<<

UNIVERSITY OF CALGARY

Neuropathogenic effects of Syncytin-1 in Multiple Sclerosis

by

Joseph Mathew Antony

A THESIS

SUBMITTED TO THE FACULTY OF GRADUATE STUDIES IN PARTIAL

FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF

PHILOSOPHY

DEPARTMENT OF MEDICAL SCIENCE

CALGARY, ALBERTA JUNE, 2006

© Joseph Mathew Antony 2006

iii

ABSTRACT

Human endogenous (HERVs) constitute 8% of the human genome and have been implicated in both health and disease. Increased HERV gene activity occurs in activated glia although the consequences of HERV expression in the nervous system remain uncertain. Relative quantification and quantitative PCR analysis of HERV envelopes (env) revealed selectively increased abundance of HERV-W-7q encoded glycoprotein, Syncytin-1 in brains but not in blood-derived leukocytes from patients with

Multiple Sclerosis (MS) relative to non-MS patients. Syncytin-1 expression in astrocytes induced the release of redox reactants, which were cytotoxic to oligodendrocytes.

Increase in Syncytin-1 expression in astrocytes in the brain white matter of MS patients was accompanied by induction of the ER stress genes, OASIS, BiP, PERK, and

GADD153. Expression of OASIS in astrocytes induced iNOS and thus, nitric oxide.

ASCT-1, a neutral amino acid transporter and Syncytin-1 receptor, was suppressed in brain white matter astrocytes of MS patients and also in astrocytes expressing Syncytin-1 or OASIS. Nitric oxide enhanced the expression of the repressor transcription factor,

Egr1, which concurrently suppressed ASCT1. Syncytin-1 mediated neuroinflammation and death of oligodendrocytes with ensuing neurobehavioral deficits were prevented by the antioxidant ferulic acid in a mouse model of MS. TNF-α implantation into a novel

Syncytin-1 transgenic mice induced ER stress, loss of ASCT1 complemented by glial activation and T cell infiltration, indicating that astrocytes actively participate in MS pathogenesis. Thus, Syncytin-1’s proinflammatory properties in the nervous system demonstrate a novel pathogenic role for an endogenous -encoded protein, which may serve as a target for future therapeutic intervention.

iv

PREFACE

Some of the work presented in this thesis has been published previously. As

required by thesis guidelines, full citations of these articles and an account of division of

labor with all co-authors are listed below. Articles are listed in the order that they are

published. As a general note, all work presented in this thesis was performed by Joseph

Mathew Antony unless explicitly stated.

1. Antony JM, van Marle G, Opii W, Butterfield DA, Mallet F, Yong VW, Wallace

JL, Deacon RM, Warren K, Power C. Human

glycoprotein-mediated induction of redox reactants causes oligodendrocyte death

and demyelination. Nat Neurosci. 2004; 7 (10): 1088-95. Copyright permission

has been obtained from Nature Publishing Group (Appendix F)

This publication comprises all of Chapter 3. I performed the majority of the work

associated with this study. Dr. Guido van Marle originally designed the SINrep5-

EGFP and SINrep5-JRFL vectors that were used as controls in this study. Wycliffe

Opii and Dr. Allan Butterfield measured protein carbonyls and 4-HNE. Dr. Francois

Mallet provided the Syncytin-1 expressing vectors and monoclonal antibody against

Syncytin-1. Dr. Wee Yong guided and provided materials for the oligodendrocyte

assays. Dr. John Wallace guided and provided materials for anti-oxidant assays. Dr.

Robert Deacon’s protocols were used for designing animal behavior protocols. Dr.

Kenneth Warren supplied clinical samples from MS patients. All experiments were

performed in the laboratory of Dr. C. Power under his supervision.

v

2. Antony JM, Izad M, Bar-Or A, Warren A, Vodjgani M, Mallet F and Power C.

Quantitative analysis of Human Endogenous Retrovirus-W env in

neuroinflammatory diseases (AIDS Research and Human Retroviruses, in Press).

This publication comprises all of Chapter 4. I did the majority of the work

associated with this study. Dr. Maryam Izad assisted in the PCR assays. DNA was

obtained from an Iranian cohort of MS patients provided by Dr. Mohammed

Vodjgani. Dr. Amit Bar-Or (cDNA from a cohort of MS patients and controls) and

Dr. Kenneth Warren (CSF and plasma) provided additional clinical samples. Dr.

Francois Mallet provided the Syncytin-1 expressing vectors and monoclonal antibody

against Syncytin-1. All experiments were performed in the laboratory of Dr. C. Power

under his supervision.

vi

3. Antony JM, Ellestad K, Shariat N, Hammond R, Imaizumi K, Mallet F and Power

C. Syncytin-1 mediates Endoplasmic Reticulum Stress in a transgenic mouse

model of Multiple Sclerosis (Manuscript submitted to Journal of Clinical

Investigation).

This publication comprises all of Chapter 5. I performed the majority of the work

associated with this study. Mr. Kristofor Ellestad designed and optimized siRNA

molecules against Syncytin-1. Ms. Neda Shariat performed immunohistochemistry

for ER stress proteins from MS patients’ brain tissue provided by Dr. Robert

Hammond. Dr. Kazunori Imaizumi provided the OASIS construct and antibody. Dr.

Kenneth Warren provided clinical samples (CSF and plasma). Dr. Francois Mallet

provided the Syncytin-1 expressing vectors and monoclonal antibody against

Syncytin-1. All experiments were performed in the laboratory of Dr. C. Power under

his supervision.

vii

ACKNOWLEDGEMENTS

I would like to extend my deepest appreciation and gratitude to my supervisor,

Dr. Christopher Power for his support and encouragement throughout the course of my graduate study at the University of Calgary and later, at the University of Alberta. His confidence and trust enabled me to transcend from one country and area of expertise to another. Also I would like to thank members of my supervisory committee, Dr. Wee

Yong, Dr. Daniel Muruve and Dr. Robert Bell, who are a part of my career process and will continue to be. In addition, a number of faculty members from various Departments made my study exciting, for which I am very appreciative. In particular, I would like to thank Dr. Fabrizio Guilliani, Dr. Peter Dickie, Dr. Suzanne Grant (University of Alberta),

Dr. Guido van Marle, Dr. John Wallace, Dr. Joseph Goren and Dr. Kamala Patel

(University of Calgary). Also, I would like to thank my course coordinators, Dr. Julie

Deans and Dr. David Severson for explaining to me the vagaries of grant and assignment writing.

Laboratory life in Calgary was the most happiest one in my life and I owe this mainly to Claudia Silva, Shigeki Tsutsui, Shuhong Liu, Gareth Jones, Andrea Sullivan,

Robyn Flynn, Qing Tang, Yu Zhu, Guido van Marle, Julie Ethier, Scot Henry, Farshid

Noorbakhsh, Neda Shariat, David Vergote, Aundria Hood (the Calgary gang); Amir

Afkhami, Ramin Sarrami, Kris Ellestad, Nicola Barsby and Martine Ooms (the

BrainPowerLab at Edmonton), members of the Wee Lab (Tiffany, Tammy, Jennifer

Wells, Viktor, Rowena, Lorraine, Angelika, Yan Fan), Zochodne Lab (Cory Toth &

James Kennedy), Patel Lab (Subhadeep, Manprit, Cory, Evelyn, Vicky, Keith & Kamala) and numerous other friends at the Health Sciences Center whose names are not

viii

mentioned here. Graduate life would not have been a smooth without the excellent help

and advise from Belinda Ibrahim, Sherry Sweeney, Rosalie Kolstad, Dr Francine Smith,

Dr. Stephen Robbins and Christine Szefer for which I am forever grateful.

My stay in Calgary and Edmonton was indeed a joyous one-my thanks to the

family of friends- Jasprit, Jose Martinez, Elena Silva, Valentine, George and Annie, Dr.

Rajan George and Deepa.

Lastly, I would like to thank all the funding agencies for the financial support and

encouragement received-Alberta Heritage Foundation for Medical Research, Multiple

Sclerosis Society of Canada and Canadian Institutes for Health Research (CIHR)-

Integrated Health Research Team (IHRT).

ix

DEDICATION

To my wife, Smitha, for what she is to me,

AND

To my parents, for their unfailing commitment to my education

x

Table of Contents

Approval Page ii

Abstract iii

Preface v

Acknowledgements viii

Dedication x

Table of Contents xi

List of Tables xvi

List of Figures xvii

Appendices xx

List of Abbreviations xxi

Epigraph xxii

CHAPTER 1: INTRODUCTION AND LITERATURE REVIEW 1

I.1. Multiple Sclerosis (MS) Pathogenesis 2

I.1.1. Clinical and demographic features of MS 2

I.1.2. Pathophysiology of MS 5

I.1.3. Neuroinflammation 8

I.1.3.1. Cellular components of neuroinflammation 9

I.1.3.1.1.Leukocytes 13

I.1.3.1.1.1. CD4+ T cells 13 I.1.3.1.1.2. CD8+ T cells 14 I.1.3.1.1.3. B cells 14 I.1.3.1.1.4. Mast cells 14 I.1.3.1.1.5. Neutrophils 15 I.1.3.1.1.6. Dendritic cells 16

xi

I.1.3.1.2. Immunoregulatory cells in MS 17 I.1.3.1.3. Resident neural cells 18 I.1.3.1.3.1. Astrocytes 18 I.1.3.1.3.2. Oligodendrocytes 20 I.1.3.1.3.3. Neurons 20 I.1.3.1.3.4. Microglia 21 I.1.3.1.4. T cell-glia interaction 22

I.1.3.2. Molecular components of neuroinflammation 24 I.1.3.2.1. Cytokines 25 I.1.3.2.2. Chemokines 27 I.1.3.2.3. Neurotrophic factors and brain repair 28 I.1.3.2.4. Proteases 31 I.1.3.2.5. Oxidative stress 33 I.1.3.2.5. Nitric oxide (NO) 34 I.1.3.2.7. ER stress 37

I.1.4. Drawbacks of EAE as a model of MS 41 I.1.5. Genetics of MS 42 I.1.6. Environmental Factors in MS 45 I.1.6.1. Infectious agents in MS pathogenesis 45 I.1.6.2. Demyelinating 46 I.1.6.3. Retroviruses and MS pathogenesis 47

