Graduate Students Alphabetical by Graduate Student Last Name

Total Page:16

File Type:pdf, Size:1020Kb

Graduate Students Alphabetical by Graduate Student Last Name Graduate Students Alphabetical by Graduate Student last name 1. Development of a mouse model to study SMN splicing and replacement therapy Presenting author: Bebee, Thomas Proximal spinal muscular atrophy (SMA) is a neuromuscular disease caused by low levels of SMN protein due to loss of the Survival Motor Neuron-1 (SMN1) gene. Humans have a duplicate gene, SMN2, that generates low levels of SMN protein due to a C>T point mutation that skips exon 7. We generated a mouse model that recapitulates SMN2 altered splicing by engineering the exon 7 C>T mutation into the homologous mouse Smn gene. The C>T mutation in Smn induces skipping of exon 7 in multiple tissues in the mouse, reduces SMN protein levels, and leads to a mild form of SMA exhibited by reduced hind limb grip strength, rearing, and activity. Increasing SMN expression by correcting splicing or increasing transcription of SMN2 are attractive therapeutic options in SMA patients. However, the timing of SMN replacement will be crucial in the treatment of SMA. Studies in zebrafish predict SMN function during embryogenesis may be important for axonal pathfinding, while mouse models and SMA disease progression in humans suggest that post-natal treatment may be sufficient to protect motor neurons. AAV9 encoding SMN has shown that early postnatal SMN expression in spinal cord motor neurons is sufficient to rescue survival though complete rescue of normal development was not observed, indicating that earlier treatment may be required for full restoration of SMN function and disease correction. To address the question of optimal therapeutic timing for SMA we have developed a temporally inducible transgenic mouse in which the expression of human SMN cDNA is under the control of tamoxifen inducible Cre-recombinase. In our inducible mouse, induced transgene recombination following tamoxifen treatment was validated in 4 week old weanling mice, PND1.5 treated neonatal mice, and E13.5 treated embryos. The temporal induction of transgene recombination and SMN expression was further validated in these mice in various tissues including the brain and spinal cord. When crossed to SMA mouse models our inducible SMN mouse model will allow for the temporal induction of SMN at varying time-points during development, both in utero and postnatally, to evaluate the time-point in which SMN replacement is required for SMA correction. The therapeutic window determined from these experiments can then be used in our Smn C>T mouse model to validate the use of drug therapies targeting splicing correction in the treatment of SMA mice, and in turn SMA patients. 2. Muscle enhancement in SMA Presenting author: Bevan, Adam K. Purpose: Spinal Muscular Atrophy (SMA) is a devastating neurological disease marked by lower motor neuron death and the subsequent atrophy of skeletal muscle. SMA patients can present with various severity of disease; SMA type 1 is very severe, usually leading to respiratory failure by 2 years, while SMA type 3 is non-fatal but patients are weak and are often wheelchair-dependent by their teenage years. While this disease primarily affects only spinal motor neurons, enhancement of skeletal muscle may be a good approach to treat patients with less-severe disease to increase their quality of life. We have therefore tested the muscle enhancing effects of follistatin gene therapy in severe (“Δ7”) and mild (“C/C”) mouse models of SMA. Methods: Newborn Δ7 (SMN2+/+, Δ7SMN+/+, Smn-/-) and C/C mice (SMN2+/+, hybSmn+/+, Smn-/-) were injected with 1e+11 particles of AAV9 carrying the follistatin transgene (AAV9.CBA.FS344) via the facial vein. Behavioral tests were performed starting at 6 days of age and continued until either 1) death in Δ7 mice or 2) at least 60 days of age in C/C mice. Muscle-related physiological measures were also performed in C/C mice at 200 days of age including muscle mass and in situ and ex vivo force generation. Transgenic follistatin expression was also confirmed by body mass, serum follistatin concentration and muscle fiber diameter measurements. Results: Both Δ7 and C/C mice showed early, positive effects of muscle enhancement, though no significant extension of survival was observed in the Δ7 group. In C/C mice, follistatin treatment had a positive effect on righting ability and, as predicted, significantly