrhage. Stroke 39, 3378-3388, doi:10.1161/STROKEAHA.107.510883 (2008). 385 Ment, L. R., Stewart, W. B., Duncan, C. C., Pitt, B. R. & Cole, J. S. Beagle puppy model of perinatal cerebral infarction. Regional cerebral prostaglandin changes during acute hypoxemia. J Neurosurg 65, 851-855, doi:10.3171/jns.1986.65.6.0851 (1986). 386 Okubo, S., Strahle, J., Keep, R. F., Hua, Y. & Xi, G. Subarachnoid hemorrhage-induced hydrocephalus in rats. Stroke 44, 547-550, doi:10.1161/STROKEAHA.112.662312 (2013). 387 Smith, A. L., Smith, D. H., Averill, D. R., Jr., Marino, J. & Moxon, E. R. Production of Haemophilus influenzae b meningitis in infant rats by intraperitoneal inoculation. Infect Immun 8, 278-290 (1973). 388 Glode, M. P., Sutton, A., Moxon, E. R. & Robbins, J. B. Pathogenesis of neonatal Escherichia coli meningitis: induction of bacteremia and meningitis in infant rats fed E. coli K1. Infect Immun 16, 75-80 (1977). 389 Moxon, E. R., Glode, M. P., Sutton, A. & Robbins, J. B. The infant rat as a model of bacterial meningitis. The Journal of infectious diseases 136 Suppl, S186-190, doi:10.1093/infdis/136.supplement.s186 (1977). 390 Dacey, R. G. & Sande, M. A. Effect of probenecid on cerebrospinal fluid concentrations of penicillin and cephalosporin derivatives. Antimicrob Agents Chemother 6, 437-441, doi:10.1128/aac.6.4.437 (1974). 391 Moxon, E. R., Smith, A. L., Averill, D. R. & Smith, D. H. Haemophilus influenzae meningitis in infant rats after intranasal inoculation. The Journal of infectious diseases 129, 154-162, doi:10.1093/infdis/129.2.154 (1974).

220 392 Greenberg, D. P., Bayer, A. S., Cheung, A. L. & Ward, J. I. Protective efficacy of protein A-specific antibody against bacteremic infection due to Staphylococcus aureus in an infant rat model. Infect Immun 57, 1113-1118 (1989). 393 Kim, K. S. Efficacy of imipenem in experimental group B streptococcal bacteremia and meningitis. 31, 304-309, doi:10.1159/000238352 (1985). 394 Kim, K. S., Manocchio, M. & Bayer, A. S. Efficacy of cefotaxime and latamoxef for Escherichia coli bacteremia and meningitis in newborn rats. Chemotherapy 30, 262-269, doi:10.1159/000238278 (1984). 395 Vogel, U., Hammerschmidt, S. & Frosch, M. Sialic acids of both the capsule and the sialylated lipooligosaccharide of Neisseria meningitis serogroup B are prerequisites for virulence of meningococci in the infant rat. Medical microbiology and immunology 185, 81-87, doi:10.1007/s004300050018 (1996). 396 Ferrieri, P., Burke, B. & Nelson, J. Production of bacteremia and meningitis in infant rats with group B streptococcal serotypes. Infect Immun 27, 1023-1032 (1980). 397 Kim, Y. S. et al. Brain injury in experimental neonatal meningitis due to group B streptococci. J Neuropathol Exp Neurol 54, 531-539, doi:10.1097/00005072-199507000- 00007 (1995). 398 Saukkonen, K. M., Nowicki, B. & Leinonen, M. Role of type 1 and S fimbriae in the pathogenesis of Escherichia coli O18:K1 bacteremia and meningitis in the infant rat. Infect Immun 56, 892-897 (1988). 399 Townsend, G. C. & Scheld, W. M. in Handbook of Animal Models of Infection : Experimental Models in Antimicrobial Chemotherapy (eds Merle A. Sande & Oto Zak) (Elsevier Science, 1999). 400 Hoiseth, S. K., Connelly, C. J. & Moxon, E. R. Genetics of spontaneous, high-frequency loss of b capsule expression in Haemophilus influenzae. Infect Immun 49, 389-395 (1985). 401 Lesse, A. J., Moxon, E. R., Zwahlen, A. & Scheld, W. M. Role of cerebrospinal fluid pleocytosis and Haemophilus influenzae type b capsule on blood brain barrier permeability during experimental meningitis in the rat. The Journal of clinical investigation 82, 102-109, doi:10.1172/JCI113556 (1988). 402 Paulson, J. N., Williams, B. L. & et.al. The Bacterial and Viral Complexity of Postinfectious Hydrocephalus in Uganda. Sci Transl Med aba0565. Accepted (2020). 403 Briese, T. et al. Virome Capture Sequencing Enables Sensitive Viral Diagnosis and Comprehensive Virome Analysis. mBio 6, e01491-01415, doi:10.1128/mBio.01491-15 (2015). 404 Nakamura, L. K. Bacillus thiaminolyticus sp. nov., nom. rev. Int J Syst Bacteriol 40, 242- 246, doi:10.1099/00207713-40-3-242 (1990). 405 Szklarczyk, D. et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Research 47, D607-D613, doi:10.1093/nar/gky1131 (2019). 406 Michaud, J. et al. Integrative analysis of RUNX1 downstream pathways and target genes. BMC genomics 9, 363, doi:10.1186/1471-2164-9-363 (2008). 407 Fabregat, A. et al. Reactome pathway analysis: a high-performance in-memory approach. BMC bioinformatics 18, 142, doi:10.1186/s12859-017-1559-2 (2017). 408 Kanehisa, M. & Goto, S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 28, 27-30, doi:10.1093/nar/28.1.27 (2000).

221 409 Warf, B. C. Hydrocephalus in Uganda: the predominance of infectious origin and primary management with endoscopic third ventriculostomy. J Neurosurg 102, 1-15, doi:10.3171/ped.2005.102.1.0001 (2005). 410 Dhenni, R. et al. Isolation and complete genome analysis of neurotropic dengue virus serotype 3 from the cerebrospinal fluid of an encephalitis patient. PLoS Negl Trop Dis 12, e0006198, doi:10.1371/journal.pntd.0006198 (2018). 411 Philip, N., Rodrigues, K. F., William, T. & John, D. V. Whole genome sequencing of Mycobacterium tuberculosis SB24 isolated from Sabah, Malaysia. Genom Data 9, 137- 139, doi:10.1016/j.gdata.2016.08.007 (2016). 412 Heintzman, N. D. & Ren, B. The gateway to transcription: identifying, characterizing and understanding promoters in the eukaryotic genome. Cellular and Molecular Life Sciences 64, 386-400, doi:10.1007/s00018-006-6295-0 (2006). 413 PrabhuDas, M. et al. Challenges in infant immunity: implications for responses to infection and vaccines. Nat Immunol 12, 189-194, doi:10.1038/ni0311-189 (2011). 414 Ygberg, S. & Nilsson, A. The developing immune system - from foetus to toddler. Acta Paediatr 101, 120-127, doi:10.1111/j.1651-2227.2011.02494.x (2012). 415 Bell, E. Innate immunity - TLR signalling. Nature Reviews Immunology 3, 692-692, doi:10.1038/nri1185 (2003). 416 Kawai, T. & Akira, S. TLR signaling. Cell death and differentiation 13, 816-825, doi:10.1038/sj.cdd.4401850 (2006). 417 Tchessalova, D., Posillico, C. K. & Tronson, N. C. Neuroimmune Activation Drives Multiple Brain States. Frontiers in Systems Neuroscience 12, doi:10.3389/fnsys.2018.00039 (2018). 418 Adiko, A. C., Babdor, J., Gutierrez-Martinez, E., Guermonprez, P. & Saveanu, L. Intracellular Transport Routes for MHC I and Their Relevance for Antigen Cross- Presentation. Frontiers in immunology 6, 335, doi:10.3389/fimmu.2015.00335 (2015). 419 Guerra, M. et al. A cell junction pathology of neural stem cells Is associated to ventricular zone disruption, hydrocephalus and neurogenesis abnormalities. Journal of Neuropathology and Experimental Neurology 74, 653-671 (2015). 420 Cheeran, M. C., Lokensgard, J. R. & Schleiss, M. R. Neuropathogenesis of congenital cytomegalovirus infection: disease mechanisms and prospects for intervention. Clin Microbiol Rev 22, 99-126, Table of Contents, doi:10.1128/CMR.00023-08 (2009). 421 Wellons, J. C., 3rd et al. Shunting outcomes in posthemorrhagic hydrocephalus: results of a Hydrocephalus Clinical Research Network prospective cohort study. J Neurosurg Pediatr 20, 19-29, doi:10.3171/2017.1.PEDS16496 (2017). 422 Yu, G. C., Wang, L. G., Han, Y. Y. & He, Q. Y. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. -a Journal of Integrative Biology 16, 284-287, doi:10.1089/omi.2011.0118 (2012). 423 Robinson, M. D. & Smyth, G. K. Moderated statistical tests for assessing differences in tag abundance. Bioinformatics 23, 2881-2887, doi:10.1093/bioinformatics/btm453 (2007). 424 Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. Bmc Bioinformatics 12, doi:10.1186/1471-2105-12- 323 (2011).

222 425 Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15-21, doi:10.1093/bioinformatics/bts635 (2013). 426 Paulson, J. N. et al. Tissue-aware RNA-Seq processing and normalization for heterogeneous and sparse data. BMC Bioinformatics 18, 437, doi:10.1186/s12859-017- 1847-x (2017). 427 Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4, 44-57, doi:10.1038/nprot.2008.211 (2009). 428 McAlister, G. C. et al. Increasing the multiplexing capacity of TMTs using reporter ion isotopologues with isobaric masses. Anal Chem 84, 7469-7478, doi:10.1021/ac301572t (2012). 429 Mertins, P. et al. Reproducible workflow for multiplexed deep-scale proteome and phosphoproteome analysis of tumor tissues by liquid chromatography-mass spectrometry. Nat Protoc 13, 1632-1661, doi:10.1038/s41596-018-0006-9 (2018). 430 Chen, Z. W., Fuchs, K., Sieghart, W., Townsend, R. R. & Evers, A. S. Deep amino acid sequencing of native brain GABAA receptors using high-resolution mass spectrometry. Mol Cell Proteomics 11, M111 011445 (2012). 431 Werner, T. et al. Ion coalescence of neutron encoded TMT 10-plex reporter ions. Anal Chem 86, 3594-3601, doi:10.1021/ac500140s (2014). 432 Perkins, D. N., Pappin, D. J., Creasy, D. M. & Cottrell, J. S. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 20, 3551-3567, doi:10.1002/(SICI)1522- 2683(19991201)20:18<3551::AID-ELPS3551>3.0.CO;2-2 (1999). 433 Koskinen, V. R., Emery, P. A., Creasy, D. M. & Cottrell, J. S. Hierarchical clustering of shotgun proteomics data. Mol Cell Proteomics 10, M110 003822, doi:10.1074/mcp.M110.003822 (2011). 434 Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43, e47, doi:10.1093/nar/gkv007 (2015). 435 Benaglia, T., Chauveau, D., Hunter, D. R. & Young, D. mixtools: AnRPackage for Analyzing Finite Mixture Models. Journal of Statistical Software 32, doi:10.18637/jss.v032.i06 (2009). 436 Wickham, H. et al. Welcome to the Tidyverse. Journal of Open Source Software 4, doi:10.21105/joss.01686 (2019). 437 Dean, R. B. & Dixon, W. J. Simplified Statistics for Small Numbers of Observations. Anal Chem 23, 636-638, doi:DOI 10.1021/ac60052a025 (1951). 438 Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118-127, doi:10.1093/biostatistics/kxj037 (2007). 439 Allin, M. P. et al. White matter and cognition in adults who were born preterm. PLoS One 6, e24525, doi:10.1371/journal.pone.0024525 (2011). 440 Tsimis, M. E. et al. Risk factors for periventricular white matter injury in very low birthweight neonates. Am J Obstet Gynecol 214, 380 e381-386, doi:10.1016/j.ajog.2015.09.108 (2016). 441 Bastiani, M. et al. Automated processing pipeline for neonatal diffusion MRI in the developing Human Connectome Project. Neuroimage 185, 750-763, doi:10.1016/j.neuroimage.2018.05.064 (2019).