I.2. Retroviruses: Introduction 51 I.2.1. Genomic and structural organization 52 I.2.2. Classification 53 I.2.3. Retroviral Biology 57 I.2.4. Retroviral pathogenesis in the nervous system 61 I.2.4.1. Retrovirus and nitric oxide 63 I.2.4.2. Retrovirus-mediated ER stress 64 I.2.5. Exogenous retroviral pathogenesis 67 I.2.5.1. Type C retrovirus 67 I.2.5.1.1. Murine leukemia (MuLV) 68 I.2.5.1.2. HTLV 69 I.2.5.2. 72 I.2.6. Endogenous retroviral pathogenesis 79 I.2.6.1. Characteristics of endogenous retroviruses 79 I.2.6.2. HERVs and evolutionary advantages 83 I.2.6.3. Murine endogenous retroviruses 84 I.2.6.4. Syncytin-1: Characteristics of protein 88 I.2.6.5. Syncytin-1: Interaction with receptors 91

xii

CONCLUSION, STATEMENT OF HYPOTHESIS AND OBJECTIVES 91

CHAPTER II: MATERIALS AND METHODS 93

II.1. Cell culture 94

II.1.1. Primary cells 94 II.1.2. Cell lines 97 II.2. Syncytin-1 constructs 97

II.2.1. Construction of SINrep5-Syncytin-1 plasmid 97 II.2.2. Preparation and titration of viral stocks 98 II.2.3. Construction of pFGH- Syncytin-1-envelope plasmid 99 II.2.4. Construction of pseudotyped virus 101 II.2.5. Soluble Syncytin-1 protein expression 101 II.3. Transfections 101

II.4. Extraction of protein from CSF and plasma 102

II.5. Antioxidant activity 102

II.6. Infection and treatment of cells 103

II.6.1. Infection of cells 103 II.6.2. Treatment with cytokines 103 II.6.3. Treatment with drugs 103 II.7. Animals and in vivo procedures 104

II.7.1. Stereotaxic implantation with SINrep5-Syncytin-1 virus 104 II.7.2. Syncytin-1 transgenic mouse 104 II.7.3. Behavior studies 108 II.7.4. Oral drug treatment 109 II.8. PCR 109

II.8.1. Isolation of DNA and RNA and cDNA preparation 109 II.8.2. Relative quantification real time RT-PCR 110 II.8.3. Quantitative real time PCR 111 II.8.4. Sequencing of virus-cell junctions 112 II.9. In vitro assays 114

II.9.1. Toxicity of astrocyte conditioned medium 114 II.9.2. Analysis of supernatant for protein carbonyls and 4-HNE 114 II.10. Microarray Analysis 116

II.11. Quantitative Immunofluorescence 117

xiii

II.12. Detection of proteins in brain homogenate and cell cultures by western blot 117

II.13. Human tissue samples 118

II.14. Immunohistochemistry and immunocytochemistry 119

II.14.1. Detection of proteins in culture, human and mouse brain tissue sections 119 II.14.2. Double label staining immunohistochemistry and microscopy 120 II.14.3. Luxol fast blue staining for myelin 121 II.14.4. Syncytia formation in astrocytes 121 II.15. Quantification of cell numbers in vivo and in vitro 121

II.16. Statistical analyses 122

CHAPTER III. SYNCYTIN-1 INDUCES NEUROINFLAMMATION 123

III.1. Introduction 124

III.2. Results 124

III.2.1. Syncytin-1 is inducible and up-regulated in MS lesions 124 III.2.2. Syncytin-1 activates pro-inflammatory molecules in glial cells 132 III.2.3. Syncytin-1 causes oligodendrocyte damage and death 135 III.2.4. Anti-oxidants prevent Syncytin-1-induced oligodendrocyte injury 138 III.2.5. Syncytin-1-induced neuroinflammation and neurobehavioral abnormalities are inhibited by ferulic acid 142

III.3. Discussion 150

CHAPTER IV. QUANTIFICATION OF SYNCYTIN-1 153

IV.1. Introduction 154

IV.2. Results 155

IV.2.1. Syncytin-1 expression is cell-and tissue type specific 155 IV.2.2. Minocycline inhibits Syncytin-1 expression 158 IV.2.3. Syncytin-1-RNA and DNA copy numbers are significantly enhanced in brain of MS patients relative to controls 158 IV.2.4. Increased Syncytin-1 DNA copies reflect un-integrated cDNA 161 IV.3. Discussion 164

xiv

CHAPTER V. SYNCYTIN-1 INDUCES ER STRESS 167

V.1. Introduction 168 V.2. Results 171 V.2.1. Syncytin-1 induces ER stress in astrocytes 171 V.2.2. OASIS down-regulates ASCT1 expression in astrocytes 174 V.2.3. Syncytin-1 diminishes oligodendrocyte viability 179 V.2.4. iNOS and Egr1 suppress ASCT1 in astrocytes 181 V.2.5. Soluble Syncytin-1 down-regulates ASCT1 in astrocytes 183 V.2.6. Syncytin-1 transgenic mice exhibit neuroinflammation 188 V.2.7. Syncytin-1 Tg animals show ER stress 193

V.3. Discussion 199

CHAPTER VI. GENERAL DISCUSSION AND CONCLUSIONS 205

VI.1. Overview 206 VI.2. Syncytin vs MSRV 206 VI.3. Syncytin-1-associated neuroinflammation 207 VI.3.1. Syncytin-1 mediates inflammation through nitric oxide intermediates 208 VI.4. Quantification of Syncytin-1 copy numbers in tissues 209 VI.4.1. Syncytin-1 RNA copy numbers in the brain 209 VI.4.2. Syncytin-1 RNA copy numbers in CSF and plasma 211 VI.4.3. Syncytin-1 DNA copy numbers and integration events 212

VI.5. Syncytin-1 induces ER stress in astrocytes 215 VI.6. Syncytin-1 causes astrocyte dysfunction 216 VI.7. Syncytin-1 indirectly regulates its receptor expression 217 VI.8. Conclusions and Future Perspectives 219

REFERENCES 227

xv

LIST OF TABLES

Table 1. Therapeutics employed in EAE 43

Table 2. Classification of HERVs in the genome 58

Table 3. List of oligonucleotide primers used in this study 106

Table 4. Syncytin-1 immunoreactivity in MS patients 130

Table 5. Stereological count in mice brains 146

Table 6. Microarray analysis of astrocytes expressing Syncytin-1- (MS related) 177