increased lean body mass and grip strength. Importantly, the ex vivo force generated by stimulation of the nerve was greater in follistatin-treated C/C mice despite having a pathological neuromuscular junction. Conclusions: While follistatin had little effect in the rapidly progressing Δ7 model, our data suggests that muscle enhancement may be a very useful therapy for patients with more mild forms of SMA. 3. Gene Therapy and Ovarian Cancer Presenting author: Bolyard, Chelsea M. Ovarian cancer is the leading cause of death due to gynecological malignancy, and a leading cause of death due to cancer in developed countries. Because patients often present in late stages of disease, aggressive multi-modal therapies may provide the most efficacious results. Within our study, we investigate how Adeno-associated virus (AAV) gene therapy can be combined with chemotherapy to treat ovarian carcinoma. Adeno- associated virus can be modified to incorporate targeting ligands into the viral capsid, facilitating AAV transduction into tumor-associated vasculature when administered systemically. These ligands bind receptors up-regulated on the angiogenic vasculature within the tumor microenvironment. AAV transduction can be increased by treatment with certain types of anti-cancer drugs, such as DNA-damaging agents (doxorubicin), histone deacetylase inhibitors (bortezomib) and proteasome inhibitors (depsipeptide). We examine the ability of TNFα-Related Apoptosis-Inducing Ligand (TRAIL) gene therapy to augment chemotherapy-mediated killing of ovarian cancer cells. Chemotherapeutics can also increase tumor cell sensitivity to TRAIL-mediated apoptosis. In this study, we will deliver TRAIL using the vasculature-targeted self-complementary AAV vector, after treatment with chemotherapeutics. Because our in vivo model of gene therapy targets the tumor-associated vasculature, our in vitro work focuses on the paracrine effects of TRAIL. When ovarian cancer cell lines were pre-treated with drugs, their sensitivity to the paracrine effects of TRAIL increased markedly. With this study, we have determined that TRAIL can be delivered within the context of the AAV genome, and that the TRAIL secreted from transduced cells maintains paracrine activity. Furthermore, the effects of chemotherapeutics can be potentiated by subsequent TRAIL treatment. This implies that with a multi-modal therapy, chemotherapeutic doses could be reduced and systemic toxicity minimized. 4. Aspirin Therapy in Neonatal Hyperoxia Presenting author: Britt, Rodney Purpose: Preterm infants are born with immature lungs, often requiring oxygen therapy to maintain homeostatic blood oxygen levels. Infants with prolonged oxygen exposure are at highest risk of developing chronic lung diseases such as Bronchopulmonary Dysplasia (BPD) and pulmonary complications later in life. Oxygen toxicity induces a pro- inflammatory response which is associated with lung injury and impairment of lung development which may persist through life. Aspirin is a nonselective cyclooxygenase (COX) inhibitor which is known to disrupt pro-inflammatory processes through multiple mechanisms. We tested the hypothesis that aspirin therapy will improve lung function and minimize airway hyperresponsiveness in lungs of adult mice following newborn hyperoxia exposure. Methods: Immediately after birth, newborn mice were exposed to room air (RA) or hyperoxia (>95% O2) and were injected daily with vehicle (PBS), 5, or 10 mg/kg aspirin (ASA). On day 7, hyperoxia exposed mice were returned to room air. Pulmonary function tests (PFT) were performed using the SCIREQ Flexivent system on day 28 and day 70. Results: Airway reactivity was assessed by measuring total resistance following intratracheal treatment with 0, 5, 10, 15, 25, and 50 mg/mL acetylmethacholine. Pressure- Volume loops (PV loops) indicated an increase in compliance in PBS/>95% O2 mice which was ameliorated in the hyperoxia exposed pups treated with aspirin. Total resistance was significantly higher in PBS/>95% O2 mice in response toacetylmethacholine challenge compared to room air controls, however both ASA/>95% O2 treatment groups exhibited only mild increase in resistance in response to 50 mg/mL acetylmethacholine (6.18±0.59 vs 2.93±0.35). Conclusions:Aspirin treatment attenuated detrimental effects of hyperoxia on lung function and airway reactivity. Aspirin may be a beneficial therapy for preterm infants who are at risk of developing BPD. 5. rAAV recombination