UNIVERSITY OF CALIFORNIA

Los Angeles

Eosinophil Infiltration in Muscular Dystrophy:

Key Characteristics and Contributions to Disease

A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy

in Molecular, Cellular & Integrative Physiology

by

Albert Chi Sek

2020

© Copyright by

Albert Chi Sek

2020

ABSTRACT OF THE DISSERTATION

Eosinophil Infiltration in Muscular Dystrophy:

Key Characteristics and Contributions to Disease

By

Albert Chi Sek

Doctor of Philosophy in Molecular, Cellular & Integrative Physiology

University of California, Los Angeles, 2020

Professor Tomas Ganz, Chair

Duchenne Muscular Dystrophy (DMD) is a genetic disorder that affects 1 in 3,500 live male births. In DMD patients, inactivating mutations in the gene encoding dystrophin prevent expression of this critical cytoskeletal protein that is essential for myofiber integrity. Mutations in the gene encoding dystrophin have also been detected in muscular dystrophy (mdx) mice, which phenocopy the disease seen in DMD patients. As a result of dystrophin deficiency, the skeletal and cardiac muscles undergo severe degeneration accompanied by infiltration with inflammatory cells. Eosinophils are prominent among the leukocytes recruited to dystrophin-deficient tissues, although their contributions to disease are incompletely understood.

Eosinophils are capable of releasing cytotoxic mediators stored in cytoplasmic granules; as such, eosinophils have been viewed largely as mediators of tissue destruction. I began this work with an evaluation of the extent and impact of eosinophil infiltration on acute muscle damage in muscular dystrophy mice. Towards this end, I generated eosinophil-deficient (mdx.PHIL) and eosinophil-overabundant (mdx.IL5tg) strains of mdx mice. Despite the varied levels of eosinophil

ii

infiltration detected in skeletal muscle tissues, my studies revealed that there were remarkably

similar levels of muscle damage in mdx, mdx.IL5tg and mdx.PHIL mice. This was evaluated by

four key measures: histopathology scoring, distribution of myofiber sizes, proportion of centrally-

nucleated myofibers and creatine kinase levels.

To evaluate the functional characteristics of eosinophils recruited to dystrophin-deficient muscle tissue, I isolated eosinophils from the quadriceps muscles of mdx, IL5tg and mdx.IL5tg mice at 4 weeks of age, extracted RNA and performed RNA-sequencing. I identified 393 transcripts that were expressed at 5-fold higher levels in the eosinophils isolated from the muscle tissues of mdx mice when compared with those from IL5tg mice. Many of these transcripts encode tissue remodeling proteins (e.g., Ctss, Ctsk, Mmp19) as well as cell surface proteins (e.g., Cd14,

Slc15a3, Trem2). By contrast, I identified 238 transcripts at 5-fold higher levels in eosinophils from the muscle tissues of IL5tg mice when compared with those from mdx mice. Many of these latter transcripts encode eosinophil-specific proteins (e.g., Ear1, Epx, Prg2) or proteins involved with cell cycle regulation.

The findings that suggest differential expression of Trem2 are particularly intriguing, as

TREM2 has been shown to promote the proliferation, activation and survival of myeloid cells in response to tissue damage. Hence, in order to evaluate the role of Trem2 in mediating eosinophil survival within dystrophin-deficient muscle tissues, we are currently generating mdx.Trem2-/- mice

and will be in a position to evaluate eosinophil infiltration and survival in the muscle tissues of

these mice. While generation of this strain is ongoing, I have determined that Trem2 is critical for

full eosinophil development in both ex vivo and in vivo experimental studies.

Taken together, these findings add to the growing body of evidence that suggests that

eosinophils do not universally promote tissue destruction. In addition, findings presented in this

dissertation highlight the unique features of eosinophils that may be uncovered through RNA-

sequencing and analysis. Among the most intriguing findings, I established that Trem2 is critical

iii for efficient eosinophil development. Further work will elucidate the regulation and functions of

TREM2 expression in eosinophils.

iv

The dissertation of Albert Chi Sek is approved.

John P. Chute

Elizabeta Nemeth

Eric Pearlman

Helene F. Rosenberg

James G. Tidball

Tomas Ganz, Committee Chair

University of California, Los Angeles

2020

v

For Mom, Dad

and Grace

vi

TABLE OF CONTENTS

Abstract of the Dissertation ii

List of tables and figures x

Acknowledgements xiii

Vita xvi

Chapter 1: Introduction 1

Duchenne Muscular Dystrophy: A Severe Genetic Disorder 2

The mdx mouse model of Duchenne Muscular Dystrophy 3

Inflammation as a critical feature of muscular dystrophy pathogenesis 5

Eosinophil infiltration in dystrophin-deficient muscle tissue 6

Study of eosinophils in vivo and ex vivo: critical reagents and mouse models 7

This dissertation 8

References 14

Chapter 2: Detection and Isolation of Mouse Eosinophils from Muscle Tissue

using Flow Cytometry 21

Preface 22

Abstract 23

Introduction 23

Methods and Results 25

Discussion 28

References 35

Chapter 3: Eosinophils Do Not Drive Acute Muscle Pathology in the mdx

Mouse Model of Duchenne Muscular Dystrophy 38

Preface 39

vii

Abstract 41

Introduction 41

Materials and Methods 42

Results 43

Discussion 47

References 48

Addendum 52

Chapter 4: Transcriptomic Profile of Eosinophils Recruited to Muscle Tissue

of Dystrophin-deficient (mdx) mice 59

Preface 60

Abstract 61

Introduction 62

Materials and Methods 63

Results 65

Discussion 70

References 79

Chapter 5: Role of TREM2 in Modulating the Development and Characteristics

of Eosinophils 82

Preface 83

Abstract 85

Introduction 85

Materials and Methods 87

Results 91

Discussion 96

References 106

viii

Chapter 6: Conclusion 109

Eosinophilic infiltration does not always promote tissue destruction 110

Eosinophil infiltration in musculoskeletal disorders 111

RNA-sequencing enables functional characterization of eosinophils 113

Future Directions – Regulation and Function of TREM2 on Eosinophils 115

Concluding Remarks 117

References 118

ix

LIST OF TABLES AND FIGURES

Chapter 1

Figure 1.1 The mdx mouse model of Duchenne Muscular Dystrophy 11

Figure 1.2 Eosinophil infiltration and histopathology in quadriceps muscle

tissue from mdx mice 12

Figure 1.3 Eosinophil characteristics: functional mediators and receptors 13

Chapter 2

Figure 2.1 Method for generating single cell suspensions from skeletal

muscle 31

Table 2.1 Antibodies used for detection and isolation of eosinophils

from skeletal muscle tissue 32

Figure 2.2 Flow cytometry strategy to identify eosinophils in skeletal

muscle tissue 33

Figure 2.3 Morphological evaluation of SigF+CD11c+ and SigF+Gr1+

populations 34

Chapter 3

Figure 3.1 Histopathology and quantitative determination of eosinophils in

the quadriceps muscle from wild-type, mdx, eosinophil-deficient

PHIL, and mdx.PHIL mice 43

Figure 3.2 Histopathology and quantitative determination of eosinophils in

the quadriceps muscle from IL-5 transgenic and mdx.IL5

transgenic mice 44

Figure 3.3 Quantitative indicators of muscle pathology 45

Figure 3.4 Eosinophil recruitment to muscle tissue 46

Figure 3.5 detected in muscle tissue lysates 47

x

Figure 3.S1 Additional images featuring lesions in quadriceps muscle from

mdx mice 50

Figure 3.S2 Detection of centrally-located nuclei 51

Figure A1. Quantification of myofiber integrity and neutrophil infiltration

in quadriceps muscle tissues from mice at 4 weeks of age 56

Figure A2. Histopathology and eosinophil infiltration in quadriceps muscle

tissues from mice at 8 weeks of age 57

Figure A3. Histopathology and quantification of myofiber integrity in

quadriceps muscle tissues from mice at 8 weeks of age 58

Chapter 4

Figure 4.1 Isolation of RNA from eosinophils from skeletal muscle tissues 74

Figure 4.2 Quality of RNA isolated from eosinophils from skeletal muscle

tissues 75

Figure 4.3 Transcripts and gene ontology pathways that are differentially

expressed in mdx vs IL5tg muscle eosinophils 76

Figure 4.4 Differentially expressed transcripts that are associated with

tissue remodeling 77

Figure 4.5 Differentially expressed transcripts that encode cell surface

proteins 78

Chapter 5

Figure 5.1 Differentiation of eosinophils ex vivo and detection of TREM2 100

Figure 5.2 Trem2 is required for efficient eosinophil expansion ex vivo 101

Figure 5.3 Trem2 gene-deletion alters eosinophil homeostasis in

the bone marrow 102

Figure 5.4 Trem2 is not critical for eosinophil recruitment in response

to a Th2-inflammatory stimulus 103

xi

Figure 5.5 Generation of mdx.Trem2-/- mice 104

Figure 5.6 Variable expression of TREM2 on human peripheral blood

eosinophils and correlation with donor BMI 105

xii

ACKNOWLEDGEMENTS

When I started graduate school in the fall of 2014, I had no idea of the non-linear, fast- moving and exponentially-rewarding journey that lay ahead. Through these experiences, I developed scientific and technical expertise, but also became a stronger writer, better presenter, and more critical thinker. I could not have imagined a better group of people to accompany and guide me along this remarkable period of personal and professional growth.

First and foremost, I am immensely grateful for the guidance that my adviser Dr. Helene

Rosenberg provided on a regular basis. Although I started this dissertation project well into my graduate training, Dr. Rosenberg encouraged me to pursue this project tenaciously and strategically. As a result of her spectacular mentorship, I was able to publish my key findings within a short time frame and to hone my communication skills, notably in writing and presenting.

I am also thankful for the technical guidance and support that other members of Dr.

Rosenberg’s research group provided. These include Caroline Percopo, who was immensely helpful in teaching me flow cytometry and was always prepared to discuss challenging experiments; Michelle Ma, who provided guidance on the molecular biology techniques; and fellow trainees including Wendy Geslewitz, Julia Krumholz, Jamie Redes, Gordon Wong, Ajinkya

Limkar and Eric Mai. In addition, I was fortunate during my graduate studies to continue to receive feedback and mentorship from my former supervisor, Dr. Kirk Druey, and members of his research group, including Dr. Sherry Xie and Dr. Eunice Chan.

I would like to thank members of my doctoral committee for facilitating my professional development and for their guidance and support during my graduate studies. Dr. Tomas Ganz and Dr. Elizabeta Nemeth encouraged me to continue with graduate training, despite early setbacks, and to approach projects with an inquisitive and creative mindset. Dr. James Tidball introduced me to the fascinating world of muscular dystrophy, while Dr. John Chute challenged me to pursue the most critical aspects of a research project. And, Dr. Eric Pearlman provided immensely helpful input on working with granulocytes and on succeeding in graduate school.

xiii

I would like to thank the terrific Molecular, Cellular and Integrative Physiology (MCIP) PhD program at UCLA, which has facilitated much of my professional development. I enjoyed the coursework, the community of scholars, and the faculty that worked so tirelessly to ensure the success of its trainees. In particular, I would like to thank Dr. Mark Frye, the program chair, and

Ms. Yesenia Rayos, graduate student affairs officer of the MCIP Program; I enjoyed their company and appreciated their genuine concern and assistance throughout graduate school.

Finally, this dissertation would not be possible without the support of family, friends and classmates who provided advice, listened to my practice presentations and, of course, offered me some fun outside the laboratory. These include my parents, James and Linda Sek; my sister,

Grace Sek; my friends (in no particular order): Steven Chan, Francois de Mets, Pipat Piewgnam,

Murto Wickens, Ninad Jog, Pop Wongpalee, Chris Shen, Frank Lin, Jalal Buckley, Thomas

Calder, Duy Tran, Michael Otto, Wei-Sheng Chen, Albert Ma, Josh Norwood, Adam Moua, Emilio

Ronquillo, and Jenny Chen Link. I am proud to have completed this academic journey with my stellar cohort within the MCIP PhD program: Shuin Park, He Iris Wang, Adam Gomez, Ivan Flores,

Michael Liu, Sara Ranjbarvaziri.

Chapter 1 contains figures that have been reprinted with permission from Sicinski, P., et al., The molecular basis of muscular dystrophy in the mdx mouse: a point mutation. Science,

1989. 244(4912): p. 1578-80; from Fairclough, R.J., M.J. Wood, and K.E. Davies, Therapy for

Duchenne muscular dystrophy: renewed optimism from genetic approaches. Nat Rev Genet,

2013. 14(6): p. 373-8; and from Rosenberg, H.F., K.D. Dyer, and P.S. Foster, Eosinophils: changing perspectives in health and disease. Nat Rev Immunol, 2013. 13(1): p. 9-22.

Chapter 2 contains findings which will be published in the upcoming paper “Detection of

Mouse Eosinophils from Tissue by Flow Cytometry and Isolation by Fluorescence-Activated Cell

Sorting (FACS)”, which will be published in Methods Molecular Biology, 2020 (in press). Caroline

M. Percopo, Ajinkya R. Limkar, Helene F. Rosenberg are co-authors. I designed experiments,

xiv

analyzed findings, and prepared data for publication. Caroline M. Percopo and Ajinkya R. Limkar

also designed and contributed experimental findings. Helene F. Rosenberg conceived project

design and wrote first draft. All authors reviewed the manuscript prior to publication.

Chapter 3 is a reprint of “Eosinophils Do Not Drive Acute Muscle Pathology in the mdx

Mouse Model of Duchenne Muscular Dystrophy” from Journal of Immunology 2019; 203(2):476-

484. Ian N. Moore, Margery G. Smelkinson, Katherine Pak, Mahnaz Minai, Roberta Smith,

Michelle Ma, Caroline M. Percopo, Helene F. Rosenberg are co-authors. I designed experiments,

performed most of the experimental work, coordinated collaborations, analyzed data, generated

the primary figures and methods section, and assisted with the first and subsequent drafts of the

manuscript. Ian N. Moore provided input on the design of the critical histology and

immunohistochemistry studies, performed the pathology scoring, provided critical descriptions of

the pathological findings, and reviewed the manuscript. Margery G. Smelkinson performed the analysis on muscle fiber size, central nuclei and PAX7+ cells, provided me with insight and instruction, wrote the methods section for this part of the manuscript, and reviewed the manuscript. Katherine Pak provided me with primary instruction on muscle biology, with information including, but not limited to, dissection, freezing and fixation, reagents in common use in the field, and reference to important literature; she has read and reviewed the manuscript.

Mahnaz Minai worked with Ian Moore to prepare tissue sections for analysis and evaluation and reviewed the manuscript. Roberta Smith performed and provided input on the serum creatine kinase assays and reviewed the manuscript. Michelle Ma and Caroline M. Percopo designed and assisted with experiments, prepared and reviewed the manuscript. Helene F. Rosenberg designed the project, reviewed the results on an ongoing basis, provided insight and direction, and wrote the first and subsequent drafts of the manuscript.

xv

VITA

Albert Chi Sek

EDUCATION

2014 – 2020 Ph.D. Candidate in Molecular, Cellular & Integrative Physiology University of California, Los Angeles National Institutes of Health Graduate Partnership Program

2008 – 2012 B.S. in Microbiology University of California, Davis

RESEARCH EXPERIENCE

2017 – present Predoctoral Fellow/PhD Candidate National Institute of Allergy and Infectious Disease (NIAID) Adviser: Helene Rosenberg, M.D., Ph.D.

2014 – 2017 Graduate Student Researcher University of California, Los Angeles Advisers: Tomas Ganz, M.D., Ph.D. & Elizabeta Nemeth, Ph.D.

2012 – 2014 Postbaccalaureate Research Fellow National Institute of Allergy and Infectious Disease Adviser: Kirk Druey, M.D.

2010 – 2012 Undergraduate Researcher University of California, Davis Adviser: Scott Dawson, Ph.D.

TEACHING & COMMUNICATION EXPERIENCE

2018 – present Staff Writer NIH Graduate Student Chronicles

2018 – 2019 Mentor to High School Students Prince George’s County, Maryland

2018 Special Volunteer Office of Strategic Planning & Evaluation, NIAID/NIH

2016 – 2017 Graduate Teaching Assistant, Department of Physiology, UCLA Foundations in Physiological Sciences, 2 sections, 17 students/year

HONORS AND AWARDS

2019 Presidential Award, Society for Leukocyte Biology

2019 NIH Graduate Student Research Award in Immunology

xvi

2019 International Eosinophil Society Travel Award

2019 NIAID Fellows Workshop Poster Award, 1st Place

2018 NIH Fellows Award in Research Excellence: Immunology (Innate)

PROFESSIONAL ACTIVITIES

2017 – 2020 National Institutes of Health Graduate Student Council Co-Chair of Graduate Student Council, 2019-2020 Chair of Mentorship, Volunteering & Outreach Committee, 2017 – 2019

2014 – 2017 Molecular, Cellular & Integrative Physiology Graduate Program Retreat Planning Committee, 2015 – 2017 Distinguished Speaker Selection Committee, 2014 – 2016

PUBLICATIONS

1. Percopo CM, Limkar AR, Sek AC, Rosenberg HF. Detection of Mouse Eosinophils from Tissue by Flow Cytometry and Isolation by Fluorescence-Activated Cell Sorting (FACS). Methods Mol Bio, ed. G. Walsh; in press, 2020.

2. Limkar AR, Mai E, Sek AC, Percopo CM, Rosenberg HF. Frontline Science: - mediated developmental phenotype of mouse eosinophils: IL5-associated expression of the Ly6G/Gr1 surface antigen. J Leukoc Biol; in press, 2019.

3. Redes JL, Basu T, Chan EC, Sek AC, Zhao M, Krishnan R, Rosenberg HF, Druey KM. Aspergillus fumigatus-secreted alkaline protease 1 (Alp1) mediates airways hyperresponsiveness in severe asthma. Immunohorizons, 2019. 3(8): pp.368-377.

4. Sek AC, Moore IN, Smelkinson MG, Pak K, Minai M, Smith R, Ma M, Percopo CM, Rosenberg HF. Eosinophils do not drive acute muscle pathology in the mdx mouse model of Duchenne Muscular Dystrophy. J Immunology, 2019. 203(2): pp. 476-484.

5. Ma M, Percopo CM, Sturdevant DE, Sek AC, Komarow HD, Rosenberg HF. Cytokine Diversity in Human Peripheral Blood Eosinophils: Profound Variability of Interleukin-16. J Immunology, 2019. 203(2): pp. 520-531.

6. Aschemeyer S, Qiao B, Stefanova D, Valore EV, Sek AC, Ruwe TA, Vieth KR, Jung G, Casu C, Rivella S, Jormakka M, Mackenzie B, Ganz T, Nemeth E. Structure-function analysis of ferroportin defines the binding site and an alternative mechanism of action of hepcidin. Blood, 2018. 131(8): pp.899-910.

7. Sek AC, Xie Z, Terai K, Long LM, Nelson C, Dudek AZ, Druey KM. Endothelial Expression of Endothelin Receptor A in the Systemic Capillary Leak Syndrome. Plos One, 2015. 10(7): pp. e0133266.

8. Hagen KD, Hirakawa MP, House SA, Schwartz CL, Pham JK, Cipriano MJ, De La Torre MJ, Sek AC, Du G, Forsythe BM, Dawson SC. Novel Structural Components of the Ventral Disc and Lateral Crest in Giardia intestinalis. Plos Negl Trop Dis, 2011. 5(12): p. e1442

xvii

CHAPTER 1

Introduction

1

Duchenne Muscular Dystrophy: A Severe Genetic Disorder

Duchenne Muscular Dystrophy (DMD) is an X-linked recessive disorder of progressive

muscle degeneration that affects 1 in 3,500 males [1, 2]. The genetic lesion underlying this

disorder is in the gene encoding dystrophin, a large protein (427 kDa) that anchors the intracellular

cytoskeleton of muscle fibers with the surrounding extracellular matrix [1, 2]. Inactivating

mutations in the dystrophin gene result in myofiber degeneration accompanied by

[1-4]. Mutations that partially preserve the dystrophin reading frame result in reduced levels of

dystrophin protein and a milder form of the disease known as Becker Muscular Dystrophy (BMD)

[1, 5, 6].

Between the ages of 2 to 5, DMD patients will begin to exhibit symptoms such as an

unsteady gait and difficulty rising from the floor [1, 2]. Often, DMD patients will need to use both their arms and legs to push themselves up, in what is known as Gower’s sign [2]. By adolescence,

at around 12-15 years of age, DMD patients are typically wheelchair-bound and often do not

survive beyond the age of 20 typically due to failure of the respiratory or cardiac muscles [1, 2].

Treatment of this devastating disorder has relied almost exclusively on oral administration

of glucocorticoids, which function broadly to inhibit inflammation in the muscle tissue [7, 8].

However, the prognosis for DMD patients, even those on treatment remains poor; patients

typically are unable to walk unassisted by 16 years of age and typically do not survive beyond 20

years of age [6, 8]. In response to this severe medical need, in 2016 the U. S. Food and Drug

Administration (FDA) approved the use of a novel therapeutic, eteplirsen (brand name: exondys

51), for the treatment of DMD characterized by mutations in exon 51 of the dystrophin gene [9].

Eteplirsen is an antisense oligonucleotide therapy designed to restore the reading frame of the

dystrophin gene by facilitating a skip over exon 51, a region that includes deleterious mutations

in 10-15% of DMD patients [8-10]. Although eteplirsen was designed to address the underlying

mutation in the defined subset of DMD patients, the therapeutic efficacy has failed to dramatically

augment the level of dystrophin protein produced in the muscle tissues of DMD patients [9, 10].

2

In a study of 11 DMD patients with more than 3 years of open-label use of eteplirsen, Sarepta

reported extraordinarily low levels of dystrophin detected in the muscle tissues of patients treated

with eteplirsen; analysis by quantitative Western Blot revealed expression of dystrophin protein

at 0.9% ± 0.8% compared to that of healthy controls [8, 10]. Furthermore, patients using eteplirsen for 24 and 48 weeks failed to show clinical benefit in a 6-minute walk test compared with patients receiving placebo [9, 10]. Although the FDA advisory committee did not recommend approval, the therapeutic has since been released eteplirsen out of the need to exercise “the greatest flexibility possible” for this lethal genetic disease with limited treatment options [9]. Hence, improved

understanding of the pathogenesis in DMD remains an area of active research and medical need.

The mdx mouse model of Duchenne Muscular Dystrophy

Mutations in the dystrophin gene have been detected in an array of non-mammalian animal models such as Caenorhabditis elegans (nematode) [11, 12], Drosophila melanogaster

(fruit fly) [13, 14] and Danio rerio (zebrafish) [15, 16]; and in mammalian animal models such as

Mus musculus (mouse), rats [17, 18], dogs [19, 20], cats [21, 22] and pigs [23, 24]. Of these, the laboratory mouse remains a popular choice in studying muscular dystrophy because of its rapid generation time and the plethora of genetic tools available [25, 26].

In the early 1980s, an inactivating mutation in the dystrophin gene arose spontaneously in a laboratory strain of C57BL/10ScSn mice [27]. These mice, later termed mdx for muscular

dystrophy on the x chromosome, exhibited abnormally high levels of serum creatine kinase (a

biomarker for muscle damage) and presented histological evidence of muscle damage and inflammation [27]. Subsequent genetic analysis revealed a single point mutation (C  T) in exon

23 of the dystrophin gene that results in a premature stop codon and prevents expression of functional, full-length dystrophin protein within the muscle tissues (Fig 1) [28-31]. Since this discovery, mdx mice (JAX Stock No. 001801) and their corresponding dystrophin-sufficient

C57BL/10ScSn parent strain (JAX Stock No. 000476) have been used widely to study the

3

pathogenesis of Duchenne Muscular Dystrophy and are a major resource used in this dissertation

[25, 26].

Skeletal muscle tissues from mdx mice exhibit distinct, well-defined periods of tissue damage, regeneration and fibrosis. Prior to three weeks of age, skeletal muscle tissues from mdx mice are histologically indistinguishable from that of dystrophin-sufficient, healthy controls [26,

32]. However, at three to four weeks of age, mdx mice exhibit the onset of acute muscle injury within the hindlimb muscles [33, 34]. This is characterized by prominent regions of myofiber

swelling and myofiber degeneration, which is accompanied by , leukocyte infiltration, and

mineralization [3, 4, 32, 33]. Following the onset of acute muscle injury, mdx mice maintain

successful regeneration of the skeletal muscle tissues during 6 to 12 weeks of their lifespan [33,

35]. At around 1 year of age, mdx mice will exhibit severe, irreversible fibrosis of the skeletal muscle tissues [33, 36].