Table 7. Microarray analysis of astrocytes expressing Syncytin-1 178

xvi

LIST OF FIGURES

Figure 1. Clinical progression of Multiple Sclerosis 4

Figure 2. Multi-step leukocyte recruitment cascade 11

Figure 3. The endoplasmic reticulum stress response 39

Figure 4. Schematic of a retrovirus 54

Figure 5. Schematic of a provirus 55

Figure 6. Schematic of retroviral classification 56

Figure 7. Life cycle of exogenous and endogenous retrovirus 59

Figure 8. Various forms of HERVs 81

Figure 9. Pathogenic potential of HERVs 82

Figure 10. Phylogenetic tree of endogenous retroviruses 87

Figure 11. Northern blot analysis of Syncytin-1 expression 90

Figure 12. Sindbis virus vector expressing Syncytin-1 100

Figure 13. Stereotaxic implantation of Sindbis virus vector 105

Figure 14. Schematic of retroviral LTR circles and formation of provirus 113

Figure 15. PCR amplification of virus-host cell junctions 115

Figure 16. Increase in Syncytin-1 mRNA in brain of MS patients 126

Figure 17. Western blot analysis of Syncytin-1 expression in MS brains 127

Figure 18. Immunohistochemical analysis of Syncytin-1 expression in brain 128

Figure 19. Mitogen-induced expression of HERV env mRNA 131

Figure 20. Overexpression of Syncytin-1 133

Figure 21. Inflammatory components of Syncytin-1 expression 134

Figure 22. Syncytin-1-mediated damage and death of human oligodendrocytes 136

xvii

Figure 23. Syncytin-1-mediated damage and death of rat oligodendrocytes 137

Figure 24. Syncytin-1 does not affect neuronal viability 139

Figure 25. Anti-oxidant activities of various drugs 140

Figure 26. Abrogation of oligodendrocyte death by anti-oxidants 141

Figure 27. Effect of various drugs on oligodendrocyte protection 143

Figure 28. Syncytin-1 induces neuroinflammation in mice 144

Figure 29. Syncytin-1 induces neurobehavioral deficits in mice 147

Figure 30. Syncytin-1 does not affect neurons 149

Figure 31. Syncytin-1 mRNA is increased in a cell-type specific manner 156

Figure 32. Transcript abundance and effect of mitogen on Syncytin-1 mRNA 157

Figure 33. Minocycline-mediated suppression of Syncytin-1 expression 159

Figure 34. Syncytin-1 RNA copy numbers in brain, CSF and plasma 160

Figure 35. Syncytin-1 DNA copy numbers in brain 162

Figure 36. Detection of circular DNA and provirus integration in brain DNA 163

Figure 37. Syncytin-1 induces ER stress in astrocytes 172

Figure 38. ER stress proteins are induced in MS 173

Figure 39. ASCT1 expression is suppressed in MS 175

Figure 40. Syncytin-1-induced ASCT1 suppression affects oligodendrocyte 180

Figure 41. Syncytin-1 and NO induce Egr1 182

Figure 42. NO regulates ASCT1 expression 184

Figure 43. Expression of functional Syncytin-1 185

Figure 44. Soluble Syncytin-1 affects ASCT1 187

Figure 45. Inhibition of iNOS and minocycline up-regulates ASCT1 expression 189

xviii

Figure 46. Development of Syncytin-1 transgenic mouse 190

Figure 47. TNF-α induces Syncytin-1 in astrocytes 191

Figure 48. TNF-α induces Syncytin-1 in transgenic mouse 192

Figure 49. TNF-α induces neuroinflammation in Syncytin-1 transgenic mouse 194

Figure 50. TNF-α induces ER stress in Syncytin-1 transgenic mouse 195

Figure 51. Syncytin-1 transgenic mouse exhibits loss of ASCT1 197

Figure 52. Model of Syncytin-1-mediated neuropathogenesis 204

xix

APPENDICES 220

APPENDIX A: SINrep5-Syncytin-1 vector 221

APPENDIX B: Quality of DNA extracted from brain tissue 222

APPENDIX C: Affymetrix GeneChip Expression Analysis 223

APPENDIX D: Autopsied samples from patients 224

APPENDIX E: Calculation of Syncytin-1 DNA copy number in the genome 225

APPENDIX F: Copyrights obtained from Publishing Houses for use of materials 226

xx

LIST OF ABBREVIATIONS

APC: Antigen presenting cells BBB: Blood brain barrier BDNF: Brain-derived neurotrophic factor BiP: immunoglobulin heavy chain-binding protein Ca2+: Calcium CD: Cluster of differentiation CNS: Central nervous system CO2: carbon dioxide CSF: cerebrospinal fluid DNA: Deoxyribonucleic acid EAAT: Excitatory amino acid transporter EDSS: Expanded Disability Status Scale ER: endoplasmic reticulum GADD153/CHOP: growth arrest- and DNA damage-inducible gene/C/EBP homologous protein gp: glycoprotein GPCR: G-protein coupled receptor Grp: glucose regulated protein HAD: HIV-associated dementia HERV: Human endogenous retrovirus HLA: human leukocyte antigen 4-HNE: 4-hydroxy-2,3-nonenal IGF-1: Insulin-like growth factor-1 IL: Interleukin kbp: kilo basepair kDa: kilo Dalton LTR: Long terminal repeats MAG: myelin-associated glycoprotein Mg2+: Magnesium MHV: Mouse hepatitis virus MIAME: Minimal Information About a Microarray Experiment MMP: Matrix metalloproteinases MOG: myelin oligodendrocyte glycoprotein MSRV Multiple sclerosis retrovirus NF-κB: nuclear factor kappa B NOS: nitric oxide synthase ONOO−: peroxynitrite PERK: PKR (pancreatic eIF2α [eukaryotic translation initiation factor2, α subunit] kinase-like ER protein kinase PLP: Proteolipid protein PP-MS: Primary Progressive Multiple Sclerosis Rag: Recombinase activating gene RANTES: Regulated upon activation, normal T cell expressed and secreted RBC: Red blood cells

xxi

RNA: Ribonucleic acid ROS: Reactive oxygen species RR-MS: Relapsing remitting Multiple Sclerosis RT-PCR: reverse transcription-polymerase chain reaction TMEV: Theiler’s Murine Encephalitis Virus TNFα: Tumor necrosis factor-alpha

xxii

Nothing in biology makes sense except in the light of evolution

Theodosius Dobzhansky, 1973 (in The American Biology Teacher)

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 1

CHAPTER 1

INTRODUCTION AND LITERATURE REVIEW

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 2

I.1. Multiple Sclerosis (MS) pathogenesis

I.1.1. Clinical and demographic features of MS

MS is a medically important disorder in Canada that afflicts the central

nervous system (CNS) of predominantly young people beyond the age of puberty.

The overall estimate of Canadian MS prevalence is 240 per 100,000, ranging from

180 in Quebec to 350 per 100,000 in Atlantic Canada 1. The overall regional

prevalence rate in the province of Alberta, Canada stands at 340 per 100,000 1. It is a

heterogeneous disorder characterized by repeated unpredictable bouts of motor

disturbances, partial paralysis, sensory abnormalities and/or visual impairment. These

variable signs and symptoms result from inflammatory processes that selectively

attacks and destroys oligodendrocytes, the cells that form the myelin sheaths around

axons in the brain and spinal cord 2. The diversity of the disease pathology and

unknown etiology have made MS among the most damaging of neurological

disorders and the least understood in terms of mechanisms of disease progression.

Indeed, some investigations have speculated that MS is not a single disease entity but

actually represents a spectrum of neuroinflammatory disorders 3. Nevertheless,

significant strides in the recent past have made a dent into unraveling the pathogenic

features of the disease. Notable among these are the use of magnetic resonance

imaging (MRI), immunotherapy using monoclonal antibodies to integrins, drugs such

as glatiramer acetate that shifts the immune response from TH (helper)-1 to TH2,

interferon (IFN)-β and minocycline 4. Exposure to viral, bacterial or other pathogens

may trigger the disease process, perhaps through a molecular mimicry mechanism

where a protein in the pathogen is similar to the host protein, myelin, eliciting an

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 3

autoimmune response 5. Several years of research in MS have implicated activated T

lymphocytes and microglia/macrophages, which can produce cytotoxic cytokines and

reactive oxygen molecules, in the destruction of oligodendrocytes 6.

MS is common among Caucasians, with 0.05-0.15% affected by this chronic

and disabling disorder of the CNS, and is less frequently observed in Asians or

Africans 7. Family members of MS patients inherit a higher risk of developing MS,

arguing for a strong genetic predisposition to this disease. MS usually begins in early

adulthood and affects women more frequently than men. MS usually starts with a

relapsing-remitting course (RR-MS) but some 20% of the cases are defined by a

primary progressive course (PP-MS) without acute relapses. Clinical and

neuropathological features are variable depending on population ethnicity. Among the

aboriginal Manitoba Cree Indians in Canada, a distinct form called neuromyelitis

optica predominates and is common among Asian populations 8. Lesions in RR-MS

patients are usually found in white matter and are characterized by disruption of the

blood-brain barrier (BBB), local edema and demyelination, typical of inflammatory

processes (Fig. 1). In PP-MS, inflammatory processes are less dominant but

progression to disability and brain atrophy evolves faster 9. Understanding the

sequence of events underlying the development of the inflammatory plaque is a

central mission in MS research. There is abundant evidence to support the hypothesis

that genetics has an important role in an individual’s vulnerability to MS, perhaps in

conjunction with trigger factors. Though various infectious agents are linked to MS,

their presence may simply provide an appropriate environment for development of

autoreactive immune response 10 directed against CNS antigens 11. Several viral

Chapter 1: Introduction and Literature Review

Preclinical Relapsing-Remitting MS Secondary Progressive MS

Brain volume

EDSS

Lesion load MRI-T1

Inflammation Demyelination/Axonal loss D Pathology

Fig. 1. Schematic representation of clinical progression of MS by clinical scale (EDSS), frequency of inflammatory events (MRI, A), lesion load (tissue damage) and brain atrophy (brain volume). Inflammation is characterized by perivascular inflammation with mononuclear cells (B), demyelinated regions (C) and axonal injury (D) (Adapted from Sospedra et al., 2005; obtained permission from NEJM and Annual Review of Immunology, See Appendix E)

4 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 5

agents including measles virus, Para influenza virus, canine distemper virus, Epstein-

Barr virus (EBV), human herpes virus (HHV)-6 and retroviruses (eg. MS retrovirus

[MSRV]) have been implicated in demyelination processes 12. I will focus this thesis

on the contribution of a human endogenous retrovirus (HERV)-encoded

envelope protein, Syncytin-1, in the pathogenesis of MS.

I.1.2. Pathophysiology of MS

The disease process in MS is predominantly located in the myelinated

regions of the brain and spinal cord. In MS-affected tissue, inflammation-induced loss

of oligodendrocytes and axonal injury are key features of the pathology 13. MS

pathogenesis involves demyelination and inflammation comprising B cells, T cells,

macrophage/microglia and astrocytes, 9 signified by plaque formation. Loss of

oligodendrocytes and neurons, astrogliosis and remyelination, leading to tissue

damage accompanies inflammatory changes. About 26% of all lesions in the cerebral

hemispheres lie outside the white matter, 5% in the cortex and 17% in the cortical

boundary 14. In this respect, a novel experimental autoimmune encephalomyelitis

(EAE) rat model of MS demonstrating cortical demyelination has been recently

established 15. Though active plaques from the same patient may be similar, histology

reveals evidence for significant heterogeneity in demyelination patterns 16. Lesions

are thus classified into 4 categories of myelin destruction 17.

Type I pattern: Toxic products of activated macrophages (eg. TNF-α, reactive

oxygen species [ROS]) destroy myelin sheath. (Also observed in EAE).

Type II pattern: Antibody-mediated demyelination induced by cooperation between

encephalitogenic T cells that induce inflammation and demyelinating anti-myelin

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 6

oligodendrocyte glycoprotein (MOG) antibodies and also complement proteins. (Also

observed in EAE).

Type III pattern: Oligodendrogliopathy-associated demyelination, commonly found

in virus-induced white matter disease in humans. Antibodies and complement

proteins are absent without any remyelination. (Not observed in EAE).

Type IV pattern: Primary oligodendrocyte degeneration and apoptosis observed.

Restricted to infrequent cases in primary-progressive disease patients. (Not observed

in EAE).

Axonal injury in MS is correlated with the extent of inflammation within

the CNS. Nevertheless, axonal damage is variable and depends on the severity of

inflammation, induction of pathogenetic mechanisms as well as diversity in host

susceptibility. Demyelination is often accompanied by significant neuronal death in

cortical and thalamic MS lesions and also death of retinal ganglion neurons 18. MRI

studies have revealed that progressive and functional deficits can be associated with

axonal loss within lesions and brain atrophy 5 (Fig. 1). Axonal injury due to toxic

products, including nitric oxide (NO) and proteases, released from macrophages,

astrocytes and major histocompatibility complex (MHC) class I-restricted T cells, is

found in active lesions during early phases of the disease 5. CD4+- and γ/δ- T cells are

pathogenic in MS lesions, but whether sufficient T cells are present in MS lesions to

induce axonal damage and neuronal death is not known. Anti-CD3 activated allogenic

and syngenic CD4+ and CD8+ T lymphocytes were found to be toxic to neurons

through a contact-dependent pathway, perhaps mediated by FasL, CD40 and LFA-1

19. Axonal loss is also observed in chronic inactive demyelinated plaques but not in

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 7

remyelinated lesions, suggesting that compromised trophic support by damaged

oligodendrocytes and astrocytes can make axons prone to destruction.

Permanent physical and cognitive disability results from cumulative axon

loss 20. Defects in axonal transport can lead to accumulation of axonal amyloid

precursor protein (APP). Using APP as a marker of early axonal damage in MS

lesions, Ferguson et al., (1997) revealed that APP is expressed in areas of acute

demyelination and inflammation, but not in chronic areas of the lesions 21. Earlier

studies indicated that in acute MS lesions, APP was detected in T cells, foamy

macrophages, activated astrocytes and microglia; also, chronic lesions displayed

APP-positive astrocytes and demyelinated axons 22. Another marker, non-

phosphorylated neurofilament (SMI-32) is abundant in neuronal cell bodies and

dendrites but is heavily phosphorylated in healthy myelinated axons. An extensive

analysis of axonal damage in 47 demyelinated lesions revealed ovoids exhibiting

intense SMI-32 immunostaining 23. Axonal ovoids are transient structures containing

axonal debris and surrounding myelin and are characteristic of degenerating axons.