223 442 Kim, S. J. et al. Effects of MR parameter changes on the quantification of diffusion anisotropy and apparent diffusion coefficient in diffusion tensor imaging: evaluation using a diffusional anisotropic phantom. Korean J Radiol 16, 297-303, doi:10.3348/kjr.2015.16.2.297 (2015). 443 Smyser, T. A. et al. Cortical Gray and Adjacent White Matter Demonstrate Synchronous Maturation in Very Preterm Infants. Cereb Cortex 26, 3370-3378, doi:10.1093/cercor/bhv164 (2016). 444 Huppi, P. S. et al. Microstructural development of human newborn cerebral white matter assessed in vivo by diffusion tensor magnetic resonance imaging. Pediatr Res 44, 584- 590, doi:10.1203/00006450-199810000-00019 (1998). 445 Wu, D. et al. Mapping the critical gestational age at birth that alters brain development in preterm-born infants using multi-modal MRI. Neuroimage 149, 33-43, doi:10.1016/j.neuroimage.2017.01.046 (2017). 446 Thompson, D. K. et al. Characterization of the corpus callosum in very preterm and full- term infants utilizing MRI. Neuroimage 55, 479-490, doi:10.1016/j.neuroimage.2010.12.025 (2011). 447 Thompson, D. K. et al. Alterations in the optic radiations of very preterm children- Perinatal predictors and relationships with visual outcomes. NeuroImage. Clinical 4, 145- 153, doi:10.1016/j.nicl.2013.11.007 (2014). 448 Bockhorst, K. H. et al. Early postnatal development of rat brain: in vivo diffusion tensor imaging. J Neurosci Res 86, 1520-1528, doi:10.1002/jnr.21607 (2008). 449 Khwaja, O. & Volpe, J. J. Pathogenesis of cerebral white matter injury of prematurity. Arch Dis Child Fetal Neonatal Ed 93, F153-161, doi:10.1136/adc.2006.108837 (2008). 450 Akbari, S. H. et al. Periventricular hyperintensity in children with hydrocephalus. Pediatr Radiol 45, 1189-1197, doi:10.1007/s00247-015-3298-8 (2015). 451 Drobyshevsky, A. et al. Serial diffusion tensor imaging detects white matter changes that correlate with motor outcome in premature infants. Dev Neurosci 29, 289-301, doi:10.1159/000105470 (2007). 452 Thompson, D. K. et al. Corpus callosum alterations in very preterm infants: Perinatal correlates and 2year neurodevelopmental outcomes. NeuroImage 59, 3571-3581, doi:10.1016/j.neuroimage.2011.11.057 (2012). 453 Sarnat, H. B. Cerebral dysgenesis : embryology and clinical expression. (Oxford University Press, 1992). 454 Maxwell, W. L. Histopathological changes at central nodes of Ranvier after stretch- injury. Microscopy research and technique 34, 522-535, doi:Doi 10.1002/(Sici)1097- 0029(19960815)34:6<522::Aid-Jemt4>3.0.Co;2-L (1996). 455 Sun, W. et al. Paranodal myelin damage after acute stretch in Guinea pig spinal cord. J Neurotrauma 29, 611-619, doi:10.1089/neu.2011.2086 (2012). 456 Song, S. K. et al. Demyelination increases radial diffusivity in corpus callosum of mouse brain. Neuroimage 26, 132-140, doi:10.1016/j.neuroimage.2005.01.028 (2005). 457 Castejon, O. J., Castejon, H. V. & Castellano, A. Oligodendroglial cell damage and demyelination in infant hydrocephalus. An electron microscopic study. J Submicrosc Cytol Pathol 33, 33-40 (2001). 458 Ayannuga, O. A. & Naicker, T. Cortical Oligodendrocytes in Kaolin-Induced Hydrocephalus in Wistar Rat: Impact of Degree and Duration of Ventriculomegaly. Ann Neurosci 24, 164-172, doi:10.1159/000477154 (2017).

224 459 Di Curzio, D. L., Buist, R. J. & Del Bigio, M. R. Reduced subventricular zone proliferation and white matter damage in juvenile ferrets with kaolin-induced hydrocephalus. Exp Neurol 248, 112-128, doi:10.1016/j.expneurol.2013.06.004 (2013). 460 Goto, J., Tezuka, T., Nakazawa, T., Sagara, H. & Yamamoto, T. Loss of Fyn tyrosine kinase on the C57BL/6 genetic background causes hydrocephalus with defects in oligodendrocyte development. Molecular and cellular neurosciences 38, 203-212, doi:10.1016/j.mcn.2008.02.009 (2008). 461 Aisengart, B., Kajiwara, J. K., Verissimo Meira, K. & da Silva Lopes, L. Morphometric analysis of the optic nerve in experimental hydrocephalus-induced rats. Pediatr Neurosurg 47, 342-348, doi:10.1159/000337728 (2011). 462 Miller, J. M. & McAllister, J. P., 2nd. Reduction of astrogliosis and microgliosis by cerebrospinal fluid shunting in experimental hydrocephalus. Cerebrospinal Fluid Res 4, 5, doi:10.1186/1743-8454-4-5 (2007). 463 Coletti, A. M. et al. Characterization of the ventricular-subventricular stem cell niche during human brain development. Development 145, doi:10.1242/dev.170100 (2018). 464 McMenamin, J. B., Shackelford, G. D. & Volpe, J. J. Outcome of neonatal intraventricular hemorrhage with periventricular echodense lesions. Ann Neurol 15, 285- 290, doi:10.1002/ana.410150315 (1984). 465 Bassan, H. et al. Periventricular hemorrhagic infarction: risk factors and neonatal outcome. Pediatr Neurol 35, 85-92, doi:10.1016/j.pediatrneurol.2006.03.005 (2006). 466 Nagashima, T., Tamaki, N., Matsumoto, S., Horwitz, B. & Seguchi, Y. Biomechanics of hydrocephalus: a new theoretical model. Neurosurgery 21, 898-904, doi:10.1227/00006123-198712000-00019 (1987). 467 Pena, A., Bolton, M. D., Whitehouse, H. & Pickard, J. D. Effects of brain ventricular shape on periventricular biomechanics: a finite-element analysis. Neurosurgery 45, 107- 116; discussion 116-108, doi:10.1097/00006123-199907000-00026 (1999). 468 Kim, H. et al. Porohyperelastic anatomical models for hydrocephalus and idiopathic intracranial hypertension. J Neurosurg 122, 1330-1340, doi:10.3171/2014.12.JNS14516 (2015). 469 Rogers, C. E. et al. Neonatal Amygdala Functional Connectivity at Rest in Healthy and Preterm Infants and Early Internalizing Symptoms. Journal of the American Academy of Child and Adolescent Psychiatry 56, 157-166, doi:10.1016/j.jaac.2016.11.005 (2017). 470 Rogers, C. E. et al. Regional white matter development in very preterm infants: perinatal predictors and early developmental outcomes. Pediatr Res 79, 87-95, doi:10.1038/pr.2015.172 (2016). 471 van Kooij, B. J. et al. Neonatal tract-based spatial statistics findings and outcome in preterm infants. AJNR Am J Neuroradiol 33, 188-194, doi:10.3174/ajnr.A2723 (2012). 472 Counsell, S. J. et al. Specific relations between neurodevelopmental abilities and white matter microstructure in children born preterm. Brain 131, 3201-3208, doi:10.1093/brain/awn268 (2008). 473 Schafer, R. J. et al. Alterations in functional connectivity for language in prematurely born adolescents. Brain 132, 661-670, doi:10.1093/brain/awn353 (2009). 474 Nosarti, C. et al. Neural substrates of letter fluency processing in young adults who were born very preterm: alterations in frontal and striatal regions. NeuroImage 47, 1904-1913, doi:10.1016/j.neuroimage.2009.04.041 (2009).

225 475 De Bruine, F. T. et al. Tractography of white-matter tracts in very preterm infants: a 2- year follow-up study. Dev Med Child Neurol 55, 427-433, doi:10.1111/dmcn.12099 (2013). 476 Kidokoro, H. et al. Brain injury and altered brain growth in preterm infants: predictors and prognosis. Pediatrics 134, e444-453, doi:10.1542/peds.2013-2336 (2014). 477 Lin, T. H. et al. Noninvasive Quantification of Axonal Loss in the Presence of Tissue Swelling in Traumatic Spinal Cord Injury Mice. J Neurotrauma, doi:10.1089/neu.2018.6016 (2019). 478 Edgin, J. O. et al. Executive functioning in preschool children born very preterm: relationship with early white matter pathology. J Int Neuropsychol Soc 14, 90-101, doi:10.1017/S1355617708080053 (2008). 479 Woodward, L. J., Anderson, P. J., Austin, N. C., Howard, K. & Inder, T. E. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med 355, 685- 694, doi:10.1056/NEJMoa053792 (2006). 480 Woodward, L. J., Clark, C. A., Bora, S. & Inder, T. E. Neonatal white matter abnormalities an important predictor of neurocognitive outcome for very preterm children. PLoS One 7, e51879, doi:10.1371/journal.pone.0051879 (2012). 481 Miller, M. Neonatal care for premature infants. The Hastings Center report 35, 4; author reply 7 (2005). 482 Boardman, J. P. et al. A common neonatal image phenotype predicts adverse neurodevelopmental outcome in children born preterm. Neuroimage 52, 409-414, doi:10.1016/j.neuroimage.2010.04.261 (2010). 483 Tissir, F. et al. Lack of cadherins Celsr2 and Celsr3 impairs ependymal ciliogenesis, leading to fatal hydrocephalus. Nat Neurosci 13, 700-707, doi:10.1038/nn.2555 (2010). 484 Sarnat, H. B. Cerebral Dysgenesis: Embryology and Clinical Expression. Vol. 1 (Oxford University Press, 1992). 485 Kinney, H. C., Karthigasan, J., Borenshteyn, N. I., Flax, J. D. & Kirschner, D. A. Myelination in the developing human brain: biochemical correlates. Neurochem Res 19, 983-996, doi:10.1007/bf00968708 (1994). 486 Soul, J. S., Eichenwald, E., Walter, G., Volpe, J. J. & du Plessis, A. J. CSF removal in infantile posthemorrhagic hydrocephalus results in significant improvement in cerebral hemodynamics. Pediatr Res 55, 872-876, doi:10.1203/01.PDR.0000119370.21770.AC (2004). 487 Hagberg, H., Peebles, D. & Mallard, C. Models of white matter injury: comparison of infectious, hypoxic-ischemic, and excitotoxic insults. Ment Retard Dev Disabil Res Rev 8, 30-38, doi:10.1002/mrdd.10007 (2002). 488 Strahle, J. M. et al. Impaired hippocampal development and outcomes in very preterm infants with perinatal brain injury. NeuroImage. Clinical 22, 101787, doi:10.1016/j.nicl.2019.101787 (2019). 489 Cuestas, E., Bas, J. & Pautasso, J. Sex differences in intraventricular hemorrhage rates among very low birth weight newborns. Gend Med 6, 376-382, doi:10.1016/j.genm.2009.06.001 (2009). 490 Tioseco, J. A., Aly, H., Essers, J., Patel, K. & El-Mohandes, A. A. Male sex and intraventricular hemorrhage. Pediatr Crit Care Med 7, 40-44, doi:10.1097/01.pcc.0000192341.67078.61 (2006).

226 491 Jezzard, P. & Balaban, R. S. Correction for geometric distortion in echo planar images from B0 field variations. Magn Reson Med 34, 65-73, doi:10.1002/mrm.1910340111 (1995). 492 Mathur, A. M., Neil, J. J., McKinstry, R. C. & Inder, T. E. Transport, monitoring, and successful brain MR imaging in unsedated neonates. Pediatr Radiol 38, 260-264, doi:10.1007/s00247-007-0705-9 (2008). 493 Woolrich, M. W. et al. Bayesian analysis of neuroimaging data in FSL. Neuroimage 45, S173-186, doi:10.1016/j.neuroimage.2008.10.055 (2009). 494 Smith, S. M. et al. Advances in functional and structural MR image analysis and implementation as FSL. Neuroimage 23 Suppl 1, S208-219, doi:10.1016/j.neuroimage.2004.07.051 (2004). 495 Andersson, J. L. R., Skare, S. & Ashburner, J. How to correct susceptibility distortions in spin-echo echo-planar images: application to diffusion tensor imaging. Neuroimage 20, 870-888, doi:10.1016/S1053-8119(03)00336-7 (2003). 496 Jenkinson, M., Bannister, P., Brady, M. & Smith, S. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage 17, 825-841, doi:10.1016/s1053-8119(02)91132-8 (2002). 497 Jenkinson, M. & Smith, S. A global optimisation method for robust affine registration of brain images. Med Image Anal 5, 143-156, doi:10.1016/s1361-8415(01)00036-6 (2001). 498 Kroenke, C. D., Bretthorst, G. L., Inder, T. E. & Neil, J. J. Modeling water diffusion anisotropy within fixed newborn primate brain using Bayesian probability theory. Magn Reson Med 55, 187-197, doi:10.1002/mrm.20728 (2006). 499 Akbari, S. H. et al. Surgical management of complex multiloculated hydrocephalus in infants and children. Childs Nerv Syst 31, 243-249, doi:10.1007/s00381-014-2596-z (2015). 500 R: A language and environment for statistical computing v. 3.5.0 (R Foundation for Statistical Computing, Vienna, Australia, 2018). 501 Wilke, G. A. & Bubeck Wardenburg, J. Role of a disintegrin and metalloprotease 10 in Staphylococcus aureus alpha-hemolysin-mediated cellular injury. Proc Natl Acad Sci U S A 107, 13473-13478, doi:10.1073/pnas.1001815107 (2010). 502 Surewaard, B. G. J. et al. alpha-Toxin Induces Platelet Aggregation and Liver Injury during Staphylococcus aureus Sepsis. Cell Host Microbe 24, 271-284 e273, doi:10.1016/j.chom.2018.06.017 (2018). 503 Dittmer, A., Hohlfeld, K., Lutzkendorf, J., Muller, L. P. & Dittmer, J. Human mesenchymal stem cells induce E-cadherin degradation in breast carcinoma spheroids by activating ADAM10. Cellular and molecular life sciences : CMLS 66, 3053-3065, doi:10.1007/s00018-009-0089-0 (2009). 504 Malinverno, M. et al. Synaptic localization and activity of ADAM10 regulate excitatory synapses through N-cadherin cleavage. J Neurosci 30, 16343-16355, doi:10.1523/JNEUROSCI.1984-10.2010 (2010). 505 Maretzky, T. et al. ADAM10-mediated E-cadherin release is regulated by proinflammatory cytokines and modulates keratinocyte cohesion in eczematous dermatitis. The Journal of investigative dermatology 128, 1737-1746, doi:10.1038/sj.jid.5701242 10.1038/sj.jid.5701242. Epub 2008 Jan 17. (2008).