Compared with DMD patients, mdx mice manifest a milder form of the disease, presenting complete ambulation, minimal wasting and fibrosis of skeletal muscle tissues, and robust

regenerative processes in the skeletal muscle tissues [26, 37]. Several hypotheses have been

raised to explain the milder disease course seen in mice, including upregulation of the compensatory, functional homologs (utrophin, α7β1 integrin) in murine muscle tissues [2, 38-40] and the background strain of mdx mice [41]. Accordingly, several research groups have generated strains of mdx mice that have mutations in modifier genes (e.g., cytidine monophosphate sialic acid hydroxylase-knockout cmah-mdx) [42], lack functional homologs (e.g., dystrophin-utrophin

double-knockout dko, α7-integrin-dystrophin double knockout α71-DkO) [43, 44], or exhibit

progressive degeneration (e.g., telomerase-deficient mdx/mTR) [45]. Despite the advent of these mdx-manipulated mouse strains, their use has not been widely adopted due to premature deaths, difficulty breeding, and the potential confounding influence of a second genetic manipulation that is not otherwise observed in DMD patients.

4

Inflammation as a critical feature of muscular dystrophy pathogenesis

Although inactivating mutations in the dystrophin gene result in myofiber fragility, increasing evidence indicates that the immune response participates in key remodeling events

that influence the course of disease [3, 46, 47]. For example, administration of inflammation-

suppressing glucocorticoids has been shown to limit leukocyte infiltration in the muscle tissue [48-

50] and to improve muscle function in DMD patients [51, 52].

Among the leukocytes recruited to dystrophin-deficient skeletal muscle tissues,

are by far the most prevalent, exceeding 3 x 104 cells per cubic millimeter of tissue

[3, 34, 53-55]. In muscular dystrophy, macrophages exert distinct and multifaceted roles in both

exacerbating and ameliorating the level of muscle damage and inflammation [3]. At the onset of

acute muscle injury, dystrophin-deficient muscle tissues are dominated by pro-inflammatory, M1-

biased macrophages that release cytolytic free radicals that lyse myofiber membranes [56].

Repeated injections of mdx mice with anti-F4/80 antibody reduced infiltrating

populations and reduced the number of muscle lesions detected in the acute injury phase [34].

Following acute muscle injury, infiltrating macrophage populations are dominated by the M2-

phenotype and are distinguished by increased expression of the anti-inflammatory cytokine

Interleukin-10 [56]. Here, M2-biased macrophages limit the level of muscle damage by competing

for arginine, a substrate required for the generation of the free radical nitric oxide [57]. M2-biased

macrophages also promote muscle regeneration through increased expression of the myogenic

factors such as Klotho and Insulin-like growth factor 1 [3, 58].

Though less well-studied, leukocytes including eosinophils [53, 59, 60], neutrophils [56,

61, 62], mast cells [53, 63-67] and lymphocytes [53, 54, 68-70] are prominent within dystrophin-

deficient skeletal muscle tissues; the role of eosinophils in muscular dystrophy is described in the

following section. Neutrophils have been shown through an antibody-based depletion study to

promote muscle damage [61], although the specific cytolytic mechanism remains to be elucidated.

Meanwhile, the impact of mast cells on dystrophic muscle damage is less clear; prior studies that

5

have inhibited mast cell degranulation [71] or have injected mast cell granules directly into the

muscle tissue [72] have not achieved consistent or physiologically-meaningful results,

respectively. Lastly, cytotoxic T-lymphocytes have been shown through an antibody-based

depletion study to drive dystrophic muscle damage due in part to the release of the cytolytic

protein perforin [68, 69].

Eosinophil infiltration in the dystrophin-deficient muscle tissue

Although eosinophils are detected only rarely in healthy muscle tissues, eosinophil

infiltration is prominent in disorders associated with severe muscle damage: eosinophilia-

syndrome [73-75], eosinophilic myositits [76-79], Limb-girdle Muscular Dystrophy type 2A

(LGMD2A) [80, 81], and Becker Muscular Dystrophy [82]. Eosinophil infiltration is also detected

in the skeletal muscle tissues of Duchenne Muscular Dystrophy patients [53, 80] and muscular

dystrophy (mdx) mice (Fig 2) [53, 60]. Earlier work by Cai and colleagues [60] described eosinophils and their degranulation products in association with damaged myofibers in mdx mice at 4 weeks of age. Interestingly, and seemingly paradoxically, Wehling-Henricks and colleagues

[53] determined that deletion of the gene encoding the eosinophil granule protein Major Basic

Protein-1 (MBP), a key cytotoxic mediator, did not ameliorate the level of muscle damage in mdx

mice.

Although eosinophils can mediate tissue damage secondary to the release of granule

proteins such as MBP, eosinophils can also exert beneficial, immunomodulatory roles. For

example, eosinophil granules contain a wealth of potent, immunomodulatory mediators such as

pre-formed cytokines, chemokines and growth factors (Fig 3); for a more comprehensive list,

please see the excellent reviews by Lee and colleagues [83] and Rosenberg and colleagues [84].

Indeed, emerging evidence in the field indicates that tissue eosinophils function primarily to

regulate the “local immunity and/or remodeling/repair” processes in what is known as the LIARR

hypothesis of eosinophil function [85].

6

As but one example, Heredia and colleagues reported that eosinophils were critical for the repair of muscle tissue following injury induced by experimental injection with snake cardiotoxin

[86]. Here, eosinophils were critical sources of the pro-regenerative cytokine interleukin-4, which

facilitated the proliferation of fibro-adipogenic precursors that support myogenesis. Eosinophil-

deficient mice exhibited impaired regeneration of the muscle tissue following damage.

Subsequent studies along these lines have confirmed that eosinophils mediate pro-regenerative

roles in response to experimentally induced damage to the liver [87] or the urogenital tract [88].

These studies have promoted our understanding of the pro-regenerative capabilities of

eosinophils responding to tissue damage. However, eosinophil functions in the dystrophin-

deficient muscle microenvironment may differ from those observed in the acute model of muscle

damage induced by injection of snake cardiotoxin. For instance, in muscular dystrophy, tissue

damage may or may not be mediated by eosinophils that sense cellular defects secondary to

dystrophin-deficiency; by contrast, eosinophils responding to cardiotoxin-induced muscle injury

are thought to be involved exclusively with the regenerative processes. Furthermore, muscle

tissues from muscular dystrophy (mdx) mice exhibit chronic tissue damage that may activate

eosinophils in a distinct manner compared with eosinophils activated by acute muscle damage.

Study of eosinophils in vivo and ex vivo: critical reagents and mouse models

In order to investigate the role of eosinophils in muscular dystrophy, I utilized several key

resources that permitted the identification and manipulation of eosinophils in vivo. First, to identify

eosinophils, I utilized an antibody against the eosinophil granule protein major basic protein-1 (α-

mMBP); this antibody was generously provided by the Lee Laboratories (Mayo Clinic). To

manipulate the levels of eosinophils in vivo, I utilized two key resources that were provided by the

Lee Laboratories (Mayo Clinic): the interleukin-5 transgenic (IL5tg/NJ.1638) and the PHIL

transgenic mouse. As interleukin-5 (IL-5) is critical to all aspects of eosinophil biology including

its development, IL5tg mice exhibit increased levels of eosinophils in the blood and bone marrow

7

as well as eosinophil infiltration into most organ systems. By contrast, the PHIL mice harbor a

transgene encoding diphtheria toxin A expression under the control of an eosinophil lineage-

specific promoter; as a result of transgene expression, eosinophil precursors commit cyto-suicide

in the bone marrow, leading to the absence of eosinophils in vivo.

This Dissertation

In this dissertation I report new findings that relate to the identification, characterization and impact of eosinophils in muscle tissue using the mdx mouse model of muscular dystrophy. As such, I have divided my key findings into the following chapters.

Chapter 2: Detection and Isolation of Mouse Eosinophils from Muscle Tissue using Flow

Cytometry details my development of a flow cytometry protocol in order to characterize

eosinophils from the muscle tissues. Among the key findings, I determined an antigen profile for eosinophils from the muscle tissue as CD45+CD11c-Gr1-MHCIIloSigF+; I also confirmed that eosinophils in the muscle tissue do not express either CD11c or Gr1 antigen. This advance permitted us to enumerate and evaluate eosinophil populations from the muscle tissue. Much of this work was featured in a “Detection of Mouse Eosinophils from Tissue by Flow Cytometry and

Isolation by Fluorescence-Activated Cell Sorting (FACS)”, a protocol in press in Methods in

Molecular Biology; for my contribution, I was listed as a co-author.

Chapter 3: Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of

Duchenne Muscular Dystrophy reports my study of the impact of eosinophil infiltration on the

acute muscle damage observed in muscular dystrophy (mdx) mice. Here, I generated eosinophil-

deficient (mdx.PHIL) and eosinophil-overabundant (mdx.IL5tg) mice and evaluated the level of

muscle damage. Among the key findings was that the degree of muscle damage did not

correspond with the degree of eosinophil infiltration in the muscle tissue. Rather, eosinophils can

8

be present in large numbers in the muscle tissue in the absence of tissue damage, as in

Interleukin 5-transgenic (IL5tg) mice. These findings are a reprint of the research originally

published in the Journal of Immunology, with permission granted by the publisher [59].

Chapter 4: Transcriptomic Profile of Eosinophils Recruited to Muscle Tissue of

Dystrophin-deficient (mdx) mice presents my study of key characteristics in eosinophils recruited to the muscle tissues of mdx mice. Here, I utilized RNA-sequencing to develop a comprehensive, unbiased understanding of transcriptional mediators in eosinophils isolated from the muscle tissues of mdx, IL5tg and mdx.IL5tg mice. I identified distinct transcriptomic profiles among eosinophils recruited to the muscle tissues of mdx mice when compared with that of IL5tg.

Among the key differences, eosinophils from the muscle tissues of mdx mice expressed high levels of transcripts encoding tissue remodeling enzymes and cell surface proteins. Increased expression of transcripts encoding cell surface proteins suggests that eosinophils sense and

actively modulate response to the dystrophin-deficient muscle microenvironment. These findings

provide further areas of investigation: in Chapter 5, I explore the expression and function of

Trem2, a transcript which is detected at 68-fold higher levels in eosinophils from the muscle

tissues of mdx mice when compared with those from IL5tg mice.

Chapter 5: Role of TREM2 in Modulating the Development and Characteristics of

Eosinophils reports my analysis of the expression and function of TREM2 in eosinophils. Here,

I identified TREM2 expression on differentiating murine eosinophils and on human peripheral blood eosinophils. Using the Trem2-/- mouse model, I established that Trem2 promotes efficient eosinophil expansion ex vivo and in vivo, but is not required for eosinophil infiltration into the airways of allergen-challenged mice. Ongoing work will evaluate the impact of Trem2 on

eosinophil characteristics, such as recruitment to dystrophin-deficient muscle tissue and

functional responses to the fungal aeroallergen Alternaria alternata. These findings on eosinophil

9

characteristics (Chapter 4) and the role of TREM2 (Chapter 5) will be prepared as a manuscript

for publication in 2020.

Chapter 6: Conclusion offers a summary and an analysis of the major findings presented in this dissertation. In addition, I discuss further promising areas of investigation revealed by this work.

10

FIGURES

Figure 1. The mdx mouse model of Duchenne Muscular Dystrophy. (A) Mdx mice harbor a

null mutation (C  T) in exon 23 of the dystrophin gene. Reprinted with permission from [28] (B)

Dystrophin is expressed in myofibers, where it links the intracellular cytoskeleton with the sarcoglycan complex at the membrane. Null mutations in the gene encoding dystrophin or in any

of the genes encoding sarcoglycan complex proteins have been associated with muscular

dystrophy. Reprinted with permission from [89].

11

Figure 2. Eosinophil infiltration and histopathology in quadriceps muscle tissue from mdx

mice. Quadriceps muscle tissue from male mdx mice at 4-weeks of age that has been stained

with (A) anti-Major Basic Protein-1 (α-mMBP) to identify eosinophils or with (B) hematoxylin and eosin to elucidate the histological features. Eosinophil infiltration was prominent in dystrophin- deficient muscle tissue and was associated with leukocyte infiltration, edema, and myofiber damage.

12

Figure 3. Eosinophil characteristics: functional mediators and receptors.

Eosinophils are distinguished by the presence of the bilobed nuclei, multiple lipid bodies, and eosin-staining granules. Eosinophil lipid bodies contain key lipid mediators, such as leukotrienes and prostaglandins. Similarly, eosinophil granules contain a wealth of pre-formed cytokines, chemokines, growth factors and enzymes. Release of key lipid mediators and granule contents is thought to mediate eosinophil functions. Eosinophils also contain many pattern recognition and chemoattractant receptors. Reprinted with permission from [84].

13

REFERENCES

1. Nowak, K.J. and K.E. Davies, Duchenne muscular dystrophy and dystrophin: pathogenesis and opportunities for treatment. EMBO Rep, 2004. 5(9): p. 872-6.

2. Allen, D.G., N.P. Whitehead, and S.C. Froehner, Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev, 2016. 96(1): p. 253-305.

3. Tidball, J.G., S.S. Welc, and M. Wehling-Henricks, Immunobiology of Inherited Muscular Dystrophies. Compr Physiol, 2018. 8(4): p. 1313-1356.

4. Rosenberg, A.S., et al., Immune-mediated pathology in Duchenne muscular dystrophy. Sci Transl Med, 2015. 7(299): p. 299rv4.

5. Blake, D.J., et al., Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev, 2002. 82(2): p. 291-329.

6. Andrews, J.G. and R.A. Wahl, Duchenne and Becker muscular dystrophy in adolescents: current perspectives. Adolesc Health Med Ther, 2018. 9: p. 53-63.

7. Henricson, E.K., et al., The cooperative international neuromuscular research group Duchenne natural history study: glucocorticoid treatment preserves clinically meaningful functional milestones and reduces rate of disease progression as measured by manual muscle testing and other commonly used clinical trial outcome measures. Muscle Nerve, 2013. 48(1): p. 55-67.

8. Mah, J.K., Current and emerging treatment strategies for Duchenne muscular dystrophy. Neuropsychiatr Dis Treat, 2016. 12: p. 1795-807.

9. Kesselheim, A.S. and J. Avorn, Approving a Problematic Muscular Dystrophy Drug: Implications for FDA Policy. JAMA, 2016. 316(22): p. 2357-2358.

10. Lim, K.R., R. Maruyama, and T. Yokota, Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des Devel Ther, 2017. 11: p. 533-545.

11. Hewitt, J.E., et al., Muscle strength deficiency and mitochondrial dysfunction in a muscular dystrophy model of Caenorhabditis elegans and its functional response to drugs. Dis Model Mech, 2018. 11(12).

12. Bessou, C., et al., Mutations in the Caenorhabditis elegans dystrophin-like gene dys-1 lead to hyperactivity and suggest a link with cholinergic transmission. Neurogenetics, 1998. 2(1): p. 61-72.

13. Shcherbata, H.R., et al., Dissecting muscle and neuronal disorders in a Drosophila model of muscular dystrophy. EMBO J, 2007. 26(2): p. 481-93.

14. Taghli-Lamallem, O., et al., Dystrophin deficiency in Drosophila reduces lifespan and causes a dilated cardiomyopathy phenotype. Aging Cell, 2008. 7(2): p. 237-49.

14

15. Bassett, D.I., et al., Dystrophin is required for the formation of stable muscle attachments in the zebrafish embryo. Development, 2003. 130(23): p. 5851-60.

16. Guyon, J.R., et al., Genetic isolation and characterization of a splicing mutant of zebrafish dystrophin. Hum Mol Genet, 2009. 18(1): p. 202-11.

17. Nakamura, K., et al., Generation of muscular dystrophy model rats with a CRISPR/Cas system. Sci Rep, 2014. 4: p. 5635.

18. Larcher, T., et al., Characterization of dystrophin deficient rats: a new model for Duchenne muscular dystrophy. PLoS One, 2014. 9(10): p. e110371.

19. Cooper, B.J., et al., The homologue of the Duchenne locus is defective in X-linked muscular dystrophy of dogs. Nature, 1988. 334(6178): p. 154-6.

20. Valentine, B.A., et al., Canine X-linked muscular dystrophy. An animal model of Duchenne muscular dystrophy: clinical studies. J Neurol Sci, 1988. 88(1-3): p. 69-81.

21. Gambino, A.N., et al., Emergent presentation of a cat with dystrophin-deficient muscular dystrophy. J Am Anim Hosp Assoc, 2014. 50(2): p. 130-5.

22. Gaschen, F. and J.M. Burgunder, Changes of skeletal muscle in young dystrophin- deficient cats: a morphological and morphometric study. Acta Neuropathol, 2001. 101(6): p. 591-600.

23. Hollinger, K., et al., Dystrophin insufficiency causes selective muscle histopathology and loss of dystrophin-glycoprotein complex assembly in pig skeletal muscle. FASEB J, 2014. 28(4): p. 1600-9.

24. Klymiuk, N., et al., Dystrophin-deficient pigs provide new insights into the hierarchy of physiological derangements of dystrophic muscle. Hum Mol Genet, 2013. 22(21): p. 4368-82.

25. Collins, C.A. and J.E. Morgan, Duchenne's muscular dystrophy: animal models used to investigate pathogenesis and develop therapeutic strategies. Int J Exp Pathol, 2003. 84(4): p. 165-72.

26. McGreevy, J.W., et al., Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis Model Mech, 2015. 8(3): p. 195-213.

27. Bulfield, G., et al., X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc Natl Acad Sci U S A, 1984. 81(4): p. 1189-92.

28. Sicinski, P., et al., The molecular basis of muscular dystrophy in the mdx mouse: a point mutation. Science, 1989. 244(4912): p. 1578-80.

29. Ryder-Cook, A.S., et al., Localization of the mdx mutation within the mouse dystrophin gene. EMBO J, 1988. 7(10): p. 3017-21.

30. Hoffman, E.P., R.H. Brown, Jr., and L.M. Kunkel, Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell, 1987. 51(6): p. 919-28.

15

31. Chamberlain, J.S., et al., Deletion screening of the Duchenne muscular dystrophy locus via multiplex DNA amplification. Nucleic Acids Res, 1988. 16(23): p. 11141-56.

32. Suelves, M., et al., uPA deficiency exacerbates muscular dystrophy in MDX mice. J Cell Biol, 2007. 178(6): p. 1039-51.

33. Villalta, S.A., et al., IFN-gamma promotes muscle damage in the mdx mouse model of Duchenne muscular dystrophy by suppressing M2 macrophage activation and inhibiting muscle cell proliferation. J Immunol, 2011. 187(10): p. 5419-28.

34. Wehling, M., M.J. Spencer, and J.G. Tidball, A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice. J Cell Biol, 2001. 155(1): p. 123-31.

35. Villalta, S.A., et al., Interleukin-10 reduces the pathology of mdx muscular dystrophy by deactivating M1 macrophages and modulating macrophage phenotype. Hum Mol Genet, 2011. 20(4): p. 790-805.

36. McDonald, A.A., et al., Disease course in mdx:utrophin+/- mice: comparison of three mouse models of Duchenne muscular dystrophy. Physiol Rep, 2015. 3(4).

37. Yucel, N., et al., Humanizing the mdx mouse model of DMD: the long and the short of it. NPJ Regen Med, 2018. 3: p. 4.

38. Matsumura, K., et al., Association of dystrophin-related protein with dystrophin- associated proteins in mdx mouse muscle. Nature, 1992. 360(6404): p. 588-91.

39. Rafael, J.A., et al., Skeletal muscle-specific expression of a utrophin transgene rescues utrophin-dystrophin deficient mice. Nat Genet, 1998. 19(1): p. 79-82.

40. Tinsley, J., et al., Expression of full-length utrophin prevents muscular dystrophy in mdx mice. Nat Med, 1998. 4(12): p. 1441-4.

41. Coley, W.D., et al., Effect of genetic background on the dystrophic phenotype in mdx mice. Hum Mol Genet, 2016. 25(1): p. 130-45.

42. Chandrasekharan, K., et al., A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy. Sci Transl Med, 2010. 2(42): p. 42ra54.

43. Deconinck, A.E., et al., Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell, 1997. 90(4): p. 717-27.

44. Grady, R.M., et al., Skeletal and cardiac in mice lacking utrophin and dystrophin: a model for Duchenne muscular dystrophy. Cell, 1997. 90(4): p. 729-38.

45. Sacco, A., et al., Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice. Cell, 2010. 143(7): p. 1059-71.

16

46. Evans, N.P., et al., Immune-mediated mechanisms potentially regulate the disease time- course of duchenne muscular dystrophy and provide targets for therapeutic intervention. PM R, 2009. 1(8): p. 755-68.

47. Tidball, J.G., Regulation of muscle growth and regeneration by the immune system. Nat Rev Immunol, 2017. 17(3): p. 165-178.

48. Hussein, M.R., et al., The effects of glucocorticoid therapy on the inflammatory and dendritic cells in muscular dystrophies. Int J Exp Pathol, 2006. 87(6): p. 451-61.

49. Kissel, J.T., et al., Mononuclear cell analysis of muscle biopsies in prednisone-treated and untreated Duchenne muscular dystrophy. CIDD Study Group. Neurology, 1991. 41(5): p. 667-72.

50. Wehling-Henricks, M., J.J. Lee, and J.G. Tidball, Prednisolone decreases cellular adhesion molecules required for inflammatory cell infiltration in dystrophin-deficient skeletal muscle. Neuromuscul Disord, 2004. 14(8-9): p. 483-90.

51. McDonald, C.M., et al., Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet, 2018. 391(10119): p. 451-461.

52. Matthews, E., et al., Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Database Syst Rev, 2016(5): p. CD003725.

53. Wehling-Henricks, M., et al., Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy. Hum Mol Genet, 2008. 17(15): p. 2280-92.

54. Engel, A.G. and K. Arahata, Mononuclear cells in myopathies: quantitation of functionally distinct subsets, recognition of antigen-specific cell-mediated cytotoxicity in some diseases, and implications for the pathogenesis of the different inflammatory myopathies. Hum Pathol, 1986. 17(7): p. 704-21.

55. Arahata, K. and A.G. Engel, Monoclonal antibody analysis of mononuclear cells in myopathies. I: Quantitation of subsets according to diagnosis and sites of accumulation and demonstration and counts of muscle fibers invaded by T cells. Ann Neurol, 1984. 16(2): p. 193-208.

56. Villalta, S.A., et al., Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy. Hum Mol Genet, 2009. 18(3): p. 482-96.

57. Wehling-Henricks, M., et al., Arginine metabolism by macrophages promotes cardiac and muscle fibrosis in mdx muscular dystrophy. PLoS One, 2010. 5(5): p. e10763.

58. Wehling-Henricks, M., et al., Macrophages escape Klotho gene silencing in the mdx mouse model of Duchenne muscular dystrophy and promote muscle growth and increase satellite cell numbers through a Klotho-mediated pathway. Hum Mol Genet, 2018. 27(1): p. 14-29.

17

59. Sek, A.C., et al., Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy. J Immunol, 2019. 203(2): p. 476-484.

60. Cai, B., et al., Eosinophilia of dystrophin-deficient muscle is promoted by perforin- mediated cytotoxicity by T cell effectors. Am J Pathol, 2000. 156(5): p. 1789-96.

61. Hodgetts, S., et al., Reduced necrosis of dystrophic muscle by depletion of host neutrophils, or blocking TNFalpha function with Etanercept in mdx mice. Neuromuscul Disord, 2006. 16(9-10): p. 591-602.

62. Arecco, N., et al., Elastase levels and activity are increased in dystrophic muscle and impair myoblast cell survival, proliferation and differentiation. Sci Rep, 2016. 6: p. 24708.

63. Gorospe, J.R., et al., A role for mast cells in the progression of Duchenne muscular dystrophy? Correlations in dystrophin-deficient humans, dogs, and mice. J Neurol Sci, 1994. 122(1): p. 44-56.

64. Helliwell, T.R., O. Gunhan, and R.H. Edwards, Mast cells in neuromuscular diseases. J Neurol Sci, 1990. 98(2-3): p. 267-76.

65. Lefaucheur, J.P., B. Gjata, and A. Sebille, Factors inducing mast cell accumulation in skeletal muscle. Neuropathol Appl Neurobiol, 1996. 22(3): p. 248-55.

66. Nahirney, P.C., P.R. Dow, and W.K. Ovalle, Quantitative morphology of mast cells in skeletal muscle of normal and genetically dystrophic mice. Anat Rec, 1997. 247(3): p. 341-9.

67. Gorospe, J.R., B.K. Nishikawa, and E.P. Hoffman, Recruitment of mast cells to muscle after mild damage. J Neurol Sci, 1996. 135(1): p. 10-7.

68. Spencer, M.J., et al., Helper (CD4(+)) and cytotoxic (CD8(+)) T cells promote the pathology of dystrophin-deficient muscle. Clin Immunol, 2001. 98(2): p. 235-43.

69. Spencer, M.J., et al., Myonuclear apoptosis in dystrophic mdx muscle occurs by perforin- mediated cytotoxicity. J Clin Invest, 1997. 99(11): p. 2745-51.

70. McDouall, R.M., M.J. Dunn, and V. Dubowitz, Nature of the mononuclear infiltrate and the mechanism of muscle damage in juvenile and Duchenne muscular dystrophy. J Neurol Sci, 1990. 99(2-3): p. 199-217.