Presenting author: Cataldi, Marcela P. The palindromic terminal repeats (TR) of adeno-associated virus (AAV) form DNA hairpins (HP) that are essential for replication and for priming the conversion of single- stranded virion DNA to double-strand. In recombinant AAV (rAAV) gene-delivery vectors, they are targets for DNA repair pathways leading to circularization, concatemerization and, infrequently, chromosomal integration. Recombination with chromosomal DNA poses a risk for genotoxicity, raising the question as to whether the hairpin structures formed by the TRs are more likely to integrate than other forms of DNA ends. Because we cannot generate rAAV viral vectors without HP ends, we investigated the contribution of AAV TR to chromosomal integration
Recommended publications
  • View Neuroscience
    Minor snRNA gene delivery improves the loss of proprioceptive synapses on SMA motor neurons Erkan Y. Osman, … , Livio Pellizzoni, Christian L. Lorson JCI Insight. 2020. https://doi.org/10.1172/jci.insight.130574. Research In-Press Preview Neuroscience Spinal muscular atrophy (SMA) is an inherited neuromuscular disorder caused by reduced expression of the survival motor neuron (SMN) protein. SMN has key functions in multiple RNA pathways, including the biogenesis of small nuclear ribonucleoproteins (snRNPs) that are essential components of both major (U2-dependent) and minor (U12-dependent) spliceosomes. Here we investigated the specific contribution of U12 splicing dysfunction to SMA pathology through selective restoration of this RNA pathway in mouse models of varying phenotypic severity. We show that viral-mediated delivery of minor snRNA genes specifically improves select U12 splicing defects induced by SMN deficiency in cultured mammalian cells as well as in the spinal cord and dorsal root ganglia of SMA mice without increasing SMN expression. This approach resulted in a moderate amelioration of several parameters of the disease phenotype in SMA mice including survival, weight gain and motor function. Importantly, minor snRNA gene delivery improved aberrant splicing of the U12 intron-containing gene Stasimon and rescued the severe loss of proprioceptive sensory synapses on SMA motor neurons, which are early signatures of motor circuit dysfunction in mouse models. Taken together, these findings establish the direct contribution of U12 splicing dysfunction to synaptic deafferentation and motor circuit pathology in SMA. Find the latest version: https://jci.me/130574/pdf Minor snRNA gene delivery improves the loss of proprioceptive synapses on SMA motor neurons Erkan Y.
    [Show full text]
  • Defining Functional Interactions During Biogenesis of Epithelial Junctions
    ARTICLE Received 11 Dec 2015 | Accepted 13 Oct 2016 | Published 6 Dec 2016 | Updated 5 Jan 2017 DOI: 10.1038/ncomms13542 OPEN Defining functional interactions during biogenesis of epithelial junctions J.C. Erasmus1,*, S. Bruche1,*,w, L. Pizarro1,2,*, N. Maimari1,3,*, T. Poggioli1,w, C. Tomlinson4,J.Lees5, I. Zalivina1,w, A. Wheeler1,w, A. Alberts6, A. Russo2 & V.M.M. Braga1 In spite of extensive recent progress, a comprehensive understanding of how actin cytoskeleton remodelling supports stable junctions remains to be established. Here we design a platform that integrates actin functions with optimized phenotypic clustering and identify new cytoskeletal proteins, their functional hierarchy and pathways that modulate E-cadherin adhesion. Depletion of EEF1A, an actin bundling protein, increases E-cadherin levels at junctions without a corresponding reinforcement of cell–cell contacts. This unexpected result reflects a more dynamic and mobile junctional actin in EEF1A-depleted cells. A partner for EEF1A in cadherin contact maintenance is the formin DIAPH2, which interacts with EEF1A. In contrast, depletion of either the endocytic regulator TRIP10 or the Rho GTPase activator VAV2 reduces E-cadherin levels at junctions. TRIP10 binds to and requires VAV2 function for its junctional localization. Overall, we present new conceptual insights on junction stabilization, which integrate known and novel pathways with impact for epithelial morphogenesis, homeostasis and diseases. 1 National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK. 2 Computing Department, Imperial College London, London SW7 2AZ, UK. 3 Bioengineering Department, Faculty of Engineering, Imperial College London, London SW7 2AZ, UK. 4 Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK.