Proteolytic enzymes, cytokines, oxidative products and free radicals released by

activated immune and glial cells, cause axonal transection. Axonal injury is also

correlated with reduced N-acetyl aspartate levels in MRI studies 23. Thus, axonal

injury in MS is a well-established clinical feature. Having described the disease in the

preceding sections, I will now attempt to elaborate on the CNS disease mechanism

(neuropathogenesis) and the neuroinflammatory components in the context of MS.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 8

I.1.3. Neuroinflammation

The CNS consists of the brain and spinal cord and is the main centre of

correlation and integration of nervous information. The brain lies in the cranial cavity

and is continuous with the spinal cord through the foramen magnum while the spinal

cord is situated within the vertebral canal of the vertebral column. Three layers of

meninges-the dura mater, the arachnoid mater and the pia mater surround both the

brain and spinal cord. The cerebrospinal fluid (CSF), synthesized in the ventricular

choroid plexus, surrounds the spinal cord and brain in the subarachnoid space 24. The

CNS is composed of large numbers of neurons and their processes (axons or nerve

fibers), which are supported by glia. The grey matter in the CNS consists of mainly

neuronal soma while the white matter consists of axons embedded in neuroglia. The

average adult human brain weighs 1300-1400 grams and consists of 100 billion

neurons. The average number of glial cells is 10-50 times the number of neurons 25.

The CNS consists of several groups of highly differentiated and complex

cells that are functionally integrated by cell-to-cell linkages and synapses. A healthy

brain enjoys an immune privileged status in that it lacks resident Natural Killer (NK)

cells, T and B-lymphocytes, lymphatic system and limited ability for capillary

endothelium to bind leukocytes. Expression of MHC class I and II is low on

neuroectodermal cells but microglia 26 (originating from mesenchymal macrophages

and monocytes during development) express MHC class I and II 27. Neurons express

high levels of MHC class I after axotomy 28 and cytokine treatment 29. The BBB is a

tight anatomic barrier that excludes proteins and cells of the blood from entering the

CNS under normal conditions 30. It consists of tight junctions between mitochondria-

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 9

rich endothelial cells in CNS vessels, basal lamina and a continuous covering

provided by extended foot processes derived from astrocytes and microglia. The BBB

has evolved to maintain homeostatic differences in the constituents of the blood and

CSF, and acts as a selective barrier to keep the ionic concentrations of the internal

environment within a narrow physiological range 31. Though the BBB separates the

CNS from circulating lymphocytes and antibodies, the circumventricular organs

allow free interaction of peripheral blood with components of the CNS 32. The

choroid plexus is a highly vascularized area containing fenestrated endothelial cells

unlike the parenchymal endothelial cells associated with a tight BBB 33.

I.1.3.1. Cellular components of neuroinflammation

The generation and selection of a diverse population of immunocompetent

but naive lymphocytes from a large number of precursor cells in the primary

lymphoid organs (PLO) and the efficient initiation of an immune response by

immunocompetent cells on antigen capture in the secondary lymphoid organs (SLO)

lead to antigen recognition and response while maintaining tolerance to self-antigens.

Lymphocyte homing and trafficking link the PLO and SLO with each other and with

the extralymphoid sites of the body 34. It is now increasingly appreciated that the CNS

is not immunologically isolated from the rest of the body, though there are

fundamental differences in the nature of the induced innate and adaptive responses.

In the event of an immune response to an antigen in the CNS,

immunological cues are sent from the CNS to the periphery for recruitment of

leukocytes to eliminate the antigen, remove debris and promote repair. However,

eradication of the antigen from the CNS leads to immunological damage, which may

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 10

be beyond repair and often fatal. Infiltration of effector lymphocytes through the BBB

and their interaction with the CNS components contribute to pathology associated

with CNS diseases. The entry of leukocytes into the CNS is generally restricted by

the tight junctions and by the lack of adhesion molecules on cerebral capillary

endothelial cells 35. However, studies have also shown that activated T cells regularly

transmigrate across the BBB as part of the normal immunological surveillance of all

tissues. Transendothelial migration of activated CD4+ T cells takes place due to

interaction of P-selectin glycoprotein ligands expressed by activated T cells with P-

selectin, which is expressed at low levels by normal cerebrovascular endothelial cells,

but levels of both these molecules increase during an inflammatory response,

increasing surveillance and migration of activated T cells 35,36.

Activated T cells can cross antigen- or cytokine-stimulated endothelial

cells of the BBB, independent of antigen specificity. The general mechanism (Fig. 2)

consists of 1. Tethering and rolling- Circulating leukocytes first tether to and then

roll on endothelial cells expressing adhesion molecules. The tethering and rolling

steps are mediated by the selectins, as well as α4 integrins. Selectins on endothelium

interact with carbohydrate-counter receptors on leukocyte surface, tethering

leukocytes that slow down and roll along the endothelium even in the presence of

shear forces that guide movement of leukocytes through the bloodstream. 2. Integrin

activation: Signaling through Gαi proteins activates integrins expressed on

leukocytes. 3. Leukocyte arrest: Firm adhesion occurs when integrins bind to their

endothelial ligands with high affinity. 4. Diapedesis: Chemokines, acting through G-

protein coupled receptors (GPCRs), mediate leukocyte transmigration into tissue 37.

Chapter 1: Introduction and Literature Review

Selectin Chemokines Integrins Chemokines Carbohydrate ligands GPCRs Adhesion molecules GPCRs of immunoglobulin superfamily

Tethering/Rolling Activation Adhesion Diapedesis

Fig. 2: Multi-step paradigm of leukocyte trafficking. Each step in recruitment can be mediated by different adhesion molecules. By combining these elements in unique ways, specific traffic of leukocytes into sites of inflammation can be achieved. Use of multiple chemoattractants increase the degree of specificity (Adapted from Ransohoff et al., 2003)

11 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 12

The importance of selectins and integrins in leukocyte trafficking through

the CNS vasculature is best elucidated in EAE mice. Though MOG-induced mice

express limited levels of P-selectin in the CNS vasculature, leukocyte rolling is

inhibited by blocking this selectin, suggesting its requirement for initial recruitment

of encephalotigenic T cells. Therapeutically, dual inhibition of α4-integrin and P-

selectin may have a synergistic effect and provide optimal benefit in human disease

38 . Blocking α4β1 integrin leads to reduced infiltration of leukocytes into the CNS

perhaps by affecting fibronectin-mediated leukocyte migration 39. Commercially

available and therapeutically viable antagonists have been derived against α4β1

integrin for treatment of MS with high therapeutic efficiency 40. However, a potential

setback to this treatment protocol was the unexpected development of progressive

multifocal leucoencephalopathy (PML). PML is a demyelinating disease caused by

the human polyomavirus JC virus, a common and widespread infection in humans

and was diagnosed in at least 3 patients who were treated with Natalizumab, a

41 recombinant humanized antibody directed to the α4 integrins (α4β1 and α4β7) . An

extensive study revealed risk of PML in approximately 1 in 1000 patients treated with

Natalizumab for 18 months, though longer treatments and the risks thereof are

unknown 42.

During an inflammatory response, leukocyte recruitment to the site of

inflammation is mediated by chemokines and is also affected by complement,

leukotrienes, prostaglandin, neuropeptides and cytokines 43. Unlike cytokines that

have pleiotropic effects, chemokines target specific leukocyte subsets. Expression of

adhesion molecules on leukocytes and endothelial cells, and chemotaxis lead to

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 13

infiltration by neutrophils and macrophages followed by chemokine-receptor

expressing lymphocytes.

I.1.3.1.1. Leukocytes

In MS, infiltrating activated T and B-lymphocytes, macrophages and

microglia induce proinflammatory cytokines and a host of other soluble factors

leading to inflammation-induced axonal and myelin damage. Oligodendrocytes

undergo damage and perhaps death through the action of several components of the

innate and adaptive immune systems in the CNS 44.

I.1.3.1.1.1. CD4+ T cells

Current experimental evidence in MS research favours the CD4+

autoreactive T cell as a crucial factor for MS pathogenesis for several reasons. These

cells are among the majority infiltrating the CNS and CSF; genetic risk to MS is

contributed by human leukocyte antigen (HLA)-DR and -DQ molecules, which when

transgenically expressed confer susceptibility to EAE; altered peptide ligands of

+ myelin basic protein (MBP)83-99 induce cross-reactive CD4 T cells exacerbating

disease in clinical trials and lastly, CD4+ T cells induce antibody production and

CD8+ maturation 5. Tissue damage caused by the initial infiltration of autoreactive T

cells release myelin antigens that promote immune responses to additional myelin

epitopes, initiating a cascade of events that culminates in chronic disease. Myelin-

reactive T cells from MS patients produce TH1-cytokines (IL-12, IL-23, IL-17, IFN-

β) whereas the same cells from healthy individuals are likely to produce TH2

cytokines (IL-4, IL-10) 11.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 14

I.1.3.1.1.2. CD8+ T cells

In the CNS, only microglia are known to express MHC class II 6. Though

astrocytes express MHC class II after IFN-γ treatment, the constitutive levels of MHC

class II are either absent or at low levels on oligodendrocytes, neurons and astrocytes.

MHC class I expression on the resident neural cells make them vulnerable to

recognition by CD8+ cells. While it has been generally accepted that CD4+ T are the

dominant encephalotigenic cells in the brain 45, there are also experimental and

clinical data showing that CD8+ T cells outnumber CD4+ T cells in MS lesions 45.

CD8+ cells are also closely apposed to oligodendrocytes and axons suggesting that

they may initiate inflammation and tissue injury in MS plaques 45.

I.1.3.1.1.3. B cells

Circulating activated memory B cells can serve as antigen presenting cells

(APCs) and skew T cell responses towards a pro-inflammatory nature. Certain B cell

clones from the CSF of MS patients have revealed receptor editing by which these

cells abrogate the ability of the host to induce autoantibodies during B cell

development. However, reactivity may be introduced to additional CNS autoantigens

46. MS patients demonstrate increased immunoglobulins in the CSF, but not in the

serum, suggesting that these myelin-specific antibodies which may contribute to

destruction of myelin 5 are produced locally in the CNS.