71. Radley, H.G. and M.D. Grounds, Cromolyn administration (to block mast cell degranulation) reduces necrosis of dystrophic muscle in mdx mice. Neurobiol Dis, 2006. 23(2): p. 387-97.

72. Gorospe, J.R., et al., Dystrophin-deficient myofibers are vulnerable to mast cell granule- induced necrosis. Neuromuscul Disord, 1994. 4(4): p. 325-33.

73. Allen, J.A., et al., Post-epidemic eosinophilia-myalgia syndrome associated with L- tryptophan. Arthritis Rheum, 2011. 63(11): p. 3633-9.

18

74. Varga, J., G.G. Maul, and S.A. Jimenez, Autoantibodies to nuclear lamin C in the eosinophilia-myalgia syndrome associated with L-tryptophan ingestion. Arthritis Rheum, 1992. 35(1): p. 106-9.

75. Varga, J., S.A. Jimenez, and J. Uitto, L-tryptophan and the eosinophilia-myalgia syndrome: current understanding of the etiology and pathogenesis. J Invest Dermatol, 1993. 100(1): p. 97S-105S.

76. Selva-O'Callaghan, A., E. Trallero-Araguas, and J.M. Grau, Eosinophilic : an updated review. Autoimmun Rev, 2014. 13(4-5): p. 375-8.

77. Harris, C., et al., A Curious Case of Proximal with Eosinophilic . Case Rep Rheumatol, 2016. 2016: p. 7810916.

78. Trueb, R.M., et al., Eosinophilic myositis/perimyositis: frequency and spectrum of cutaneous manifestations. J Am Acad Dermatol, 1997. 37(3 Pt 1): p. 385-91.

79. Kaufman, L.D., et al., The spectrum of eosinophilic myositis. Clinical and immunopathogenic studies of three patients, and review of the literature. Arthritis Rheum, 1993. 36(7): p. 1014-24.

80. Schroder, T., et al., Eosinophils in hereditary and inflammatory myopathies. Acta Myol, 2013. 32(3): p. 148-53.

81. Luo, Y.B., et al., Limb-girdle Muscular Dystrophy Type 2A with Muscular Eosinophilic Infiltration in a Chinese Patient. Chin Med J (Engl), 2018. 131(17): p. 2133-2134.

82. Weinstock, A., et al., Becker muscular dystrophy presenting as eosinophilic inflammatory in an infant. J Child Neurol, 1997. 12(2): p. 146-7.

83. Lee, J.J., et al., Human versus mouse eosinophils: "that which we call an eosinophil, by any other name would stain as red". J Allergy Clin Immunol, 2012. 130(3): p. 572-84.

84. Rosenberg, H.F., K.D. Dyer, and P.S. Foster, Eosinophils: changing perspectives in health and disease. Nat Rev Immunol, 2013. 13(1): p. 9-22.

85. Lee, J.J., et al., Eosinophils in health and disease: the LIAR hypothesis. Clin Exp Allergy, 2010. 40(4): p. 563-75.

86. Heredia, J.E., et al., Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell, 2013. 153(2): p. 376-88.

87. Goh, Y.P., et al., Eosinophils secrete IL-4 to facilitate liver regeneration. Proc Natl Acad Sci U S A, 2013. 110(24): p. 9914-9.

88. Vicetti Miguel, R.D., et al., IL-4-secreting eosinophils promote endometrial stromal cell proliferation and prevent Chlamydia-induced upper genital tract damage. Proc Natl Acad Sci U S A, 2017.

19

89. Fairclough, R.J., M.J. Wood, and K.E. Davies, Therapy for Duchenne muscular dystrophy: renewed optimism from genetic approaches. Nat Rev Genet, 2013. 14(6): p. 373-8.

20

CHAPTER 2

Detection and Isolation of Mouse Eosinophils

From Muscle Tissue using Flow Cytometry

21

CHAPTER 2 PREFACE Flow Cytometry is a powerful technique that facilitates simultaneous, quantitative

measurements of antigen expression within a given population of cells. In addition, flow cytometry

and fluorescence-activated cell sorting (FACS) allows analysis and isolation, respectively, of

subpopulations based on antigen expression.

Earlier studies indicated that eosinophils exhibited substantial diversity in the expression of antigens, depending on the tissue and physiological context. As but two relevant examples,

Percopo and colleagues [1] identified a population of SiglecF+Gr1hi eosinophils in the lungs of allergen-challenged mice; this population exhibited a distinct cytokine profile when compared with the more numerous and morphologically-indistinguishable SiglecF+Gr1- eosinophils.

Furthermore, in more recent work (Limkar, Mai, Sek and colleagues [2]), we demonstrated that

the eosinophil-activating cytokine, interleukin-5, drives the expression of the Gr1 antigen on

eosinophils.

This chapter reports the development of a protocol to identify muscle eosinophils by flow

cytometry. Towards this end, we adapted a protocol to prepare muscle single cell suspensions

from Liu et al. [3] and we ultimately identified muscle eosinophils as live CD45+CD11c-Gr1-

MHCIIloSigF+ cells. Subsequently, Kastenschmidt and colleagues [4] published a protocol in which eosinophils in dystrophic muscle were identified as live F4/80+CD11b+SigF+ cells.

The protocol described in this chapter was among those included in a recent publication that I co-authored, Percopo CM, et al. Detection of Mouse Eosinophils from Tissue by Flow

Cytometry and Isolation by Fluorescence-Activated Cell Sorting (FACS). Methods Mol Bio, ed. G.

Walsh; in press, 2020.

22

ABSTRACT

While eosinophil infiltration in the muscle tissue is a prominent feature of Duchenne muscular

dystrophy, the contributions of eosinophils to disease pathology are not fully understood. Earlier

studies using the dystrophin-deficient mdx mouse model suggested that eosinophils could

promote characteristic muscle damage, but this hypothesis had not been addressed directly by

an evaluation of dystrophin-deficiency in mice lacking eosinophils. As a first and essential step

towards the study of eosinophils and their role in this disorder, we devised a means to detect,

quantify, and isolate eosinophils from infiltrates identified in quadriceps muscles of mdx mice. To

do this, we generated single cell suspensions from muscle tissue and adapted the flow cytometric

protocol currently in use in our lab for identification of eosinophils in lung and bone marrow. Using

this method, we determined that eosinophils from muscle tissue can be defined as CD45+CD11c-

Gr1-SigF+MHC-IIlo cells. When stained with modified Wright-Giemsa dyes, > 99% of the cells with this antigen profile exhibited ring-shaped nuclei and eosin-staining cytoplasmic granules characteristic of eosinophils. At four weeks of age, leukocyte infiltrates in quadriceps muscles of mdx mice contain a significant fraction of eosinophils (7.7% ± 1.9% of total CD45+ cells). There is also a very small population of SigF+Gr1+ cells (0.11 ± 0.05% of total CD45+ cells), which is similar to the population previously identified in the lungs of allergen-challenged mice; by contrast, no eosinophils were detected in the SigF+CD11c+ fraction. Taken together, these findings represent a crucial technical advance in our ability to enumerate eosinophils and to perform

downstream analyses, including characterization of eosinophil contents and gene expression in

this unique tissue microenvironment.

INTRODUCTION Although rarely detected in healthy muscle tissue, eosinophil infiltration is prominent in

disorders with significant muscle damage (e.g., muscular dystrophy [5-8], eosinophil-myalgia syndrome [9-11]) and in experimental animal models (e.g., injection with cobra cardiotoxin [12]).

23

Earlier work examining eosinophil function in the muscle tissue has relied in large part on

immunohistochemical detection using an antibody against the eosinophil granule protein, major

basic protein-1 (α-mMBP1). While this technique is important for determining eosinophil localization and their association with muscle lesions in situ, it is not suited for evaluating the cellular and biochemical properties of these leukocytes.

Multiple studies have demonstrated the heterogeneous nature of tissue eosinophils, including distinct granule contents [1, 13, 14], transcriptomic profiles [15], and even unique expression of cell surface antigens [1, 15-18]. It is thought that cues provided by the tissue

microenvironment, such as cytokines, chemokines and damage-associated molecular patterns

(DAMPs), prime eosinophils to assume unique, functional characteristics. These cues are

abundantly detected in the muscle tissue of dystrophin-deficient mdx mice [19-22] and may influence the characteristics of infiltrating eosinophils.

Flow cytometry is a powerful technique to quantify populations expressing specific antigen profiles and to analyze characteristics such as the level of immunoreactive protein expression.

Similarly, fluorescence-activated cell sorting (FACS) can facilitate isolation of cells with unique antigen profiles for downstream analyses. When applied to the study of eosinophils in muscular dystrophy, flow cytometry and fluorescence-activated cell sorting can be used to i) quantify the level of eosinophils in the muscle tissue across disease models; ii) measure the expression of specific proteins in eosinophils; and iii) isolate eosinophils for downstream applications. Although eosinophils in the bone marrow and in the lung have been identified as CD45+CD11c-Gr1-

SigF+MHC-IIlo cells [23-25], eosinophils have been shown to express distinct cell surface antigens depending on their activation state and tissue source. For example, eosinophils in the thymus and gastrointestinal tract express the CD11c antigen [26, 27] and a subset of eosinophils isolated from the lungs of allergen-challenged mice express the Gr1 antigen [1]. At the start of this study, the

antigen profile of eosinophils infiltrating the muscle tissue had not been fully characterized.

24

Here, we describe a protocol that facilitates preparation of single cell suspensions from

muscle tissue followed by procedures to identify and to isolate eosinophils by flow cytometry and

FACS, respectively. We provide a preliminary characterization of eosinophils from the muscle tissue, including their prevalence, antigen profile and cytological features. Finally, we note the impact that this technical advance has on our ability to perform subsequent characterization of eosinophils in the muscle tissue.

METHODS AND RESULTS

Mice mdx mice (C57BL/10ScSn-Dmdmdx/J, 001801) were obtained from Jackson laboratory and bred

under pathogen-free conditions in the National Institute of Allergy and Infectious Diseases (NIAID)

14B South Animal Facility. All experimental procedures were conducted in accordance with

approved animal study protocol LAD 7.

Preparation of Single Cell Suspension of Muscle Tissue

Mice were anesthetized, exsanguinated, and exposed to a lethal dose of isoflurane via inhalation

prior to cervical dislocation. Hindlimb skeletal muscle tissues were carefully dissected to remove

the surrounding fascia and excess fat. Muscle tissues were stored in a 6-well plate containing

Roswell Park Memorial Institute (RPMI) 1640 media, on ice, until completion of tissue collection.

As outlined in Figure 1, muscle tissues were then minced using dissection scissors and transferred to a 50 mL conical tube containing 10 mL of collagenase-containing muscle

dissociation media [RPMI, 10% fetal bovine serum, 1000 U/mL type II Collagenase, 1x Penicillin-

Streptomycin]; tubes were incubated in a rotating incubator at 37°C for 90 minutes. Cells were

pelleted and re-suspended in 10 mL of collagenase and dispase-containing muscle dissociation

media [RPMI, 10% fetal bovine serum, 100 U/mL type II Collagenase, 1 U/mL Dispase, 1x

Penicillin-Streptomycin]; tubes were returned to the rotating incubator and incubated for 37oC for

25

30 minutes. Muscle fragments were subjected to 10-successive passages through a 20-gauge

needle before filtration using a cell strainer with 40 µM pores. Cells were examined and counted

using a Luna-Fl Dual Fluorescent Counter (Logos Biosystems). Most cells appeared well-

separated and exhibited > 90% viability when assessed by propidium iodide-uptake. For

fluorescence-activated cell sorting (FACS), single cell suspensions were stained as described in

the following section; for flow cytometry, single-cell suspensions were frozen at 1-10 x 106

cells/mL in freezing media (90% FBS, 10% DMSO) prior to staining and analysis.

Flow Cytometric Analysis of Leukocyte Populations in Muscle Tissue To identify eosinophils in skeletal muscle tissue, we adapted our existing flow cytometry

strategy which had been used previously to identify eosinophils in the lung and bone marrow. Our

strategy utilizes a viability dye (Live/Dead Aqua), the pan-leukocyte marker (CD45), and a specific marker for mature mouse eosinophils (Siglec-F); the evaluation also includes markers commonly found on macrophages, dendritic cell and neutrophils (MHC-II, CD11c, and Gr1). Detailed information regarding antibody concentrations used and vendor information is shown in Table 1.

Fluorescent-labeled cells in suspension were evaluated using an LSR-II flow cytometer

(BD Biosciences) and further analyzed using the FlowJo software, version 10.6 (TreeStar). A

minimum of 200,000 events were recorded per sample. As shown in Figure 2, we set gates for

(i) single cells using a combination of forward scatter-height and forward scatter-area; (ii) non-

debris cells that were >50,000 side scatter-area and >50,000 forward scatter-area; (iii) live cells;

and (iv) CD45+ leukocytes. Within the CD45+ leukocyte population, we identified eosinophils from the (v) myeloid cells, which have relatively higher granularity, using forward scatter-area and side scatter-area; (vi) CD11c-Gr1- cells; (vii) and MHC-IIloSiglec-F+ cells. Although we identified muscle

eosinophils as CD11c-Gr1-SigF+ cells, we also explored the possibility that the smaller populations

of SigF+ CD11c+ or SigF+Gr1+ cells might be eosinophil variants; these findings are presented in the following section.

26

Wright-Giemsa Staining Provides Cytochemical confirmation of CD45+CD11c-Gr1-

SigF+MHC-IIlo cells as Eosinophils

Recent studies have demonstrated remarkable heterogeneity in eosinophil features,

including variations in antigen expression [1, 27, 28] and cytokine content [1, 15, 25] that relate

in part to the activation state and tissue source. Indeed, level of Siglec-F expression has been

considered to be a marker of eosinophil activation [29-31] and has been associated with altered cytokine profiles [15, 32], changes in eosinophil morphology [25], and antigen expression [15].

Likewise, eosinophils from the gastrointestinal tract and thymus express the antigen CD11c [26,

27], and, a subset of lung eosinophils from allergen-challenged mice express the antigen Gr1 [1].

We therefore considered the possibility that some or all the SigF+CD11c+ and/or SigF+Gr1+ cells infiltrating dystrophin-deficient skeletal muscle might also be eosinophils.

Fluorescence-activated cell sorting (FACS) is a specialized application of flow cytometry that permits isolation of cells expressing unique antigen profiles. We utilized FACS to isolate and evaluate the SigF+CD11c-Gr1-, SigF+CD11c+, SigF+Gr1+ populations from the muscle tissues of mdx mice at 4 weeks of age. As shown in Figure 2, panel viii and in Figure 3A, virtually all

(>99%) of the SigF+CD11c-Gr1- cells exhibited prominent eosinophil features when stained with modified Wright-Giemsa dyes (Diff-Quik, Thermo Fisher Scientific), including eosin staining of the granules and ring-shaped nuclei [31, 33, 34]. Flow cytometric analysis revealed robust levels of

Siglec-F in CD11c-Gr1-SigF+ cells, with a median fluorescence intensity (MFI) of 2250. This predominant population of Siglec-F+ cells was detected among the leukocytes infiltrating the muscle tissues of mdx mice at 4 weeks of age (7.7 ± 1.9% of total CD45+ cells). By contrast,

CD11c+SigF+ cells were detected at significantly lower levels within the dystrophic muscle tissue

(Figure 3B, 0.21 ± 0.12% of total CD45+ cells) and did not exhibit eosin staining of the granules

and lacked ring-shaped nuclei.

Interestingly, SigF+Gr1+ cells (0.11 ± 0.05% of total CD45+ cells, Figure 3C) exhibited

segmented nuclei but lacked robust granule staining. While nuclear segmentation is a feature that

27

eosinophils share with neutrophils, Percopo et al. [1] demonstrated that a subset of lung

eosinophils from allergen-challenged mice express the Gr1 antigen; unlike those identified in

muscle, these lung eosinophils were morphologically indistinguishable from the dominant

SigF+Gr1- population. Likewise, Limkar and colleagues [2] found that the Th2 cytokine, IL-5, drives

expression of Gr1 on eosinophils isolated from mouse bone marrow. Thus, by visual inspection

alone, we are unable to define the lineage of the SigF+Gr1+ cells. It may be possible that SigF+Gr1+

cells are a minor population of eosinophils that have undergone degranulation, and thus stain poorly with anionic eosin dyes.

DISCUSSION

In this study, we developed a protocol to identify eosinophils from skeletal muscle infiltrates detected in the mdx mouse model of Duchenne muscular dystrophy. We adapted a protocol from Liu et al. [3] for preparing single cell suspensions of muscle tissue. Using flow cytometry, we identified eosinophils isolated from the skeletal muscle tissue as liveCD45+CD11c-

Gr1-SigF+MHC-IIlo and identified eosinophils as a prominent component of the leukocyte infiltration in muscle tissue from mdx mice at 4 weeks of age. We confirmed that this population exhibited cytological features characteristic of eosinophils and we explored the possibility of eosinophils within the smaller SigF+Gr1+ and SigF+CD11c+ cell populations in the muscle tissue.

More recently, Kastenschmidt et al. [4] identified eosinophil populations from mdx muscle

infiltrates as F4/80+CD11b+SigF+ cells; further study of the cellular features of this population, such

as antigen expression, eosin staining, morphology, is needed to clarify potential similarities and

differences with the antigen profile we have described in this chapter.

Our strategy to identify eosinophils utilizes an antibody targeting the Sialic acid-binding

immunoglobulin-type lectin F (Siglec-F). Expressed on all mouse eosinophils, Siglec-F is a

convenient target for identification of eosinophils by flow cytometry [23]. However, administration

of antibodies targeting Siglec-F have been linked to reduced eosinophil levels in vivo [35] and

28

eosinophil apoptosis ex vivo [36]; a monoclonal antibody targeting the functional paralog in humans, Siglec 8, is currently under clinical exploration as a therapeutic agent in eosinophil- associated disorders [37-39]. Hence, use of an antibody targeting Siglec-F may affect eosinophil viability. As such, we ultimately identified alternative means for identifying eosinophils in lungs from IL5tg and allergen-challenged mice by gating on a combination of granularity, as determined by side scatter, and the antigens CD11c, Gr1, and MHC-II [40]. Unfortunately, this strategy was unsuccessful in identifying eosinophils in the muscle tissues (data not shown). Alternative markers for the identification of eosinophils, such as the IL-5 receptor alpha-subunit (CD125) and the chemokine receptor CCR3, were not ubiquitously or highly co-expressed on SigF+ eosinophils detected in the muscle tissue (data not shown). Hence, use of anti-Siglec-F appears to be necessary to discriminate between eosinophils and other, more numerous leukocyte populations detected in mdx muscle infiltrates. Despite the use of Siglec-F in our flow cytometry and fluorescence-activated cell sorting strategy, eosinophils (CD45+CD11c-Gr1-SigF+MHC-IIlo cells)

were largely viable and appeared intact morphologically (Figure 2).

Eosinophil expression of the CD11c and Gr1 antigens has been demonstrated in specific

tissue contexts. For example, eosinophils residing the thymus and gastrointestinal tract express

the CD11c antigen [26, 27] while a subset of eosinophils recruited to the airways of allergen-

challenged mice express the Gr1 antigen [1]. In this study, we identified a prominent population

of SigF+ eosinophils recruited to the muscle tissue (7.7 ± 1.9% of CD45+ cells); this population did not express the CD11c or Gr1 antigens. By contrast, we detected two minor populations of SigF+

cells that express the CD11c (0.21 ± 0.12% of CD45+ cells) or Gr1 antigens (0.11 ± 0.05% of

CD45+ cells). While the SigF+CD11c+ cells lacked clear cytological evidence of eosinophil features, the SigF+Gr1+ presented some features that have been observed in eosinophils and,

more broadly, in granulocytes. Cytological examination suggests that this population of SigF+Gr1+

cells may belong the neutrophil lineage, a hypothesis that is supported by the association of the

Gr1 antigen with the neutrophil lineage [41]. Definitive exclusion of this minor population of

29

SigF+Gr1+ from the eosinophil lineage may be obtained through genetic analyses, for example,

by measuring the expression of genes specific for neutrophils (e.g., myeloperoxidase, mpo; neutrophil elastase, elane) and eosinophils (e.g., eosinophil peroxidase, epx; major basic protein, prg2). Taken together, our findings indicate that the predominant population of eosinophils in the

muscle tissue lack expression of the CD11c and Gr1 antigens and can be identified as

CD45+CD11c-Gr1-SigF+MHC-IIlo cells.

Critically, flow cytometry and fluorescence-activated cell sorting (FACS) have enabled us

to quantify the level of eosinophil infiltration, to characterize the expression of immunoreactive

antigens on eosinophils, and to isolate eosinophils for downstream analyses. As described in

Chapter 3, we utilized this method to evaluate the level of eosinophil infiltration in the muscle

tissue of eosinophil-deficient and eosinophil-overabundant mdx mouse models; and we utilized

this method to evaluate the expression of the chemokine receptor, CCR3, on eosinophils [7].

Subsequently, in Chapter 4, we described use of this method to isolate eosinophil RNA for

sequencing; this approach has been instrumental in identifying unique, functional characteristics

in this infiltrating leukocyte population. Finally, in Chapter 5, we utilized this approach to

characterize expression of a novel cell surface protein that has not been previously reported on

eosinophils, the triggering receptor expressed on myeloid cells -2 (TREM-2).

30

FIGURES

Figure 1. Method for generating single cell suspensions from mouse skeletal muscle. As described in [3], single cell suspensions were generated from skeletal muscle tissue by enzymatic digestion using 1000 U/mL of type II collagenase followed by a digestion with 1000 U/mL type II collagenase and 11 U/mL dispase. Samples were further separated by 10 successive passages through a 20-gauge needle before filtration with a 40-µM cell strainer.

31

Antibody Vendor & Antibody Fluorophore Target Details Target Catalog Number Dilution

Membrane-permeable dye Thermo Fisher, Live/Dead Aqua 1:100 that enters nonviable cells #L34957

Fc Block BD Biosciences, none non-specific Fc binding 1:100 (CD16/32) #553142 Thermo Fisher, CD45 eF450 detected on leukocytes 1:100 #48-0451-82 detected usually on Thermo Fisher, CD11c Alexa 700 macrophage, dendritic 1:100 #56-0114-82 cells detected primarily on BD Biosciences, Gr1 APC neutrophils but also on 1:100 #553129 some eosinophils

BD Biosciences, SigF PE detected on eosinophils 1:100 #552126

detected primarily on dendritic cells, Thermo Fisher, MHC-II Pe-Cy7 1:100 macrophages, B-cells, T- #25-5321-82 cells

Table 1. Antibodies used for detection and isolation of eosinophils from skeletal muscle tissue.

32

Figure 2. Flow cytometry strategy to identify eosinophils in skeletal muscle tissue.

Eosinophils were identified in the skeletal muscle tissue as LiveCD45+CD11c-Gr1-SigF+MHC-IIlo

cells. Briefly, single cell suspensions of skeletal muscle tissues were gated (1) on forward scatter-

height and forward scatter-area to identify single cells and (2) side scatter-area and forward scatter-area to eliminate debris. Cells were then gated for (3) no staining with the membrane-

permeable Live/Dead dye, (4) presence of the leukocyte marker CD45 and (5) granularity based

on side scatter-area. Of the myeloid population, we identified eosinophils as (6) lacking CD11c,

Gr1 and (7) expressing Siglec-F and low levels of MHC-II. (8) Staining this population with modified Wright-Giemsa dyes (Diff-Quik) confirmed hallmark eosinophil characteristics such as eosin staining of the granules and the ring-form nuclear morphology. Figure reprinted with permission from [7].

33

Figure 3. Morphological evaluation of SigF+CD11c+ and SigF+Gr1+ populations. Specific populations of mdx muscle cells were isolated using fluorescence-activated cell sorting (FACS) and stained with modified Wright-Giemsa (Diff-Quik, Thermo Fisher Scientific). (A) SiglecF+ cells

that lack both the CD11c and Gr1 antigens (CD45+CD11c-Gr1-SigF+MHC-IIlo) comprised the dominant population (7.7% ± 1.9% of CD45+ cells) and exhibited hallmark eosinophil features, such as eosin staining and a ring-shaped nucleus. (B) By contrast, very few cells were detected as SigF+CD11c+Gr1- (0.21 ± 0.12% of total CD45+) and those that were detected were not morphologically eosinophils . (C) Likewise, the small population of SiglecF+Gr1+ cells (0.11 ±

0.05% of total CD45+ cells) exhibited ring-shaped nuclei but only minimal eosin staining of the

granules.

34

REFERENCES

1. Percopo, C.M., et al., SiglecF+Gr1hi eosinophils are a distinct subpopulation within the lungs of allergen-challenged mice. J Leukoc Biol, 2017. 101(1): p. 321-328.