227 506 Reyat, J. S. et al. ADAM10-Interacting Tetraspanins Tspan5 and Tspan17 Regulate VE- Cadherin Expression and Promote T Lymphocyte Transmigration. J Immunol 199, 666- 676, doi:10.4049/jimmunol.1600713 (2017). 507 Solanas, G., Cortina, C., Sevillano, M. & Batlle, E. Cleavage of E-cadherin by ADAM10 mediates epithelial cell sorting downstream of EphB signalling. Nat Cell Biol 13, 1100- 1107, doi:10.1038/ncb2298 (2011). 508 Roessmann, U., Velasco, M. E., Sindely, S. D. & Gambetti, P. Glial fibrillary acidic protein (GFAP) in ependymal cells during development. An immunocytochemical study. Brain research 200, 13-21, doi:10.1016/0006-8993(80)91090-2 (1980). 509 Bruni, J., Delbigio, M. & Clattenburg, R. Ependyma: Normal and pathological. A review of the literature☆. Brain Research Reviews 9, 1-19, doi:10.1016/0165-0173(85)90016-5 (1985). 510 Sarnat, H. B. Regional differentiation of the human fetal ependyma: immunocytochemical markers. J Neuropathol Exp Neurol 51, 58-75, doi:10.1097/00005072-199201000-00008 (1992). 511 Reiss, K. et al. ADAM10 cleavage of N-cadherin and regulation of cell-cell adhesion and beta-catenin nuclear signalling. EMBO J 24, 742-752, doi:10.1038/sj.emboj.7600548 (2005). 512 Barsoum, I. B. et al. Hypoxia induces escape from innate immunity in cancer cells via increased expression of ADAM10: role of nitric oxide. Cancer Res 71, 7433-7441, doi:10.1158/0008-5472.CAN-11-2104 (2011). 513 Vincent, B. Regulation of the alpha-secretase ADAM10 at transcriptional, translational and post-translational levels. Brain Res Bull 126, 154-169, doi:10.1016/j.brainresbull.2016.03.020 (2016). 514 Endres, K. & Fahrenholz, F. Regulation of alpha-secretase ADAM10 expression and activity. Experimental brain research 217, 343-352, doi:10.1007/s00221-011-2885-7 (2012). 515 Maretzky, T. et al. ADAM10 mediates E-cadherin shedding and regulates epithelial cell- cell adhesion, migration, and beta-catenin translocation. Proceedings of the National Academy of Sciences of the United States of America 102, 9182-9187, doi:10.1073/pnas.0500918102 (2005). 516 Powers, M. E., Kim, H. K., Wang, Y. & Bubeck Wardenburg, J. ADAM10 mediates vascular injury induced by Staphylococcus aureus alpha-hemolysin. J Infect Dis 206, 352-356, doi:10.1093/infdis/jis192 (2012). 517 Fukumizu, M., Takashima, S. & Becker, L. E. Glial reaction in periventricular areas of the brainstem in fetal and neonatal posthemorrhagic hydrocephalus and congenital hydrocephalus. Brain & development 18, 40-45 (1996). 518 Guerra, M. M. et al. Cell Junction Pathology of Neural Stem Cells Is Associated With Ventricular Zone Disruption, Hydrocephalus, and Abnormal Neurogenesis. J Neuropathol Exp Neurol 74, 653-671, doi:10.1097/NEN.0000000000000203 (2015). 519 Delgehyr, N. et al. Ependymal cell differentiation, from monociliated to multiciliated cells. Methods Cell Biol 127, 19-35, doi:10.1016/bs.mcb.2015.01.004 (2015). 520 Gould, S. J., Howard, S. & Papadaki, L. The development of ependyma in the human fetal brain: an immunohistological and electron microscopic study. Brain research. Developmental brain research 55, 255-267, doi:10.1016/0165-3806(90)90207-f (1990).

228 521 Feldner, A. et al. Loss of Mpdz impairs ependymal cell integrity leading to perinatal- onset hydrocephalus in mice. 9, 890-905, doi:10.15252/emmm.201606430 (2017). 522 Klezovitch, O., Fernandez, T. E., Tapscott, S. J. & Vasioukhin, V. Loss of cell polarity causes severe brain dysplasia in Lgl1 knockout mice. Genes & development 18, 559-571, doi:10.1101/gad.1178004 (2004). 523 Rasin, M. R. et al. Numb and Numbl are required for maintenance of cadherin-based adhesion and polarity of neural progenitors. Nature neuroscience 10, 819-827, doi:10.1038/nn1924 (2007). 524 Jouannet, S. et al. TspanC8 tetraspanins differentially regulate the cleavage of ADAM10 substrates, Notch activation and ADAM10 membrane compartmentalization. Cellular and molecular life sciences : CMLS 73, 1895-1915, doi:10.1007/s00018-015-2111-z (2016). 525 Mechtersheimer, S. et al. Ectodomain shedding of L1 adhesion molecule promotes cell migration by autocrine binding to integrins. The Journal of cell biology 155, 661-673, doi:10.1083/jcb.200101099 (2001). 526 Saftig, P. & Reiss, K. The "A Disintegrin And Metalloproteases" ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol 90, 527-535, doi:10.1016/j.ejcb.2010.11.005 (2011). 527 Morales, D. M. et al. Alterations in protein regulators of neurodevelopment in the cerebrospinal fluid of infants with posthemorrhagic hydrocephalus of prematurity. Molecular & cellular proteomics : MCP 11, M111.011973, doi:10.1074/mcp.M111.011973 (2012). 528 Limbrick, D. D., Jr. et al. Cerebrospinal fluid biomarkers of infantile congenital hydrocephalus. PLoS One 12, e0172353, doi:10.1371/journal.pone.0172353 (2017). 529 Muniz-Talavera, H. & Schmidt, J. V. The mouse Jhy gene regulates ependymal cell differentiation and ciliogenesis. 12, e0184957, doi:10.1371/journal.pone.0184957 (2017). 530 Wolforth, J. Methods of Blood Collection in the Mouse. Lab Animal 29, 47-53 (2000). 531 Xue, M., Balasubramaniam, J., Buist, R. J., Peeling, J. & Del Bigio, M. R. Periventricular/intraventricular hemorrhage in neonatal mouse cerebrum. J Neuropathol Exp Neurol 62, 1154-1165 (2003). 532 Zhu, W. et al. Mouse models of intracerebral hemorrhage in ventricle, cortex, and hippocampus by injections of autologous blood or collagenase. PLoS One 9, e97423, doi:10.1371/journal.pone.0097423 (2014). 533 Aquilina, K., Chakkarapani, E., Love, S. & Thoresen, M. Neonatal rat model of intraventricular haemorrhage and post-haemorrhagic ventricular dilatation with long-term survival into adulthood. Neuropathol Appl Neurobiol 37, 156-165 (2011). 534 Chen, Z. et al. Neuroprotective effects of edaravone after intraventricular hemorrhage in rats. Neuroreport 25, 635-640, doi:10.1097/WNR.0000000000000050 (2014). 535 Garton, T. P. et al. Hemoglobin-induced neuronal degeneration in the hippocampus after neonatal intraventricular hemorrhage. Brain research 1635, 86-94, doi:10.1016/j.brainres.2015.12.060 (2016). 536 Gu, W., Li, F., Zhang, W. & Jia, P. Expression and significance of aquaporin protein in sprague-dawley rats after experimental intraventricular hemorrhage. Cell Mol Biol (Noisy-le-grand) 62, 59-62 (2016).

229 537 Sugimoto, T., Yasuhara, A. & Matsumura, T. Intracranial hemorrhage following administration of sodium bicarbonate in rabbits. Brain Dev 3, 297-303 (1981). 538 Lorenzo, A. V., Welch, K. & Conner, S. Spontaneous germinal matrix and intraventricular hemorrhage in prematurely born rabbits. J Neurosurg 56, 404-410, doi:10.3171/jns.1982.56.3.0404 (1982). 539 Karwacki, Z., Kowianski, P., Morys, J., Dziewiatkowski, J. & Suchorzewska, J. The influence of the intraventricular hemorrhage on the intracranial pressure and hemodynamic changes in the experimental intracerebral hemorrhage in the rabbits. Folia Neuropathol 38, 55-59 (2000). 540 Dohare, P. et al. AMPA-Kainate Receptor Inhibition Promotes Neurologic Recovery in Premature Rabbits with Intraventricular Hemorrhage. J Neurosci 36, 3363-3377, doi:10.1523/JNEUROSCI.4329-15.2016 (2016). 541 Rudy, T. A., Westergaard, J. L. & Yaksh, T. L. Hyperthermia produced by simulated intraventricular hemorrhage in the cat. Exp Neurol 58, 296-310 (1978). 542 Ment, L. R., Stewart, W. B. & Duncan, C. C. Local cerebral glucose utilization in the beagle puppy model of intraventricular hemorrhage. J Neurosurg 57, 384-389, doi:10.3171/jns.1982.57.3.0384 (1982). 543 Ment, L. R., Stewart, W. B., Duncan, C. C. & Lambrecht, R. Beagle puppy model of intraventricular hemorrhage. J Neurosurg 57, 219-223, doi:10.3171/jns.1982.57.2.0219 (1982). 544 Ment, L. R., Stewart, W. B., Duncan, C. C., Scott, D. T. & Lambrecht, R. Beagle puppy model of intraventricular hemorrhage. Effect of indomethacin on cerebral blood flow. J Neurosurg 58, 857-862, doi:10.3171/jns.1983.58.6.0857 (1983). 545 Ment, L. R., Stewart, W. B. & Duncan, C. C. Beagle puppy model of intraventricular hemorrhage: ethamsylate studies. Prostaglandins 27, 245-256 (1984). 546 Ment, L. R., Stewart, W. B. & Duncan, C. C. Beagle puppy model of intraventricular hemorrhage. Effect of superoxide dismutase on cerebral blood flow and prostaglandins. J Neurosurg 62, 563-569, doi:10.3171/jns.1985.62.4.0563 (1985). 547 Johnson, D. L., Getson, P., Shaer, C. & O'Donnell, R. Intraventricular hemorrhage in the newborn beagle puppy. A limited model of intraventricular hemorrhage in the premature infant. Pediatr Neurosci 13, 78-83 (1987). 548 Ment, L. R., Stewart, W. B., Petroff, O. A. & Duncan, C. C. Thromboxane synthesis inhibitor in a beagle pup model of perinatal asphyxia. Stroke 20, 809-814 (1989). 549 Farstad, T., Odden, J. P. & Bratlid, D. Effect of intraventricular hemorrhage on pulmonary function in newborn piglets. Biol Neonate 66, 238-246 (1994). 550 Tang, T. et al. In vivo quantification of intraventricular hemorrhage in a neonatal piglet model using an EEG-layout based electrical impedance tomography array. Physiol Meas 37, 751-764, doi:10.1088/0967-3334/37/6/751 (2016). 551 Clancy, B., Finlay, B. L., Darlington, R. B. & Anand, K. J. Extrapolating brain development from experimental species to humans. NeuroToxicology 28, 931-937 (2007). 552 Workman, A. D., Charvet, C. J., Clancy, B., Darlington, R. B. & Finlay, B. L. Modeling transformations of neurodevelopmental sequences across mammalian species. J Neurosci 33, 7368-7383, doi:10.1523/JNEUROSCI.5746-12.2013 (2013). 553 Chua, C. O. et al. Consequences of intraventricular hemorrhage in a rabbit pup model. Stroke 40, 3369-3377 (2009).