I.1.3.1.1.4. Mast cells

Mast cells arise from CD34+/c-kit+/CD13+/FcεRI- hematopoietic

progenitor cells that differentiate in the presence of stem cell factor, a c-kit ligand,

which in the CNS is synthesized by glial cells. These effector cells of the innate

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 15

immune system are not abundant in the brain, but an increase in their numbers is

associated with a variety of neurological disorders including MS 47. Degranulated

mast cells in the brains of EAE animals release histamines, which alter BBB

permeability 48. Moreover, mast cell-deficient mice (W/Wv) exhibited significantly

reduced disease onset and clinical scores in a MOG-induced model of EAE 49. In

addition to increased numbers of mast cells in plaques, elevated levels of tryptase 50

and histamine 51 in the CSF of MS patients suggest that mast cells may play an

important role in the pathogenesis. This is also important because tryptase activates

proteinase-activated receptor [PAR-2], which is involved in MS pathogenesis

(discussed later). The increase in mast cell numbers is thought to be due to

chemoattractants such as RANTES (regulated upon activation, normal T cell

expressed and secreted) released by inflammatory cells, and hence could be a

secondary event 52.

I.1.3.1.1.5. Neutrophils

Recruitment of neutrophils from the blood into the injured tissue is a

typical inflammatory response and is mostly mediated by CXCL8 (IL-8), which, in

addition to neutrophils, is also produced by a wide variety of other cells 53. The

chemotactic effects of CXCL8 are mediated by binding to its receptors CXCR1 and

CXCR2, both of which are highly expressed on glial cells in acute and chronic brain

lesions of MS patients. However, both neutrophils and eosinophils are rarely

observed in MS lesions, except in cases of fulminant variations of MS such as

Devic’s or Marburg’s syndromes 54. The high levels of CXCL8 in the peripheral

blood of MS patients therefore suggests that high serum CXCL8 levels may be

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 16

preventing the neutrophils from transmigrating into the brain. Nevertheless,

neutrophils may have increased activity prior to the clinical phase of MS, where it

may promote the production and synthesis of matrix metalloproteinases (MMPs),

which play a role in tissue inflammation in MS 55.

I.1.3.1.1.6. Dendritic cells (DCs)

Dendritic cells (DCs) are highly specialized APCs and act as sentinels of

the innate immune system and, due to their intrinsic ability to produce cytokines and

stimulate NK and naïve T cells, DCs link the innate and adaptive arms of the immune

system 56. Under non-inflammatory conditions, the brain parenchyma lacks DCs but

high numbers in the brain and CSF are seen following inflammatory conditions. They

originate from the choroid plexus and meninges or arise from a subpopulation of

activated microglia 35 after antigen uptake. DCs secrete antiviral IFN-γ and also

stimulate the adaptive immune system 57. CD4+ CD11c- type 2 DC precursor cells are

similar to systemic type 1 IFN producing cells. These precursors generate large

quantities of IFN-α/β and TNF-α, which, through an autocrine fashion, stimulate

these cells to mature into DCs that can stimulate adaptive immunity 58. DCs

accumulate in the CNS and CSF in MS and EAE and are detected using the marker

DC-SIGN/CD209, a C-type lectin receptor expressed by immature DCs in non-

lymphoid tissue, but also expressed by mature DCs in secondary lymphoid tissue 59.

DCs have been shown to engulf myelin components and interact predominantly with

CD8+ T lymphocytes in MS lesions. By virtue of chemokines and cytokines released,

DCs mature and activate T lymphocytes in the CNS and also migrate to draining

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 17

lymph nodes where they initiate a new wave of autoreactive T cells to infiltrate the

CNS 59.

I.1.3.1.2. Immunoregulatory cells in MS

CD4+ immunoregulatory T cells (Tregs) express high levels of CD25 and

the transcription factor, Forkhead Box-P3 (FOXP3) 60. These cells suppress T cell

proliferation by both cell-cell contact and cytokine-mediated mechanisms,

particularly producing anti-inflammatory cytokines IL-10, transforming growth factor

(TGF)-β and IL-4 60. Since MS patients show reduced numbers of Tregs, the

subsequent lack of IL-10, TGF-β and IL-4 may further contribute to the pathogenesis.

In an interesting study, Tregs were isolated from MS patients who were vaccinated

with irradiated MBP-reactive T cells. Isolated Tregs were found to reduce the

proliferative response of autologous MBP-reactive T cells 61. These data emphasize

the relevance of Tregs in MS and efforts should be made to harness the major benefit

of Tregs, namely to suppress encephalitogenic T effector cells.

NK cells may also exert an immunoregulatory role in MS by targeted lysis

of encephalotigenic cells through perforin and/or TNF-related apoptosis-inducing

ligand (TRAIL)-dependent mechanisms. Natural Killer (NK)-T cells are another

group of immunoregulatory cells that act through induction of IL-4 and IFN-γ, the

former cytokine being selectively up-regulated by binding the CD1d protein on NKT

cells 5. In humans, NKT cells express a conserved T cell receptor (TCR)α chain,

Vα24JαQ, paired with a selected Vβ11 segment and, in MS, there is a considerable

decrease in the number of these immunoregulatory cells 62.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 18

I.1.3.1.3. Resident neural cells

I.1.3.1.3.1. Astrocytes

There are 3 types of glial cells in the mature CNS-astrocytes,

oligodendrocytes and microglia. Astrocytes are the most abundant cells in the CNS

and are restricted to the brain and spinal cord and have elaborate star-like processes.

There are 2 types of astrocytes in the CNS. The fibrous type with many thin processes

forms a scaffold throughout the grey matter, while the protoplasmic type is short,

with thicker processes and form a continuous covering around blood vessels and both

respond to injury 63. Camillio Golgi’s observations at the end of the 19th century that

astrocytes connect blood vessels and neurons set the precedence for investigations

into the complex roles played by astrocytes 64. Astrocytes maintain the appropriate

chemical environment for neuronal signaling. There are several conditions involving

defective astrocytes, chief among which is the white matter disorder, Alexander’s

disease, characterized by the accumulation of Rosenthal fibers, which are protein

aggregates in astrocytes due to mutations in the glial fibrillary acidic protein (GFAP)

gene. The astrocytes also contain αB-crystallin and heat shock protein (Hsp)-27 65.

Mice that lack GFAP are viable but exhibit morphological and functional alterations

and decreased myelination, suggesting a link between astrocyte function and

maintenance of myelination 66. Astrocytes produce soluble factors, particularly

growth factors, that promote functioning of otherwise defective oligodendrocytes 67,

suggesting extensive cross-talk between astrocytes and oligodendrocytes. Evidence

for a pathogenic role of astrocytes in MS is gaining attention, which is the theme

of my thesis. Activated astrocytes exhibited complement activation in pre-

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 19

demyelinating lesions from MS brains 68, release glutamate leading to neuronal and

oligodendrocyte death 69, express chemokines that lure dendritic cells to sample the

antigens in the CNS 70 and a host of other functions that enhance neuropathogenesis

of demyelination. Astrocyte can also secrete serine and glutamate 71. Glutamate

produced by active neurons is removed by astrocytes through glia-specific

transporters-excitatory amino acid transporter 1 (EAAT1, [rodent, GLAST]) and

EAAT2 [rodent, GLT1]. Astrocyte gap junctions are formed by connexins 43 and 30,

through which these glial cells dissipate glutamate, in addition to propagation of the

intercellular Ca2+ waves. In response to CNS injury, astrocytes become activated,

involving cellular hypertrophy, changes in gene expression and cellular proliferation

forming the classical ‘scar tissue’. However the scar-forming reactive astrocytes are

essential for spatial and temporal regulation of inflammation post CNS injury. Loss of

astrocytes leads to accumulation of extracellular glutamate with subsequent

excitotoxicity of neurons and oligodendrocytes 72. Reactive astrocytes produce

cytotoxins such as NO radicals and ROS that can damage neural cells and contribute

to secondary degeneration after CNS insults 73 and can also inhibit axonal

regeneration 74. Thus astrocytes are able to exert harmful and beneficial effects. It

seems that evolutionary adaptations favoured mechanisms to contain local injuries

rather than those to deal with large injuries. Restricting the injury within insulated

local regions allows robust leukocyte- and microglia-mediated inflammatory

reactions to occur. This allows the damaged tissue to recover and also prevent these

inflammatory cells from invading adjacent healthy tissue. However, during this

process axonal guidance pathways are perturbed and can slow down remyelination74.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 20

I.1.3.1.3.2. Oligodendrocytes

Oligodendrocytes and astrocytes are both cells of neuroectodermal origin

75 like neurons. Oligodendrocytes are restricted to the CNS and form a layer of lipid-

rich and laminated layer called myelin around most axons. Myelin plays an important

role in the conduction of action potentials. In MS, myelin degenerates and

oligodendrocyte die causing a functional short-circuiting of exposed axonal

processes, leading to demyelination 76. Mice deficient in 2’3’ cyclic nucleotide

phosphodiesterase (CNPase), a gene expressed in oligodendrocytes, exhibit motor

deficits due to axonal damage even though normal-looking myelin is formed 77,

suggesting that it could be altered trophic support from oligodendrocytes that

contribute to axonal damage rather than demyelination that leads to axonal damage 14.

The NG2 glycoprotein is a membrane protein expressed in the developing and adult

CNS by subpopulations of glia including oligodendroglial precursor cells (OPCs).

NG2-positive cells are observed in MS lesions and, since they are precursors to

oligodendrocytes during development, the presence of NG2-positive cells in MS

lesions may suggest the possibility of on-going remyelination 78.

I.1.3.1.3.3. Neurons

The neuronal population is established shortly after birth and mature

neurons do not divide. They can be excitatory, inhibitory or modulatory in their effect

and motor, secretory or sensory in their function 79. Neurons range from the small,

globular granule cells with a diameter of 6-8 μm to the pear-shaped Purkinje cells and

star-shaped anterior horn cells which range from 60-80 μm in humans. Neuronal

nuclei are large and spherical with a large nucleolus. The perikaryon or the neuronal

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 21

body comprises the Nissl substance (an intracytoplasmic basophilic mass and extends

along the dendrites), neurotubules (aligned longitudinally along axons and dendrites

and involved in axoplasmic transport) and neurofilaments (found mostly in axons,

maintain neuronal form and axoplasmic transport), in addition to other common

organelles. Importantly, axons emerge from the neuron as a slender thread and are

frequently myelinated. The dendrites are afferent components of neurons and are

arranged in a stellate fashion and lack neurofilaments. The synapse is a specialized

junction where axons and dendrites emerging from different neurons

intercommunicate 80. Neurons also play a modulatory role in inflammation, wherein

healthy neurons actively suppress immune function of microglia by interaction

between CD200 receptor on neurons and its ligand on microglia. CD200-/- (deficient)

mice showed activated microglia with increased expression of CD45, CD11b and

inducible nitric oxide synthase (iNOS) during EAE 81. The pathological features of

axonal demyelination are described in section I.1.2.