2. Limkar, A.R., et al., Frontline Science: Cytokine-mediated developmental phenotype of mouse eosinophils: IL-5-associated expression of the Ly6G/Gr1 surface Ag. J Leukoc Biol, 2019.

3. Liu, L., et al., Isolation of skeletal muscle stem cells by fluorescence-activated cell sorting. Nat Protoc, 2015. 10(10): p. 1612-24.

4. Kastenschmidt, J.M., I. Avetyan, and S.A. Villalta, Characterization of the Inflammatory Response in Dystrophic Muscle Using Flow Cytometry. Methods Mol Biol, 2018. 1687: p. 43-56.

5. Wehling-Henricks, M., et al., Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy. Hum Mol Genet, 2008. 17(15): p. 2280-92.

6. Cai, B., et al., Eosinophilia of dystrophin-deficient muscle is promoted by perforin- mediated cytotoxicity by T cell effectors. Am J Pathol, 2000. 156(5): p. 1789-96.

7. Sek, A.C., et al., Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy. J Immunol, 2019. 203(2): p. 476-484.

8. Schroder, T., et al., Eosinophils in hereditary and inflammatory myopathies. Acta Myol, 2013. 32(3): p. 148-53.

9. Allen, J.A., et al., Post-epidemic eosinophilia-myalgia syndrome associated with L- tryptophan. Arthritis Rheum, 2011. 63(11): p. 3633-9.

10. Martin, R.W., et al., The clinical spectrum of the eosinophilia-myalgia syndrome associated with L-tryptophan ingestion. Clinical features in 20 patients and aspects of pathophysiology. Ann Intern Med, 1990. 113(2): p. 124-34.

11. Hertzman, P.A., et al., Association of the eosinophilia-myalgia syndrome with the ingestion of tryptophan. N Engl J Med, 1990. 322(13): p. 869-73.

12. Heredia, J.E., et al., Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell, 2013. 153(2): p. 376-88.

13. Chen, H.H., et al., Eosinophils from Murine Lamina Propria Induce Differentiation of Naive T Cells into Regulatory T Cells via TGF-beta1 and Retinoic Acid. PLoS One, 2015. 10(11): p. e0142881.

14. Sugawara, R., et al., Small intestinal eosinophils regulate Th17 cells by producing IL-1 receptor antagonist. J Exp Med, 2016. 213(4): p. 555-67.

15. Mesnil, C., et al., Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest, 2016. 126(9): p. 3279-95.

35

16. Johansson, M.W., et al., Up-regulation and activation of eosinophil integrins in blood and airway after segmental lung antigen challenge. J Immunol, 2008. 180(11): p. 7622-35.

17. Sedgwick, J.B., et al., Comparison of airway and blood eosinophil function after in vivo antigen challenge. J Immunol, 1992. 149(11): p. 3710-8.

18. Kroegel, C., et al., Blood and bronchoalveolar eosinophils in allergic subjects after segmental antigen challenge: surface phenotype, density heterogeneity, and prostanoid production. J Allergy Clin Immunol, 1994. 93(4): p. 725-34.

19. Pelosi, L., et al., Increased levels of interleukin-6 exacerbate the dystrophic phenotype in mdx mice. Hum Mol Genet, 2015. 24(21): p. 6041-53.

20. Villalta, S.A., et al., Interleukin-10 reduces the pathology of mdx muscular dystrophy by deactivating M1 macrophages and modulating macrophage phenotype. Hum Mol Genet, 2011. 20(4): p. 790-805.

21. Coles, C.A., et al., Expression profiling in exercised mdx suggests a role for extracellular proteins in the dystrophic muscle immune response. Hum Mol Genet, 2019.

22. Tidball, J.G., S.S. Welc, and M. Wehling-Henricks, Immunobiology of Inherited Muscular Dystrophies. Compr Physiol, 2018. 8(4): p. 1313-1356.

23. Dyer, K.D., et al., Antigen profiles for the quantitative assessment of eosinophils in mouse tissues by flow cytometry. J Immunol Methods, 2011. 369(1-2): p. 91-7.

24. Geslewitz, W.E., C.M. Percopo, and H.F. Rosenberg, Eosinophil persistence in vivo and sustained viability ex vivo in response to respiratory challenge with fungal allergens. Clin Exp Allergy, 2018. 48(1): p. 29-38.

25. Abdala Valencia, H., et al., Phenotypic plasticity and targeting of Siglec-F(high) CD11c(low) eosinophils to the airway in a murine model of asthma. Allergy, 2016. 71(2): p. 267-71.

26. Throsby, M., et al., CD11c+ eosinophils in the murine thymus: developmental regulation and recruitment upon MHC class I-restricted thymocyte deletion. J Immunol, 2000. 165(4): p. 1965-75.

27. Jung, Y., et al., IL-1beta in eosinophil-mediated small intestinal homeostasis and IgA production. Mucosal Immunol, 2015. 8(4): p. 930-42.

28. Weller, P.F. and L.A. Spencer, Functions of tissue-resident eosinophils. Nat Rev Immunol, 2017. 17(12): p. 746-760.

29. Voehringer, D., N. van Rooijen, and R.M. Locksley, Eosinophils develop in distinct stages and are recruited to peripheral sites by alternatively activated macrophages. J Leukoc Biol, 2007. 81(6): p. 1434-44.

30. Diny, N.L., et al., Eosinophil-derived IL-4 drives progression of myocarditis to inflammatory dilated cardiomyopathy. J Exp Med, 2017. 214(4): p. 943-957.

36

31. Abdala-Valencia, H., et al., Shaping eosinophil identity in the tissue contexts of development, homeostasis, and disease. J Leukoc Biol, 2018. 104(1): p. 95-108.

32. Ong, S., et al., Natural killer cells limit cardiac inflammation and fibrosis by halting eosinophil infiltration. Am J Pathol, 2015. 185(3): p. 847-61.

33. Dyer, K.D., et al., Protocols for identifying, enumerating, and assessing mouse eosinophils. Methods Mol Biol, 2013. 1032: p. 59-77.

34. Percopo, C.M., et al., Impact of eosinophil-peroxidase (EPX) deficiency on eosinophil structure and function in mouse airways. J Leukoc Biol, 2019. 105(1): p. 151-161.

35. Zimmermann, N., et al., Siglec-F antibody administration to mice selectively reduces blood and tissue eosinophils. Allergy, 2008. 63(9): p. 1156-63.

36. Zhang, M., et al., Defining the in vivo function of Siglec-F, a CD33-related Siglec expressed on mouse eosinophils. Blood, 2007. 109(10): p. 4280-7.

37. Youngblood, B.A., et al., AK002, a Humanized Sialic Acid-Binding Immunoglobulin-Like Lectin-8 Antibody that Induces Antibody-Dependent Cell-Mediated Cytotoxicity against Human Eosinophils and Inhibits Mast Cell-Mediated Anaphylaxis in Mice. Int Arch Allergy Immunol, 2019. 180(2): p. 91-102.

38. Youngblood, B.A., et al., Siglec-8 antibody reduces eosinophils and mast cells in a transgenic mouse model of eosinophilic gastroenteritis. JCI Insight, 2019. 4(19).

39. Legrand, F., et al., Sialic acid-binding immunoglobulin-like lectin (Siglec) 8 in patients with eosinophilic disorders: Receptor expression and targeting using chimeric antibodies. J Allergy Clin Immunol, 2019. 143(6): p. 2227-2237 e10.

40. Geslewitz, W.E., C.M. Percopo, and H.F. Rosenberg, FACS isolation of live mouse eosinophils at high purity via a protocol that does not target Siglec F. J Immunol Methods, 2018. 454: p. 27-31.

41. Lee, P.Y., et al., Ly6 family proteins in neutrophil biology. J Leukoc Biol, 2013. 94(4): p. 585-94.

37

CHAPTER 3

Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy

38

Chapter Preface In this chapter, I present my findings on the contribution of eosinophils to acute muscle

damage that is a characteristic of muscular dystrophy. This includes a reprint of our study that

was originally published in the Journal of Immunology.

Although eosinophil infiltration has been observed as a prominent feature of muscular

dystrophy, their contribution to disease pathophysiology is not well-understood. Traditionally, eosinophils have been viewed as tissue-destructive; this perception arises from their infiltration in pathophysiological states and from the presence of cationic granule contents that have characterized cytotoxicity in in vitro experimental systems. However, recent studies have illustrated beneficial immunomodulatory roles for these granulocytes in an array of disease contexts [35, 36, 49, 50].

Given this reconsideration of eosinophil function, we investigated their contribution to acute muscle damage in mdx mice, which model Duchenne Muscular Dystrophy by harboring null mutations in the dystrophin gene. Towards this end, we generated novel strains of dystrophin- deficient mdx mice, including those in which eosinophils are genetically-ablated (mdx.PHIL) or are present in large numbers (mdx.IL5tg). We also examined factors that might promote eosinophil infiltration in dystrophin-deficient muscle tissue.

Our major finding was that eosinophil infiltration did not drive the acute muscle damage in quadriceps muscle tissue in the setting of dystrophin-deficiency. We determined that eosinophil infiltration in this model was not associated with systemic eosinophilia or with any detectable type

2 immune response. However, we detected increased expression of canonical eosinophil chemo- attractants within the dystrophin-deficient skeletal muscle. As eosinophils are potent immunomodulatory cells, their contribution to muscle repair and inflammation in muscular dystrophy remains to be further elucidated.

These findings provided the impetus for subsequent investigations into the functional characteristics of eosinophils recruited to dystrophin-deficient skeletal muscle tissue (Chapter 4)

39 and into the mechanisms by which eosinophils are recruited to dystrophin-deficient skeletal muscle tissue (Chapter 6).

40

41

42

43

44

45

46

47

48

49

50

51

ADDENDUM

During the oral defense of the dissertation, I received excellent feedback regarding the contributions of eosinophil infiltration and its role in promoting muscle damage in the mdx mouse model of muscular dystrophy. Many of these points can be clarified by additional studies that have already been performed but were not included in the Sek AC et al. J Immunol, 2019 manuscript contained within this chapter. Hence, findings from these studies are included here as an addendum to this chapter.

Further evaluation of myofiber integrity and inflammation during the acute injury phase of muscular dystrophy

Measuring the distribution of myofiber sizes provides a quantitative assessment of the proportion of myofibers that have been damaged. In our initial assessment (Figure 3B, 3C of this chapter), we observed a normal bell-shaped distribution of myofiber sizes in quadriceps muscle tissues from wild-type and eosinophil-deficient PHIL mice at 4 weeks of age. By contrast, muscle tissues from mdx and mdx.PHIL mice did not exhibit the normal bell-shaped distribution of myofiber sizes. When compared with their respective controls, muscle tissues from mdx and mdx.PHIL mice included sizable proportions of enlarged (> 5000 µm2) or shrunken (< 1000 µm2) myofibers, indicating significant myofiber damage in both groups. Accordingly, we presented our data by comparing the distribution of myofiber sizes in mdx mice and wild-type controls and, separately, and presented the distribution of myofiber sizes in mdx.PHIL and PHIL controls.

These comparisons facilitated a binary understanding of myofiber damage that occurs both in the presence (mdx) and absence (mdx.PHIL) of eosinophil infiltration in dystrophin- deficient muscle tissues. However, these comparisons did not facilitate direct analysis of the reduction in myofiber damage that might accompany eosinophil depletion in muscular dystrophy.

To address this issue, we compared directly the distribution of myofiber sizes in quadriceps muscle tissues from male mdx and mdx.PHIL mice at 4 weeks of age (Figure A1a). The

52

distributions of myofiber sizes were remarkably similar between mdx and mdx.PHIL genotypes

and there were no statistically significant differences in any of the myofiber size bins. In order to

quantify the impact of eosinophil depletion on myofiber damage, we evaluated the proportion of

myofibers within the normal size range observed in quadriceps muscle tissues from wild-type

mice (1001 – 5000 µm2, 87 ± 3.5% of myofibers). Myofibers occurring within this size range can be distinguished from damaged myofibers that are swollen (>5000 µm2) or shrunken (<1000 µm2).

Using this metric, we detected similar proportions of myofibers of normal size in mdx and mdx.PHIL mice (62 ± 5.5 % vs 56 ± 4.6%, Figure A1b). The proportion of myofibers of normal sizes in mdx and mdx.PHIL mice differed significantly from that of their respective wild-type (87 ±

3.5% of myofibers, p = 0.0054) and PHIL (79 ± 6.6, p = 0.0041) controls. Intriguingly, the proportions of myofibers within this normal size range were similar to the proportions of healthy

myofibers detected by nuclear positioning (Figure 3D of this chapter), which were 72 ± 2.3% in mdx mice, 70 ± 1.8% in mdx.PHIL mice. Taken together, these data indicate that eosinophil infiltration has no apparent impact on quadriceps muscle damage at 4 weeks of age in dystrophin- deficient mdx mice.

Next, we asked if manipulation of the eosinophil lineage might have an impact on the other leukocyte populations recruited to dystrophin-deficient muscle tissue. We focused our attention on neutrophils, a subset of myeloid cells that shares many features in common with eosinophils.

As shown in Figure A1c, neutrophils (CD45+CD11c-Gr1+SigF-) were detected at similar levels in preparations of skeletal muscle tissues from mdx (0.90 ± 0.45% of live cells), mdx.PHIL (0.90 ±

1.0% of live cells) and mdx.IL5tg (0.58 ± 0.41% of live cells) mice at 4 weeks of age. Although these data suggest that eosinophil infiltration does not affect the degree of neutrophil infiltration in dystrophin-deficient muscle tissues, eosinophil infiltration may have an impact on the infiltration

of other leukocyte populations, including mast cells and T-lymphocytes.

53

Eosinophil Infiltration during the regenerative phase of muscular dystrophy

Muscular dystrophy (mdx) mice exhibit well-defined stages of skeletal muscle

pathophysiology; these include acute muscle injury at 3-4 weeks of age and ongoing regeneration

at 6-12 weeks of age. In our earlier work, we focused exclusively on the 4-week timepoint in order to evaluate the impact of eosinophil infiltration on acute muscle damage in mdx mice. Since eosinophils have been described as critical to successful tissue regeneration in several experimental models, we evaluated the impact of eosinophil infiltration during the regenerative phase of muscular dystrophy. Quadriceps muscle tissues from mdx mice at 8 weeks of age exhibited histological features consistent with ongoing regeneration, including regions with intact myofibers of intermediate size and regions with limited myofiber necrosis (Figure A2a). Major basic protein-1 (MBP)-positive eosinophils were detected in quadriceps muscle tissues from mdx mice at 8 weeks of age (Figure A2b), often near sites of myofiber necrosis. Flow cytometric analysis identified populations of eosinophils (CD45+CD11c-Gr1-SigF+MHC-IIlo) among

preparations of skeletal muscle tissues from mdx and eosinophil-enriched mdx.IL5tg mice at 8

weeks of age; unsurprisingly, eosinophils were rarely detected in preparations of skeletal muscle

tissues from eosinophil-deficient mdx.PHIL mice (Figure A2c). Interestingly, the fraction of eosinophils (% of live cells) was significantly lower in preparations of skeletal muscle tissues from mdx mice at 8 weeks of age compared with 4 weeks of age (0.58 ± 0.31% of live cells versus 1.2

± 0.34% of live cells, respectively; p = 0.007).

Evaluation of muscle pathology during the regenerative phase of muscular dystrophy

As eosinophil infiltration is detected in skeletal muscle tissues from mdx mice during the regenerative phase (8 weeks of age), I asked if eosinophil infiltration might contribute to the successful regeneration of damaged dystrophin-deficient muscle tissue. Towards this end, I evaluated the histopathology of quadriceps muscle tissues from mdx, eosinophil-deficient mdx.PHIL, and eosinophil-enriched mdx.IL5tg mice at 8 weeks of age. Histological evaluation of

54

quadriceps muscle tissue from eosinophil-deficient mdx.PHIL mice at 8 weeks of age revealed

prominent myofiber regeneration, indicated by central positioning of nuclei within myofibers, along

with limited edema and myofiber necrosis (Figure A3a). Similarly, quadriceps muscle tissue from

eosinophil-enriched mdx.IL5tg mice at 8 weeks of age revealed prominent myofiber regeneration,

as indicated by central positioning of nuclei within myofibers, along with limited myofiber necrosis

and robust leukocyte infiltration (Figure A3b).

In order to evaluate quantitatively the impact of eosinophil infiltration on the regeneration

of damaged, dystrophin-deficient muscle tissues, I assessed features such as myofiber integrity

and myofiber regeneration. Quadriceps muscle tissues from eosinophil-deficient mdx.PHIL mice

at 8 weeks of age exhibited similar distribution of myofiber sizes when compared with the mdx

parent strain at 8 weeks of age (Figure A3c), including similar proportions of small myofibers less

than 1000 µm2 (15 ± 3.2 % vs 15 ± 1.5%, respectively) and large myofibers greater than 5000

µm2 (19 ± 5.7% vs 19 ± 3.1%, respectively). Compared with the normal myofiber size range at 8 weeks of age (1001 – 6000 µm2, 80 ± 2.7% of wild-type myofibers), substantial proportions of

myofibers of normal size were detected in mdx (70 ± 1.6%) and mdx.PHIL (70 ± 3.2%) mice;

these data indicate ongoing myofiber regeneration within the muscle tissues of both mdx and

mdx.PHIL mice at 8 weeks of age. Furthermore, quadriceps muscle tissues from eosinophil-

deficient mdx.PHIL and eosinophil-enriched mdx.IL5tg mice at 8 weeks of age exhibited similar

proportions of centrally-nucleated myofibers when compared with the mdx parent strain at 8

weeks of age (Figure A3d; 37 ± 3.9%, 41 ± 7.34% vs 37 ± 3.5%, respectively). Of note, the proportions of centrally-nucleated myofibers detected across dystrophin-deficient mouse strains at 8 weeks were slightly higher than those at 4 weeks of age (Figure 3D; 27 ± 2.4%, mdx; 30 ±

1.9%, mdx.PHIL). Taken together, these data indicate that the presence of eosinophil had no significant impact on the integrity or regeneration of myofibers in 8-week mice during the regenerative phase of muscular dystrophy.

55

FIGURES

Figure A1. Quantification of myofiber integrity and neutrophil infiltration in quadriceps

muscle tissues from mice at 4 weeks of age. (A) Distribution of myofiber sizes in quadriceps muscle tissue; data reproduced from Figures 3B and 3C, with modifications. n = 3.0 X 104

myofibers (mdx), 3.2 X 104 myofibers (mdx.PHIL); 2-3 mice per genotype. None of the differences

observed were statistically significant between mdx and mdx.PHIL groups, two-way ANOVA with

multiple comparisons. (B) Quantification of myofibers of intermediate size (1001 – 5000 µm2) in

quadriceps muscle tissues; data reproduced from Figures 3B and 3C with modifications. n = 2-3 mice per genotype, ** p < 0.01, one-way ANOVA with multiple comparisons. (C) Neutrophils

(CD45+CD11c-Gr1+SigF-) as a percent of the total live cells in preparations of quadriceps muscle tissues from male mice at 4 weeks of age. n = 2 – 15 mice per genotype, n.s. = no significant difference, one-way ANOVA with multiple comparisons.

56

Figure A2. Histopathology and eosinophil infiltration in quadriceps muscle tissues from

mice at 8 weeks of age. (A) H&E-stained cross-section of quadriceps muscle tissue from a male

mdx mouse at 8 weeks of age. (B) Immunohistochemical staining with anti-MBP documenting

eosinophil infiltration in quadriceps muscle tissue from a male mdx mouse at 8 weeks of age. (C)

Eosinophils (CD45+CD11c-Gr1-SigF+MHC-IIlo) as a percent of the total live cells in preparations

of quadriceps muscle tissues from male mice at 8 weeks of age. n = 4 – 5 mice per genotype, **** p < 0.0001, n.s. = no significant difference, one-way ANOVA with multiple comparisons.

57

Figure A3. Histopathology and quantification of myofiber integrity in quadriceps muscle

tissues from mice at 8 weeks of age. (A) H&E-stained cross-section of quadriceps muscle

tissue from a male mdx.PHIL mouse at 8 weeks of age. (B) H&E-stained longitudinal section of quadriceps muscle tissue from a male mdx.IL5tg mouse at 8 weeks of age. (C) Distribution of

myofiber sizes in quadriceps muscle tissues from mdx and mdx.PHIL mice at 8 weeks of age; n

= 3 per genotype. None of the myofiber sizes were statistically significant between mdx and

mdx.PHIL groups, two-way ANOVA with multiple comparisons. (D) Proportion of centrally

nucleated myofibers in quadriceps muscle tissues from male mice at 8 weeks of age. ** p < 0.01,

*** p < 0.001, one-way ANOVA with multiple comparisons.

58

CHAPTER 4

Transcriptomic Profile of Eosinophils Recruited to Muscle Tissue of Dystrophin-deficient (mdx) mice

59

CHAPTER 4 PREFACE

RNA-sequencing is a powerful technique that allows for simultaneous detection and

quantification of all transcripts contained within a population of cells. As such, RNA-sequencing

provides a comprehensive, unbiased view of the genetic processes underlying key pathways and

functional mediators within cells.

To date, there have been fewer than 7 publications that have utilized RNA-sequencing to

interrogate eosinophil characteristics [1-6]. There are several reasons for the paucity of studies utilizing this approach; chief among them is the perception that eosinophils do not respond transcriptionally to the tissue microenvironment. However, increasing evidence indicates that eosinophils exhibit unique properties, including distinct transcriptomic profiles, that depend in part on the tissue from which they are isolated [1, 4, 5, 7-9]. It is thought that cues provided by the

tissue microenvironment prime eosinophils to assume distinct characteristics that potentiate their function [10].

In muscular dystrophy (mdx) mice, eosinophils are detected in inflammatory infiltrates that are associated with severe muscle damage [11-13]. To develop a comprehensive understanding of the characteristics of eosinophils recruited to skeletal muscle and to explore the role of these cells in the pathogenesis of muscular dystrophy, we examined gene expression with RNA-sequencing.

This chapter represents our successful efforts towards obtaining high-quality RNA from eosinophils, followed by sequencing and analysis of genes that are differentially expressed in eosinophils from distinct microenvironments. Among the differentially expressed genes, we detected high levels of the transcript encoding Triggering Receptor Expressed on Myeloid Cells-

2 (TREM2) in eosinophils from mdx muscle tissue; investigation into the regulation and function of TREM2 on eosinophils forms the basis of subsequent investigation (Chapter 5).

60

ABSTRACT

Eosinophils are among the leukocytes recruited to the muscle lesions in muscular dystrophy

(mdx) mice, although their contribution to disease remains incompletely understood. In our earlier

work, we demonstrated that eosinophils do not drive the acute muscle damage in mdx mice but

do contribute immunomodulatory mediators including IL-4 and IL-1RA. To uncover additional

mediators of eosinophil function in the muscle tissue, we isolated and sequenced eosinophil RNA

from the muscle tissues of mdx, IL5tg and mdx.IL5tg mice at 4 weeks of age. Despite abundant

ribonucleases contained within eosinophil granules, we were able to isolate high-quality RNA

(RIN: 8.2 – 9.7) for sequencing. Analysis of our sequencing results revealed distinct transcriptomic profiles among eosinophils isolated from the muscle tissues of mdx and IL5tg mice. We identified

393 unique transcripts that were detected at significantly higher levels in eosinophils from muscle tissues from mdx mice when compared with those from IL5tg; by contrast, we detected 238 unique transcripts that were detected at significantly higher levels in eosinophils from muscle tissue from

IL5tg mice when compared with those from the mdx strain. Among the set of 393 transcripts increased in eosinophils from mdx muscle tissue, we detected many transcripts encoding enzymes associated with tissue remodeling (Ctsk, Ctss, Mmp2, Mmp12) as well as transcripts encoding cell surface receptors (Cd14, Slc15a3, Trem2). Gene ontology pathway confirmed that the set of 393 transcripts elevated in eosinophils from mdx muscle tissue was significantly associated with tissue remodeling pathways such as serine-type peptidase activity (p = 2.2e-07) and collagen binding (p = 9.4e-10). Taken together, these findings challenge the perception of eosinophils as static and end-stage and highlight unique features for further investigation. As such, ongoing work will elucidate the impact of receptors, such as TREM2, on the functions and characteristics of eosinophils in health and disease.

61

INTRODUCTION

Eosinophils are granulocytes that are recruited to dystrophin-deficient muscle tissue, where their contributions to disease pathophysiology remain incompletely understood. Recently, we demonstrated that eosinophil infiltration does not drive the acute muscle damage in muscular dystrophy (mdx) mice [13]. These findings are consistent with earlier work by Wehling-Henricks and colleagues, who demonstrated that deletion of the gene encoding the cytotoxic eosinophil granule protein Major Basic Protein-1 (MBP-1), did not have any impact on the level of muscle damage in mdx mice [12].