230 554 Scafidi, J., Fagel, D. M., Ment, L. R. & Vaccarino, F. M. Modeling premature brain injury and recovery. Int J Dev Neurosci 27, 863-871, doi:10.1016/j.ijdevneu.2009.05.009 (2009). 555 Christian, E. A. et al. Trends in hospitalization of preterm infants with intraventricular hemorrhage and hydrocephalus in the United States, 2000-2010. J Neurosurg Pediatr 17, 260-269, doi:10.3171/2015.7.PEDS15140 (2016). 556 Fukumizu, M., Takashima, S. & Becker, L. E. Neonatal posthemorrhagic hydrocephalus: neuropathologic and immunohistochemical studies. Pediatric Neurology 13, 230-234 (1995). 557 Fukumizu, M., Takashima, S. & Becker, L. E. Glial reaction in periventricular areas of the brainstem in fetal and neonatal posthemorrhagic hydrocephalus and congenital hydrocephalus. Brain and Development 18, 40-45 (1996). 558 Ballabh, P. Pathogenesis and prevention of intraventricular hemorrhage. Clin Perinatol 41, 47-67, doi:10.1016/j.clp.2013.09.007 (2014). 559 Ballabh, P. Intraventricular hemorrhage in premature infants - mechanism of disease. Pediatric Research 67, 1-8 (2010). 560 Guerra, M. et al. Defects in cell-cell junctions lead to neuroepithelial/ependymal denudation in the telencephalon of human hydrocephalic foetuses. Cerebrospinal Fluid Research 7, S56, doi:10.1186/1743-8454-7-s1-s56 (2010). 561 Di Curzio, D. L., Buist, R. J. & Del Bigio, M. R. Reduced subventricular zone proliferation and white matter damage in juvenile ferrets with kaolin-induced hydrocephalus. Exp Neurol 248C, 112-128, doi:10.1016/j.expneurol.2013.06.004 (2013). 562 Cherian, S. S. et al. Posthemorrhagic ventricular dilation in the neonate: development and characterization of a rat model. J Neuropathol Exp.Neurol. 62, 292-303 (2003). 563 Di Curzio, D. L., Turner-Brannen, E., Mao, X. & Del Bigio, M. R. Magnesium sulfate treatment for juvenile ferrets following induction of hydrocephalus with kaolin. Fluids Barriers CNS 13, 7, doi:10.1186/s12987-016-0031-4 (2016). 564 McAllister, J. P., 2nd et al. An update on research priorities in hydrocephalus: overview of the third National Institutes of Health-sponsored symposium "Opportunities for Hydrocephalus Research: Pathways to Better Outcomes". J Neurosurg 123, 1163-1166, doi:10.3171/2014.12.JNS132352 (2015). 565 Dorner, R. A., Burton, V. J., Allen, M. C., Robinson, S. & Soares, B. P. Preterm neuroimaging and neurodevelopmental outcome: a focus on intraventricular hemorrhage, post-hemorrhagic hydrocephalus, and associated brain injury. J Perinatol, doi:10.1038/s41372-018-0209-5 (2018). 566 Papile, L. A., Munsick-Bruno, G. & Schaefer, A. Relationship of cerebral intraventricular hemorrhage and early childhood neurologic handicaps. J Pediatr 103, 273-277, doi:10.1016/s0022-3476(83)80366-7 (1983). 567 Hasan, K. M. et al. Quantitative diffusion tensor imaging and intellectual outcomes in spina bifida: laboratory investigation. J Neurosurg Pediatr 2, 75-82, doi:10.3171/PED/2008/2/7/075 (2008). 568 McAllister, J. P., 2nd, Abdolvahabi, R. M., Walker, M. L., Mitchell, J. A. & Jones, H. C. Effects of congenital hydrocephalus on the hypothalamic gonadotrophin-releasing hormone system. Neurosurg Focus 22, E4, doi:10.3171/foc.2007.22.4.5 (2007). 569 McAllister, J. P. I. et al. Ventricular zone disruption in human neonates with intraventricular hemorrhage. J Neuropathol Exp Neurol in review (2016).

231 570 Rubio, D. M. et al. Defining translational research: implications for training. Acad Med 85, 470-475, doi:10.1097/ACM.0b013e3181ccd618 (2010). 571 Dagi, T. F. Commentary: Translational Medicine and Neurosurgery: Chance and the Prepared Mind. Neurosurgery 83, E177-E178, doi:10.1093/neuros/nyy352 (2018). 572 Gao, P., Sultan, K. T., Zhang, X. J. & Shi, S. H. Lineage-dependent circuit assembly in the neocortex. Development 140, 2645-2655, doi:10.1242/dev.087668 (2013). 573 Alexander, E., Jr. & Botterell, E. H. Unilateral hydrocephalus resulting from occlusion of foramen of Monro; complication of radical removal of brain abscess. J Neurosurg 6, 197- 206, doi:10.3171/jns.1949.6.3.0197 (1949). 574 Arnold, J. G. Primary ependymitis. Subacute type with occlusion of the foramina of Monro and hydrocephalus of the lateral ventricles. Arch. Neurol. Psychiat., Chicago 32, 143-153 (1934). 575 Johnson, R. T. & Johnson, K. P. Hydrocephalus following viral infection: the pathology of aqueductal stenosis developing after experimental mumps virus infection. J Neuropathol Exp Neurol 27, 591-606 (1968). 576 Johnson, R. T. & Johnson, K. P. Hydrocephalus as a Sequela of Experimental Myxovirus Infections. Exp Mol Pathol 10, 68-&, doi:Doi 10.1016/0014-4800(69)90049-5 (1969). 577 Pang, D., Sclabassi, R. J. & Horton, J. A. Lysis of intraventricular blood clot with urokinase in a canine model: Part 2. In vivo safety study of intraventricular urokinase. Neurosurgery 19, 547-552, doi:10.1227/00006123-198610000-00009 (1986). 578 Emmert, A. S. et al. Impaired neural differentiation and glymphatic CSF flow in the Ccdc39 rat model of neonatal hydrocephalus: genetic interaction with L1cam. Dis Model Mech 12, doi:10.1242/dmm.040972 (2019). 579 Del Bigio, M. R. Hydrocephalus-induced changes in the composition of cerebrospinal fluid. Neurosurgery 25, 416-423, doi:10.1097/00006123-198909000-00016 (1989). 580 Gram, M. et al. Extracellular hemoglobin - mediator of inflammation and cell death in the choroid plexus following preterm intraventricular hemorrhage. J Neuroinflammation 11, 200, doi:10.1186/s12974-014-0200-9 (2014). 581 Miyake, K. Innate immune sensing of pathogens and danger signals by cell surface Toll- like receptors. Semin Immunol 19, 3-10, doi:10.1016/j.smim.2006.12.002 (2007). 582 Sokol, B. et al. Soluble Toll-Like Receptors 2 and 4 in Cerebrospinal Fluid of Patients with Acute Hydrocephalus following Aneurysmal Subarachnoid Haemorrhage. PLoS One 11, e0156171, doi:10.1371/journal.pone.0156171 (2016). 583 Noguchi, M. et al. Interleukin-2 Receptor gamma Chain Mutation Results in X-Linked Severe Combined Immunodeficiency in Humans. Journal of Immunology 181, 5817- 5827 (2008). 584 Noguchi, M. et al. Interleukin-2 Receptor Gamma Chain Mutation Results in X-Linked Severe Combined Immunodeficiency in Humans. Cell 73, 147-157, doi:Doi 10.1016/0092-8674(93)90167-O (1993). 585 de Oliveira, Y. P. A., Pontes-de-Carvalho, L. C., Couto, R. D. & Noronha-Dutra, A. A. Oxidative stress in sepsis. Possible production of free radicals through an erythrocyte- mediated positive feedback mechanism. Brazilian Journal of Infectious Diseases 21, 19- 26, doi:10.1016/j.bjid.2016.11.004 (2017). 586 Zia, M. T. et al. Oxidative-nitrosative stress in a rabbit pup model of germinal matrix hemorrhage: role of NAD(P)H oxidase. Stroke 40, 2191-2198, doi:10.1161/STROKEAHA.108.544759 (2009).

232 587 Savman, K., Nilsson, U. A., Blennow, M., Kjellmer, I. & Whitelaw, A. Non-protein- bound iron is elevated in cerebrospinal fluid from preterm infants with posthemorrhagic ventricular dilatation. Pediatr Res 49, 208-212, doi:10.1203/00006450-200102000-00013 (2001). 588 Lehner, C. et al. Oxidative Stress and Blood-Brain Barrier Dysfunction Under Particular Consideration of Matrix Metalloproteinases. Antioxidants & redox signaling 15, 1305- 1323, doi:10.1089/ars.2011.3923 (2011). 589 Gofton, T. E. & Young, G. B. Sepsis-associated encephalopathy. Nat Rev Neurol 8, 557- 566, doi:10.1038/nrneurol.2012.183 (2012). 590 Widmann, C. N. & Heneka, M. T. Long-term cerebral consequences of sepsis. Lancet Neurol 13, 630-636, doi:10.1016/s1474-4422(14)70017-1 (2014). 591 Nwafor, D. C. et al. Targeting the Blood-Brain Barrier to Prevent Sepsis-Associated Cognitive Impairment. J Cent Nerv Syst Dis 11, 1179573519840652-1179573519840652, doi:10.1177/1179573519840652 (2019). 592 Braun, A. et al. Paucity of pericytes in germinal matrix vasculature of premature infants. J Neurosci 27, 12012-12024, doi:10.1523/jneurosci.3281-07.2007 (2007). 593 Ballabh, P. Intraventricular hemorrhage in premature infants: mechanism of disease. Pediatr Res 67, 1-8, doi:10.1203/PDR.0b013e3181c1b176 (2010). 594 Kamei, A., Houdou, S., Mito, T., Konomi, H. & Takashima, S. Developmental change in type VI collagen in human cerebral vessels. Pediatric Neurology 8, 183-186, doi:10.1016/0887-8994(92)90065-7 (1992). 595 Weller, R. O. & Mitchell, J. Cerebrospinal fluid edema and its sequelae in hydrocephalus. Adv Neurol 28, 111-123 (1980). 596 Del Bigio, M. R., Slobodian, I., Schellenberg, A. E., Buist, R. J. & Kemp-Buors, T. L. Magnetic resonance imaging indicators of blood-brain barrier and brain water changes in young rats with kaolin-induced hydrocephalus. Fluids and Barriers of the CNS 8, 22, doi:10.1186/2045-8118-8-22 (2011). 597 Schaad, U. B., Kaplan, S. L. & McCracken, G. H., Jr. Steroid therapy for bacterial meningitis. Clin Infect Dis 20, 685-690, doi:10.1093/clinids/20.3.685 (1995). 598 McIntyre, P. B. et al. Dexamethasone as adjunctive therapy in bacterial meningitis. A meta-analysis of randomized clinical trials since 1988. JAMA 278, 925-931, doi:10.1001/jama.278.11.925 (1997). 599 Coyle, P. K. Glucocorticoids in central nervous system bacterial infection. Arch Neurol 56, 796-801, doi:10.1001/archneur.56.7.796 (1999). 600 Goodman, S., Sprung, C. L. & International Sepsis, F. The International Sepsis Forum's controversies in sepsis: corticosteroids should be used to treat septic shock. Crit Care 6, 381-383, doi:10.1186/cc1537 (2002). 601 Schaad, U. B. et al. Dexamethasone therapy for bacterial meningitis in children. Swiss Meningitis Study Group. Lancet 342, 457-461, doi:10.1016/0140-6736(93)91592-a (1993). 602 Syrogiannopoulos, G. A. et al. Dexamethasone therapy for bacterial meningitis in children: 2- versus 4-day regimen. The Journal of infectious diseases 169, 853-858, doi:10.1093/infdis/169.4.853 (1994). 603 King, S. M. et al. Dexamethasone therapy for bacterial meningitis: Better never than late? Can J Infect Dis 5, 210-215, doi:10.1155/1994/257198 (1994).

233 604 Wald, E. R. et al. Dexamethasone therapy for children with bacterial meningitis. Meningitis Study Group. Pediatrics 95, 21-28 (1995). 605 Kilpi, T., Peltola, H., Jauhiainen, T. & Kallio, M. J. Oral glycerol and intravenous dexamethasone in preventing neurologic and audiologic sequelae of childhood bacterial meningitis. The Finnish Study Group. Pediatr Infect Dis J 14, 270-278, doi:10.1097/00006454-199504000-00005 (1995). 606 Kanra, G. Y. et al. Beneficial effects of dexamethasone in children with pneumococcal meningitis. Pediatr Infect Dis J 14, 490-494, doi:10.1097/00006454-199506000-00005 (1995). 607 Lebel, M. H. et al. Dexamethasone therapy for bacterial meningitis. Results of two double-blind, placebo-controlled trials. N Engl J Med 319, 964-971, doi:10.1056/NEJM198810133191502 (1988). 608 Lebel, M. H. et al. Magnetic resonance imaging and dexamethasone therapy for bacterial meningitis. Am J Dis Child 143, 301-306, doi:10.1001/archpedi.1989.02150150055017 (1989). 609 Odio, C. M. et al. The beneficial effects of early dexamethasone administration in infants and children with bacterial meningitis. N Engl J Med 324, 1525-1531, doi:10.1056/NEJM199105303242201 (1991). 610 Weiss, M. H. & Nulsen, F. E. The effect of glucocorticoids on CSF flow in dogs. J Neurosurg 32, 452-458, doi:10.3171/jns.1970.32.4.0452 (1970). 611 Sato, O. The effect of dexamethasone on cerebrospinal fluid production rate in the dog. No To Shinkei 19, 485-492 (1967). 612 Whitelaw, A., Christie, S. & Pople, I. Transforming growth factor-beta1: a possible signal molecule for posthemorrhagic hydrocephalus? Pediatr Res 46, 576-580, doi:10.1203/00006450-199911000-00014 (1999). 613 Galbreath, E., Kim, S. J., Park, K., Brenner, M. & Messing, A. Overexpression of TGF- beta 1 in the central nervous system of transgenic mice results in hydrocephalus. J Neuropathol Exp Neurol 54, 339-349, doi:10.1097/00005072-199505000-00007 (1995). 614 Wyss-Coray, T. et al. Increased central nervous system production of extracellular matrix components and development of hydrocephalus in transgenic mice overexpressing transforming growth factor-beta 1. Am J Pathol 147, 53-67 (1995). 615 Kanaji, M., Tada, T. & Kobayashi, S. A murine model of communicating hydrocephalus: Role of TGF-beta1. J Clin Neurosci 4, 51-56, doi:10.1016/s0967-5868(97)90011-4 (1997). 616 Tada, T., Kanaji, M. & Kobayashi, S. Induction of communicating hydrocephalus in mice by intrathecal injection of human recombinant transforming growth factor-beta 1. J Neuroimmunol 50, 153-158, doi:10.1016/0165-5728(94)90041-8 (1994). 617 Tada, T. et al. Intraventricular administration of hepatocyte growth factor treats mouse communicating hydrocephalus induced by transforming growth factor beta1. Neurobiol Dis 21, 576-586, doi:10.1016/j.nbd.2005.09.002 (2006). 618 Hilton, D. J. et al. Cloning and characterization of a binding subunit of the interleukin 13 receptor that is also a component of the interleukin 4 receptor. Proc Natl Acad Sci U S A 93, 497-501, doi:10.1073/pnas.93.1.497 (1996). 619 Jiang, H., Harris, M. B. & Rothman, P. IL-4/IL-13 signaling beyond JAK/STAT. The Journal of allergy and clinical immunology 105, 1063-1070, doi:10.1067/mai.2000.107604 (2000).