    [Show full text]
  • Systemic Restoration of UBA1 Ameliorates Disease in Spinal Muscular Atrophy
    Systemic restoration of UBA1 ameliorates disease in spinal muscular atrophy Rachael A. Powis, … , Mimoun Azzouz, Thomas H. Gillingwater JCI Insight. 2016;1(11):e87908. https://doi.org/10.1172/jci.insight.87908. Research Article Neuroscience Therapeutics The autosomal recessive neuromuscular disease spinal muscular atrophy (SMA) is caused by loss of survival motor neuron (SMN) protein. Molecular pathways that are disrupted downstream of SMN therefore represent potentially attractive therapeutic targets for SMA. Here, we demonstrate that therapeutic targeting of ubiquitin pathways disrupted as a consequence of SMN depletion, by increasing levels of one key ubiquitination enzyme (ubiquitin-like modifier activating enzyme 1 [UBA1]), represents a viable approach for treating SMA. Loss of UBA1 was a conserved response across mouse and zebrafish models of SMA as well as in patient induced pluripotent stem cell–derive motor neurons. Restoration of UBA1 was sufficient to rescue motor axon pathology and restore motor performance in SMA zebrafish. Adeno- associated virus serotype 9–UBA1 (AAV9-UBA1) gene therapy delivered systemic increases in UBA1 protein levels that were well tolerated over a prolonged period in healthy control mice. Systemic restoration of UBA1 in SMA mice ameliorated weight loss, increased survival and motor performance, and improved neuromuscular and organ pathology. AAV9-UBA1 therapy was also sufficient to reverse the widespread molecular perturbations in ubiquitin homeostasis that occur during SMA. We conclude that UBA1 represents a safe and effective therapeutic target for the treatment of both neuromuscular and systemic aspects of SMA. Find the latest version: https://jci.me/87908/pdf RESEARCH ARTICLE Systemic restoration of UBA1 ameliorates disease in spinal muscular atrophy Rachael A.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Mitochondrial Heat Shock Protein 60: Evaluation of Its Role As a Neuroprotectant in Familial ALS and Its Mutation As a Cause of Hereditary Spastic Paraplegia
    Mitochondrial Heat Shock Protein 60: Evaluation of its role as a neuroprotectant in familial ALS and its mutation as a cause of hereditary spastic paraplegia By Laura A. Cooper Integrated Program in Neuroscience McGill University, Montreal June 2011 A thesis submitted to McGill University in partial fulfillment of the requirements of the degree of master’s in science © Laura A. Cooper, 2011 TABLE OF CONTENTS ABSTRACT…………………………………………………………………………….vi RÉSUMÉ……………………………………………………………………………….viii ACKNOWLEDGMENTS……………………………………………………………..xi LIST OF FIGURES……………………………………………………………………xiii ABBREVIATIONS………………………………………………………………….....xv INTRODUCTION……………………………………………………………………..xx CHAPTER 1 – Literature Review 1.1 Amyotrophic Lateral Sclerosis 1.1.1 Clinical Overview..................................................................................................1 1.1.2 Sporadic ALS…………………………………………………………………….2 1.1.3 Familial ALS……………………………………………………………………..4 1.1.4 Cu/Zn Superoxide Dismutase Mutation and fALS……………………………….4 1.1.5 Protein Aggregation in SOD1-Related fALS…………………………………….5 1.1.6 Mitochondrial Abnormalities in Mouse Models of SOD1-Related fALS………..7 1.1.7 Relationship Between Mutant SOD1 and Mitochondrial Abnormalities………..8 1.2 Heat Shock Proteins 1.2.1 Normal Function in the Central Nervous System and Relevance to ALS……….9 1.2.2 Heat Shock Proteins as a Therapeutic Target in ALS………………………….10 1.3 Mitochondrial Heat Shock Protein Hsp60 1.3.1 Structure and Function………………………………………………………....12 ii 1.3.2 Mitochondrial Hsp60 in Neuroprotection…………………………………….