Rather, we uncovered elevated levels of immunomodulatory mediators associated with

eosinophil infiltration in dystrophin-deficient muscle tissue; these include the pro-regenerative

cytokine interleukin-4 (IL-4) as well as the anti-inflammatory mediator interleukin-1 receptor antagonist (IL-1RA) [13]. Increased expression and release of these eosinophil-derived cytokines

may affect the pathophysiology in muscular dystrophy. While important, IL-4 and IL-1RA

represent but a small fraction of the known repertoire of mediators that are contained within

eosinophil granules and lipid bodies [14]. As such, IL-4 and IL-1RA may work in conjunction with

other eosinophil-derived cytokines to mediate key eosinophil functions in muscular dystrophy.

Along these lines, recent studies have reported unique functional characteristics in

eosinophils responding to pathophysiological contexts. For example, Arnold and colleagues [1]

described distinct transcriptomic profiles in eosinophils isolated from the gastrointestinal tract

following bacterial infection when compared with eosinophils from the gastrointestinal tracts of

uninfected mice. Furthermore, eosinophils from the gastrointestinal tract of infected mice

exhibited higher levels of programmed death ligand-1 (PDL-1) which directly suppressed the

bacterially induced Th1/Th17 response [1].

By extension, eosinophils recruited to the dystrophin-deficient muscle microenvironment may

exhibit unique, as yet undetermined, characteristics that help promote its function within muscular

dystrophy. To uncover these characteristics, we utilized an unbiased, comprehensive approach

62

by sequencing eosinophil RNA isolated from the muscle tissues of mdx, IL5tg, and mdx.IL5tg. We compared the transcriptomic profiles of eosinophils isolated from the muscle tissues of mdx mice with those from IL5tg in order to identify key functional mediators for further investigation.

MATERIALS AND METHODS

Mice

Mdx mice (C57BL/10ScSn-Dmdmdx/J, 001801) were obtained from Jackson Laboratory. IL5tg mice were obtained from the Lee Laboratories (Mayo Clinic). Mdx-IL5tg mice were generated by crossing female mdx mice with male IL5tg mice; male offspring were serotyped to determine IL-

5 expression. Only male mice were used in all experiments. All mice were bred and maintained under pathogen-free conditions in the National Institute of Allergy and Infectious Diseases (NIAID)

14B South Animal Facility; all experimental procedures were conducted in accordance with approved animal study protocol LAD7.

Isolation of Eosinophils by Fluorescent-Activated Cell Sorting

As described in Chapter 2, single cell suspensions were generated from hind limb muscle tissue.

These cells were stained with a viability dye (Live/Dead Aqua, ThermoFisher), an Fc receptor blocking antibody (anti-CD16/32, BD Biosciences) and fluorochrome-conjugated antibodies including anti-CD45 and anti-CD11c (eBioscience); anti-Gr1 and anti-Siglec-F (BD Biosciences).

Eosinophils (liveCD45+CD11c-Gr1-SigF+MHC-IIlo cells) were isolated and sorted directly into

TRIZOL-LS (Life Technologies) by fluorescence-activated cell sorting (FACS).

Isolation and Purification of Eosinophil RNA

RNA was isolated from eosinophils that had been FACS sorted into TRIZOL-LS (Life

Technologies). Following the manufacturer’s protocol, the 3:1 ratio of TRIZOL-LS to sample was maintained prior to addition of chloroform. RNA was purified from the aqueous phase, combined

63

with equal volumes of 70% ethanol, and subjected to the RNeasy cleanup kit (Qiagen) to remove

potential contaminants. RNA samples were resuspended in 20-25 µL of RNase-free water and

stored at -80oC prior to quality control analysis.

RNA Quality

RNA samples were loaded onto an RNA Pico Chip (Agilent Technologies) and the electrophoretic trace was analyzed using an the 2100 Bioanalyzer (Agilent Technologies). Based on the ratio of the 28S:18S RNA detected, the 2100 Bioanalyzer assigned RNA integrity number (RIN) scores, which range from 0 (most degraded) to 10 (most intact).

RNA-Sequencing and Analysis

RNA libraries were prepared using the NEBnext Ultra Low Input Total RNA Library Prep Kit (New

England BioLabs) following the manufacturer’s instructions. Paired end-reads of the RNA libraries were sequenced using an Illumina HiSeq 4000 Next Generation sequencer, with more than 100

Million reads per sample. Using the Cutadapt software, reads were trimmed to remove adapters and low-quality sequences before aligning to the mouse reference genome mm10. Transcript abundance was quantified using the RSEM software package and reported as fragments per kilobase of transcript per million reads (FPKM).

Identification of Differentially Expressed Genes

Voom-normalized gene counts from eosinophils from mdx muscle tissue were compared against those from eosinophils from muscle tissue from IL5tg mice. Transcripts that were significantly upregulated more than 5-fold in eosinophils from the muscle tissues of mdx vs. IL5tg mice were identified; statistical significance was determined using limma empirical bayes t-test with multiple

test correction.

64

RESULTS

Successful isolation of high-quality RNA from muscle eosinophils

Within their cytoplasmic granules, eosinophils contain abundant ribonucleases that can

complicate the isolation of high-quality RNA for sequencing. As eosinophils represent a

subpopulation of the leukocytes that are recruited to skeletal muscle tissue, efforts to isolate RNA

from this population must be specific for the eosinophil population and rapid to avoid RNA

degradation secondary to endogenous ribonuclease activity. Here, eosinophil RNA was obtained

by isolating eosinophils (CD45+CD11c-Gr1-SigF+MHC-IIlo cells; described in Chapter 2) from the muscle tissues and subsequently extracting RNA from this population using the phenol-based

TRIZOL-LS reagent (Figure 1A). To remove contaminants, eosinophil RNA was further purified using the RNeasy Mini Kit (Qiagen); RNA purification has been utilized in prior studies examining

eosinophil transcriptomes [6, 15, 16]. Furthermore, all experimental procedures were conducted on the same day to limit potential changes to the transcriptome, as well as nucleic acid degradation, that can be associated with prolonged experimental procedures.

A total of 9 RNA samples were collected; these samples represent biological replicates from each of the three relevant genotypes (mdx, IL5tg, mdx.IL5tg). Although 3 - 5 mice were used for each biological sample, the number of eosinophils varied depending on the genotype. For example, approximately 400,000 eosinophils were collected from each mdx sample while >

1,000,000 eosinophils were collected for each mdx.IL5tg sample (Figure 1B). Therefore, the total

RNA yield per sample varied from 12 ng to 547 ng (Figure 1B); these yields were similar to those reported by Wacht and colleagues who successfully isolated ~100 ng of RNA from the peripheral blood eosinophils of healthy volunteers [15].

Eosinophil RNA isolated from the muscle tissues using the method outlined above exhibited minimal degradation. All RNA samples submitted for sequencing had an RNA Integrity Number

(RIN) greater than 8.20, with most samples having a RIN greater than 9.0 (Figure 2). These RIN

65

values are similar to those of Wacht et al. [15] and Barnig et al. [16] who reported RIN scores

between 9.2 - 9.4 in RNA isolated from peripheral blood eosinophils from healthy volunteers.

Distinct transcriptomic profiles in muscle eosinophils

Because eosinophils contain pre-formed cytokines within their cytoplasmic granules,

eosinophils were viewed traditionally as end-stage effector cells incapable of responding

transcriptionally to distinct stimuli. However, analysis of the transcriptome in isolated eosinophils

indicated abundant expression of transcripts: in eosinophils from mdx muscle tissue, a total of

9,525 distinct transcripts were detected at ≥ 2 fragments per kilobase of transcript per million

reads (FPKM); similarly, 9,258 transcripts were detected in eosinophils from muscle tissue from

IL5tg mice and 8,870 transcripts were detected in eosinophils from mdx.IL5tg muscle tissue.

To identify transcripts that may encode key eosinophil characteristics, we compared the transcriptome of eosinophils from the skeletal muscle tissues of mdx mice against the transcriptome of eosinophils from the skeletal muscle tissues of IL5tg mice. We reasoned that eosinophils detected in the presence of severe muscle damage associated with dystrophin- deficiency would differ significantly from eosinophils detected in the absence of muscle damage, as in IL5tg mice [13]. Consistent with our hypothesis, eosinophils from the mdx muscle tissue expressed 393 unique transcripts that were detected at >5-fold higher levels when compared with

eosinophils from the muscle tissues of IL5tg mice (mdx > IL5tg; Figure 3A). Many of the mdx >

IL5tg transcripts encode proteins involved with tissue remodeling such as the cysteine protease

family of cathepsins (e.g., Ctsb, Ctsk, Ctss) and the zinc endopeptidase family of matrix

metalloproteinases (e.g., Mmp19, Mmp14, Mmp2). In addition, many of the mdx > IL5tg

transcripts highly detected in eosinophils from the mdx muscle tissue encode cell surface proteins

such as inflammatory and chemotactic receptors (e.g., Cd14, Ccr3, Nlrp3, Trem2) as well as

transporters (e.g., Slc15a3, Slc22a4). Analysis of the mdx > IL5tg set of transcripts indicated

significant association with tissue remodeling pathways (Figure 3B), such as collagen binding

66

(GO ID: 0005518, p = 9.41e-10), extracellular matrix structural constituent (GO ID: 0005201, p =

3.17e-07) and metalloendopeptidase activity (GO ID: 0004222, p = 3.60e-04).

By contrast, eosinophils from the muscle tissues of IL5tg mice expressed 238 unique

transcripts that were detected at >5-fold higher levels when compared with eosinophils from the

muscle tissues of mdx mice (IL5tg > mdx; Figure 3A). Many of the IL5tg > mdx transcripts encode

eosinophil-specific proteins such as eosinophil-associated ribonucleases (e.g., Ear1, Ear2, Ear6) and eosinophil granule proteins such as eosinophil peroxidase (Epx) and major basic protein-1

(Prg2). In addition, many of the IL5tg > mdx transcripts encode proteins involved with cell cycle processes such as histones (e.g., Hist1h2bn, Hist1h3c). Analysis of the IL5tg > mdx set of transcripts indicated significant association with eosinophil-specific pathways such as defense response to nematode (GO ID: 0002215, p = 7.54e-05); as well as cellular proliferation pathways such as chromatin assembly (GO ID: 0031497, p = 1.71e-04).

Eosinophils recruited to mdx muscle tissue exhibit increased expression of transcripts associated with tissue remodeling

Given the association between the mdx transcriptome and tissue remodeling, we examined the level of key transcripts in eosinophils from the muscle tissues of mdx, IL5tg and mdx.IL5tg mice (Figure 4). We identified a set of 32 key transcripts that encode proteins involved with tissue remodeling. In eosinophils from the muscle tissues of mdx mice, these transcripts were detected at relatively abundant levels; this may have profound physiologic impact. For example, 23 of these transcripts were expressed at FPKM values >10 in eosinophils from the muscle tissues of mdx mice; by contrast, only 4 of these transcripts were expressed at FPKM values >10 in eosinophils from the muscle tissues of IL5tg mice.

Of the 32 key transcripts, many encoded enzymes associated with tissue remodeling, including the cysteine protease family of cathepsins as well as the zinc endopeptidase family of matrix metalloproteinases (MMPs). For example, transcripts encoding cathepsin S (Ctss), were

67

detected at 198-fold higher levels in eosinophils from the mdx muscle tissue when compared with

that of IL5tg (p = 1.57e-03). Similarly, transcripts encoding the enzyme, matrix metalloproteinase

12 (Mmp12), were detected at 172-fold higher levels in mdx eosinophils when compared with that

of IL5tg eosinophils (p = 5.17e-04). Both cathepsin S and matrix metalloproteinase 12 have been

associated with the degradation of components of the extracellular matrix [17-21].

In addition, many of the key transcripts encode structural components of the extracellular

matrix in muscle tissue. For example, transcripts of Col22a1, which encodes the alpha 1 chain of type XXII collagen, were detected at 142-fold higher levels in eosinophils from mdx mice when compared with that of IL5tg (p = 2.41e-05). Similarly, transcripts of Col3a1, which encode the alpha 1 chain of type III collagen, were detected at 11-fold higher levels in eosinophils from mdx mice when compared with that that of IL5tg (p = 6.76e-04). Collagens, together with laminins, fibronectins and proteoglycans, form the extracellular matrix that surrounds myofibers [22].

Eosinophils recruited to mdx muscle tissue exhibit increased expression of transcripts encoding cell surface proteins

Given the association between the mdx transcriptome and cell surface proteins, we examined the expression of key transcripts in eosinophils from the muscle tissues of mdx, IL5tg and mdx.IL5tg mice (Figure 5). We identified 17 key transcripts that encode cell surface proteins.

Expression of these transcripts were relatively high: 9 of the transcripts were expressed at FPKM values >10 in eosinophils from the muscle tissues of mdx mice; by contrast, only 1 of the transcripts was expressed at FPKM values >10 in eosinophils from the muscle tissues of IL5tg.

The relative abundance of these key transcripts in eosinophils from the muscle tissues of mdx mice suggests they may encode proteins at level sufficient to produce a physiologically relevant impact.

Of the 17 key transcripts, many encoded inflammatory receptors that may potentiate eosinophil characteristics within dystrophin-deficient muscle tissue. For example, transcripts of

68

Cd14, which encode the CD14 receptor, were detected at 6.8-fold higher levels in eosinophils

from the muscle tissues of mdx mice when compared with eosinophils from the muscle tissues of

IL5tg mice (p = 3.81e-04). Expression of the CD14 receptor has not been previously detected on

eosinophils [23]; CD14 has been reported as a co-receptor for toll-like receptor 4 (TLR4) on

macrophages [24, 25]. Similarly, transcripts of Nlrp3, which encodes the NALP3 inflammasome

receptor, were detected at 18.51-fold higher levels in eosinophils from the muscle tissues of mdx

mice when compared with that of IL5tg (p = 2.22e-03). In macrophages, NALP3 receptor

recognizes damage-associated molecular patterns to initiate the release of the pro-inflammatory

cytokine interleukin-1 beta (IL1β) [26]; however, expression and function of NALP3 on eosinophils

has not been reported previously.

In addition to inflammatory receptors, many of the key transcripts encoded transporters that

may have an impact on the bioenergetic state of eosinophils in the muscle tissues of mdx mice.

For example, transcripts encoding the peptide/histidine transporter SLC15A3 (Slc15a3) were

detected at 15.5-fold higher levels in eosinophils from the muscle tissues of mdx mice when

compared with that of IL5tg (p = 4.74e-04). Expression of SLC15A3 has not been previously

reported on eosinophils; however, increased expression of SLC15A3 has been reported in

macrophages following stimulation of the TLR2, TLR4, TLR7 or TLR9 signaling pathways [27].

Similarly, transcripts encoding the ergothioneine transporter SLC22A4 (Slc22a4), were detected

at 11.3-fold higher levels in eosinophils from the muscle tissues of mdx mice when compared with that of IL5tg (p = 3.35e-04). Expression of SLC22A4 transporter has not been previously reported on eosinophils; furthermore, the physiological relevance to transport of ergothioneine, a key antioxidant, remains unclear [28].

Intriguingly, levels of the transcript encoding TREM2 (Trem2) were detected at 68-fold higher levels in eosinophils from the muscle tissues of mdx mice when compared with that detected in the IL5tg (p = 5.23 e-03). Trem2 encodes the triggering receptor expressed on myeloid cells-2

(Trem2) which has been linked to the activation, proliferation, and survival of microglial cells

69

encountering tissue damage [29]. Expression of Trem2 has not been reported on eosinophils;

detailed examination of the function and regulation of this receptor is presented in Chapter 5.

Discussion

In this study, we examined the transcriptional profile of eosinophils recruited to the muscle

tissues of mdx, IL5tg and mdx.IL5tg mice at 4 weeks of age. Although eosinophils contain abundant ribonucleases within their cytoplasmic granules, we were able to isolate high-quality

RNA for sequencing (RIN Scores: 8.2-9.7). Analysis of our RNA-sequencing experiment identified distinct transcriptomic features among eosinophils recruited to the muscle tissues of mdx and

IL5tg mice, including 393 unique transcripts that were detected at significantly higher levels in eosinophils from the muscle tissues of mdx mice and 238 unique transcripts that were detected at significantly higher levels in eosinophils from the muscle tissues of IL5tg mice. Many of the 393 unique transcripts identified in eosinophils from the muscle tissues of mdx mice encode proteins involved with tissue remodeling, suggesting a potential role for eosinophils in the remodeling of muscle damage associated with dystrophin-deficiency. Intriguingly, many of the 393 unique transcripts encode cell surface receptors, suggesting that eosinophils may be actively sensing and responding to cues provided by the dystrophin-deficient muscle microenvironment.

Our findings challenge the perception of eosinophils as static and end-stage by demonstrating that eosinophils recruited to dystrophin-deficient (mdx) muscle tissue exhibit distinct transcriptomic features compared with eosinophils recruited to hyper-eosinophilic (IL5tg) muscle tissue. These findings add to the growing body of evidence that demonstrate heterogeneity in the characteristics of eosinophils. For example, Mesnil and colleagues used mRNA microarrays to identify differential expression of transcripts among populations of inflammatory and resident eosinophils in allergen-challenged airways [8]. In another instance, Barnig and colleagues used mRNA microarrays to describe distinct transcriptomic features among peripheral blood eosinophils from asthmatic patients compared with that of healthy donors [16].

70

The distinct transcriptomic features observed in eosinophils isolated from the muscle tissues

of mdx and IL5tg mice may derive in part from unique signals provided by the tissue microenvironment. In mdx mice, the dystrophin-deficient muscle microenvironment contains

abundant damage-associated molecular patterns (DAMPs) such as the high mobility group box 1

(HMGB1, [30]); and also contains abundant cytokines and chemokines. By contrast, eosinophils

isolated from the skeletal muscle tissues of IL5-transgenic mice may have been primed by

constitutively high levels of the eosinophilopoietic cytokine, IL-5. Extensive work among

eosinophil researchers has identified a wealth of receptors that are expressed on the surface of

eosinophils; activation of these receptors may contribute to initiation of key pathways involved

with gene transcription. Among the receptors described on eosinophils are adhesion receptors

(e.g., CD44, CVLA4) and pattern recognition receptors (e.g., TLR2, TLR4) as well as receptors

that modulate responses to chemoattractants (e.g., CCR3, CCR5), lipid mediators (e.g.,

Leukotriene B4 receptor, EP4 prostaglandin receptor), and cytokines (e.g., IL-4R, IL-5R); a more

comprehensive list of receptors expressed on eosinophils was compiled by Lee and colleagues

[31].

Current understanding of eosinophil transcriptional pathways that occur downstream of

receptor activation is limited by the relatively few studies examining this topic. Stokes and

colleagues demonstrated that the transcription factor stat6, which is activated by IL-4/IL-13

signaling, is required for eosinophil migration to allergen-challenged airways; however, loss of

stat6 did not affect the immunoreactive levels of key cytokines in eosinophils [32]. Changes in the

eosinophil transcriptome that occur as a result of receptor activation may be mediated through

other transcription factors. For example, Fairfax and colleagues identified 9 transcription factors

(e.g., Ikzf3, Nod2, Bcl3, Foxo1) present in mature eosinophils [33]; activation of any combination

of these transcription factors may in turn propagate and impart the unique transcriptomic features

observed in eosinophils isolated from the muscle tissues of mdx and IL5tg mice. Despite limited

understanding of transcriptional regulation in eosinophils, this remains an area of active research

71 with important clinical relevance. Glucocorticoids, which are a mainstay in the treatment of eosinophilic and inflammatory disorders, affect the transcriptional profile of eosinophils [2].

Our findings indicate that eosinophils recruited to dystrophin-deficient muscle tissue express higher levels of transcripts that encode proteins associated with tissue remodeling. These include enzymes that remodel components of the extracellular matrix such as matrix metalloproteinases

2, 12, 14, and 19; as well as cathepsin b, k, s. This is consistent with earlier reports indicating that eosinophils express matrix metalloproteinase and cathepsin family members within eosinophil granules [14, 34, 35]. However, our findings indicate significantly higher levels of these remodeling transcripts in eosinophils recruited to, and possibly activated by, the dystrophin- deficient muscle microenvironment. Furthermore, our findings identify expression of two key remodeling enzymes that have not been previously reported in eosinophil granules or associated with eosinophil disorders: matrix metalloproteinase 19 and cathepsin K. Highlighting their complexity in vivo, expression of cathepsin and matrix metalloproteinase family members have been shown both to limit and to promote muscle damage in muscular dystrophy mice [18, 21].

In addition, we detected increased expression of transcripts encoding collagen subunits in mdx eosinophils; these factors are critical components of the extracellular matrix. Increased collagen deposition also results in tissue fibrosis, which have been linked with specific instances of eosinophil infiltration. However, eosinophil-mediated tissue fibrosis has been described through the release of the pro-fibrotic factor Transforming Growth Factor-Beta (TGF-β) [36]; we are unaware of any earlier reports documenting the storage and secretion of collagen from eosinophil granules. Hence, we investigated the possibility that mdx eosinophils may be engulfing exogeneous nucleic acids, such as those provided by the myofibers. Through electron microscopic studies of eosinophils from mdx mice, there was no ultrastructural evidence for the bulk uptake of exogeneous nucleic acids (Elizabeth R. Fischer, RML/NIAID, personal communication).

72

Intriguingly, we detected increased expression of transcripts encoding cell surface proteins in

eosinophils from mdx mice. These include the inflammatory receptors CD14 and NLRP3, the

peptide/histidine transporter SLC15A3 and the ergothioneine transporter SLC22A4. Increased

expression of these cell surface proteins may potentiate the functional characteristics of

eosinophils in dystrophin-deficient muscle tissue. For example, increased expression of

transporters may promote the uptake of metabolites critical for prolonged cell survival, especially

in the presence of severe extracellular damage seen in dystrophin-deficient muscle tissue.

Similarly, expression of inflammatory receptors may potentiate eosinophil characteristics, for

example, by promoting transcription and/or release of key mediators from the granules. Given the

role of inflammatory receptors in mediating key eosinophil characteristics, we focus in chapter 5

on the function and regulation of a key receptor, TREM2, that is expressed at significantly higher

levels in mdx muscle eosinophils.

In summary, we extracted and sequenced eosinophil RNA from the muscle tissues of mdx,

IL5tg, and mdx.IL5tg mice. Eosinophils from the mdx muscle tissue exhibited distinct

transcriptomic features when compared with eosinophils from the IL5tg muscle tissue. In mdx muscle eosinophils, increased expression of transcripts associated with tissue remodeling as well as transcripts encoding cell surface proteins may potentiate specific eosinophil functional characteristics. As such, further studies will evaluate the impact of eosinophil infiltration and eosinophil-derived mediators on the regeneration of damaged, dystrophin-deficient muscle tissue.

73

FIGURES

Figure 1. Isolation of RNA from eosinophils from skeletal muscle tissues. (A) As described

in Chapter 2, hind limb skeletal muscles were enzymatically digested to generate single cell suspensions that were subjected to fluorescence-activated cell sorting (FACS). Isolated eosinophils (CD45+CD11c-Gr1-SigF+MHC-IIlo cells) were sorted directly into TRIZOL-LS and RNA

was isolated following the manufacturer’s protocol. To remove potential contaminants, isolated

RNA was subjected to cleanup using the Qiagen RNeasy Kit. Following library preparation, RNA

was sequenced using an Illumina HiSeq 4000 Next Generation sequencer with more than 100

Million reads per sample. Trimmed reads were aligned to the mouse reference genome and

transcript abundance quantified using the RSEM program. (B) A total of 9 eosinophil RNA

samples were submitted for sequencing: 3 from mdx, 3 from IL5tg, and 3 from mdx.IL5tg mice.

RNA samples ranged from 12 – 547 ng and were collected from 3-5 male mice at 4 weeks of age;

between 329,000 and 4,940,0000 eosinophils were used for RNA isolation.

74

Figure 2. Quality of RNA isolated from eosinophils from skeletal muscle tissues. The electrophoretic trace of each RNA was analyzed using an RNA PicoChip and Agilent BioAnalyzer

2100, which calculates the RNA Integrity Number (RIN) score based on the ratio of the 28s:18s ribosomal RNA subunits. RIN scores range from 0 (most degraded) to 10 (most intact). All RNA samples submitted for sequencing exhibited two distinct electrophoretic peaks and had RIN scores ≥ 8.20.

75

Figure 3. Transcripts and gene ontology pathways that are differentially expressed in mdx

vs IL5tg muscle eosinophils. (A) In black circle, transcripts that were detected at significantly

higher levels (> 5-fold) in eosinophils from mdx muscle tissue when compared with that of IL5tg.

Many of the mdx > IL5tg transcripts encode proteins involved with tissue remodeling (e.g., collagens, cathepsins, and matrix metalloproteinases) or cell surface proteins (e.g., CD74,

SLC15A3, TREM2). In blue circle, transcripts that were detected at significantly higher levels ( >

5-fold) in eosinophils from IL5tg muscle tissue when compared with that of mdx. Many of the IL5tg

> mdx transcripts encode eosinophil-specific proteins (e.g., eosinophil-associated ribonucleases, eosinophil peroxidase) or cell cycle processes. (B) Select gene ontology terms that were significantly associated with mdx > IL5tg transcripts. Gene ontology terms shown here were those associated with tissue remodeling processes.