234 620 Callard, R. E., Matthews, D. J. & Hibbert, L. IL-4 and IL-13 receptors: are they one and the same? Immunology Today 17, 108-110, doi:10.1016/0167-5699(96)80600-1 (1996). 621 Minty, A. et al. Interleukin-13 is a new human lymphokine regulating inflammatory and immune responses. Nature 362, 248-250, doi:10.1038/362248a0 (1993). 622 Mori, S., Maher, P. & Conti, B. Neuroimmunology of the Interleukins 13 and 4. Brain Sci 6, doi:10.3390/brainsci6020018 (2016). 623 Won, S. Y., Kim, S. R., Maeng, S. & Jin, B. K. Interleukin-13/Interleukin-4-induced oxidative stress contributes to death of prothrombinkringle-2 (pKr-2)-activated microglia. J Neuroimmunol 265, 36-42, doi:10.1016/j.jneuroim.2013.09.014 (2013). 624 Deboy, C. A. et al. Immune-mediated neuroprotection of axotomized mouse facial motoneurons is dependent on the IL-4/STAT6 signaling pathway in CD4(+) T cells. Exp Neurol 201, 212-224, doi:10.1016/j.expneurol.2006.04.028 (2006). 625 Yang, M. S. et al. Interleukin-13 and -4 induce death of activated microglia. Glia 38, 273-280, doi:10.1002/glia.10057 (2002). 626 Nam, J. H. et al. Interleukin-13/-4-induced oxidative stress contributes to death of hippocampal neurons in abeta1-42-treated hippocampus in vivo. Antioxidants & redox signaling 16, 1369-1383, doi:10.1089/ars.2011.4175 (2012). 627 Park, K. W., Baik, H. H. & Jin, B. K. Interleukin-4-induced oxidative stress via microglial NADPH oxidase contributes to the death of hippocampal neurons in vivo. Curr Aging Sci 1, 192-201, doi:10.2174/1874609810801030192 (2008). 628 Cauley, K. A. & Cataltepe, O. Axial diffusivity of the corona radiata correlated with ventricular size in adult hydrocephalus. AJR Am J Roentgenol 203, 170-179, doi:10.2214/AJR.12.10009 (2014). 629 Kulkarni, A. V., Donnelly, R., Mabbott, D. J. & Widjaja, E. Relationship between ventricular size, white matter injury, and neurocognition in children with stable, treated hydrocephalus. J Neurosurg-Pediatr 16, 267-274, doi:10.3171/2015.1.Peds14597 (2015). 630 Pruessmeyer, J. & Ludwig, A. The good, the bad and the ugly substrates for ADAM10 and ADAM17 in brain pathology, inflammation and cancer. Semin Cell Dev Biol 20, 164- 174, doi:10.1016/j.semcdb.2008.09.005 (2009). 631 Chubinskaya, S., Mikhail, R., Deutsch, A. & Tindal, M. H. ADAM-10 protein is present in human articular cartilage primarily in the membrane-bound form and is upregulated in osteoarthritis and in response to IL-1alpha in bovine nasal cartilage. J Histochem Cytochem 49, 1165-1176, doi:10.1177/002215540104900910 (2001). 632 Shackleton, B., Crawford, F. & Bachmeier, C. Inhibition of ADAM10 promotes the clearance of Abeta across the BBB by reducing LRP1 ectodomain shedding. Fluids Barriers CNS 13, 14, doi:10.1186/s12987-016-0038-x (2016). 633 Shackleton, B., Crawford, F. & Bachmeier, C. Inhibition of ADAM10 promotes the clearance of Aβ across the BBB by reducing LRP1 ectodomain shedding. Fluids and Barriers of the CNS 13, 14, doi:10.1186/s12987-016-0038-x (2016). 634 Endres, K. & Fahrenholz, F. Upregulation of the alpha-secretase ADAM10--risk or reason for hope? FEBS J 277, 1585-1596, doi:10.1111/j.1742-4658.2010.07566.x (2010). 635 Lee, H. R. et al. Cilostazol suppresses beta-amyloid production by activating a disintegrin and metalloproteinase 10 via the upregulation of SIRT1-coupled retinoic acid receptor- beta. J Neurosci Res 92, 1581-1590, doi:10.1002/jnr.23421 (2014).

235 636 Morales, D. M. et al. Lumbar cerebrospinal fluid biomarkers of posthemorrhagic hydrocephalus of prematurity: amyloid precursor protein, soluble amyloid precursor protein alpha, and L1 cell adhesion molecule. Neurosurgery In Press (2016). 637 Del Bigio, M. R. Calcium-mediated proteolytic damage in white matter of hydrocephalic rats? J Neuropathol Exp Neurol 59, 946-954, doi:10.1093/jnen/59.11.946 (2000). 638 Vinukonda, G. et al. Epidermal growth factor preserves myelin and promotes astrogliosis after intraventricular hemorrhage. Glia 64, 1987-2004, doi:10.1002/glia.23037 (2016). 639 Vinukonda, G. et al. Hyaluronidase and Hyaluronan Oligosaccharides Promote Neurological Recovery after Intraventricular Hemorrhage. J Neurosci 36, 872-889, doi:10.1523/JNEUROSCI.3297-15.2016 (2016). 640 Vose, L. R. et al. Treatment with thyroxine restores myelination and clinical recovery after intraventricular hemorrhage. J Neurosci 33, 17232-17246, doi:10.1523/JNEUROSCI.2713-13.2013 (2013). 641 Dummula, K. et al. Bone morphogenetic protein inhibition promotes neurological recovery after intraventricular hemorrhage. J Neurosci 31, 12068-12082, doi:10.1523/JNEUROSCI.0013-11.2011 (2011). 642 Vinukonda, G. et al. Neuroprotection in a rabbit model of intraventricular haemorrhage by cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-alpha inhibition. Brain 133, 2264-2280, doi:10.1093/brain/awq107 (2010). 643 Rodríguez, E. M. et al. A cell junction pathology of neural stem cells leads to abnormal neurogenesis and hydrocephalus. Biological research 45, 231-242, doi:10.4067/s0716- 97602012000300005 (2012).

236 8 Appendix

1. Copyright license agreement with Elsevier Science and Technology Journals for use of

article titled “MR diffusion changes in the perimeter of the lateral ventricles demonstrate

periventricular injury in post-hemorrhagic hydrocephalus of prematurity”231

2. Confirmation of a non-exclusive permission granted by the Journal of Neurosurgery-

Pediatrics to use the article titled “Feasibility of fast brain diffusion MRI to quantify

white matter injury in pediatric hydrocephalus”292

3. Copyright license agreement with Oxford University Press for use of the article titled

“MR diffusion changes in the perimeter of the lateral ventricles demonstrate

periventricular injury in post-hemorrhagic hydrocephalus of prematurity”231

4. Confirmation of open access license PLOS ONE through CC-BY for use of the article

titled “Age-specific global epidemiology of hydrocephalus: Systematic review,

metanalysis and global birth surveillance”32

5. Copyright license agreement with Springer for use of article titled “Cerebrospinal Fluid

Biomarkers of Hydrocephalus in Cerebrospinal Fluid Disorders: Lifelong

Implications”248

237 7/23/20, 5:11 AM

Elsevier Science & Technology Journals - License Terms and Conditions

This is a License Agreement between Albert Isaacs ("You") and Elsevier Science & Technology Journals ("Publisher") provided by Copyright Clearance Center ("CCC"). The license consists of your order details, the terms and conditions provided by Elsevier Science & Technology Journals, and the CCC terms and conditions.

All payments must be made in full to CCC.

Order Date 23-Jul-2020 Type of Use Republish in a Order license ID 1050282-1 thesis/dissertation ISSN 2213-1582 Publisher Elsevier Inc Portion Chapter/article

LICENSED CONTENT

Publication Title NeuroImage : Clinical Rightsholder Elsevier Science & Technology Journals Article Title MR di!usion changes in the perimeter of the Publication Type e-Journal lateral ventricles Start Page 102031 demonstrate periventricular injury in Volume 24 post-hemorrhagic hydrocephalus of prematurity. Date 12/31/2011 Language English Country United States of America

REQUEST DETAILS

Portion Type Chapter/article Rights Requested Main product Page range(s) 1-12 Distribution Worldwide Total number of pages 12 Translation Original language of publication Format (select all that Print, Electronic apply) Copies for the disabled? No Who will republish the Academic institution Minor editing privileges? Yes content? Incidental promotional No Duration of Use Life of current edition use? Lifetime Unit Quantity Up to 499 Currency CAD

NEW WORK DETAILS

https://marketplace.copyright.com/rs-ui-web/mp/license/ee483d92-0dd1-454c-b4f3-843b89bf323d/d93cb273-51af-43e9-a4f1-2cb8cdb2acd1 Page 1 of 5 7/23/20, 5:11 AM

Title Pathophysiology and Institution name University of Calgary and Proteogenomics of Washington University in Postinfectious and St. Louis Posthemorrhagic Expected presentation 2020-07-31 Hydrocephalus in Infants date Instructor name Albert Isaacs

ADDITIONAL DETAILS

Order reference number N/A The requesting person / Albert Isaacs organization to appear on the license

REUSE CONTENT DETAILS

Title, description or MR di!usion changes in Title of the MR di!usion changes in numeric reference of the the perimeter of the article/chapter the the perimeter of the portion(s) lateral ventricles portion is from lateral ventricles demonstrate demonstrate periventricular injury in periventricular injury in post-hemorrhagic post-hemorrhagic hydrocephalus of hydrocephalus of prematurity. prematurity. Editor of portion(s) Limbrick, David D.; Author of portion(s) Limbrick, David D.; Shimony, Joshua S.; Yan, Shimony, Joshua S.; Yan, Yan; Rogers, Cynthia E.; Yan; Rogers, Cynthia E.; Zimbalist, Sophia A.; Neil, Zimbalist, Sophia A.; Neil, Je!rey J.; Han, Rowland H.; Je!rey J.; Han, Rowland H.; Alexopoulos, Dimitrios; Alexopoulos, Dimitrios; Lean, Rachel E.; Smyser, Lean, Rachel E.; Smyser, Christopher D.; Isaacs, Christopher D.; Isaacs, Albert M. Albert M. Volume of serial or 24 Issue, if republishing an N/A monograph article from a serial Page or page range of 102031 (1-12) Publication date of 2018-12-31 portion portion

PUBLISHER TERMS AND CONDITIONS

Elsevier publishes Open Access articles in both its Open Access journals and via its Open Access articles option in subscription journals, for which an author selects a user license permitting certain types of reuse without permission. Before proceeding please check if the article is Open Access on http://www.sciencedirect.com and refer to the user license for the individual article. Any reuse not included in the user license terms will require permission. You must always fully and appropriately credit the author and source. If any part of the material to be used (for example, "gures) has appeared in the Elsevier publication for which you are seeking permission, with credit or acknowledgement to another source it is the responsibility of the user to ensure their reuse complies with the terms and conditions determined by the rights holder. Please contact [email protected] with any queries.

CCC Republication Terms and Conditions

https://marketplace.copyright.com/rs-ui-web/mp/license/ee483d92-0dd1-454c-b4f3-843b89bf323d/d93cb273-51af-43e9-a4f1-2cb8cdb2acd1 Page 2 of 5 7/23/20, 5:11 AM

1. Description of Service; De"ned Terms. This Republication License enables the User to obtain licenses for republication of one or more copyrighted works as described in detail on the relevant Order Con"rmation (the "Work(s)"). Copyright Clearance Center, Inc. ("CCC") grants licenses through the Service on behalf of the rightsholder identi"ed on the Order Con"rmation (the "Rightsholder"). "Republication", as used herein, generally means the inclusion of a Work, in whole or in part, in a new work or works, also as described on the Order Con"rmation. "User", as used herein, means the person or entity making such republication.

2. The terms set forth in the relevant Order Con"rmation, and any terms set by the Rightsholder with respect to a particular Work, govern the terms of use of Works in connection with the Service. By using the Service, the person transacting for a republication license on behalf of the User represents and warrants that he/she/it (a) has been duly authorized by the User to accept, and hereby does accept, all such terms and conditions on behalf of User, and (b) shall inform User of all such terms and conditions. In the event such person is a "freelancer" or other third party independent of User and CCC, such party shall be deemed jointly a "User" for purposes of these terms and conditions. In any event, User shall be deemed to have accepted and agreed to all such terms and conditions if User republishes the Work in any fashion.

3. Scope of License; Limitations and Obligations.

3.1. All Works and all rights therein, including copyright rights, remain the sole and exclusive property of the Rightsholder. The license created by the exchange of an Order Con"rmation (and/or any invoice) and payment by User of the full amount set forth on that document includes only those rights expressly set forth in the Order Con"rmation and in these terms and conditions, and conveys no other rights in the Work(s) to User. All rights not expressly granted are hereby reserved.