    [Show full text]
  • Supplementary Table S1. Correlation Between the Mutant P53-Interacting Partners and PTTG3P, PTTG1 and PTTG2, Based on Data from Starbase V3.0 Database
    Supplementary Table S1. Correlation between the mutant p53-interacting partners and PTTG3P, PTTG1 and PTTG2, based on data from StarBase v3.0 database. PTTG3P PTTG1 PTTG2 Gene ID Coefficient-R p-value Coefficient-R p-value Coefficient-R p-value NF-YA ENSG00000001167 −0.077 8.59e-2 −0.210 2.09e-6 −0.122 6.23e-3 NF-YB ENSG00000120837 0.176 7.12e-5 0.227 2.82e-7 0.094 3.59e-2 NF-YC ENSG00000066136 0.124 5.45e-3 0.124 5.40e-3 0.051 2.51e-1 Sp1 ENSG00000185591 −0.014 7.50e-1 −0.201 5.82e-6 −0.072 1.07e-1 Ets-1 ENSG00000134954 −0.096 3.14e-2 −0.257 4.83e-9 0.034 4.46e-1 VDR ENSG00000111424 −0.091 4.10e-2 −0.216 1.03e-6 0.014 7.48e-1 SREBP-2 ENSG00000198911 −0.064 1.53e-1 −0.147 9.27e-4 −0.073 1.01e-1 TopBP1 ENSG00000163781 0.067 1.36e-1 0.051 2.57e-1 −0.020 6.57e-1 Pin1 ENSG00000127445 0.250 1.40e-8 0.571 9.56e-45 0.187 2.52e-5 MRE11 ENSG00000020922 0.063 1.56e-1 −0.007 8.81e-1 −0.024 5.93e-1 PML ENSG00000140464 0.072 1.05e-1 0.217 9.36e-7 0.166 1.85e-4 p63 ENSG00000073282 −0.120 7.04e-3 −0.283 1.08e-10 −0.198 7.71e-6 p73 ENSG00000078900 0.104 2.03e-2 0.258 4.67e-9 0.097 3.02e-2 Supplementary Table S2.
    [Show full text]
  • The Ubiquitin Proteasome System in Neuromuscular Disorders: Moving Beyond Movement
    International Journal of Molecular Sciences Review The Ubiquitin Proteasome System in Neuromuscular Disorders: Moving Beyond Movement 1, , 2, 3,4 Sara Bachiller * y , Isabel M. Alonso-Bellido y , Luis Miguel Real , Eva María Pérez-Villegas 5 , José Luis Venero 2 , Tomas Deierborg 1 , José Ángel Armengol 5 and Rocío Ruiz 2 1 Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Sölvegatan 19, 221 84 Lund, Sweden; [email protected] 2 Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla/Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Sevilla, Spain; [email protected] (I.M.A.-B.); [email protected] (J.L.V.); [email protected] (R.R.) 3 Unidad Clínica de Enfermedades Infecciosas, Hospital Universitario de Valme, 41014 Sevilla, Spain; [email protected] 4 Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, 29071 Universidad de Málaga, Spain 5 Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, 41013 Sevilla, Spain; [email protected] (E.M.P.-V.); [email protected] (J.Á.A.) * Correspondence: [email protected] These authors contributed equally to the work. y Received: 14 July 2020; Accepted: 31 August 2020; Published: 3 September 2020 Abstract: Neuromuscular disorders (NMDs) affect 1 in 3000 people worldwide. There are more than 150 different types of NMDs, where the common feature is the loss of muscle strength. These disorders are classified according to their neuroanatomical location, as motor neuron diseases, peripheral nerve diseases, neuromuscular junction diseases, and muscle diseases. Over the years, numerous studies have pointed to protein homeostasis as a crucial factor in the development of these fatal diseases.