76

Figure 4. Differentially expressed transcripts that are associated with tissue remodeling.

Expression of select transcripts that were detected at significantly higher levels in eosinophils from the mdx muscle tissue when compared with that of IL5tg (mdx > IL5tg, > 5-fold). Transcripts shown here encode structural components of the extracellular matrix or encode proteins involved with remodeling of the muscle tissue. Transcript abundance is expressed as fragments per kilobase of transcript per million reads (FPKM); values are represented as mean ± standard deviation. n = 3 per genotype.

77

Figure 5. Differentially expressed transcripts that encode cell surface proteins. Expression of select transcripts that were detected at significantly higher levels in eosinophils from the mdx muscle tissue when compared with that of IL5tg (mdx > IL5tg, > 5-fold). Transcripts shown here encode cell surface proteins. Transcript abundance is expressed as fragments per kilobase of transcript per million reads (FPKM); values are represented as mean ± standard deviation. n = 3 per genotype.

78

REFERENCES

1. Arnold, I.C., et al., Eosinophils suppress Th1 responses and restrict bacterially induced gastrointestinal inflammation. J Exp Med, 2018. 215(8): p. 2055-2072.

2. Gadkari, M., et al., Transcript- and protein-level analyses of the response of human eosinophils to glucocorticoids. Sci Data, 2018. 5: p. 180275.

3. Bouffi, C., et al., Transcription Factor Repertoire of Homeostatic Eosinophilopoiesis. J Immunol, 2015. 195(6): p. 2683-95.

4. Bouffi, C., et al., IL-33 markedly activates murine eosinophils by an NF-kappaB-dependent mechanism differentially dependent upon an IL-4-driven autoinflammatory loop. J Immunol, 2013. 191(8): p. 4317-25.

5. Grace, J.O., et al., Reuse of public, genome-wide, murine eosinophil expression data for hypotheses development. J Leukoc Biol, 2018. 104(1): p. 185-193.

6. Shen, Z.J., et al., RNA Seq profiling reveals a novel expression pattern of TGF-beta target genes in human blood eosinophils. Immunol Lett, 2015. 167(1): p. 1-10.

7. Percopo, C.M., et al., SiglecF+Gr1hi eosinophils are a distinct subpopulation within the lungs of allergen-challenged mice. J Leukoc Biol, 2017. 101(1): p. 321-328.

8. Mesnil, C., et al., Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest, 2016. 126(9): p. 3279-95.

9. Abdala-Valencia, H., et al., Shaping eosinophil identity in the tissue contexts of development, homeostasis, and disease. J Leukoc Biol, 2018. 104(1): p. 95-108.

10. Lee, J.J., et al., Eosinophils in health and disease: the LIAR hypothesis. Clin Exp Allergy, 2010. 40(4): p. 563-75.

11. Cai, B., et al., Eosinophilia of dystrophin-deficient muscle is promoted by perforin- mediated cytotoxicity by T cell effectors. Am J Pathol, 2000. 156(5): p. 1789-96.

12. Wehling-Henricks, M., et al., Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy. Hum Mol Genet, 2008. 17(15): p. 2280-92.

13. Sek, A.C., et al., Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy. J Immunol, 2019. 203(2): p. 476-484.

14. Rosenberg, H.F., K.D. Dyer, and P.S. Foster, Eosinophils: changing perspectives in health and disease. Nat Rev Immunol, 2013. 13(1): p. 9-22.

15. Wacht, G., et al., FACS - based isolation of human eosinophils allows purification of high quality RNA. J Immunol Methods, 2018. 463: p. 47-53.

16. Barnig, C., et al., Circulating Human Eosinophils Share a Similar Transcriptional Profile in Asthma and Other Hypereosinophilic Disorders. PLoS One, 2015. 10(11): p. e0141740.

79

17. Olson, O.C. and J.A. Joyce, Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat Rev Cancer, 2015. 15(12): p. 712-29.

18. Tjondrokoesoemo, A., et al., Cathepsin S Contributes to the Pathogenesis of Muscular Dystrophy in Mice. J Biol Chem, 2016. 291(19): p. 9920-8.

19. Fanjul-Fernandez, M., et al., Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochim Biophys Acta, 2010. 1803(1): p. 3-19.

20. Liu, S.L., et al., Matrix metalloproteinase-12 is an essential mediator of acute and chronic arterial stiffening. Sci Rep, 2015. 5: p. 17189.

21. Ogura, Y., et al., Therapeutic potential of matrix metalloproteinases in Duchenne muscular dystrophy. Front Cell Dev Biol, 2014. 2: p. 11.

22. Hantai, D., et al., Fibronectin, laminin, type I, III and IV collagens in Duchenne's muscular dystrophy, congenital muscular dystrophies and congenital myopathies: an immunocytochemical study. Connect Tissue Res, 1985. 13(4): p. 273-81.

23. Meerschaert, J., et al., CD14(+) cells are necessary for increased survival of eosinophils in response to lipopolysaccharide. Am J Respir Cell Mol Biol, 2000. 23(6): p. 780-7.

24. He, Z., et al., CD14 Is a Co-Receptor for TLR4 in the S100A9-Induced Pro-Inflammatory Response in Monocytes. PLoS One, 2016. 11(5): p. e0156377.

25. Arroyo-Espliguero, R., et al., CD14 and toll-like receptor 4: a link between infection and acute coronary events? Heart, 2004. 90(9): p. 983-8.

26. Rada, B., et al., NLRP3 inflammasome activation and interleukin-1beta release in macrophages require calcium but are independent of calcium-activated NADPH oxidases. Inflamm Res, 2014. 63(10): p. 821-30.

27. Song, F., et al., Regulation and biological role of the peptide/histidine transporter SLC15A3 in Toll-like receptor-mediated inflammatory responses in macrophage. Cell Death Dis, 2018. 9(7): p. 770.

28. Grundemann, D., et al., Discovery of the ergothioneine transporter. Proc Natl Acad Sci U S A, 2005. 102(14): p. 5256-61.

29. Ulland, T.K. and M. Colonna, TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol, 2018. 14(11): p. 667-675.

30. Giordano, C., et al., Toll-like receptor 4 ablation in mdx mice reveals innate immunity as a therapeutic target in Duchenne muscular dystrophy. Hum Mol Genet, 2015. 24(8): p. 2147- 62.

31. Lee, J.J., et al., Human versus mouse eosinophils: "that which we call an eosinophil, by any other name would stain as red". J Allergy Clin Immunol, 2012. 130(3): p. 572-84.

32. Stokes, K., et al., Cutting edge: STAT6 signaling in eosinophils is necessary for development of allergic airway inflammation. J Immunol, 2015. 194(6): p. 2477-81. 80

33. Fairfax, K.A., et al., Transcriptional profiling of eosinophil subsets in interleukin-5 transgenic mice. J Leukoc Biol, 2018. 104(1): p. 195-204.

34. Kuwabara, Y., et al., Role of Matrix Metalloproteinase-2 in Eosinophil-Mediated Airway Remodeling. Front Immunol, 2018. 9: p. 2163.

35. Schwingshackl, A., et al., Human eosinophils release matrix metalloproteinase-9 on stimulation with TNF-alpha. J Allergy Clin Immunol, 1999. 104(5): p. 983-9.

36. Aceves, S.S. and S.J. Ackerman, Relationships between eosinophilic inflammation, tissue remodeling, and fibrosis in eosinophilic esophagitis. Immunol Allergy Clin North Am, 2009. 29(1): p. 197-211, xiii-xiv.

81

CHAPTER 5

Role of TREM2 in Modulating The

Development and Characteristics of Eosinophils

82

CHAPTER 5 PREFACE

In our earlier work (Chapter 4), we described the use of RNA-sequencing to identify transcripts that were differentially expressed in eosinophils isolated from the muscle tissues of mdx, IL5tg and mdx.IL5tg mice. Among our key findings, we detected high levels of transcripts encoding tissue remodeling enzymes and cell surface receptors in eosinophils isolated from the muscle tissues of mdx mice; by contrast, eosinophils from the muscle tissues of IL5tg mice exhibited relatively low levels of these transcripts.

We were intrigued by increased expression of transcripts encoding cell surface proteins, as it suggests that eosinophils recruited to the dystrophin-deficient muscle tissue may be promoting mechanisms to enable them to sense and to adapt to this unique microenvironment. We focused our attention on the differential expression of these transcripts in eosinophils isolated from the muscle tissues of mdx, IL5tg and mdx.IL5tg mice. In general, we observed that eosinophils from mdx mice exhibit high levels of these transcripts while eosinophils from mdx.IL5tg and IL5tg mice exhibited intermediate and low expression of these transcripts, respectively. These trends in transcript expression indicate that eosinophils respond transcriptionally and can increase expression of specific pathways or classes of genes. Furthermore, it is likely that cues provided by the dystrophin-deficient and/or IL5-overabundant microenvironment mediate the distinct transcriptional profiles observed in these eosinophils.

One transcript in particular caught our attention – Trem2, which was detected at 68-fold higher levels in eosinophils from the muscle tissues of mdx mice when compared with eosinophils from the IL5tg mice. Trem2 encodes the Triggering Receptor Expressed on Myeloid cell-2 (TREM2) receptor that is expressed on myeloid cells such as microglia and osteoclasts [1-3]. Importantly,

TREM2 has been shown to mediate the activation, proliferation and survival of myeloid cells, particularly in response to tissue damage [3-5]. To the best of our knowledge, there are no published reports documenting expression of Trem2 in either mouse or human eosinophils at the transcript or at the protein level. Given the function of TREM2 in other myeloid populations, we

83 hypothesized that increased expression of Trem2 may be critical for the activation, proliferation and survival of eosinophils recruited to dystrophin-deficient (mdx) muscle tissue. In this chapter, we explore the expression and regulation of TREM2 on eosinophils, as well as the impact on key eosinophil characteristics, such as development and proliferation.

84

ABSTRACT

Eosinophils exhibit a wealth of cell surface proteins that modulate key responses to the tissue

microenvironment. Through RNA-sequencing, we identified relatively high expression of Trem2

in eosinophils from dystrophin-deficient (mdx) muscle tissue. Trem2 encodes the Triggering

Receptor Expressed on Myeloid Cell-2 (TREM2), which modulates the activation, proliferation

and survival of myeloid cells including osteoclasts and microglia. Expression and function of

Trem2 has not been reported previously in eosinophils. In this study, we investigated the

expression and function of TREM2 in eosinophils in health and in disease. Using an ex vivo

differentiation system, we detected expression of TREM2 on 4 – 21% of bone marrow-derived

cultured eosinophils. Expression reached a maximum at day 8, a timepoint that corresponded

with a 5-fold increase in the level of eosinophils in culture. Interestingly, Trem2-deficient cultures

failed to expand and instead exhibited increased cell death. Loss of Trem2 reduced the

homeostatic levels of eosinophils in the bone marrow but did not abrogate eosinophil recruitment

to the airways of allergen-challenged mice. In addition, we are generating a mouse model

(mdx.Trem2-/-) to evaluate the impact of Trem2-deficiency on eosinophil recruitment in a Th2- independent setting, such as that seen in muscular dystrophy. Expression of TREM2 was highly variable on peripheral blood eosinophils from healthy donors and was correlated with the body mass index (BMI) of the donor. Taken together, these findings demonstrate that TREM2 is expressed on human and murine eosinophils and that expression of this receptor mediates key characteristics such as proliferation and survival. Ongoing work will elucidate the regulation of

TREM2 expression on eosinophils as well as the impact of TREM2 receptor activation on eosinophil characteristics.

INTRODUCTION

Despite the perception of eosinophils as merely ‘end-stage effector cells’ [6], emerging evidence has highlighted remarkable heterogeneity in the characteristics and functions of

85

eosinophils [7-10]. In line with these studies, we identified distinct transcriptomic profiles among eosinophils recruited to the muscle tissues of mdx, IL5tg and mdx.IL5tg mice (Chapter 4). These include differential expression of transcripts encoding tissue remodeling enzymes and, intriguingly, differential expression of transcripts encoding cell surface proteins. We noted that eosinophils isolated from the muscle tissues of mdx mice exhibited 68-fold higher levels of the

Trem2 transcript when compared with eosinophils from IL5tg mice.

Trem2 encodes Triggering Receptor Expressed on Myeloid Cells-2 (TREM-2), a single

transmembrane receptor of the immunoglobulin family that signals through the DAP10 and

DAP12 adapters [5]. Variants in the Trem2 gene are associated with increased risk of developing

Alzheimer’s Disease; these variants result in decreased expression of TREM2 on the cell surface

as well as decreased signaling response [5]. Inactivating mutations in the Trem2 gene result in

Nasu-Hakola Disease, an autosomal recessive disorder distinguished by the presence of early-

onset dementia and recurrent bone fractures [5, 11]. As a result of these disease associations,

Trem2 has been studied extensively in myeloid cells involved with bone resorption (i.e.,

osteoclasts) and with the clearance of Aβ plaques (i.e., microglial cells).

Expression of Trem2 has been shown to promote the activation, proliferation and survival of

myeloid cells. For example, in the 5XFAD mouse model of Alzheimer’s Disease, Trem2-deficiency

was associated with reduced viability, recruitment and activation of microglia in response to Aβ

plaques [4, 12]. Furthermore, Trem2-/-5XFAD mice exhibited increased Aβ burden in the hippocampus, suggesting that Trem2 mediates beneficial processes that limit the pathology in

Alzheimer’s Disease [12]. Accordingly, monoclonal antibody-based therapeutics that seek to augment the activity of TREM2 in microglial cells are currently being explored for the treatment of

Alzheimer’s Disease; examples include the compound AL002 developed jointly by AbbVie, Inc. and Alector, Inc. (www.abbvie.com/our-science/pipeline/al002.html) as well as the compound

ATV:TREM2 developed jointly by Denali Therapeutics, Inc. and Takeda Pharmaceutical

Company, Ltd. (https://denalitherapeutics.com/pipeline).

86

TREM2 can be activated in vitro by a range of phospholipids such as phosphatidic acid (PA),

phosphatidylinositol (PI) and phosphatidylserine (PS) [12]. However, the relevance of these

ligands in vivo remains to be determined; detection of phosphatidylserine displayed on apoptotic

cells may activate the TREM2 receptor in vivo [5, 12]. Nonetheless, activation of TREM2 results in enhanced cellular energetics and activation of key pathways such as mechanistic target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) [4, 5, 13]. Interestingly, loss of

Trem2 in macrophages results in enhanced cytokine release following treatment with diverse stimuli such as bacterial lipopolysaccharides, zymosan and CpG oligodeoxynucleotides [14].

Together, these findings indicate critical roles for TREM2 in mediating intracellular characteristics and in modulating the release of inflammatory cytokines.

The pleiotropic roles of Trem2 suggest that its expression in eosinophils may mediate critical functions. To our knowledge, there are no prior reports of Trem2 expression in eosinophils or, more broadly, in granulocytes. Similar to the role described in other myeloid populations, Trem2 may mediate the activation, proliferation and survival of eosinophils, particularly in response to tissue damage such as that seen in muscular dystrophy. In this study, we evaluated the expression patterns of TREM2 in eosinophils as well as the impact of this receptor on eosinophil- mediated processes in health and disease. Central to our exploration of Trem2 function in eosinophils is the use of the Trem2-/- mouse model, which contains a 175 base pair deletion in

the Trem2 gene that results in a premature stop codon (www.jax.org/strain/027197).

MATERIALS AND METHODS

Mice

All mice were bred and maintained under pathogen-free conditions in the National Institute of

Allergy and Infectious Diseases (NIAID) Animal Facility 14b South. mdx (C57BL/10ScSn-

Dmdmdx/J, 001801) and Trem2-/- mice (C57BL/6J-TREM2em2Adiuj, 027197) were obtained from

Jackson Laboratory and wild-type controls (C57BL/6NCr, 556) were obtained from Charles River 87

Laboratory. To generate muscular dystrophy mice that lack Trem2 (mdx.Trem2-/-), parental mdx and Trem2-/- strains were intercrossed to generate F1 hybrids that were backcrossed onto the

Trem2-/- parental strain. The presence of the mdx and Trem2 mutations were confirmed by

polymerase chain reaction (PCR) as recommended by Jackson Laboratory.

Ex vivo culturing of Bone Marrow-Derived Eosinophils

As described in [15], bone marrow cells were collected by the femurs and tibiae of adult

mice with RPMI-1640 medium (Thermo Fisher). Bone marrow cells were subjected to hypotonic

lysis to remove erythrocytes before culturing in media at 1 x 106 cells/mL. Cells were cultured in

RPMI-1640 medium containing 20% Fetal Bovine Serum (Atlanta Biologicals), 25 mM HEPES

(Life Technologies), 100 IU/mL penicillin and 10 µg/mL streptomycin (Cellgro), 2 mM glutamine

(Life Technologies), 1 X nonessential amino acids, 1 mM sodium pyruvate (Life Technologies),

50 µM β-mercaptoethanol (Sigma-Aldrich).

From day 0 to 4, growth media was supplemented with 100 ng/mL recombinant mouse Stem Cell

Factor (rm-CSF) and recombinant mouse FMS-like tyrosine kinase 3 ligand (rm-FLT3L). Starting on day 4, growth media was supplemented with 10 ng/mL of recombinant mouse Interleukin-5

(rm-IL5) only. Every 2 days, half of the growth medium was replaced with fresh growth medium containing supplemented factors (rm-CSF and rm-FLT3L; or rm-IL5) and cells were maintained at 1e6/mL. Cell Number and viability were assessed using the Luna-FL dual fluorescence cell counter (Logos Bio).

Evaluation of Bone Marrow-Derived Eosinophils

The morphological features of cultured cells were evaluated using Wright-Giemsa staining. As described in [15], approximately 50,000 cells were centrifuged onto slides using the Shandon

Cytospin 4 (Thermo Fisher). Cells were fixed with methanol before staining with modified Giemsa

88

Dyes (Diff-Quik). Images were acquired using the Leica DMI 4000B microscope equipped with

Image Pro Plus software (Media Cybernetics).

TREM2 expression and eosinophil maturity were evaluated by flow cytometric analysis. Aliquots

of cells (1 x 106) were collected every two days of culture and frozen at -80°C in 90% fetal bovine

serum, 10% dimethylsulfoxide. Samples were stained with an Fc receptor blocking antibody (anti-

CD16/32, BD Biosciences), a viability dye (Live/Dead Aqua, ThermoFisher) and a fluorochrome-

conjugated antibody against TREM2 (R&D Systems, Cat No. FAB17291A). Samples were

evaluated using the LSR-II Flow Cytometer (BD Biosciences), with > 150,000 events were

recorded per sample; flow cytometry data was analyzed using FlowJo (Tree Star). Mature

eosinophils were identified as live SSC-Ahi SigF+ cells and were expressed as a percent of the

total live cells in each sample. TREM2+ populations were identified using fluorescent minus one

(FMO) and isotype controls. TREM2+ populations were expressed as a percent of the total number of eosinophils (live SSC-Ahi SigF+ cells) in each sample.

Evaluation of Eosinophils in Bone Marrow

Bone marrow cells were collected by flushing the tibia and femur of mice with RPMI-1640 media.

Following hypotonic lysis, bone marrow cells were enumerated and frozen at -80°C at a concentration of 1-10 x 106 cells/mL of freezing media (90% fetal bovine serum, 10%

dimethylsulfoxide). As described in Chapter 2, samples were stained with a viability dye

(Live/Dead Aqua, ThermoFisher), an Fc receptor blocking antibody (anti-CD16/32, BD

Biosciences) and fluorochrome-conjugated antibodies including anti-CD45 and anti-CD11c

(eBioscience); anti-Gr1 and anti-Siglec-F (BD Biosciences). Samples were evaluated using the

LSR-II Flow Cytometer (BD Biosciences), with > 400,000 events were recorded per sample; flow

cytometry data was analyzed using FlowJo (Tree Star). Eosinophils were identified as

liveCD45+CD11c-Gr1-SigF+MHC-IIlo cells and expressed as a percent of the total live cells in each sample.

89

Alternaria alternata challenge and evaluation of disease parameters

Wild-type and Trem2-/- mice were inoculated intranasally with a filtrate of Alternaria alternata

(Greer Allergy Immunotherapy; 50 µg/mL, 50 µL per mouse) at day 0, 3, 6, 10. At day 14, mice

were euthanized and the bronchoalveolar lavage fluid (BALf) was collected using 0.8-1.2 mL of

PBS containing 0.1% bovine serum albumin. Cells recovered from the BALf of each mouse were enumerated using the Luna-FL dual fluorescence cell counter (Logos Bio). Approximately 50,000 cells from the BALf fluid were used to evaluate the fraction of eosinophils. Briefly, cells were centrifuged onto slides (Shandon Cytospin 4, ThermoFisher), fixed in methanol and stained with modified Wright-Giemsa dyes (Diff-Quik). Eosinophils were identified based on cytological features including eosin staining of the granules and ring-shaped nuclei. Eosinophils were enumerated relative to other cell types and expressed as a percent of the total cells detected in the BALf fluid for each sample. Total number of eosinophils in the BALf was calculated using the total number of cells recovered in the BALf and the proportion of eosinophils detected among

BALf cells.

Blood Donors

Samples of whole, EDTA-anti-coagulated blood were obtained from the National Institutes of

Health Clinical Center (Protocol Identifier NCT00001846). All donors consented to donation of blood sample and met the eligibility criteria: ≥ 18 years of age, ≥ 110 pounds, no known history of heart, lung, kidney, hematological disorders, and were not pregnant at time of donation. Donor samples were de-identified by the National Institutes of Health Clinical Center and all samples used in this study were negative for cytomegalovirus.

90

Isolation of Human Blood Eosinophils and Detection of TREM2

Eosinophils were isolated from human blood samples via negative selection using the Miltenyi

Human Eosinophil Isolation Kit (Catalog Number 130-104-466) as described previously [7].

Following the manufacturer’s protocol, each blood sample was combined with a cocktail of

particle-bound antibodies that target non-eosinophil populations. Subsequently, each sample was subjected to a magnetic field using the MACSxpress Separator (Catalog Number 130-098-308), which impeded the passage of antibody-bound leukocytes. Isolated eosinophils were stained with modified Wright-Giemsa Dyes (Diff-Quik, Thermo Fisher Scientific) and analyzed for eosin staining and nuclear morphology; staining confirmed that eosinophils isolated using this protocol were ≥ 98% pure.

Isolated eosinophils were stained with an Fc receptor blocking antibody (anti-CD16/32, BD

Biosciences), a viability dye (Live/Dead Aqua, ThermoFisher) and a fluorochrome-conjugated

antibody against TREM2 (R&D Systems, Cat No. FAB17291A) prior to analysis via flow

cytometry. Samples were evaluated using the LSR-II Flow Cytometer (BD Biosciences), with >

60,000 events were recorded per sample; flow cytometry data was analyzed using FlowJo (Tree

Star). TREM2+ populations were identified by comparing TREM2-staining with isotype and

fluorescent minus one (FMO) controls; TREM2+ eosinophils were expressed as a percent of the

total number of eosinophils (live SSC-Ahi SigF+ cells) in each sample.

RESULTS

TREM2 Expression Patterns on Bone Marrow-Derived Eosinophils

TREM2 has been detected on myeloid cells including microglia and osteoclasts, where it has

been shown to mediate cell survival through the activation of the Wnt/β-Catenin pathway [1, 3,

16]. As the expression and function of TREM2 on eosinophils has not been reported, we began

with an ex vivo system to investigate the expression of TREM2 during the differentiation and

proliferation of eosinophils. In this protocol [15], unselected bone marrow cells are cultured for 4

91

days in media containing Stem Cell Factor (SCF) and FMS-like Tyrosine Kinase-3 Ligand (Flt3l)

before differentiation in media containing Interleukin-5 (IL-5). As a result, cultures exhibit an

expansion in the number of mature eosinophils, from approximately 8% of the cultured cells at

day 0 to more than 94% of the cultured cells at day 16 (Fig 1A). Eosinophils differentiated ex vivo

exhibit characteristic morphologic features, such as eosin staining of the granules and ring-form

nuclei (Fig 1B).

Because TREM2 is an inducible receptor, we examined its expression patterns in cultures of

bone marrow-derived eosinophils sampled every 2 days; this allowed us to evaluate TREM2

during eosinophil development and maturation. Immunoreactive TREM2 was detected on the

surface of a small population (~5%) of eosinophils at the earliest time points during differentiation

in response to IL-5 (days 4-6 of culture). However, a prominent population of eosinophils (20 ±

1.4%, red arrow) expressed immunoreactive TREM2 at day 8; expression then decreased, returning to baseline levels (~5% of the total eosinophils) by day 12 (Fig 1C). Interestingly, the peak expression of TREM2 at day 8 coincided with the 5-fold expansion in the number of eosinophils: at day 6, eosinophils represent ~5% of the cultured cells while at day 8, eosinophils represent ~25% of the cultured cells (Fig 1A, red arrow). Increased expression of TREM2 during eosinophil expansion suggests that this receptor may be associated with eosinophil expansion and proliferation, similar to the role described in osteoclasts and microglial cells [1, 3, 4, 11, 12,

16].