3.2. General Payment Terms: You may pay by credit card or through an account with us payable at the end of the month. If you and we agree that you may establish a standing account with CCC, then the following terms apply: Remit Payment to: Copyright Clearance Center, 29118 Network Place, Chicago, IL 60673-1291. Payments Due: Invoices are payable upon their delivery to you (or upon our notice to you that they are available to you for downloading). After 30 days, outstanding amounts will be subject to a service charge of 1-1/2% per month or, if less, the maximum rate allowed by applicable law. Unless otherwise speci"cally set forth in the Order Con"rmation or in a separate written agreement signed by CCC, invoices are due and payable on "net 30" terms. While User may exercise the rights licensed immediately upon issuance of the Order Con"rmation, the license is automatically revoked and is null and void, as if it had never been issued, if complete payment for the license is not received on a timely basis either from User directly or through a payment agent, such as a credit card company.

3.3. Unless otherwise provided in the Order Con"rmation, any grant of rights to User (i) is "one-time" (including the editions and product family speci"ed in the license), (ii) is non-exclusive and non-transferable and (iii) is subject to any and all limitations and restrictions (such as, but not limited to, limitations on duration of use or circulation) included in the Order Con"rmation or invoice and/or in these terms and conditions. Upon completion of the licensed use, User shall either secure a new permission for further use of the Work(s) or immediately cease any new use of the Work(s) and shall render inaccessible (such as by deleting or by removing or severing links or other locators) any further copies of the Work (except for copies printed on paper in accordance with this license and still in User's stock at the end of such period).

3.4. In the event that the material for which a republication license is sought includes third party materials (such as photographs, illustrations, graphs, inserts and similar materials) which are identi"ed in such material as having been used by permission, User is responsible for identifying, and seeking separate licenses (under this Service or otherwise) for, any of such third party materials; without a separate license, such third party materials may not be used.

https://marketplace.copyright.com/rs-ui-web/mp/license/ee483d92-0dd1-454c-b4f3-843b89bf323d/d93cb273-51af-43e9-a4f1-2cb8cdb2acd1 Page 3 of 5 7/23/20, 5:11 AM

3.5. Use of proper copyright notice for a Work is required as a condition of any license granted under the Service. Unless otherwise provided in the Order Con"rmation, a proper copyright notice will read substantially as follows: "Republished with permission of [Rightsholder's name], from [Work's title, author, volume, edition number and year of copyright]; permission conveyed through Copyright Clearance Center, Inc. " Such notice must be provided in a reasonably legible font size and must be placed either immediately adjacent to the Work as used (for example, as part of a by-line or footnote but not as a separate electronic link) or in the place where substantially all other credits or notices for the new work containing the republished Work are located. Failure to include the required notice results in loss to the Rightsholder and CCC, and the User shall be liable to pay liquidated damages for each such failure equal to twice the use fee speci"ed in the Order Con"rmation, in addition to the use fee itself and any other fees and charges speci"ed.

3.6. User may only make alterations to the Work if and as expressly set forth in the Order Con"rmation. No Work may be used in any way that is defamatory, violates the rights of third parties (including such third parties' rights of copyright, privacy, publicity, or other tangible or intangible property), or is otherwise illegal, sexually explicit or obscene. In addition, User may not conjoin a Work with any other material that may result in damage to the reputation of the Rightsholder. User agrees to inform CCC if it becomes aware of any infringement of any rights in a Work and to cooperate with any reasonable request of CCC or the Rightsholder in connection therewith.

4. Indemnity. User hereby indemni"es and agrees to defend the Rightsholder and CCC, and their respective employees and directors, against all claims, liability, damages, costs and expenses, including legal fees and expenses, arising out of any use of a Work beyond the scope of the rights granted herein, or any use of a Work which has been altered in any unauthorized way by User, including claims of defamation or infringement of rights of copyright, publicity, privacy or other tangible or intangible property.

5. Limitation of Liability. UNDER NO CIRCUMSTANCES WILL CCC OR THE RIGHTSHOLDER BE LIABLE FOR ANY DIRECT, INDIRECT, CONSEQUENTIAL OR INCIDENTAL DAMAGES (INCLUDING WITHOUT LIMITATION DAMAGES FOR LOSS OF BUSINESS PROFITS OR INFORMATION, OR FOR BUSINESS INTERRUPTION) ARISING OUT OF THE USE OR INABILITY TO USE A WORK, EVEN IF ONE OF THEM HAS BEEN ADVISED OF THE POSSIBILITY OF SUCH DAMAGES. In any event, the total liability of the Rightsholder and CCC (including their respective employees and directors) shall not exceed the total amount actually paid by User for this license. User assumes full liability for the actions and omissions of its principals, employees, agents, a#liates, successors and assigns.

6. Limited Warranties. THE WORK(S) AND RIGHT(S) ARE PROVIDED "AS IS". CCC HAS THE RIGHT TO GRANT TO USER THE RIGHTS GRANTED IN THE ORDER CONFIRMATION DOCUMENT. CCC AND THE RIGHTSHOLDER DISCLAIM ALL OTHER WARRANTIES RELATING TO THE WORK(S) AND RIGHT(S), EITHER EXPRESS OR IMPLIED, INCLUDING WITHOUT LIMITATION IMPLIED WARRANTIES OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. ADDITIONAL RIGHTS MAY BE REQUIRED TO USE ILLUSTRATIONS, GRAPHS, PHOTOGRAPHS, ABSTRACTS, INSERTS OR OTHER PORTIONS OF THE WORK (AS OPPOSED TO THE ENTIRE WORK) IN A MANNER CONTEMPLATED BY USER; USER UNDERSTANDS AND AGREES THAT NEITHER CCC NOR THE RIGHTSHOLDER MAY HAVE SUCH ADDITIONAL RIGHTS TO GRANT.

7. E!ect of Breach. Any failure by User to pay any amount when due, or any use by User of a Work beyond the scope of the license set forth in the Order Con"rmation and/or these terms and conditions, shall be a material breach of the license created by the Order Con"rmation and these terms and conditions. Any breach not cured within 30 days of written notice thereof shall result in immediate termination of such license without further notice. Any unauthorized (but licensable) use of a Work that is terminated immediately upon notice thereof may be liquidated by payment of the Rightsholder's ordinary license price therefor; any unauthorized (and unlicensable) use that is not terminated immediately for any reason (including, for example, because materials containing the Work cannot

https://marketplace.copyright.com/rs-ui-web/mp/license/ee483d92-0dd1-454c-b4f3-843b89bf323d/d93cb273-51af-43e9-a4f1-2cb8cdb2acd1 Page 4 of 5 7/23/20, 5:11 AM

reasonably be recalled) will be subject to all remedies available at law or in equity, but in no event to a payment of less than three times the Rightsholder's ordinary license price for the most closely analogous licensable use plus Rightsholder's and/or CCC's costs and expenses incurred in collecting such payment.

8. Miscellaneous.

8.1. User acknowledges that CCC may, from time to time, make changes or additions to the Service or to these terms and conditions, and CCC reserves the right to send notice to the User by electronic mail or otherwise for the purposes of notifying User of such changes or additions; provided that any such changes or additions shall not apply to permissions already secured and paid for.

8.2. Use of User-related information collected through the Service is governed by CCC's privacy policy, available online here:https://marketplace.copyright.com/rs-ui-web/mp/privacy-policy

8.3. The licensing transaction described in the Order Con"rmation is personal to User. Therefore, User may not assign or transfer to any other person (whether a natural person or an organization of any kind) the license created by the Order Con"rmation and these terms and conditions or any rights granted hereunder; provided, however, that User may assign such license in its entirety on written notice to CCC in the event of a transfer of all or substantially all of User's rights in the new material which includes the Work(s) licensed under this Service.

8.4. No amendment or waiver of any terms is binding unless set forth in writing and signed by the parties. The Rightsholder and CCC hereby object to any terms contained in any writing prepared by the User or its principals, employees, agents or a#liates and purporting to govern or otherwise relate to the licensing transaction described in the Order Con"rmation, which terms are in any way inconsistent with any terms set forth in the Order Con"rmation and/or in these terms and conditions or CCC's standard operating procedures, whether such writing is prepared prior to, simultaneously with or subsequent to the Order Con"rmation, and whether such writing appears on a copy of the Order Con"rmation or in a separate instrument.

8.5. The licensing transaction described in the Order Con"rmation document shall be governed by and construed under the law of the State of New York, USA, without regard to the principles thereof of con$icts of law. Any case, controversy, suit, action, or proceeding arising out of, in connection with, or related to such licensing transaction shall be brought, at CCC's sole discretion, in any federal or state court located in the County of New York, State of New York, USA, or in any federal or state court whose geographical jurisdiction covers the location of the Rightsholder set forth in the Order Con"rmation. The parties expressly submit to the personal jurisdiction and venue of each such federal or state court.If you have any comments or questions about the Service or Copyright Clearance Center, please contact us at 978-750- 8400 or send an e-mail to [email protected].

v 1.1

https://marketplace.copyright.com/rs-ui-web/mp/license/ee483d92-0dd1-454c-b4f3-843b89bf323d/d93cb273-51af-43e9-a4f1-2cb8cdb2acd1 Page 5 of 5 From: permissions [email protected] Subject: Fwd: Permissions Date: July 23, 2020 at 5:07 AM To: [email protected]

[EXTERNAL]

Dr. Isaacs, Non-exclusive permission is granted at no charge for the use you describe, provided proper credit is given as determined by style guidelines of the publisher of the new work, or by some accepted style such as AP or Chicago. Please save this communica@on as proof of permission grant.

Best of luck on your dissertation!

Gillian Shasby

[email protected]

Begin forwarded message:

From: [email protected] Subject: Permissions Date: July 22, 2020 at 5:22:55 PM EDT To: [email protected]

A user submitted the Permissions form with the following values:

Permission Cat: Original Author Seeking Permission to Use Content in non-AANS Publication Article Title: Feasibility of fast brain diffusion MRI to quantify white matter injury in pediatric hydrocephalus Authors: 7. Isaacs AM, Shimony JS, Morales DM, Castaneyra-Ruiz L, Hartman A, Cook M, Smyser CD, Strahle J, Smyth MD, Yan Y, McAllister JP, McKinstry RC, Limbrick DD Publication: Journal of Neurosurgery: Pediatrics Pub Month: Jul Pub Year: 2019 Volume: 24 Issue: 4 Page Range: 353-479 Permission Note: ALL - this paper was published as part of my PhD work. I am hoping to get permission to incorporate the paper in my dissertation Media Type: Print and Electronic Media: Not listed/other Language: English (as originally published) Title Edition: Pathophysiology and Proteogenomics of Post-infectious and Post-hemorrhagic Hydrocephalus in Infants Repub Date: July 31, 2020 Quantity: 10 Publisher: Albert M. Isaacs Translation: Permission Note: this paper was published as part of my PhD work. I am hoping to get permission to incorporate the paper in my dissertation Ref Number: Title: Dr. First Name: Albert Middle Name: M Last Name: Isaacs My Title: Resident - Neurosurgery Institution: Washington University in St. Louis & University of Calgary Address1: 1403 - 29 St NW Address2: City: Calgary State: AB Zip Code: T2N2T9 Country: CA Country: CA Phone: 3149144011 Email: [email protected] Fax: Website: Other Company: Custom Form Id: Permissions Thank you. 7/23/20, 7:29 AM

Oxford University Press - Journals - License Terms and Conditions

This is a License Agreement between Albert Isaacs ("You") and Oxford University Press - Journals ("Publisher") provided by Copyright Clearance Center ("CCC"). The license consists of your order details, the terms and conditions provided by Oxford University Press - Journals, and the CCC terms and conditions.

All payments must be made in full to CCC.

Order Date 23-Jul-2020 Type of Use Republish in a Order license ID 1050324-1 thesis/dissertation ISSN 0022-3069 Publisher American Association of Neuropathologists, Inc. Portion Chapter/article

LICENSED CONTENT

Publication Title Journal of neuropathology Rightsholder Oxford University Press - and experimental Journals neurology Publication Type Journal Article Title Blood Exposure Causes Start Page 803 Ventricular Zone Disruption and Glial End Page 813 Activation In Vitro. Issue 9 Author/Editor American Association of Volume 77 Neuropathologists. Date 12/31/1941 Language English Country United States of America

REQUEST DETAILS

Portion Type Chapter/article Rights Requested Main product Page range(s) 803–813 Distribution Worldwide Total number of pages 10 Translation Original language of publication Format (select all that Print, Electronic apply) Copies for the disabled? No Who will republish the Academic institution Minor editing privileges? Yes content? Incidental promotional No Duration of Use Life of current edition use? Lifetime Unit Quantity Up to 499 Currency CAD

NEW WORK DETAILS

https://marketplace.copyright.com/rs-ui-web/mp/license/24a8bcaf-c6a8-4e40-8baa-ed75875979ff/5ef9fbf5-0500-44d2-8b88-6381e552d9cf Page 1 of 5 7/23/20, 7:29 AM

Title Pathophysiology and Institution name University of Calgary and Proteogenomics of Washington University in Postinfectious and St. Louis Posthemorrhagic Expected presentation 2020-07-31 Hydrocephalus in Infants date Instructor name Albert Isaacs

ADDITIONAL DETAILS

Order reference number N/A The requesting person / Albert Isaacs organization to appear on the license

REUSE CONTENT DETAILS

Title, description or Blood Exposure Causes Title of the Blood Exposure Causes numeric reference of the Ventricular Zone article/chapter the Ventricular Zone portion(s) Disruption and Glial portion is from Disruption and Glial Activation In Vitro. Activation In Vitro. Editor of portion(s) Strahle, Jennifer M; Author of portion(s) Strahle, Jennifer M; Morales, Diego M; Morales, Diego M; McAllister, James P; McAllister, James P; Limbrick, David D; Isaacs, Limbrick, David D; Isaacs, Albert M; Dahiya, Sonika Albert M; Dahiya, Sonika M; Castaneyra-Ruiz, M; Castaneyra-Ruiz, Leandro; Brody, Steven L Leandro; Brody, Steven L Volume of serial or 77 Issue, if republishing an 9 monograph article from a serial Page or page range of 803-813 Publication date of 2018-08-31 portion portion

CCC Republication Terms and Conditions

1. Description of Service; De!ned Terms. This Republication License enables the User to obtain licenses for republication of one or more copyrighted works as described in detail on the relevant Order Con!rmation (the "Work(s)"). Copyright Clearance Center, Inc. ("CCC") grants licenses through the Service on behalf of the rightsholder identi!ed on the Order Con!rmation (the "Rightsholder"). "Republication", as used herein, generally means the inclusion of a Work, in whole or in part, in a new work or works, also as described on the Order Con!rmation. "User", as used herein, means the person or entity making such republication.