I.1.3.1.3.4. Microglia

Mononuclear phagocytes comprise an essential component of the

innate immune system, playing important roles in the defense against infectious

agents. Microglia, forming 12% of the CNS cells, demonstrate low turn over rate and

can be induced to express the surface molecules, CD80, CD86 and MHC class II,

necessary for antigen presentation. The perivascular and meningeal macrophages that

line the endothelia of the cerebral blood vessels are bone-marrow-derived cells. In the

resting state, microglia have a ‘ramified’ morphology, with flattened or angular

nuclei, scanty cytoplasm that accumulates at both poles of the cells and are terminally

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 22

non-dividing cells. Activated microglia assume different shapes and could become

bushy with abundant cytoplasm and ramified thick processes lying adjacent to

neurons, and are thus called perineuronal microglia. They also appear as rod-shaped

microglia with a fusiform, elongated body with small, thin processes. Resting

microglia are postulated to have several roles including sensing and reacting to injury,

regulating blood flow and vasculogenesis, phagocytosis, regulating astrogliosis,

neurodevelopment by secreting trophic factors and neuroendocrine functions 82. The

activated microglia express complement type 3 receptor (CR3), MAC-1, MHC class I

and II antigens and lectin-binding protein 82. In MS, microglia may present antigens,

including myelin-derived peptides through MHC class II molecules to cytotoxic T

cells, leading to disease exacerbation and also recruit further monocytes from the

blood stream, enhancing the cytopathic response. However, due to their phagocytic

nature, microglia may also rid the brain of damaged cells and thus contribute to

resolution of inflammation 82.

I.1.3.1.4. T cell-glia interaction

T cell-secreted IFN-γ activates MHC class II-expressing

microglia/macrophages and perhaps astrocytes, which in turn phagocytose other cells

and lytic debris 83. Microglia also modulate the immune response by cytokine-

mediated polarization of T cells and further enhancement of MHC class II expression.

Microglia phagocytose specific antigens in the CNS and present them on MHC class I

and/or II while also producing cytokines and toxic molecules that compromise neural

cell function and survival. These actions induce further recruitment of inflammatory

and immunologically active cells. In MS lesions, reactive astrogliosis and abundant

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 23

macrophages induce peroxynitrite (OONO-) (indicated by nitrotyrosine

immunoreactivity), up-regulation of adhesion molecules, FasL, TNF-α, IL-1β,

complement and ROS, all of which mediate oligodendrocyte cytotoxicity leading to

CNS damage 84. When peripheral macrophages are selectively depleted by

intravenous injection of mannosylated liposomes containing dichloromethylene

diphosphonate (clodronate), EAE-induced demyelination is reduced in rodents

passively transferred with MBP-reactive T cells 85. Autoreactive and proinflammatory

cytokine-producing CD4+ T cells that activate microglia are also associated with

production of neurotrophic factors suggesting that the same cells play different roles

at various stages of the lesion.

Oligodendrocytes are vulnerable to perforin 86 through MHC class I

effector pathway 87 and CD4+ T cell-mediated lytic damage 88 through Fas/FasL or

TNF-receptor (TNF-R) via the MHC class II elimination pathway 87. Importantly, T

lymphocytes induce considerable damage to oligodendrocytes through a host of

soluble secreted factors. Oligodendrocytes may be injured by several means.

Recently, N-methyl-D-aspartate (NMDA) receptors on oligodendrocytes and

glutamate-mediated killing of these cells were discovered 89, suggesting the use of

NMDA receptor antagonists for preventing myelin damage. It has been established

that chemokines direct TH1 polarized cells into the CNS; however, recently it has

been shown that TH2 polarized cells (producing IL-4, IL-5, IL-13, and high levels of

IL-10) induce EAE in B and T cell deficient (Rag1-/- and TCRα-/-) mice. Normal (i.e.,

+/+ Rag1 ) mice were resistant to EAE induction by TH2 cells but not to induction by

TH1 cells. Moreover, an unusually high percentage of neutrophils and mast cells were

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 24

-/- present in the CNS of Rag1 mice into which TH2 cells had been transferred but this

-/- 90,91 was not the case in Rag1 mice that had received TH1 cells . This suggests that T

cell dynamics in the CNS are far more complex than thought with regard to initiating

demyelination as well as resolution of inflammation. This calls for reassessment of

facts regarding the role of T cells in autoimmunity.

I.1.3.2. Molecular components of MS

A persistent question within the MS research community is whether MS

pathogenesis is initiated when autoreactive T cells are generated in the systemic

compartment (immune-initiated hypothesis) or whether events within the CNS initiate

the disease process (neural initiated hypothesis) 9. Accumulation of activated T cells

in early MS lesions and surrounding normal appearing white matter indicate the

importance of cell-mediated immunity in the pathogenesis of MS. Autoimmune

activated T cells in the CNS of MS patients recognize myelin sheath. This has been

demonstrated in the EAE model, where the disease process is initiated by systemic

immunization with neural autoantigens (eg. myelin) or by passively transferring

activated myelin-specific T cells to induce an active disease, reinforcing the concept

that T-cell autoimmune reactions of myelin may be involved in the inflammatory

response in the CNS 92. Adoptive transfer of neural antigen sensitized CD4+ T cells is

performed as described below. After harvesting spleen and lymph nodes from myelin

(related protein)-injected mice and controls, single cell suspensions are prepared. The

purified lymphocytes are counted, and immediately thereafter, 10, 20 or 50 millions

of either spleen or lymph node cells are injected into age-matched mice through the

tail-vein. One week after the cell transfer, an EAE immunization is carried out. T

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 25

cells that secrete TH1 cytokines (IFN-γ, TNF-α, IL-2) are more likely to transfer the

disease than other myelin-specific T cells. T cells secreting IL-4, -5, -10 and -13

protect animals from EAE. Thus EAE is a prototypic autoimmune disorder caused by

a TH1 cell response to myelin antigens. However, heterogeneity within various EAE

models with regard to lesion topography and extent of demyelination/axonal

disruption, indicate the need to define mechanisms linking neuroinflammation and

actual tissue injury 9. The concerns with EAE as a model for MS will be discussed

later. The molecular components of inflammation are extensive to dissect.

Nevertheless, some of these and their contribution to pathogenesis in MS will be

discussed below.

I.1.3.2.1. Cytokines

Cytokines are small, secreted proteins which mediate and regulate

immunity, inflammation and hematopoiesis. Of these cytotoxic cytokines, TNF-α and

IL-1β are harmful to oligodendrocytes. IL-1β is a potent regulator of MMPs

implicated in demyelination and axonal loss in CNS diseases. IL-1β reduces

expression of glutamate receptors in astrocytes and impairs glutamate uptake. This

results in glutamate dysregulation through α-amino-3-hydroxy-5-methyl-4-

isoxazolepropionic acid (AMPA)/kainate receptors and subsequent glutamate toxicity

in oligodendrocytes, when co-cultured with astrocytes and microglia 93. Though

oligodendrocytes express receptors for IL-2, IL-1α/β, TNF-α, IL-6 and

neurotrophins, contradictory findings exist with regard to their trophic or lethal

functions upon ligand binding. During neuroinflammation, TNF-receptors (-RI and -

RII) are up-regulated on oligodendrocytes and microglia while Fas is increased on

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 26

oligodendrocytes. Ligand binding to TNF-RI and IL-1β receptor and nerve growth

factor (NGF) binding in the absence of its high affinity receptor, TrkA (in mature

oligodendrocytes) can induce synthesis of intracellular ceramide, a sphingolipid

generated by sphingomyelinase. This pathway activates c-Jun-NH2-terminal kinase

(JNK) leading to apoptotic cell death 94.

Recently, the contribution of TRAIL to oligodendrocyte death was

investigated in primary cultures of adult human oligodendrocytes. By activating JNK

pathway 95, TRAIL selectively targeted and killed oligodendrocytes, but not

microglia, through interaction with the receptors, TRAIL-R1 and -R2. However, this

process occurred in the absence of non-signaling decoy receptors, -R3 and -R4, since

most cell types express -R1 and -R2, but microglia express -R3 96. Inconsistencies

with regard to TNF-α-mediated oligodendrocyte killing suggest that this cytokine

may have both lethal as well as neuroprotective effects through induction of trophic

factors, insulin-like growth factor (IGF)-1 and ciliary neurotrophic factor (CNTF) 97.

Though IFN-γ amplifies the inflammatory response, it does not alter oligodendrocyte

survival, which nevertheless is more vulnerable to Fas-mediated apoptosis

precipitated by TNF-α 98. Furthermore, TNF-α blockade in EAE reduces pathology

and TNF-α overexpression increases severity of demyelination 99. However, TNF-α

neutralization worsened MS signs 100 contradicting findings in EAE mice. Since TNF-

α induces protective autoimmunity (discussed later), an innate immune response-

mediated induction of TNF-α may be protective whereas TH1-induced release of IFN-

γ may be detrimental to myelin 101.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 27

I.1.3.2.2. Chemokines

Chemokines are a superfamily of low molecular weight (8-15 kDa),

secreted, heparan-binding molecules that serve as potent chemoattractants for

immune cells. Chemokines bind to GPCRs expressed on most immune cells.

Conserved serine and threonine residues in the short cytoplasmic tail of the

chemokine receptor are important for signaling through phosphorylation by various

kinases. Leukocyte migration and their interaction with neural cells require

chemokine-mediated signaling. Signaling from chemokine receptors is mediated by

Gαi, Gαq and G16 proteins that associate with chemokine receptors. Upon receptor

activation, phosphatidylinositol [PI]-3-kinase-γ is activated and the protein kinase C

(PKC) pathway is triggered by Gβγ subunit by generation of inositol-1,4,5

2+ triphosphate (IP3) and diacylglycerol (DAG). IP3 mobilizes release of [Ca ]i from

intracellular stores activating conventional PKC isoforms that regulate downstream

signaling. Ca2+ flux, generation of lipid mediators and ROS mediate changes in cell

shape, membrane ruffling, actin polymerization as well as extension and retraction of

lamellipodia that propel leukocytes towards the site of inflammation. Receptor

dimerization associated with chemokine binding also activates the JAK/STAT (Janus

kinase/signal transducer and activator of transcription) pathway mediated by a

conserved DRY motif in the intracytoplasmic tails. JAK phosphorylation leads to

STAT recruitment to the receptor followed by its activation and translocation to the

nucleus to activate or repress gene expression 102. Chemokines play an important role

in leukocyte recruitment to inflammatory lesions. They also regulate proliferation and

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 28

migration of neural progenitor cells to sites of CNS trauma and confer trophic activity

on neurons that survive initial insult.