    [Show full text]
  • UBA1: at the Crossroads Of
    Review UBA1: At the Crossroads of Ubiquitin Homeostasis and Neurodegeneration 1,2 1,2, Ewout J.N. Groen and Thomas H. Gillingwater * Neurodegenerative diseases are a leading cause of disability and early death. A Trends common feature of these conditions is disruption of protein homeostasis. Disruption of protein homeostasis is an Ubiquitin-like modifier activating enzyme 1 (UBA1), the E1 ubiquitin-activating important feature of many neurode- generative diseases. The E1 ubiqui- enzyme, sits at the apex of the ubiquitin cascade and represents an important tin-activating enzyme UBA1 sits at regulator of cellular protein homeostasis. Critical contributions of UBA1-depen- the apex of ubiquitin pathways, playing a critical role in regulating protein dent pathways to the regulation of homeostasis and degeneration in the nervous homeostasis. UBA1 regulates a system are emerging, including specific disruption of UBA1 in spinal muscular diverse range of cellular processes in atrophy (SMA) and Huntington's disease (HD). In this review we discuss recent the nervous system. findings that put UBA1 at the centre of cellular homeostasis and neurodegen- UBA1 contributes to the pathogenesis eration, highlighting the potential for UBA1 to act as a promising therapeutic of several neurodegenerative diseases, target for a range of neurodegenerative diseases. including SMA and HD. In SMA, decreased UBA1 expression leads to perturbations in ubiquitin homeostasis, aberrant accumulation of downstream Disruption of Protein Homeostasis in Neurodegenerative Disease target proteins, and neuromuscular Neurodegenerative diseases are a common cause of disability and early death throughout global degeneration. In HD, UBA1 expression populations [1,2]. Although our understanding of the underlying pathogenic mechanisms has decreases over time, leading to selec- tive accumulation of toxic forms of hun- improved greatly over recent years, most neurodegenerative diseases currently remain untreat- tingtin protein in the brain.
    [Show full text]
  • FARE2021WINNERS Sorted by Institute
    FARE2021WINNERS Sorted By Institute Swati Shah Postdoctoral Fellow CC Radiology/Imaging/PET and Neuroimaging Characterization of CNS involvement in Ebola-Infected Macaques using Magnetic Resonance Imaging, 18F-FDG PET and Immunohistology The Ebola (EBOV) virus outbreak in Western Africa resulted in residual neurologic abnormalities in survivors. Many case studies detected EBOV in the CSF, suggesting that the neurologic sequelae in survivors is related to viral presence. In the periphery, EBOV infects endothelial cells and triggers a “cytokine stormâ€. However, it is unclear whether a similar process occurs in the brain, with secondary neuroinflammation, neuronal loss and blood-brain barrier (BBB) compromise, eventually leading to lasting neurological damage. We have used in vivo imaging and post-necropsy immunostaining to elucidate the CNS pathophysiology in Rhesus macaques infected with EBOV (Makona). Whole brain MRI with T1 relaxometry (pre- and post-contrast) and FDG-PET were performed to monitor the progression of disease in two cohorts of EBOV infected macaques from baseline to terminal endpoint (day 5-6). Post-necropsy, multiplex fluorescence immunohistochemical (MF-IHC) staining for various cellular markers in the thalamus and brainstem was performed. Serial blood and CSF samples were collected to assess disease progression. The linear mixed effect model was used for statistical analysis. Post-infection, we first detected EBOV in the serum (day 3) and CSF (day 4) with dramatic increases until euthanasia. The standard uptake values of FDG-PET relative to whole brain uptake (SUVr) in the midbrain, pons, and thalamus increased significantly over time (p<0.01) and positively correlated with blood viremia (p≤0.01).