2. The terms set forth in the relevant Order Con!rmation, and any terms set by the Rightsholder with respect to a particular Work, govern the terms of use of Works in connection with the Service. By using the Service, the person transacting for a republication license on behalf of the User represents and warrants that he/she/it (a) has been duly authorized by the User to accept, and hereby does accept, all such terms and conditions on behalf of User, and (b) shall inform User of all such terms and conditions. In the event such person is a "freelancer" or other third party independent of User and CCC, such party shall be deemed jointly a "User" for purposes of these terms and conditions. In any event, User shall be deemed to have accepted and agreed to all such terms and conditions if User republishes the Work in any fashion.

https://marketplace.copyright.com/rs-ui-web/mp/license/24a8bcaf-c6a8-4e40-8baa-ed75875979ff/5ef9fbf5-0500-44d2-8b88-6381e552d9cf Page 2 of 5 7/23/20, 7:29 AM

3. Scope of License; Limitations and Obligations.

3.1. All Works and all rights therein, including copyright rights, remain the sole and exclusive property of the Rightsholder. The license created by the exchange of an Order Con!rmation (and/or any invoice) and payment by User of the full amount set forth on that document includes only those rights expressly set forth in the Order Con!rmation and in these terms and conditions, and conveys no other rights in the Work(s) to User. All rights not expressly granted are hereby reserved.

3.2. General Payment Terms: You may pay by credit card or through an account with us payable at the end of the month. If you and we agree that you may establish a standing account with CCC, then the following terms apply: Remit Payment to: Copyright Clearance Center, 29118 Network Place, Chicago, IL 60673-1291. Payments Due: Invoices are payable upon their delivery to you (or upon our notice to you that they are available to you for downloading). After 30 days, outstanding amounts will be subject to a service charge of 1-1/2% per month or, if less, the maximum rate allowed by applicable law. Unless otherwise speci!cally set forth in the Order Con!rmation or in a separate written agreement signed by CCC, invoices are due and payable on "net 30" terms. While User may exercise the rights licensed immediately upon issuance of the Order Con!rmation, the license is automatically revoked and is null and void, as if it had never been issued, if complete payment for the license is not received on a timely basis either from User directly or through a payment agent, such as a credit card company.

3.3. Unless otherwise provided in the Order Con!rmation, any grant of rights to User (i) is "one-time" (including the editions and product family speci!ed in the license), (ii) is non-exclusive and non-transferable and (iii) is subject to any and all limitations and restrictions (such as, but not limited to, limitations on duration of use or circulation) included in the Order Con!rmation or invoice and/or in these terms and conditions. Upon completion of the licensed use, User shall either secure a new permission for further use of the Work(s) or immediately cease any new use of the Work(s) and shall render inaccessible (such as by deleting or by removing or severing links or other locators) any further copies of the Work (except for copies printed on paper in accordance with this license and still in User's stock at the end of such period).

3.4. In the event that the material for which a republication license is sought includes third party materials (such as photographs, illustrations, graphs, inserts and similar materials) which are identi!ed in such material as having been used by permission, User is responsible for identifying, and seeking separate licenses (under this Service or otherwise) for, any of such third party materials; without a separate license, such third party materials may not be used.

3.5. Use of proper copyright notice for a Work is required as a condition of any license granted under the Service. Unless otherwise provided in the Order Con!rmation, a proper copyright notice will read substantially as follows: "Republished with permission of [Rightsholder's name], from [Work's title, author, volume, edition number and year of copyright]; permission conveyed through Copyright Clearance Center, Inc. " Such notice must be provided in a reasonably legible font size and must be placed either immediately adjacent to the Work as used (for example, as part of a by-line or footnote but not as a separate electronic link) or in the place where substantially all other credits or notices for the new work containing the republished Work are located. Failure to include the required notice results in loss to the Rightsholder and CCC, and the User shall be liable to pay liquidated damages for each such failure equal to twice the use fee speci!ed in the Order Con!rmation, in addition to the use fee itself and any other fees and charges speci!ed.

3.6. User may only make alterations to the Work if and as expressly set forth in the Order Con!rmation. No Work may be used in any way that is defamatory, violates the rights of third parties (including such third parties' rights of copyright, privacy, publicity, or other tangible or intangible property), or is otherwise

https://marketplace.copyright.com/rs-ui-web/mp/license/24a8bcaf-c6a8-4e40-8baa-ed75875979ff/5ef9fbf5-0500-44d2-8b88-6381e552d9cf Page 3 of 5 7/23/20, 7:29 AM

illegal, sexually explicit or obscene. In addition, User may not conjoin a Work with any other material that may result in damage to the reputation of the Rightsholder. User agrees to inform CCC if it becomes aware of any infringement of any rights in a Work and to cooperate with any reasonable request of CCC or the Rightsholder in connection therewith.

4. Indemnity. User hereby indemni!es and agrees to defend the Rightsholder and CCC, and their respective employees and directors, against all claims, liability, damages, costs and expenses, including legal fees and expenses, arising out of any use of a Work beyond the scope of the rights granted herein, or any use of a Work which has been altered in any unauthorized way by User, including claims of defamation or infringement of rights of copyright, publicity, privacy or other tangible or intangible property.

5. Limitation of Liability. UNDER NO CIRCUMSTANCES WILL CCC OR THE RIGHTSHOLDER BE LIABLE FOR ANY DIRECT, INDIRECT, CONSEQUENTIAL OR INCIDENTAL DAMAGES (INCLUDING WITHOUT LIMITATION DAMAGES FOR LOSS OF BUSINESS PROFITS OR INFORMATION, OR FOR BUSINESS INTERRUPTION) ARISING OUT OF THE USE OR INABILITY TO USE A WORK, EVEN IF ONE OF THEM HAS BEEN ADVISED OF THE POSSIBILITY OF SUCH DAMAGES. In any event, the total liability of the Rightsholder and CCC (including their respective employees and directors) shall not exceed the total amount actually paid by User for this license. User assumes full liability for the actions and omissions of its principals, employees, agents, a"liates, successors and assigns.

6. Limited Warranties. THE WORK(S) AND RIGHT(S) ARE PROVIDED "AS IS". CCC HAS THE RIGHT TO GRANT TO USER THE RIGHTS GRANTED IN THE ORDER CONFIRMATION DOCUMENT. CCC AND THE RIGHTSHOLDER DISCLAIM ALL OTHER WARRANTIES RELATING TO THE WORK(S) AND RIGHT(S), EITHER EXPRESS OR IMPLIED, INCLUDING WITHOUT LIMITATION IMPLIED WARRANTIES OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. ADDITIONAL RIGHTS MAY BE REQUIRED TO USE ILLUSTRATIONS, GRAPHS, PHOTOGRAPHS, ABSTRACTS, INSERTS OR OTHER PORTIONS OF THE WORK (AS OPPOSED TO THE ENTIRE WORK) IN A MANNER CONTEMPLATED BY USER; USER UNDERSTANDS AND AGREES THAT NEITHER CCC NOR THE RIGHTSHOLDER MAY HAVE SUCH ADDITIONAL RIGHTS TO GRANT.

7. E#ect of Breach. Any failure by User to pay any amount when due, or any use by User of a Work beyond the scope of the license set forth in the Order Con!rmation and/or these terms and conditions, shall be a material breach of the license created by the Order Con!rmation and these terms and conditions. Any breach not cured within 30 days of written notice thereof shall result in immediate termination of such license without further notice. Any unauthorized (but licensable) use of a Work that is terminated immediately upon notice thereof may be liquidated by payment of the Rightsholder's ordinary license price therefor; any unauthorized (and unlicensable) use that is not terminated immediately for any reason (including, for example, because materials containing the Work cannot reasonably be recalled) will be subject to all remedies available at law or in equity, but in no event to a payment of less than three times the Rightsholder's ordinary license price for the most closely analogous licensable use plus Rightsholder's and/or CCC's costs and expenses incurred in collecting such payment.

8. Miscellaneous.

8.1. User acknowledges that CCC may, from time to time, make changes or additions to the Service or to these terms and conditions, and CCC reserves the right to send notice to the User by electronic mail or otherwise for the purposes of notifying User of such changes or additions; provided that any such changes or additions shall not apply to permissions already secured and paid for.

8.2. Use of User-related information collected through the Service is governed by CCC's privacy policy, available online here:https://marketplace.copyright.com/rs-ui-web/mp/privacy-policy

https://marketplace.copyright.com/rs-ui-web/mp/license/24a8bcaf-c6a8-4e40-8baa-ed75875979ff/5ef9fbf5-0500-44d2-8b88-6381e552d9cf Page 4 of 5 7/23/20, 7:29 AM

8.3. The licensing transaction described in the Order Con!rmation is personal to User. Therefore, User may not assign or transfer to any other person (whether a natural person or an organization of any kind) the license created by the Order Con!rmation and these terms and conditions or any rights granted hereunder; provided, however, that User may assign such license in its entirety on written notice to CCC in the event of a transfer of all or substantially all of User's rights in the new material which includes the Work(s) licensed under this Service.

8.4. No amendment or waiver of any terms is binding unless set forth in writing and signed by the parties. The Rightsholder and CCC hereby object to any terms contained in any writing prepared by the User or its principals, employees, agents or a"liates and purporting to govern or otherwise relate to the licensing transaction described in the Order Con!rmation, which terms are in any way inconsistent with any terms set forth in the Order Con!rmation and/or in these terms and conditions or CCC's standard operating procedures, whether such writing is prepared prior to, simultaneously with or subsequent to the Order Con!rmation, and whether such writing appears on a copy of the Order Con!rmation or in a separate instrument.

8.5. The licensing transaction described in the Order Con!rmation document shall be governed by and construed under the law of the State of New York, USA, without regard to the principles thereof of con$icts of law. Any case, controversy, suit, action, or proceeding arising out of, in connection with, or related to such licensing transaction shall be brought, at CCC's sole discretion, in any federal or state court located in the County of New York, State of New York, USA, or in any federal or state court whose geographical jurisdiction covers the location of the Rightsholder set forth in the Order Con!rmation. The parties expressly submit to the personal jurisdiction and venue of each such federal or state court.If you have any comments or questions about the Service or Copyright Clearance Center, please contact us at 978-750- 8400 or send an e-mail to [email protected].

v 1.1

https://marketplace.copyright.com/rs-ui-web/mp/license/24a8bcaf-c6a8-4e40-8baa-ed75875979ff/5ef9fbf5-0500-44d2-8b88-6381e552d9cf Page 5 of 5 From: plosone [email protected] Subject: Re: Permission to use Date: July 24, 2020 at 5:50 AM To: [email protected]

[EXTERNAL]

Dear Dr Isaacs,

Thank you for your message. PLOS ONE publishes all of the content in the articles under an open access license called “CC-BY.” This license allows you to download, reuse, reprint, modify, distribute, and/or copy articles or images in PLOS journals, so long as the original creators are credited (e.g., including the article’s citation and/or the image credit). Additional permissions are not required. You can read about our open access license here: http://journals.plos.org/plosone/s/licenses-and-copyright

There are many ways to access our content, including HTML, XML, and PDF versions of each article. Higher resolution versions of figures can be downloaded directly from the article.

Thank you for your interest in PLOS ONE and for your continued support of the Open Access model. Please do not hesitate to be in touch with any additional questions.

Kind regards, Eoin O'Connor

Editorial Office PLOS | plos.org Empowering researchers to transform science 1160 Battery Street, Suite 225, San Francisco, CA 94111

Case Number: 06722973 ref:_00DU0Ifis._5004P1DJysV:ref

------Original Message ------From: Albert Isaacs [[email protected]] Sent: 23/07/2020 14:12 To: [email protected] Cc: [email protected] Subject: Permission to use

Good morning,

As the first author of the citation below, which was published in PLOS One during my PhD, my thesis advisory committee gave me permission to include the paper (in sections or entirety) in my dissertation. I was wondering if you would be so kind to advise me on how to get your permission to use such material for my thesis. It will not be published by another publisher but will be submitted to the Washington University School of Medicine and the University of Calgary upon completion (July 31, 2020).

https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0204926

Best regards,

Albert M Isaacs, MD PhD Division of Neurosurgery University of Calgary, Calgary, AB

Eoin O'Connor

Editorial Office PLOS | plos.org Empowering researchers to transform science 1160 Battery Street, Suite 225, San Francisco, CA 94111 7/23/20, 4:58 AM

Springer - License Terms and Conditions

This is a License Agreement between Albert M. Isaacs ("You") and Springer ("Publisher") provided by Copyright Clearance Center ("CCC"). The license consists of your order details, the terms and conditions provided by Springer, and the CCC terms and conditions.