Microglia, astrocytes and endothelial cells express CCR7 receptor that

leads CCL21/SLC and CCL19/ECL-expressing B lymphocytes to migrate to the CNS

where they may destroy myelin. Active MS lesions have increased amounts of

CCL2/MCP-1, CCL8/MCP-2 and CCL7/MCP-3; CCR2, CCR3 and CCR5 on

macrophages, microglia and T cells; CCR3 and CCR5 on reactive astrocytes and

CXCR3 receptor for CXCL10/IP10 is present on all perivascular T cells and

astrocytes in active lesions. The chemokines CCL3/MIP1α, CCL5/RANTES,

CXCL9/MIG and CXCL10/IP-10 are found in the CSF of MS patients. This

chemokine profile suggests that the expression of these chemokines by lesion-

associated cells may be involved in the recruitment of specific T cells expressing

CXCR3 or CCR5 103.

I.1.3.2.3. Neurotrophic factors and brain repair

It has been observed that neurotrophic factors are induced by the

inflammatory cytokines, TNF-α and IL-1β within parenchymal microglia. Microglia-

derived IL-1β induces NGF, CNTF and IGF-1, which repair the injured CNS. TNF-α

plays a dual role in demyelination (TNF-R-I) and remyelination (TNF-R-II), by

interacting with the receptor available 101. TGFβ also plays a dual role by down-

regulating proinflammatory cytokines but unfortunately induces astrocytic activation

and glial scar formation that inhibits oligodendrocyte differentiation and

remyelination 104. IL-6, IL-1β and chemokines (CCL2, CCL3 and CCL5) are essential

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 29

for axonal growth and oligodendrocyte differentiation but also promote inflammatory

cell recruitment and survival 104. IL-1β binds to its receptor on astrocytes leading to

the production of growth factors (Neurotrophin [NT], NGF, CNTF) that act on neural

progenitors and favour repair of oligodendrocytes and induce remyelination 104.

Interestingly, TNF-α binds to TNF-RII (p75) on oligodendrocyte progenitors and

promote their differentiation into mature and functional oligodendrocytes 101. Similar

to IL-6-, CNTF-, oncostatin-M- and IL-11-mediated survival of neurons, leukemia

inhibitory factor (LIF)-mediated signaling through its receptor in mice infected with

Theiler’s murine encephalomyelitis virus (TMEV) 105, can also potentiate

oligodendrocyte survival 106. Thus, an innate immune response is also needed for

repair of the CNS following injury.

Brain repair takes place following injury by neurotrophins (NGF, Brain-

derived neurotrophic factor [BDNF], NT-3, 4/5) and their receptors (Trk A, B & C

and the low affinity p75NTR) as well as growth factors (platelet-derived growth factor

[PDGF], fibroblast growth factor [FGF]-2, IGF-1). These are re-expressed by T and B

cells, macrophages, astrocytes and mast cells in the brain lesions. Growth factors

binding to their receptors lead to receptor clustering and autophosphorylation of

tyrosine residues in the cytoplasmic tail of the receptors. This opens up docking sites

for SH2 domain-containing adaptor proteins such as Shc and/or Grb2 and to

activation of the Ras/Raf/MEK/ERKs cascade 107. Expression of extracellular matrix

or cell surface proteins, (chondroitin sulphate proteoglycan (CSP), integrin, ephrin,

tenascins and semaphorins) and oligodendrocyte proteins (Nogo (Neurite outgrowth

inhibitor), myelin-associated glycoprotein [MAG], MOG, Notch-1) may contribute to

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 30

neurogenesis, gliogenesis, synaptogenesis, migration etc. Re-expression of Jagged

and Notch-1 is important for axonal growth and remyelination. Neurocan, a

chondroitin sulphate proteoglycan, can bind to FGF-2 and promote neural-cell-

precursor differentiation and bind neural cell adhesion molecule on inflammatory

cells, allowing their entry into the brain 104. Astrocytic IGF-1 and FGF-2 are essential

growth factors for oligodendrocyte precursor proliferation and for maturation into

myelin producing oligodendrocytes. High affinity FGF receptors present on

oligodendrocytes in demyelinated lesions are stimulated upon ligand binding, leading

to colonization of oligodendrocytes in lesions. Most importantly, upon activation,

astrocyte-derived growth factors and neurotrophic factors promote neuronal survival

and regeneration 108. Astrocytes mediate synaptogenesis wherein high affinity

receptors on astrocytes bind to vasoactive intestinal peptides (VIPs) 109. Also VIP-

stimulated astrocytes produce activity-dependent neurotrophic factor that promotes

survival of spinal cord and cortical neurons 110. Thus, astrocytes are thought to be just

more than ‘brain glue’ 64 and contribute to several dynamic processes in the CNS.

Undifferentiated adult neural stem cells (NSC) can alter developmental

and differentiation programs in response to injury. Subependymal-zone-restricted

adult neural stem cells (NSC) can selectively migrate into areas of demyelination and

differentiate into myelin-producing cells. These cells constitutively express adhesion

molecules, cytokines and their receptors to guide their migration. Adult NSC

transplanted into brain lesions can differentiate into myelin producing

oligodendrocytes and promote functional recovery 104. Since astrocyte-derived FGF-2

can stimulate NSC proliferation, adult NSC enhance neurogenesis when co-cultured

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 31

with hippocampal astrocytes 111. Failure of brain repair could be due to the ageing of

the brain repair mechanism, as young animals have better capabilities of regenerating

axons and remyelination when compared to older ones. Also, extensive degeneration

may damage the rostral migratory stream used by adult NSC to migrate and colonize

damaged areas 104,112.

I.1.3.2.4. Proteases

Proteases are involved in several aspects of the nervous system including

cell growth, neurite outgrowth, cell survival and pathogenicity. The major classes of

peptide-cleaving enzymes include the cysteine proteases, aspartyl proteases and the

metalloproteases. The major cysteine proteases, calpain and cathepsin, degrade

myelin proteins 113 114. An essential enzyme that cleaves the retroviral gag-pol

polyprotein essential for formation of infectious particles is an aspartyl protease and

is thus, a target of several protease inhibitors 115. The metalloproteases catalyze

reactions involving tissue remodeling and degradation. Of these, MMPs influence

growth and survival of neurons and oligodendrocytes. MMPs can disrupt myelin and

cause demyelination 116. Cellular sources of MMP in the diseased brain include

infiltrating leukocytes (lymphocytes and macrophages) and intrinsic cells (microglia,

astrocytes and neurons). MMPs have both beneficial as well as detrimental qualities

in the context of CNS diseases. Thus, caution must be exercised when using MMP

inhibitors without understanding their specificity with regard to the type of MMP

targeted as well as the phase of CNS disease when treatment is given. Nevertheless,

the damage caused by MMPs is immense and leads to a variety of inflammatory and

neoplasic disorders where MMP production is significantly increased at various

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 32

stages of the disease 116. Presumably by different mechanisms, MMP-9 and -12

mediate oligodendrocyte process extension, remyelination and maturation of

precursor cells into myelinating oligodendrocytes. MMP-9 does so by removing the

injury-induced deposition of inhibitory NG2 proteoglycan 117, that inhibits axon

regeneration 104. MMP-12, which is expressed on phagocytic macrophages, active MS

lesions, oligodendrocytes and to a lesser extent in astrocytes, is essential for process

extension as well as maturation of oligodendrocytes and it may possibly do so by

processing neuregulin, IGF-1 or IGF binding proteins and thyroid hormones 118.

MMP-7 and membrane type (MT)-4-MMP can also perform the role of TNF-α-

converting enzyme (TACE), converting the TNF-α precursor to its active form, thus

perpetuating inflammation. MMP-1 119 and MMP-2 120 are neurotoxic, directly or

indirectly by MMP-2 cleavage of CXCL12/SDF-1α releasing a neurotoxic form of

the chemokine 121. Also MMPs degrade laminin, the extracellular matrix substrate

leading to loss of integrin signaling and death due to cell detachment (anoikis) 122.

Another protease, myelencephalon-specific protease, abundant in neurons and

oligodendrocytes can affect neurite growth and affect oligodendrocyte survival 123.

In the nervous system, proteinase-activated receptors (PARs), which are a

family of GPCRs, are widely expressed on glial cells and neurons. PARs are activated

by proteolytic cleavage of their extracellular amino terminus unmasking a tethered

ligand that binds intramolecularly to the receptor, initiating a signal transduction

event. When expressed on CNS-derived cells, PAR2 contributes to the pathogenesis

of ischemia and neurodegeneration 124. However, enhanced PAR2 expression is

observed in MS and EAE, chiefly on perivascular macrophages and astrocytes. Mice

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 33

lacking PAR2 showed diminished disease severity, associated with lower T cell

reactivity and neuroinflammation 125. Thus PARs have diverse roles in various

diseases and specific antagonists to PAR2 may be beneficial in MS.

I.1.3.2.5. Oxidative stress

Several common pathways leading to oligodendrocyte damage can be

elucidated. Of these, oxidative stress plays an important role in death of

oligodendrocytes and neurons. Unregulated production of free radicals such as

hydrogen peroxide, NO, superoxide and highly reactive hydroxyl ions lead to cellular

oxidative stress 126. ROS in cells and tissue causes lipid peroxidation when free

radicals attack double bonds of unsaturated fatty acids such as linoleic acid and

arachidonic acid generating highly reactive lipid peroxy radicals that initiate a chain

of events leading to the formation of breakdown products including 4-hydroxy-2,3-

nonenal (4-HNE), acrolein, malonaldehyde and F2 isoprostanes which are increased

in neurodegenerative disorders. Acrolein, a lipid peroxidation product, down

regulates glutamate and glucose uptake, and 4-HNE inhibits the glutamate

transporter, EAAT1/GLT-1 127. DNA bases are also susceptible to oxidative damage

leading to mutations and forming 8-hydroxyguanine and 8-hydroxy-2-

deoxyguanosine suggesting selective attack on guanine bases by hydroxy radicals 128.