Trem2-Deficiency Limits Expansion of Eosinophils ex vivo and in vivo

Given that TREM2 can be detected on eosinophils cultured ex vivo, we evaluated the impact of Trem2 on the differentiation, proliferation, and survival of eosinophils. We utilized the ex vivo system described in Fig 1 to examine eosinophil expansion and proliferation in Trem2-/- mice.

While similar numbers of cells were observed between day 0 and day 10, cultures from Trem2-/-

bone marrow did not exhibit similar robust expansion observed in cultures from wild-type bone

92

marrow (Fig 2A). Furthermore, between day 2 and day 17, cultures of Trem2-/- bone marrow- derived eosinophils exhibited slightly lower viability when compared with those derived from the wild-type (Fig 2B). Nonetheless, when evaluated at day 14, cultures of Trem2-/- cells exhibited

features characteristic of eosinophils, including eosin staining of the granules and ring-form nuclei,

but also exhibited increased evidence of cell-death (Fig 2C, arrows).

Next, we asked if Trem2 might modulate the levels of eosinophils at homeostasis. Towards this end, we evaluated the level of eosinophils in the bone marrow of wild-type and Trem2-/- mice.

While eosinophils constitute approximately 6% of the bone marrow cells in wild-type mice, we detected significantly lower levels of eosinophils in the bone marrow of Trem2-/- mice (Fig 3A; 3.7

± 0.25%, ** p < 0.01). Furthermore, eosinophils in the bone marrow of Trem2-/- expressed

significantly lower median fluorescent intensity (MFI) for Siglec-F when compared with those of

wild-type (Fig 3B; 727 ± 14 vs 857 ± 16, respectively; ****p < 0.0001). Taken together, these findings indicate that Trem2 is not required for the differentiation of eosinophils, but suggest that it may contribute to the potential for proliferation and their ongoing survival.

Trem2 Is Not Required for Eosinophil Recruitment to the Airways of Allergen-challenged mice

As Trem2 is critical for expansion of eosinophils ex vivo and in vivo, we asked if Trem2 might

contribute to the efficient expansion and mobilization of eosinophils associated with

pathophysiological states. Accordingly, we utilized a mouse model of airway inflammation, in

which eosinophils are recruited to the airways following challenge with the fungal allergens such

as Alternaria alternata. We challenged both wild-type and Trem2-/- mice with four doses of

Alternaria alternata (day 0, 3, 6, 10) and then evaluated eosinophil infiltration at day 14 (Fig 4A).

Surprisingly, we detected similar levels of total cells (Fig 4B) and eosinophils (Fig 4C) recruited

to the airways of wild-type and Trem2-/- mice. In addition, eosinophils recruited to the airways of

Trem2-/- mice appeared morphologically indistinguishable from those recruited to the airways of

93

wild-type mice (Fig 4D); we did not observe any evidence for increased cell death associated with

eosinophils recruited to the airways of Trem2-/- mice. Taken together, these findings indicate that

Trem2 gene-deletion does not result in impaired recruitment of eosinophils to the airways of

allergen-challenged mice. Rather, signals associated with Alternaria alternata challenge may be

sufficient to overcome the decreased eosinophil proliferation associated with Trem2-deficiency.

Generation of Trem2-deficient muscular dystrophy mice (mdx.Trem2-/-)

We also initiated a study to evaluate the impact of Trem2 on the persistence and characteristics of eosinophils detected in muscle infiltrates as a feature of muscular dystrophy. As described in Chapter 4, we detected a striking, 68-fold increase in the expression of Trem2 transcripts in eosinophils isolated from the muscle tissues of mdx mice when compared with eosinophils from the muscle tissues of IL5tg mice. Increased expression of Trem2 in these eosinophils may promote an activated state in which eosinophils exhibit increased persistence and also exhibit unique functional characteristics. Hence, in order to evaluate the impact of Trem2

on eosinophil persistence and characteristics in muscular dystrophy, we generated muscular

dystrophy mice that lack Trem2 (mdx.Trem2-/-) by intercrossing the mdx (C57BL/10ScSn-

Dmdmdx/J, 001801) and Trem2-/- strains (C57BL/6J-TREM2em2Adiuj, 027197).

As shown in Figure 5, F1 offspring generated during the first cross include female mice that are heterozygous for both the mdx and the Trem2 mutations. Subsequently, F1 hybrid females

(mdx+/-Trem2+/-) were bred with Trem2-/- males to generate male offspring that are hemizygous for the mdx mutation and homozygous for the Trem2 mutation (mdxx/-Trem2-/-); these mice

represent our desired genotype and are expected to occur at the Mendelian frequency of 1:4 male

mice. Subsequent mating between F2 mice will most likely augment the expected frequency of

the desired genotype to 1:2 in the F3 male offspring, while mating of the F3 mice can produce

offspring that exclusively express both alleles of the mdx and Trem2 mutations (mdx-/-Trem2-/-).

94

While generation of these mice is ongoing, we have successfully obtained F2 offspring in

January 2020; here, the desired genotype (mdxx/-Trem2-/-) is to be expected in 1:4 males.

Previously, we have utilized polymerase chain reaction (PCR) to confirm the genotypes of the mdx and Trem2-/- parental strains; we expect that confirmation of the mdx and Trem2 mutations should proceed seamlessly in F2 mice. Using PCR-confirmed mdx.Trem2-/- mice, we will evaluate

the degree of eosinophil infiltration, the functional characteristics of eosinophils in the muscle

tissue and the impact on muscle pathology. We note here that while loss of Trem2 did not

abrogate eosinophil recruitment to the airways of allergen-challenged mice, dystrophin-deficient

muscle tissue represents a distinct context in which eosinophils are recruited and activated.

Eosinophil infiltration in muscular dystrophy occurs in the absence of a systemic, Th2-mediated

response [17] in contrast to the Th2-driven, allergen-challenged model we utilized (Fig 4; [18,

19]); hence, investigation of the potential impact of Trem2 on eosinophils recruited in muscular

dystrophy may yield intriguing insights into the effects of Trem2 activity in non Th2-mediated

settings.

Variability of surface TREM2 expression on Human Peripheral Blood Eosinophils

Given the association between variants in the Trem2 gene and disorders such as Alzheimer’s

Disease [20-22] and Nasu-Hakola disease (NHD) [23, 24], we asked if TREM2 might also be

detected on human eosinophils. Towards this end, we isolated eosinophils from the peripheral

blood of healthy donors using negative selection, in which antibodies bound and selectively

removed non-eosinophil populations. Eosinophils were isolated at high purity (>98%) and

analyzed for the expression of surface TREM2 by flow cytometry. We detected TREM2

expression on the surface of 0.7 – 63% of peripheral blood eosinophils from normal human

subjects; the MFI varied from 343 – 538 (Fig 6A). In Figure 6B, representative flow cytometry

plots are shown from donors with low (donor 15) or high (donor 5) proportions of TREM2+

eosinophils.

95

Next, we asked if the variability in TREM2 expression on peripheral blood eosinophils might

correlate with any of the clinical parameters provided on de-identified reports. Intriguingly, we

observed a significant correlation between the proportion of TREM2 expressing eosinophils and

the body mass index (BMI) of the donor (R2 = 0.56, p = 0.01; Fig 6C). Taken together, these findings provide evidence of TREM2 expression on human eosinophils and indicate that

expression may be affected by unique pathophysiological conditions such as adiposity or

variations in diet.

DISCUSSION

In this study, we evaluated the expression of TREM2 on human and murine eosinophils, and

identified a key role for TREM2 in the expansion of the eosinophil lineage ex vivo. We extended

these findings in vivo and determined that Trem2-deficient mice exhibited a reduced number of

eosinophils in bone marrow at baseline. Despite lower levels of eosinophils in the bone marrow

at baseline, Trem2-/- mice mobilized eosinophils to the airways following challenge with a Th2 stimulus; these results suggest that Trem2-mediated signaling may have an impact on eosinophils via a mechanism that is independent of Th2 signaling. We intend to investigate Trem2-mediated

effects on eosinophil recruitment in a non Th2-mediated signaling such as muscular dystrophy

[17]; as such, we are actively generating muscular dystrophy mice that lack Trem2 (mdx.Trem2-/-

). Finally, we described variable expression of TREM2 on peripheral blood eosinophils that correlated with increased body mass index of the donor.

These findings, while intriguing, represent an initial exploration towards elucidating the function and regulation of Trem2 in eosinophils. As such, these data invite many new areas of

investigation that are discussed below.

Expression and function of TREM2 has been identified on multiple cell types, nearly all of

which belong to the monocyte/macrophage lineage: osteoclasts [1, 11], microglia [3, 4, 12, 25],

Kupffer cells [26], alveolar macrophages [27, 28], adipose macrophages [29] and dendritic cells

96

[30]. Hence, to the best of our knowledge, this is the first report of TREM2 expression on

eosinophils and, more broadly, on cells of the granulocyte lineage. Earlier work demonstrated that

neutrophilic infiltrates express high levels of TREM1 – a receptor thought to oppose the activity

of TREM2 – in response to pathogens such as bacteria and fungi [25, 31]. In addition, several groups reported an inverse relationship between Trem1 and Trem2 expression in macrophages and microglia, notably in response to bacterial lipopolysaccharides or interleukin-4 (IL-4) [25, 32].

These seminal studies, while important, have led to the perception that cells of the monocyte/macrophage lineage largely express TREM2 but can also express TREM1 under specific inflammatory circumstances; and that granulocytes, including eosinophils, largely express TREM1 but not TREM2 [2].

Our findings identify TREM2 expression on eosinophils differentiated ex vivo and demonstrate that Trem2 is required for the efficient development of eosinophils under ex vivo and in vivo conditions. These findings are consistent with earlier reports, in which Trem2 has been shown to promote the survival of microglia and osteoclasts through activation of the β-catenin/Wnt pathway

[1, 3, 16] and increased intracellular metabolism [4, 33]. Ongoing work will evaluate these features in wild-type and Trem2-/- eosinophils as potential mechanisms by which Trem2 exerts pro-survival and proliferative effects.

We detected peak TREM2 expression on eosinophils at day 8 of ex vivo culturing, a timepoint that corresponded with the 5-fold expansion in the number of eosinophils in culture. Increased expression of TREM2 at this timepoint highlights the potential role of TREM2 in mediating eosinophil proliferation. However, we note that eosinophils continue to proliferate following day 8 and that Trem2-deficiency abrogates the dramatic eosinophil expansion observed at later timepoints, notably at day 13. One possibility is that Trem2 might be acting very early to promote the survival and expansion of eosinophils, the effects of which are magnified after several cycles of cell proliferation. Another possibility is that while peak TREM2 is detected at day 8, its effects may be mediated at subsequent timepoints through the actions of cleaved or soluble Trem2

97

(sTREM2) [34]. Indeed, multiple reports have demonstrated that TREM2 is cleaved by proteases such as ADAM10/17 [35-38] and that sTREM2 promotes the survival of cells such as microglia and alveolar macrophages [27, 39, 40]. Ongoing work will evaluate the extent to which sTREM2 can be detected in the supernatants of eosinophil cultures and the effect that sTREM2 depletion

– for example, through specific protease inhibitors – might have on eosinophil proliferation.

Finally, our findings raise intriguing questions about the regulation and function of TREM2 in disease. As described in Chapter 4, we noted a 68-fold increase in the level of Trem2 transcripts detected in eosinophils from the muscle tissues of mdx mice when compared with eosinophils from the muscle tissues of IL5tg mice. Increased expression of Trem2 in eosinophils may be attributed to activation by factors found in dystrophin-deficient muscle microenvironment such as

IL-4, IL-1RA and Eotaxin-1/CCL11. In support of this, Turnball and colleagues [14] demonstrated that administration of IL-4 or thioglycollate augments the level of TREM2 expressed on peritoneal macrophages. As such, we have initiated aspects of this study to identify factors that promote

TREM2 expression on eosinophils; similar to the findings by Turnball and colleagues [14], we observed dramatic upregulation of TREM2 expression on bone marrow-derived eosinophils stimulated with IL-4 (data not shown).

Along these lines, we observed increased expression of TREM2 on peripheral blood eosinophils that was correlated with the body mass index (BMI) of the donor. Since adiposity is associated with a chronic inflammatory state [41, 42], circulating factors in donors with higher

BMIs may promote the expression of TREM2 on eosinophils. Interestingly, Jaitin and colleagues

[29] reported increased expression of TREM2 on macrophages recruited to the adipose tissue of mice fed a high-fat diet. Although Trem2 was not required for macrophage infiltration into the adipose tissue, Trem2 mediated a key transcriptional program in recruited macrophages. Hence, further work is needed to investigate the functional differences between TREM2+ and TREM2- eosinophils and their impact in metabolic syndromes such as adiposity.

98

In summary, we evaluated TREM2 expression patterns on human and mouse eosinophils and identified contexts in which TREM2 expression is increased. Among these are eosinophils undergoing expansion, eosinophils recruited to dystrophin-deficient muscle tissue and eosinophils from donors with increased body mass index. Ongoing work will evaluate the role of specific factors in promoting TREM2 expression as well as the intracellular pathways by which

TREM2 regulate eosinophil expansion.

99

FIGURES

Figure 1. Differentiation of eosinophils ex vivo and detection of TREM2. (A) Eosinophils can undergo ex vivo expansion and differentiation from unselected mouse bone marrow cells using our published protocol [15]. Following expansion of eosinophil progenitors by Stem Cell Factor

(SCF) and FMS-like tyrosine kinase-3 ligand (Flt3l), IL-5 results in the generation of mature eosinophils (SSChiSigF+) which have expanded to ~25% of the cells in culture by day 8 as indicated by the red arrow. Mature eosinophils constitute > 80% of the cultured cells by day 12, and > 94% by day 16. (B) At day 14, cultured cells exhibited features characteristic of eosinophils, including eosin staining of the granules and ring-form nuclei. (C) Immunoreactive TREM2 was

detected on 4 - 7% of bone marrow-derived eosinophils (SSChiSigF+) on days 4 - 6 and days 12

- 16 of culture. Immunoreactive TREM2 was detected on 18 - 21% of bone marrow-derived

eosinophils at day 8 of culture as indicated by the red arrow. (A, C) Mean ± Standard Deviation

of three separate cultures.

100

Figure 2. Trem2 is required for efficient eosinophil expansion ex vivo. (A) Total live cells and

(B) viability in ex vivo bone marrow cultures from wild-type (black bars) and Trem2-/- mice (blue bars). Cultured cells were differentiated in stem cell factor (SCF), Fms-like tyrosine kinase-3

Ligand (Flt3l) and Interleukin-5 (IL-5) as indicated. Mean ± Standard Deviation of three separate cultures; ** p < 0.01, * p < 0.05, 2-way ANOVA with multiple comparisons. (C) At day 14, cultured cells from Trem2-/- mice were morphologically eosinophils, and displayed eosin staining within cytoplasmic granules and ring-form nuclei. Cells undergoing apoptosis are indicated by the black

arrows.

101

Figure 3. Trem2 gene-deletion alters eosinophil homeostasis in the bone marrow. (A)

Levels of mature eosinophils (CD45+CD11c-Gr1-SigF+MHC-IIlo cells) in the bone marrow of wild- type and Trem2-/- mice. (B) Median Fluorescent Intensity (MFI) of Siglec-F expression on eosinophils in the bone marrow of wild-type and Trem2-/- mice; data shown are mean ± standard

deviation, n= 4 per genotype; **** p < 0.0001, ** p < 0.01, unpaired t-test.

102

Figure 4. Trem2 is not critical for eosinophil recruitment in response to a Th2-inflammatory stimulus. (A) Wild-type and Trem2-/- mice were challenged with four inoculations of a filtrate of

Alternaria alternata on days 0, 3, 6, 10 (50 µg / mL, 50 µL per mouse) and evaluated on day 14 as previously described [19]. Similar number of (B) cells and (C) eosinophils were recovered from the bronchoalveolar lavage fluid of wild-type and Trem2-/- mice at day 14 post challenge. (D) At day 14 post challenge, eosinophils recovered from the bronchoalveolar lavage fluid of wild-type

and Trem2-/- mice were morphologically indistinguishable; n = 6 per genotype; n.s., no significant

differences.

103

Figure 5. Generation of mdx.Trem2-/- mice. Male Trem2-/- mice (C57BL/6J-TREM2em2Adiuj,

027197) were crossed with female mdx mice (C57BL/10ScSn-Dmdmdx/J, 001801) to generate

female mice heterozygous for mutations in both the mdx and Trem2 genes. Mdx+/-Trem2+/- female

mice were crossed with male Trem2-/- mice to produce offspring in which 25% of the males are

hemizygous for the mdx mutation and homozygous for the Trem2 mutation. Skeletal muscle

tissues from male mdx.Trem2-/- mice will be evaluated for the degree of eosinophil infiltration and the level of muscle damage.

104

Figure 6. Variable expression of TREM2 on human peripheral blood eosinophils and correlation with donor BMI. (A) Peripheral blood eosinophils from healthy donors exhibit variable expression of TREM2 (0.7 – 63.7%) and variable median fluorescence intensity (MFI;

343 – 538). Significant correlation (p = 0.05, R2 = 0.39) between the MFI of TREM2 and percent of eosinophils expressing TREM2. (B) Flow cytometric analysis of TREM2 expression on peripheral blood eosinophils from a donor with low TREM2 expression (0.7%, Donor #5) compared to a donor with high TREM2 expression (63%, Donor #15). SSC-A, side scatter area.

(C) Significant correlation (p = 0.012, R2 = 0.57) between percent of TREM2+ eosinophils and the body mass index of donor.

105

REFERENCES

1. Otero, K., et al., TREM2 and beta-catenin regulate bone homeostasis by controlling the rate of osteoclastogenesis. J Immunol, 2012. 188(6): p. 2612-21.

2. Colonna, M., TREMs in the immune system and beyond. Nat Rev Immunol, 2003. 3(6): p. 445-53.

3. Zheng, H., et al., TREM2 Promotes Microglial Survival by Activating Wnt/beta-Catenin Pathway. J Neurosci, 2017. 37(7): p. 1772-1784.

4. Ulland, T.K., et al., TREM2 Maintains Microglial Metabolic Fitness in Alzheimer's Disease. Cell, 2017. 170(4): p. 649-663 e13.

5. Ulland, T.K. and M. Colonna, TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol, 2018. 14(11): p. 667-675.

6. Lee, J.J., et al., Eosinophils in health and disease: the LIAR hypothesis. Clin Exp Allergy, 2010. 40(4): p. 563-75.

7. Ma, M., et al., Cytokine Diversity in Human Peripheral Blood Eosinophils: Profound Variability of IL-16. J Immunol, 2019. 203(2): p. 520-531.

8. Percopo, C.M., et al., SiglecF+Gr1hi eosinophils are a distinct subpopulation within the lungs of allergen-challenged mice. J Leukoc Biol, 2017. 101(1): p. 321-328.

9. Abdala Valencia, H., et al., Phenotypic plasticity and targeting of Siglec-F(high) CD11c(low) eosinophils to the airway in a murine model of asthma. Allergy, 2016. 71(2): p. 267-71.

10. Abdala-Valencia, H., et al., Shaping eosinophil identity in the tissue contexts of development, homeostasis, and disease. J Leukoc Biol, 2018. 104(1): p. 95-108.

11. Paloneva, J., et al., DAP12/TREM2 deficiency results in impaired osteoclast differentiation and osteoporotic features. J Exp Med, 2003. 198(4): p. 669-75.

12. Wang, Y., et al., TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model. Cell, 2015. 160(6): p. 1061-71.

13. Peng, Q., et al., TREM2- and DAP12-dependent activation of PI3K requires DAP10 and is inhibited by SHIP1. Sci Signal, 2010. 3(122): p. ra38.

14. Turnbull, I.R., et al., Cutting edge: TREM-2 attenuates macrophage activation. J Immunol, 2006. 177(6): p. 3520-4.

15. Dyer, K.D., et al., Functionally competent eosinophils differentiated ex vivo in high purity from normal mouse bone marrow. J Immunol, 2008. 181(6): p. 4004-9.

16. Otero, K., et al., Macrophage colony-stimulating factor induces the proliferation and survival of macrophages via a pathway involving DAP12 and beta-catenin. Nat Immunol, 2009. 10(7): p. 734-43.

106

17. Sek, A.C., et al., Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy. J Immunol, 2019. 203(2): p. 476-484.

18. Ma, M., et al., Alternaria alternata challenge at the nasal mucosa results in eosinophilic inflammation and increased susceptibility to influenza virus infection. Clin Exp Allergy, 2018. 48(6): p. 691-702.

19. Geslewitz, W.E., C.M. Percopo, and H.F. Rosenberg, Eosinophil persistence in vivo and sustained viability ex vivo in response to respiratory challenge with fungal allergens. Clin Exp Allergy, 2018. 48(1): p. 29-38.

20. Guerreiro, R., et al., TREM2 variants in Alzheimer's disease. N Engl J Med, 2013. 368(2): p. 117-27.

21. Jonsson, T., et al., Variant of TREM2 associated with the risk of Alzheimer's disease. N Engl J Med, 2013. 368(2): p. 107-16.

22. Sims, R., et al., Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial- mediated innate immunity in Alzheimer's disease. Nat Genet, 2017. 49(9): p. 1373-1384.

23. Paloneva, J., et al., Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet, 2002. 71(3): p. 656-62.

24. Klunemann, H.H., et al., The genetic causes of basal ganglia calcification, dementia, and bone cysts: DAP12 and TREM2. Neurology, 2005. 64(9): p. 1502-7.

25. Owens, R., et al., Divergent Neuroinflammatory Regulation of Microglial TREM Expression and Involvement of NF-kappaB. Front Cell Neurosci, 2017. 11: p. 56.

26. Goncalves, L.A., et al., TREM2 governs Kupffer cell activation and explains belr1 genetic resistance to malaria liver stage infection. Proc Natl Acad Sci U S A, 2013. 110(48): p. 19531-6.

27. Wu, K., et al., TREM-2 promotes macrophage survival and lung disease after respiratory viral infection. J Exp Med, 2015. 212(5): p. 681-97.

28. Koth, L.L., et al., DAP12 is required for macrophage recruitment to the lung in response to cigarette smoke and toward CCL2. J Immunol, 2010. 184(11): p. 6522-8.

29. Jaitin, D.A., et al., Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell, 2019. 178(3): p. 686-698 e14.

30. Bouchon, A., et al., A DAP12-mediated pathway regulates expression of CC chemokine receptor 7 and maturation of human dendritic cells. J Exp Med, 2001. 194(8): p. 1111- 22.

31. Bouchon, A., J. Dietrich, and M. Colonna, Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol, 2000. 164(10): p. 4991-5.

107

32. Liu, Q., et al., Peripheral TREM1 responses to brain and intestinal immunogens amplify stroke severity. Nat Immunol, 2019. 20(8): p. 1023-1034.

33. Nugent, A.A., et al., TREM2 Regulates Microglial Cholesterol Metabolism upon Chronic Phagocytic Challenge. Neuron, 2019.

34. Zhong, L. and X.F. Chen, The Emerging Roles and Therapeutic Potential of Soluble TREM2 in Alzheimer's Disease. Front Aging Neurosci, 2019. 11: p. 328.

35. Schlepckow, K., et al., An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function. EMBO Mol Med, 2017. 9(10): p. 1356-1365.

36. Thornton, P., et al., TREM2 shedding by cleavage at the H157-S158 bond is accelerated for the Alzheimer's disease-associated H157Y variant. EMBO Mol Med, 2017. 9(10): p. 1366-1378.

37. Feuerbach, D., et al., ADAM17 is the main sheddase for the generation of human triggering receptor expressed in myeloid cells (hTREM2) ectodomain and cleaves TREM2 after Histidine 157. Neurosci Lett, 2017. 660: p. 109-114.

38. Kleinberger, G., et al., The FTD-like syndrome causing TREM2 T66M mutation impairs microglia function, brain perfusion, and glucose metabolism. EMBO J, 2017. 36(13): p. 1837-1853.

39. Zhong, L., et al., Soluble TREM2 induces inflammatory responses and enhances microglial survival. J Exp Med, 2017. 214(3): p. 597-607.

40. Zhong, L., et al., Soluble TREM2 ameliorates pathological phenotypes by modulating microglial functions in an Alzheimer's disease model. Nat Commun, 2019. 10(1): p. 1365.

41. Ellulu, M.S., et al., Obesity and inflammation: the linking mechanism and the complications. Arch Med Sci, 2017. 13(4): p. 851-863.

42. Lumeng, C.N. and A.R. Saltiel, Inflammatory links between obesity and metabolic disease. J Clin Invest, 2011. 121(6): p. 2111-7.