    [Show full text]
  • SPINAL MUSCULAR ATROPHY: PATHOLOGY, DIAGNOSIS, CLINICAL PRESENTATION, THERAPEUTIC STRATEGIES & TREATMENTS Content
    SPINAL MUSCULAR ATROPHY: PATHOLOGY, DIAGNOSIS, CLINICAL PRESENTATION, THERAPEUTIC STRATEGIES & TREATMENTS Content 1. DISCLAIMER 2. INTRODUCTION 3. SPINAL MUSCULAR ATROPHY: PATHOLOGY, DIAGNOSIS, CLINICAL PRESENTATION, THERAPEUTIC STRATEGIES & TREATMENTS 4. BIBLIOGRAPHY 5. GLOSSARY OF MEDICAL TERMS 1 SPINAL MUSCULAR ATROPHY: PATHOLOGY, DIAGNOSIS, CLINICAL PRESENTATION, THERAPEUTIC STRATEGIES & TREATMENTS Disclaimer The information in this document is provided for information purposes only. It does not constitute advice on any medical, legal, or regulatory matters and should not be used in place of consultation with appropriate medical, legal, or regulatory personnel. Receipt or use of this document does not create a relationship between the recipient or user and SMA Europe, or any other third party. The information included in this document is presented as a synopsis, may not be exhaustive and is dated November 2020. As such, it may no longer be current. Guidance from regulatory authorities, study sponsors, and institutional review boards should be obtained before taking action based on the information provided in this document. This document was prepared by SMA Europe. SMA Europe cannot guarantee that it will meet requirements or be error-free. The users and recipients of this document take on any risk when using the information contained herein. SMA Europe is an umbrella organisation, founded in 2006, which includes spinal muscular atrophy (SMA) patient and research organisations from across Europe. SMA Europe campaigns to improve the quality of life of people who live with SMA, to bring effective therapies to patients in a timely and sustainable way, and to encourage optimal patient care. SMA Europe is a non-profit umbrella organisation that consists of 23 SMA patients and research organisations from 22 countries across Europe.
    [Show full text]
  • Supplementary Material Contents
    Supplementary Material Contents Immune modulating proteins identified from exosomal samples.....................................................................2 Figure S1: Overlap between exosomal and soluble proteomes.................................................................................... 4 Bacterial strains:..............................................................................................................................................4 Figure S2: Variability between subjects of effects of exosomes on BL21-lux growth.................................................... 5 Figure S3: Early effects of exosomes on growth of BL21 E. coli .................................................................................... 5 Figure S4: Exosomal Lysis............................................................................................................................................ 6 Figure S5: Effect of pH on exosomal action.................................................................................................................. 7 Figure S6: Effect of exosomes on growth of UPEC (pH = 6.5) suspended in exosome-depleted urine supernatant ....... 8 Effective exosomal concentration....................................................................................................................8 Figure S7: Sample constitution for luminometry experiments..................................................................................... 8 Figure S8: Determining effective concentration .........................................................................................................
    [Show full text]
  • Hnrnp C1/C2 May Regulate Exon 7 Splicing in the Spinal Muscular Atrophy Gene SMN1
    Kobe J. Med. Sci., Vol. 54, No. 5, pp. E227-E236, 2008 HnRNP C1/C2 May Regulate Exon 7 Splicing in the Spinal Muscular Atrophy Gene SMN1 SANAE IRIMURA1, KEIKO KITAMURA1,2,3, NOZOMU KATO1,2,3, KAYOKO SAIKI2,3, ATSUKO TAKEUCHI3, GUNADI1, MASAFUMI MATSUO2, HISAHIDE NISHIO1,2 and MYEONG JIN LEE1,* 1Department of Genetic Epidemiology, 2Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan; 3Kobe Pharmaceutical University, Kobe, Japan; Received 26 December 2008/ Accepted 28 December 2008 Key Words: Spinal muscular atrophy, Alternative pre-mRNA splicing, SMN genes, Exon 7, HnRNP C1/C2 Spinal muscular atrophy (SMA) is caused by loss of SMN1. A nearly identical gene, SMN2, fails to compensate for the loss of SMN1 because SMN2 produces mainly an exon 7-skipped product. The +6C in SMN1 exon 7 proceeds to include exon 7 into mRNA, while the +6U in SMN2 causes skipping of exon 7. Here, ∼45kD proteins bound to the SMN exon 7 RNA probe was found, and identified as hnRNP C1/C2. In gel-shift assay, hnRNP C1/C2 had a greater affinity for the RNA probe with +6C than for the RNA probe with +6U. In vitro splicing assay showed that anti-hnRNP C1/C2 antibody hampered splicing of SMN1 exon 7, but did not affect splicing of SMN2 exon 7. In conclusion, we showed the possibility that hnRNP C1/C2 enhanced SMN1 exon 7 splicing specifically. Spinal muscular atrophy (SMA) is a common autosomal recessive neuromuscular disorder that is characterized by degeneration of the anterior horn cells of the spinal cord, which leads to the axial and limb weakness associated with muscle atrophy.
    [Show full text]