All payments must be made in full to CCC.

Order Date 22-Jul-2020 Type of Use Republish in a Order license ID 1050126-1 thesis/dissertation ISBN-13 9783319979274 Publisher Springer International Publishing; Springer International Publishing Portion Chapter/article

LICENSED CONTENT

Publication Title Cerebrospinal Fluid Country Switzerland Disorders : Lifelong Rightsholder Springer Implications Publication Type Book Date 11/19/2018 Language English

REQUEST DETAILS

Portion Type Chapter/article Rights Requested Main product Page range(s) 47-70 Distribution Worldwide Total number of pages 23 Translation Original language of publication Format (select all that Electronic apply) Copies for the disabled? No Who will republish the Academic institution Minor editing privileges? No content? Incidental promotional No Duration of Use Life of current edition use? Lifetime Unit Quantity Up to 499 Currency CAD

NEW WORK DETAILS

Title Pathophysiology and Institution name University of Calgary and Proteogenomics of Post- Washington University in infectious and Post- St. Louis hemorrhagic Expected presentation 2020-07-31 Hydrocephalus in Infants date Instructor name Albert Isaacs

https://marketplace.copyright.com/rs-ui-web/mp/license/680059fb-6ec8-4465-b389-c0718543b84b/db3a1fd8-6dbd-43b1-8262-23e802edf130 Page 1 of 5 7/23/20, 4:58 AM

ADDITIONAL DETAILS

Order reference number N/A The requesting person / Albert M. Isaacs organization to appear on the license

REUSE CONTENT DETAILS

Title, description or Cerebrospinal Fluid Title of the Cerebrospinal Fluid numeric reference of the Biomarkers of article/chapter the Biomarkers of portion(s) Hydrocephalus portion is from Hydrocephalus Editor of portion(s) Limbrick DD & Leonard JR Author of portion(s) Albert M. Isaacs Volume of serial or 1 Issue, if republishing an N/A monograph article from a serial Page or page range of 47-70 Publication date of 2018-11-19 portion portion

PUBLISHER TERMS AND CONDITIONS

A maximum of 10% of the content may be licensed for republication. The user is responsible for identifying and seeking separate licenses for any third party materials that are identi!ed anywhere in the work. Without a separate license, such third party materials may not be reused. If you are placing a request on behalf of/for a corporate organization, please use RightsLink.For further information visit http://www.nature.com/reprints/permission-requests.html and https://www.springer.com/gp/rights-permissions/obtaining-permissions/882

CCC Republication Terms and Conditions

1. Description of Service; De!ned Terms. This Republication License enables the User to obtain licenses for republication of one or more copyrighted works as described in detail on the relevant Order Con!rmation (the "Work(s)"). Copyright Clearance Center, Inc. ("CCC") grants licenses through the Service on behalf of the rightsholder identi!ed on the Order Con!rmation (the "Rightsholder"). "Republication", as used herein, generally means the inclusion of a Work, in whole or in part, in a new work or works, also as described on the Order Con!rmation. "User", as used herein, means the person or entity making such republication.

2. The terms set forth in the relevant Order Con!rmation, and any terms set by the Rightsholder with respect to a particular Work, govern the terms of use of Works in connection with the Service. By using the Service, the person transacting for a republication license on behalf of the User represents and warrants that he/she/it (a) has been duly authorized by the User to accept, and hereby does accept, all such terms and conditions on behalf of User, and (b) shall inform User of all such terms and conditions. In the event such person is a "freelancer" or other third party independent of User and CCC, such party shall be deemed jointly a "User" for purposes of these terms and conditions. In any event, User shall be deemed to have accepted and agreed to all such terms and conditions if User republishes the Work in any fashion.

3. Scope of License; Limitations and Obligations.

3.1. All Works and all rights therein, including copyright rights, remain the sole and exclusive property of the Rightsholder. The license created by the exchange of an Order Con!rmation (and/or any invoice) and payment by User of the full amount set forth on that document includes only those rights expressly set

https://marketplace.copyright.com/rs-ui-web/mp/license/680059fb-6ec8-4465-b389-c0718543b84b/db3a1fd8-6dbd-43b1-8262-23e802edf130 Page 2 of 5 7/23/20, 4:58 AM

forth in the Order Con!rmation and in these terms and conditions, and conveys no other rights in the Work(s) to User. All rights not expressly granted are hereby reserved.

3.2. General Payment Terms: You may pay by credit card or through an account with us payable at the end of the month. If you and we agree that you may establish a standing account with CCC, then the following terms apply: Remit Payment to: Copyright Clearance Center, 29118 Network Place, Chicago, IL 60673-1291. Payments Due: Invoices are payable upon their delivery to you (or upon our notice to you that they are available to you for downloading). After 30 days, outstanding amounts will be subject to a service charge of 1-1/2% per month or, if less, the maximum rate allowed by applicable law. Unless otherwise speci!cally set forth in the Order Con!rmation or in a separate written agreement signed by CCC, invoices are due and payable on "net 30" terms. While User may exercise the rights licensed immediately upon issuance of the Order Con!rmation, the license is automatically revoked and is null and void, as if it had never been issued, if complete payment for the license is not received on a timely basis either from User directly or through a payment agent, such as a credit card company.

3.3. Unless otherwise provided in the Order Con!rmation, any grant of rights to User (i) is "one-time" (including the editions and product family speci!ed in the license), (ii) is non-exclusive and non-transferable and (iii) is subject to any and all limitations and restrictions (such as, but not limited to, limitations on duration of use or circulation) included in the Order Con!rmation or invoice and/or in these terms and conditions. Upon completion of the licensed use, User shall either secure a new permission for further use of the Work(s) or immediately cease any new use of the Work(s) and shall render inaccessible (such as by deleting or by removing or severing links or other locators) any further copies of the Work (except for copies printed on paper in accordance with this license and still in User's stock at the end of such period).

3.4. In the event that the material for which a republication license is sought includes third party materials (such as photographs, illustrations, graphs, inserts and similar materials) which are identi!ed in such material as having been used by permission, User is responsible for identifying, and seeking separate licenses (under this Service or otherwise) for, any of such third party materials; without a separate license, such third party materials may not be used.

3.5. Use of proper copyright notice for a Work is required as a condition of any license granted under the Service. Unless otherwise provided in the Order Con!rmation, a proper copyright notice will read substantially as follows: "Republished with permission of [Rightsholder's name], from [Work's title, author, volume, edition number and year of copyright]; permission conveyed through Copyright Clearance Center, Inc. " Such notice must be provided in a reasonably legible font size and must be placed either immediately adjacent to the Work as used (for example, as part of a by-line or footnote but not as a separate electronic link) or in the place where substantially all other credits or notices for the new work containing the republished Work are located. Failure to include the required notice results in loss to the Rightsholder and CCC, and the User shall be liable to pay liquidated damages for each such failure equal to twice the use fee speci!ed in the Order Con!rmation, in addition to the use fee itself and any other fees and charges speci!ed.

3.6. User may only make alterations to the Work if and as expressly set forth in the Order Con!rmation. No Work may be used in any way that is defamatory, violates the rights of third parties (including such third parties' rights of copyright, privacy, publicity, or other tangible or intangible property), or is otherwise illegal, sexually explicit or obscene. In addition, User may not conjoin a Work with any other material that may result in damage to the reputation of the Rightsholder. User agrees to inform CCC if it becomes aware of any infringement of any rights in a Work and to cooperate with any reasonable request of CCC or the Rightsholder in connection therewith.

https://marketplace.copyright.com/rs-ui-web/mp/license/680059fb-6ec8-4465-b389-c0718543b84b/db3a1fd8-6dbd-43b1-8262-23e802edf130 Page 3 of 5 7/23/20, 4:58 AM

4. Indemnity. User hereby indemni!es and agrees to defend the Rightsholder and CCC, and their respective employees and directors, against all claims, liability, damages, costs and expenses, including legal fees and expenses, arising out of any use of a Work beyond the scope of the rights granted herein, or any use of a Work which has been altered in any unauthorized way by User, including claims of defamation or infringement of rights of copyright, publicity, privacy or other tangible or intangible property.

5. Limitation of Liability. UNDER NO CIRCUMSTANCES WILL CCC OR THE RIGHTSHOLDER BE LIABLE FOR ANY DIRECT, INDIRECT, CONSEQUENTIAL OR INCIDENTAL DAMAGES (INCLUDING WITHOUT LIMITATION DAMAGES FOR LOSS OF BUSINESS PROFITS OR INFORMATION, OR FOR BUSINESS INTERRUPTION) ARISING OUT OF THE USE OR INABILITY TO USE A WORK, EVEN IF ONE OF THEM HAS BEEN ADVISED OF THE POSSIBILITY OF SUCH DAMAGES. In any event, the total liability of the Rightsholder and CCC (including their respective employees and directors) shall not exceed the total amount actually paid by User for this license. User assumes full liability for the actions and omissions of its principals, employees, agents, a"liates, successors and assigns.

6. Limited Warranties. THE WORK(S) AND RIGHT(S) ARE PROVIDED "AS IS". CCC HAS THE RIGHT TO GRANT TO USER THE RIGHTS GRANTED IN THE ORDER CONFIRMATION DOCUMENT. CCC AND THE RIGHTSHOLDER DISCLAIM ALL OTHER WARRANTIES RELATING TO THE WORK(S) AND RIGHT(S), EITHER EXPRESS OR IMPLIED, INCLUDING WITHOUT LIMITATION IMPLIED WARRANTIES OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. ADDITIONAL RIGHTS MAY BE REQUIRED TO USE ILLUSTRATIONS, GRAPHS, PHOTOGRAPHS, ABSTRACTS, INSERTS OR OTHER PORTIONS OF THE WORK (AS OPPOSED TO THE ENTIRE WORK) IN A MANNER CONTEMPLATED BY USER; USER UNDERSTANDS AND AGREES THAT NEITHER CCC NOR THE RIGHTSHOLDER MAY HAVE SUCH ADDITIONAL RIGHTS TO GRANT.

7. E#ect of Breach. Any failure by User to pay any amount when due, or any use by User of a Work beyond the scope of the license set forth in the Order Con!rmation and/or these terms and conditions, shall be a material breach of the license created by the Order Con!rmation and these terms and conditions. Any breach not cured within 30 days of written notice thereof shall result in immediate termination of such license without further notice. Any unauthorized (but licensable) use of a Work that is terminated immediately upon notice thereof may be liquidated by payment of the Rightsholder's ordinary license price therefor; any unauthorized (and unlicensable) use that is not terminated immediately for any reason (including, for example, because materials containing the Work cannot reasonably be recalled) will be subject to all remedies available at law or in equity, but in no event to a payment of less than three times the Rightsholder's ordinary license price for the most closely analogous licensable use plus Rightsholder's and/or CCC's costs and expenses incurred in collecting such payment.

8. Miscellaneous.

8.1. User acknowledges that CCC may, from time to time, make changes or additions to the Service or to these terms and conditions, and CCC reserves the right to send notice to the User by electronic mail or otherwise for the purposes of notifying User of such changes or additions; provided that any such changes or additions shall not apply to permissions already secured and paid for.

8.2. Use of User-related information collected through the Service is governed by CCC's privacy policy, available online here:https://marketplace.copyright.com/rs-ui-web/mp/privacy-policy

8.3. The licensing transaction described in the Order Con!rmation is personal to User. Therefore, User may not assign or transfer to any other person (whether a natural person or an organization of any kind) the license created by the Order Con!rmation and these terms and conditions or any rights granted hereunder; provided, however, that User may assign such license in its entirety on written notice to CCC in the event of a transfer of all or substantially all of User's rights in the new material which includes the Work(s) licensed under this Service.

https://marketplace.copyright.com/rs-ui-web/mp/license/680059fb-6ec8-4465-b389-c0718543b84b/db3a1fd8-6dbd-43b1-8262-23e802edf130 Page 4 of 5 7/23/20, 4:58 AM

8.4. No amendment or waiver of any terms is binding unless set forth in writing and signed by the parties. The Rightsholder and CCC hereby object to any terms contained in any writing prepared by the User or its principals, employees, agents or a"liates and purporting to govern or otherwise relate to the licensing transaction described in the Order Con!rmation, which terms are in any way inconsistent with any terms set forth in the Order Con!rmation and/or in these terms and conditions or CCC's standard operating procedures, whether such writing is prepared prior to, simultaneously with or subsequent to the Order Con!rmation, and whether such writing appears on a copy of the Order Con!rmation or in a separate instrument.

8.5. The licensing transaction described in the Order Con!rmation document shall be governed by and construed under the law of the State of New York, USA, without regard to the principles thereof of con$icts of law. Any case, controversy, suit, action, or proceeding arising out of, in connection with, or related to such licensing transaction shall be brought, at CCC's sole discretion, in any federal or state court located in the County of New York, State of New York, USA, or in any federal or state court whose geographical jurisdiction covers the location of the Rightsholder set forth in the Order Con!rmation. The parties expressly submit to the personal jurisdiction and venue of each such federal or state court.If you have any comments or questions about the Service or Copyright Clearance Center, please contact us at 978-750- 8400 or send an e-mail to [email protected].

v 1.1

https://marketplace.copyright.com/rs-ui-web/mp/license/680059fb-6ec8-4465-b389-c0718543b84b/db3a1fd8-6dbd-43b1-8262-23e802edf130 Page 5 of 5