108

CHAPTER 6

Conclusion

109

Eosinophil infiltration does not always promote tissue destruction

The view of eosinophils as primary mediators of tissue destruction arose initially from their

association with innate immunity and host defense [1]. Eosinophils can release granule proteins

that have prominent anti-parasitic [2, 3], anti-viral [4-7] and bactericidal [8, 9] properties. While

the contributions of eosinophils to host defense in vivo remains an area of active investigation,

the most recent findings are highly controversial. For example, while eosinophil granule proteins

are toxic to the schistosomulae and larvae of the helminth S. mansoni in experiments carried out

in vitro, eosinophil ablation did not significantly alter disease progression in vivo [10]. As but

another example, eosinophils were shown to promote the growth of Trichinella spiralis larvae by

suppressing inflammation and promoting glucose uptake in the muscle tissue [11, 12].

Eosinophil infiltration into the tissues in the absence of known pathogens was viewed as an

extension of their role in host defense [1]. In this paradigm, recruited eosinophils release cytotoxic

mediators such as the cationic protein major basic protein-1 (MBP-1) and reactive oxygen species

(ROS) by eosinophil peroxidase (EPX) and the NADPH oxidase system. Supporting this view are

a number of associations between eosinophils and tissue damage, for instance, in asthma [13,

14] and in the autoimmune skin disease bullous pemphigoid [15].

Despite their association with tissue damage, eosinophils have also been detected in the

absence of tissue damage, notably in the gastrointestinal tract and in the mammary gland. Here,

eosinophils have been shown to mediate critical homeostatic functions; eosinophils maintain the mucosal barrier by secreting Immunoglobulin-A (IgA) [16, 17] and facilitate ductal branching of the mammary gland [18]. These associations indicate that eosinophils are not universally associated with tissue destruction and that their infiltration into the tissues should not necessarily be regarded as pathological features of the disease process.

In this dissertation, we determined that eosinophil infiltration is associated with, but does not drive, acute muscle damage in muscular dystrophy (mdx) mice. We utilized eosinophil overabundant (mdx.IL5tg) and eosinophil-deficient (mdx.PHIL) mice to demonstrate that the

110 degree of eosinophil infiltration did not correspond with the degree of acute muscle damage. For instance, muscle tissues from eosinophil-sufficient (mdx) and eosinophil-deficient (mdx.PHIL) mice exhibited similar proportions of centrally-nucleated myofibers (27 ± 2.3% vs 29.5 ± 1.8%, respectively) at 4 weeks of age; central positioning of myofiber nuclei identifies recently-damaged myofibers undergoing regeneration (Chapter 3). In fact, we observed that eosinophils were present in high numbers in the absence of any muscle damage in hyper-eosinophilic IL5tg parent mouse strain.

These findings add to the growing body of evidence that challenges the view of eosinophils as purely tissue-destructive. Rather, eosinophils may be recruited to sites of existing tissue damage by elevated levels of damage-associated molecular patterns (DAMPs) such as sphingosine 1-phosphate (S1P) and high mobility group box 1 (HMGB1); indeed, eosinophils exhibit chemotactic properties towards HMGB1 [19] and S1P [20]. Once recruited, eosinophils may modulate the local immunity and/or tissue repair processes, an idea first advanced by Lee and colleagues [21]. Consistent with this idea, recent studies have demonstrated that eosinophil infiltration is critical for the remodeling of acutely damaged tissues in the muscle [22], liver [23], or urogenital tract [24]. Similarly, eosinophils recruited to dystrophin-deficient muscle tissue may mediate one or more aspects of tissue repair; of note, we have detected prominent expression of transcripts encoding tissue remodeling proteins in eosinophils recruited to the muscle tissues of mdx mice (Chapter 4).

Eosinophil infiltration in musculoskeletal disorders

Although we established that eosinophils do not promote muscle damage in mdx mice

(Chapter 3, [25]), the roles of eosinophils in DMD as well as in other musculoskeletal disorders remain to be determined. Earlier work by Wehling-Henricks and colleagues [26] demonstrated that CCR3 antibody-based depletion of eosinophils mitigated the level of muscle damage in mdx mice; however, CCR3 expression has also been detected on lymphocytes [27-30], which are

111 recruited to dystrophin-deficient muscle tissues [26, 31]. More specific targeting of the eosinophil population, for example through the deletion of the gene encoding the eosinophil granule protein

Major Basic Protein-1, did not ameliorate the level of muscle damage in muscular dystrophy mice

[26].

Eosinophil infiltration is prominent in the skeletal muscle tissues of individuals with eosinophilia-myalgia syndrome, a multisystem disorder distinguished by peripheral eosinophilia, myalgia, and the absence of infectious or neoplastic etiology. Epidemiological evidence has linked cases of eosinophilia-myalgia syndrome with the consumption of contaminated L-tryptophan, for example, 1,1’-ethylidenebis L-tryptophan (EBT) [32-34]. However, the mechanisms by which contaminated L-tryptophan lead to myalgia, as well as the role of eosinophils in this disorder, remain unclear. Accordingly, several groups have tried unsuccessfully to replicate features of eosinophilia-myalgia syndrome in animal models [35]. Notably, Silver and colleagues [36, 37] administered contaminated tryptophan (EBT) to wild-type mice, which exhibited prominent inflammation and fibrosis in response to EBT; however, features such as peripheral eosinophilia were conspicuously absent, in contrast with the disease seen in humans. The absence of a suitable animal model suggests that other factors, in addition to the consumption of contaminated

L-tryptophan, may contribute to eosinophil infiltration that is a characteristic of this disorder.

Eosinophil infiltration in skeletal muscle tissues has been reported in patients with limb girdle muscular dystrophy type 2A (LGMD2A) [38-42], an inherited disorder of muscle wasting caused by mutations in the gene encoding calpain-3 (CAPN3) [43]. Expression of calpain-3 is limited to skeletal muscle tissues, where it binds structural proteins including titin [43]. Recent studies indicate that calpain-3 is not required for maintaining the integrity of the sarcolemma but may be critical for the expansion in muscle mass following periods of atrophy [44]. Furthermore, histological analysis revealed rare and limited necrosis in the skeletal muscle tissues of LGMD2A patients and Capn3-/- mice [45-47]. The contributions of eosinophils to the pathophysiology of

LGMD2A remains unclear. Given current concepts that feature a role for eosinophils in

112

remodeling damaged muscle tissue [22], it is reasonable to speculate that recruited eosinophils might promote the expansion of pro-myogenic populations in calpain-3-deficient muscle tissue.

Together, these disorders illustrate the distinct etiology and pathophysiology accompanying disorders associated with eosinophil infiltration in the muscle tissue. As such, infiltrating eosinophils may exhibit distinct features and modulate unique functions within the muscle tissue that would merit further investigation. In the following section, we review our use of RNA- sequencing methods in evaluating the characteristics of eosinophils recruited in muscular dystrophy; use of this approach may reveal unique insights into the role of eosinophils recruited in other musculoskeletal disorders.

RNA-sequencing enables functional characterization of eosinophils

In Chapter 4 of this dissertation, we provided evidence indicating that eosinophils isolated from the muscle tissues of mdx, IL5tg, and mdx.IL5tg mice exhibit distinct transcriptomic features.

We note that eosinophils from the muscle tissues of mdx mice exhibit robust expression of transcripts encoding remodeling enzymes and cell surface proteins; by contrast, eosinophils from the muscle tissues of IL5tg mice preferentially express transcripts encoding eosinophil-specific and cell cycle-related proteins. Taken together, these findings are consistent with current

concepts regarding eosinophils and their capacity to respond transcriptionally to the tissue

microenvironment in order to modify their cellular features and functionality.

These findings directly challenge the perception of eosinophils as purely “end-stage effector

cells” with limited transcriptional activity. This view derived in large part to the dominant presence

of pre-formed granule contents, which would abrogate the need for any de novo synthesis of

functional mediators. Furthermore, isolating intact, high-quality RNA from eosinophils is

complicated by limited abundance and the high concentration of ribonucleases stored in the

cytoplasmic granules. As a result of these factors, only a limited number of studies (<7

publications) have pursued RNA-sequencing to analyze eosinophil characteristics.

113

While recognizing these challenges, we believe that our findings indicate that sequencing and

analysis of the eosinophil transcriptome provides an invaluable approach for the characterization

of these leukocytes. First, although eosinophils contain pre-formed mediators stored within their

granules, there may be a need to replenish granule contents through de novo synthesis.

Eosinophil granules may be rapidly depleted in response to inflammatory insults and/or increased

persistence within tissues. For example, eosinophils recruited to the gastrointestinal tract exhibit

prolonged survival (t1/2 = 6 days) when compared with eosinophils in circulation (t1/2 = 3-24 hours)

[48]. Hence, analysis of the transcriptomic profiles in eosinophils will help to identify unique cellular characteristics and potential mediators of eosinophil function within the tissues.

By utilizing RNA-sequencing, we were able to develop a comprehensive, unbiased view of the characteristics of eosinophils from three distinct microenvironments. Analysis of the eosinophil transcriptome revealed differential expression of several transcripts encoding proinflammatory cytokines and chemokines (e.g., Il15, Ccl7, Ccl8), but the most prominent class of transcripts that were differentially expressed in eosinophils were those encoding tissue remodeling and cell surface proteins. As such, these findings are consistent with the view that eosinophils recruited to the muscles of mdx mice function primarily to regulate remodeling and local immune responses.

Similarly, use of RNA-sequencing in other settings may further challenge preconceived notions regarding eosinophil function and orient investigators towards critical themes for subsequent investigation.

Analysis of our RNA-sequencing results revealed differential expression of the transcript encoding Trem2, which was detected at relatively higher levels in eosinophils recruited to the muscle tissues of mdx mice. In Chapter 5 of this dissertation, we engaged in critical studies aimed at elucidating the expression and function of Trem2 in eosinophils. Trem2 transcripts encode

Triggering Receptor Expressed on Myeloid Cells-2 (TREM2), a receptor that has been shown to promote the proliferation, activation and survival of myeloid cells in the setting of tissue damage

[49-53]. Importantly, we identified a key role for Trem2 in modulating the expansion of the

114

eosinophil lineage; as such, expression of TREM2 may be critical for the expansion and

persistence of eosinophils in dystrophin-deficient muscle tissue. As discussed in the following

section, these findings raise intriguing questions about the regulation of Trem2 expression as well

as its potential role in mediating eosinophil activation.

Future Directions – Regulation and Function of TREM2 on Eosinophils

In this dissertation, we identified prominent expression of Trem2 transcripts in eosinophils

isolated from the muscle tissues of mdx mice (Chapter 4) as well as prominent expression of

immunoreactive TREM2 on mouse eosinophils cultured ex vivo and on their human eosinophil

counterparts isolated from peripheral blood (Chapter 5). These findings are both novel and

intriguing, as expression of TREM2 was widely considered to be limited to cells of the

monocyte/macrophage lineage [54, 55]. Moving forward, it will be critical to elucidate the signals

that promote TREM2 expression in eosinophils. Earlier work has demonstrated that pro-

inflammatory mediators (e.g., bacterial lipopolysaccharides) limit, while anti-inflammatory

mediators (e.g., interleukin-4) promote, expression of immunoreactive TREM2 in macrophages

and microglial cells [56, 57]. Given the increased expression of Trem2 in eosinophils isolated from the muscle tissues of dystrophin-deficient (mdx) mice, we will evaluate the impact of specific inflammatory mediators on the expression of TREM2 on eosinophils; notably, we will examine the impact of IL-4, IL-1RA, Eotaxin-1/CCL11 and RANTES/CCL5, all of which are elevated in dystrophin-deficient muscle tissue (Chapter 3, [25]) and have known impact on mouse

eosinophils. As part of our investigation into the regulation of TREM2 expression in eosinophils,

we will also examine the expression of other TREM and TREM-like family members (e.g., Trem1,

Trem3, Trem4, Trem5, Treml1, Treml2) in cultured eosinophils at baseline and in response to

stimuli.

Our findings indicate that Trem2 is required for efficient development of eosinophils at

baseline, but is not required for their mobilization into the airways in response to allergen- 115

challenge. Intriguingly, Trem2-/- mice challenged intranasally with components of the fungal allergen, Alternaria alternata exhibited similar levels of eosinophils in both the airways and in the

bone marrow when compared with findings generated in wild-type mice (data not shown). These

results suggest that conditions secondary to allergen challenge, such as Th2-mediated

inflammation, may overcome defects in eosinophil development associated with Trem2-

deficiency. As such, it will be critical to evaluate the inter-connecting pathways and cellular

features critical to Trem2-mediated development of eosinophils. To start, we will evaluate

activation of key signaling pathways (e.g., mTOR, Akt) as well as the activity of key cellular

processes (e.g., autophagic flux, metabolic activity) in cultures of wild-type and Trem2-/-

eosinophils; these features are impaired in Trem2-/- microglial cells [51, 53]. Furthermore, we will

evaluate the impact of Th2 cytokines (e.g., IL-4, IL-13) on these cellular features; findings from

this study may help explain the eosinophil recruitment and expansion observed in both wild-type

and Trem2-/- mice in response to challenge with Alternaria alternata.

Finally, we intend to investigate the role of Trem2 in disorders associated with eosinophil

infiltration. Because Trem2 is detected at high levels in eosinophils isolated from the muscle

tissues of mdx mice, we have generated mdx.Trem2-/- mice and will utilize these mice to analyze

the degree of eosinophil infiltration in the muscle tissues. Given the decreased viability that is a

characteristic of Trem2-/- eosinophils cultured ex vivo, we expect mdx.Trem2-/- mice to exhibit

decreased eosinophil infiltration in the muscle tissues when compared with that observed in mdx

mice. In addition, we intend to continue our investigation of Trem2-mediated functions in

association with Alternaria alternata-driven airways hyperreactivity. In our earlier work (Chapter

5), we established that Trem2-deficiency did not abrogate eosinophil recruitment to the airways.

However, it may have an impact on eosinophil function in this microenvironment. Further work will elucidate the impact of Trem2-/- eosinophils on airway histopathology following allergen

challenge.

116

Concluding Remarks

This dissertation examined the function and characteristics of eosinophils recruited to the skeletal muscle tissues of muscular dystrophy (mdx) mice. Towards this end, I developed novel protocols to identify and to isolate eosinophils from muscle tissue and to isolate eosinophil RNA for sequencing and analysis. These tools may be applicable to the characterization of eosinophils in other disorders; for examples, eosinophils are prominent in the muscle tissues associated with

Limb-Girdle Muscular Dystrophy Type 2A (LGMD2A) and eosinophilia-myalgia syndrome. Using these tools, I established that eosinophils do not drive the acute muscle damage in muscular dystrophy (mdx) mice, and that eosinophils exhibit unique features that may potentiate their function. In particular, I focused on Trem2, which encodes a receptor critical for the efficient development of eosinophils. Further work is needed in order to elucidate the regulation and functions of Trem2 in eosinophils and eosinophil-mediated disorders.

117

REFERENCES

1. Lee, J.J. and N.A. Lee, Eosinophil degranulation: an evolutionary vestige or a universally destructive effector function? Clin Exp Allergy, 2005. 35(8): p. 986-94.

2. McLaren, D.J., et al., Morphological studies on the killing of schistosomula of Schistosoma mansoni by human eosinophil and neutrophil cationic proteins in vitro. Parasite Immunol, 1981. 3(4): p. 359-73.

3. Hamann, K.J., et al., In vitro killing of microfilariae of Brugia pahangi and Brugia malayi by eosinophil granule proteins. J Immunol, 1990. 144(8): p. 3166-73.

4. Domachowske, J.B., et al., Recombinant human eosinophil-derived neurotoxin/RNase 2 functions as an effective antiviral agent against respiratory syncytial virus. J Infect Dis, 1998. 177(6): p. 1458-64.

5. Domachowske, J.B., et al., Eosinophil cationic protein/RNase 3 is another RNase A- family ribonuclease with direct antiviral activity. Nucleic Acids Res, 1998. 26(14): p. 3358-63.

6. Phipps, S., et al., Eosinophils contribute to innate antiviral immunity and promote clearance of respiratory syncytial virus. Blood, 2007. 110(5): p. 1578-86.

7. Rugeles, M.T., et al., Ribonuclease is partly responsible for the HIV-1 inhibitory effect activated by HLA alloantigen recognition. AIDS, 2003. 17(4): p. 481-6.

8. Lehrer, R.I., et al., Antibacterial properties of eosinophil major basic protein and eosinophil cationic protein. J Immunol, 1989. 142(12): p. 4428-34.

9. Rosenberg, H.F., Recombinant human eosinophil cationic protein. Ribonuclease activity is not essential for cytotoxicity. J Biol Chem, 1995. 270(14): p. 7876-81.

10. Swartz, J.M., et al., Schistosoma mansoni infection in eosinophil lineage-ablated mice. Blood, 2006. 108(7): p. 2420-7.

11. Huang, L., et al., Eosinophils mediate protective immunity against secondary nematode infection. J Immunol, 2015. 194(1): p. 283-90.

12. Huang, L., et al., Eosinophils and IL-4 Support Nematode Growth Coincident with an Innate Response to Tissue Injury. PLoS Pathog, 2015. 11(12): p. e1005347.

13. Filley, W.V., et al., Identification by immunofluorescence of eosinophil granule major basic protein in lung tissues of patients with bronchial asthma. Lancet, 1982. 2(8288): p. 11-6.

14. Gleich, G.J., et al., The eosinophilic leukocyte and the pathology of fatal bronchial asthma: evidence for pathologic heterogeneity. J Allergy Clin Immunol, 1987. 80(3 Pt 2): p. 412-5.

15. Lin, L., et al., Eosinophils Mediate Tissue Injury in the Autoimmune Skin Disease Bullous Pemphigoid. J Invest Dermatol, 2018. 138(5): p. 1032-1043.

118

16. Chu, V.T., et al., Eosinophils promote generation and maintenance of immunoglobulin-A- expressing plasma cells and contribute to gut immune homeostasis. Immunity, 2014. 40(4): p. 582-93.

17. Jung, Y., et al., IL-1beta in eosinophil-mediated small intestinal homeostasis and IgA production. Mucosal Immunol, 2015. 8(4): p. 930-42.

18. Gouon-Evans, V., M.E. Rothenberg, and J.W. Pollard, Postnatal mammary gland development requires macrophages and eosinophils. Development, 2000. 127(11): p. 2269-82.

19. Lotfi, R., et al., Eosinophils oxidize damage-associated molecular pattern molecules derived from stressed cells. J Immunol, 2009. 183(8): p. 5023-31.

20. Roviezzo, F., et al., Human eosinophil chemotaxis and selective in vivo recruitment by sphingosine 1-phosphate. Proc Natl Acad Sci U S A, 2004. 101(30): p. 11170-5.

21. Lee, J.J., et al., Eosinophils in health and disease: the LIAR hypothesis. Clin Exp Allergy, 2010. 40(4): p. 563-75.

22. Heredia, J.E., et al., Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell, 2013. 153(2): p. 376-88.

23. Goh, Y.P., et al., Eosinophils secrete IL-4 to facilitate liver regeneration. Proc Natl Acad Sci U S A, 2013. 110(24): p. 9914-9.

24. Vicetti Miguel, R.D., et al., IL-4-secreting eosinophils promote endometrial stromal cell proliferation and prevent Chlamydia-induced upper genital tract damage. Proc Natl Acad Sci U S A, 2017.

25. Sek, A.C., et al., Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy. J Immunol, 2019. 203(2): p. 476-484.

26. Wehling-Henricks, M., et al., Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy. Hum Mol Genet, 2008. 17(15): p. 2280-92.

27. Gerber, B.O., et al., Functional expression of the eotaxin receptor CCR3 in T lymphocytes co-localizing with eosinophils. Curr Biol, 1997. 7(11): p. 836-43.

28. Jinquan, T., et al., CCR3 expression induced by IL-2 and IL-4 functioning as a death receptor for B cells. J Immunol, 2003. 171(4): p. 1722-31.

29. Jinquan, T., et al., Eotaxin activates T cells to chemotaxis and adhesion only if induced to express CCR3 by IL-2 together with IL-4. J Immunol, 1999. 162(7): p. 4285-92.

30. Francis, J.N., et al., T lymphocytes expressing CCR3 are increased in allergic rhinitis compared with non-allergic controls and following allergen immunotherapy. Allergy, 2007. 62(1): p. 59-65.

119

31. Morrison, J., et al., T-cell-dependent fibrosis in the mdx dystrophic mouse. Lab Invest, 2000. 80(6): p. 881-91.

32. Allen, J.A., et al., Post-epidemic eosinophilia-myalgia syndrome associated with L- tryptophan. Arthritis Rheum, 2011. 63(11): p. 3633-9.

33. Hertzman, P.A., et al., Association of the eosinophilia-myalgia syndrome with the ingestion of tryptophan. N Engl J Med, 1990. 322(13): p. 869-73.

34. Mayeno, A.N., et al., Characterization of Peak-E, a Novel Amino-Acid Associated with Eosinophilia-Myalgia-Syndrome. Science, 1990. 250(4988): p. 1707-1708.

35. Clauw, D.J., Animal models of the eosinophilia-myalgia syndrome. J Rheumatol Suppl, 1996. 46: p. 93-7; discussion 92, 97-8.

36. Silver, R.M., et al., A murine model of the eosinophilia-myalgia syndrome induced by 1,1'-ethylidenebis (L-tryptophan). J Clin Invest, 1994. 93(4): p. 1473-80.

37. Suzuki, S., et al., A contaminant of L-tryptophan enhances expression of dermal collagen in a murine model of eosinophilia myalgia syndrome. Proc Assoc Am Physicians, 1996. 108(4): p. 315-22.

38. Schroder, T., et al., Eosinophils in hereditary and inflammatory myopathies. Acta Myol, 2013. 32(3): p. 148-53.

39. Luo, Y.B., et al., Limb-girdle Muscular Dystrophy Type 2A with Muscular Eosinophilic Infiltration in a Chinese Patient. Chin Med J (Engl), 2018. 131(17): p. 2133-2134.

40. Mahmood, O.A., X. Jiang, and Q. Zhang, Limb-girdle muscular dystrophy subtypes: First-reported cohort from northeastern China. Neural Regen Res, 2013. 8(20): p. 1907- 18.

41. Krahn, M., et al., CAPN3 mutations in patients with idiopathic eosinophilic myositis. Ann Neurol, 2006. 59(6): p. 905-11.

42. Krahn, M., et al., Eosinophilic infiltration related to CAPN3 mutations: a pathophysiological component of primary calpainopathy? Clin Genet, 2011. 80(4): p. 398-402.

43. Zatz, M. and A. Starling, Calpains and disease. N Engl J Med, 2005. 352(23): p. 2413- 23.

44. Kramerova, I., et al., Calpain 3 and CaMKIIbeta signaling are required to induce HSP70 necessary for adaptive muscle growth after atrophy. Hum Mol Genet, 2018. 27(9): p. 1642-1653.

45. Kramerova, I., et al., Null mutation of calpain 3 (p94) in mice causes abnormal sarcomere formation in vivo and in vitro. Hum Mol Genet, 2004. 13(13): p. 1373-88.

46. Fanin, M., et al., Loss of calpain-3 autocatalytic activity in LGMD2A patients with normal protein expression. Am J Pathol, 2003. 163(5): p. 1929-36.

120

47. Vainzof, M., et al., The effect of calpain 3 deficiency on the pattern of muscle degeneration in the earliest stages of LGMD2A. J Clin Pathol, 2003. 56(8): p. 624-6.

48. Marichal, T., C. Mesnil, and F. Bureau, Homeostatic Eosinophils: Characteristics and Functions. Front Med (Lausanne), 2017. 4: p. 101.

49. Ulland, T.K. and M. Colonna, TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol, 2018. 14(11): p. 667-675.

50. Zheng, H., et al., TREM2 in Alzheimer's Disease: Microglial Survival and Energy Metabolism. Front Aging Neurosci, 2018. 10: p. 395.

51. Ulland, T.K., et al., TREM2 Maintains Microglial Metabolic Fitness in Alzheimer's Disease. Cell, 2017. 170(4): p. 649-663 e13.

52. Jaitin, D.A., et al., Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell, 2019. 178(3): p. 686-698 e14.

53. Wang, Y., et al., TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model. Cell, 2015. 160(6): p. 1061-71.

54. Colonna, M., TREMs in the immune system and beyond. Nat Rev Immunol, 2003. 3(6): p. 445-53.

55. Deming, Y., et al., Triggering receptor expressed on myeloid cells 2 (TREM2): a potential therapeutic target for Alzheimer disease? Expert Opin Ther Targets, 2018. 22(7): p. 587-598.

56. Owens, R., et al., Divergent Neuroinflammatory Regulation of Microglial TREM Expression and Involvement of NF-kappaB. Front Cell Neurosci, 2017. 11: p. 56.

57. Turnbull, I.R., et al., Cutting edge: TREM-2 attenuates macrophage activation. J Immunol, 2006. 177(6): p. 3520-4.

121