Protein oxidation impairs glutamine synthetase, superoxide dismutase and leads to

protein carbonyl formation that is toxic to cells 129. Oxidative stress products impair

cellular functions due to the formation of toxic species such as peroxides, alcohols,

aldehydes and ketones that are toxic to lymphocytes and blood vessel macrophages

130.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 34

Oligodendrocytes are vulnerable to a variety of mediators of cell death

including oxidative stress and glutamate excitotoxicity. When exposed to ROS,

oligodendrocytes are more vulnerable to injury than microglia or astrocytes.

Interestingly, lower endogenous antioxidant levels in oligodendrocytes may make

these cell types more vulnerable to oxidative stress than neurons 131. Free radicals

from virus-infected or Fc-receptor or intercellular cell adhesion molecule (ICAM)-1-

crosslinked macrophage/microglia are also cytotoxic to oligodendrocytes 132,133.

Moreover, mature oligodendrocytes are less sensitive to effects of NO than immature

oligodendrocytes 134. Thus, several mechanisms of oligodendrocyte death in MS have

been reported but the outcome of cell death in MS-necrosis or apoptosis remains

undefined.

I.1.3.2.6. Nitric oxide (NO)

NO is an essential intracellular and intercellular signaling molecule

participating in regulation of diverse pathophysiological mechanisms in

cardiovascular, nervous and immunological systems. NO acts as a neurotransmitter, a

host defense effector molecule and also as a cytotoxic free radical. It is

biosynthesized from L-arginine and molecular oxygen utilizing nicotinamide adenine

dinucleotide phosphate (NADPH) as a terminal electron donor and heme, flavin

mononucleotide (FMN), flavin-adenine dinucleotide (FAD) and tetrahydrobiopterin

as cofactors 135. NO production is catalyzed by nitric oxide synthases (NOSs) (130-

160 kDa) whose isoforms include, neuronal nitric oxide synthase (nNOS; NOS I),

inducible nitric oxide synthase (iNOS or NOS II) and endothelial nitric oxide

synthase (eNOS; NOS III). A mitochondrial or mtNOS has also been reported 136.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 35

The iNOS promoter has the binding sites for transcription factors, NF-κB,

and STAT 136. Activation of NF-κB regulates inflammatory gene expression,

including iNOS transcription. Physiologically relevant concentrations of NO

contribute to the balance between pro- and anti- apoptotic activity. High

concentrations of NO produced by iNOS can overwhelm cellular protection systems

and shift towards apoptosis whereas low concentrations (generated by eNOS and

nNOS) may be anti-inflammatory and anti-apoptotic. When biological systems have

an unequal balance between endogenous antioxidants and ROS, the resulting

oxidative stress can cause molecular damage and apoptosis, through p38 mitogen-

activated protein kinase (MAPK) activation and induction of p53 137.

Different isoenzymes of NOS are specific to certain cell-types of the body

and they each produce different redox states of NO. In the brain, glial iNOS

expression has been described after injury and pathology with both deleterious and

protective effects, and particularly in MS, iNOS immunoreactivity has been observed

in plaques 138. eNOS and nNOS are constitutively produced in resting cells and are

activated by Ca2+ and calmodulin while iNOS is induced by immunostimulation and

is Ca2+-independent. NO involved in pathogenicity is produced by all isoforms of

NOS 139. Macrophages isolated from active MS lesions display immunoreactivity for

all three NOS isoforms, eNOS, nNOS and iNOS, supporting NO production. NO

contributes to the cytotoxicity of oligodendrocytes and the destruction of myelin in

MS brain and spinal cord 140. iNOS is implicated in the pathogenesis of EAE, TMEV-

induced demyelination and MS. In chronic MS, astrocytes adjacent to inflammatory

lesions occasionally express iNOS. Other cells expressing iNOS include CD64+ and

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 36

CD14+ microglia/macrophages. Expression of iNOS contributes to tissue injury, BBB

breakdown, plaque formation and contribute to glutamate-induced neuronal

excitotoxicity. Endothelial cells are damaged by peroxynitrite as indicated by

nitrotyrosine formation. iNOS could thus be a part of a complex interaction between

components of the innate and inflammatory responses which contribute to

demyelination 84. nNOS is also produced by astrocytes 141,142 in the spinal cord and is

involved in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity in a

mouse model of Parkinson’s Disease 143. Thus, the contribution of NO by eNOS as

well as nNOS must be considered when studying the pathogenicity of NO using iNOS

deficient mice models.

The electrons in the outer molecular orbital of NO exist in 2 excitation

states: a low energy state called the triplet state and the high-energy state or the

singlet state. Singlet NO tends to react with thiol groups to form disulfide and

hydroxylamine, whereas triplet NO favours reaction with superoxide radicals,

producing peroxynitrite (ONOO−) 144. When nitrosative and/or oxidative stress is

coupled with decreased antioxidant levels, normal cellular metabolites can cause

deleterious effects that signal apoptotic cell death 144,145. High concentrations of NO

during inflammation can react with oxygen generating reactive nitrogen oxide

- intermediates (RNOIs) such as nitrogen dioxide (NO2), NO and ONOO , which react

with cellular proteins, lipids and carbohydrates resulting in oxidative damage

characterized by reduced levels of antioxidants such as glutathione, ascorbic acid and

uric acid 136.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 37

NO mediates pathophysiology primarily by altering the function of

biological macromolecules through covalent modifications. One of these metabolites,

protein-linked 3-nitrotyrosine (3-NT), is markedly elevated in various diseases

including MS 146. 3-NT, formed by nitration of tyrosine by reactive nitrogen species

derived from NO and nitrite are indicators of NO formation. Nitrogen dioxide can

convert hypochlorous acid (HOCl) to nitryl chloride that can form 3-NT, which is a

footprint of peroxynitrite-induced nitration. Free 3-NT is taken up by mammalian

cells and irreversibly incorporated only into α-tubulin, through a post-translational

mechanism catalyzed by tubulin-tyrosine ligase, a process that alters microtubule

function 147. Tyrosine nitration can induce structural (in neurons, it may disrupt

neurofilaments) and enzymatic alterations as it interferes with tyrosine

phosphorylation and hence, signaling. Close proximity of 3-NT as well as iNOS

immunoreactivity to the vessels may contribute to BBB damage, a cardinal feature of

active MS lesions, HIV-associated dementia (HAD) patients and disorders of striatal

neurodegeneration 148.

I.1.3.2.7. ER stress

Most secretory proteins and extracellular domains of transmembrane

proteins are cotranslationally transported in an unfolded state into the lumen of the

endoplasmic reticulum (ER), where resident enzymatic activities prevent these

nascent proteins from aggregating as they fold into their native conformations 149. If

demand exceeds the protein folding capacity, unfolded proteins accumulate in the ER

(called ER stress), which activates an ER-to-nucleus signaling pathway called the

unfolded protein response (UPR). This leads to transcriptional induction of

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 38

chaperones, oxido-reductases, phospholipid biosynthetic enzymes, ER-associated

degradation (ERAD) components and secretory proteins (Fig. 3). The aggregate

effect of UPR activation is containment and reversal of ER stress. UPR is associated

with global repression of translation through phosphorylation of eukaryotic initiation

factor (eIF) 2α, mediated by PERK (double-stranded RNA-activated protein kinase-

like ER kinase), while mRNA vital for adaptation to ER stress becomes

translationally privileged. The end effect of these responses is to afford proteins

passing through the ER a chance to fold to their native state, reduce the load on the

ER, disposal of unretrievable unfolded polypeptides through ERAD and increase

capacity for ER export and transport. If the ER stress is not contained, the UPR

directs the cell to an apoptotic pathway 149.

IFN-γ, produced by T lymphocytes and NK cells, is detectable during the

symptomatic phase of MS. Recently, it has come to light that the deleterious effects

of IFN-γ on developmental myelination are partly mediated by ER stress in

oligodendrocytes. Remyelination failure was modulated by PERK, an upstream

transducer of ER stress 150. Activation of PERK leads to phosphorylation of eIF2α at

serine 51 resulting in inhibition of general protein synthesis in order to reduce the

protein load in the ER. However, eIF2α phosphorylation also leads to activation of

activating transcription factor (ATF)-4, which induces the expression of growth

arrest- and DNA damage-inducible gene/C/EBP homologous protein

(GADD153/CHOP). The latter is a dominant-negative inhibitor of the

CCAAT/enhancer-binding proteins (C/EBP) and is a negative regulator of the anti-

apoptotic protein, Bcl-2 and its expression facilitates apoptosis 151. Another ER stress

Chapter 1: Introduction and Literature Review

UPR target genes Transcription factor

Nucleus P P PERK P P Transcription factor

IRE1

BiP Correctly Folded proteins Misfolded proteins

ER lumen ERAD ATF6 Proteasome

Fig. 3: The proximal ER stress transducers ATF6, IRE1 and PERK associate with BiP in the active state. Upon accumulation of unfolded/misfolded proteins in the ER lumen, these sensors are activated leading to expression of genes that encode proteins that enhance ER protein folding capacity, while ERAD is accelerated to remove terminally misfolded proteins (Adapted from Wu, J and Kaufman RJ, 2006)

39 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 40

sensor, ATF6, resides in the ER membrane with a cytosolic amino-terminal domain

and an ER luminal carboxy terminal domain. Upon activation, the amino-terminal is

proteolytically cleaved and the cleaved portion translocates to the nucleus,

cooperating with other proteins to form complexes that induce the expression of

chaperones. One of the up-regulated genes is X-box-binding protein (XBP1), a

substrate of high inositol-requiring enzyme 1 (IRE1). IRE1 is a protein with an ER

luminal amino-terminal domain, a transmembrane domain, a serine/threonine kinase

domain and a carboxyl-terminal endonuclease domain in the cytoplasm. Under ER

stress, IRE1 oligomerises, autophosphorylates, and removes an intron from the XBP1

mRNA, resulting in a transcription factor that activates target genes 151. IRE1

transmits a signal via apoptosis signaling kinase, JNK and also recruits TNF-

receptor-associated factor (TRAF)-2. This cascade triggers caspase-12 activation and

subsequent apoptosis 152.

In mammalian cells, phosphorylation of eIF2α leads to up-regulation of

the master chaperone, glucose regulated protein/immunoglobulin heavy chain-binding

protein (Grp78/BiP), which binds to properly folded and misfolded proteins. In

normal cells, Grp78/BiP associates with luminal domains of its mediators, PERK,

ATF6 and IRE1 but under stress conditions, Grp78/BiP is sequestered to proteins in

the ER where its mediators are released. These components serve to reduce the levels

of newly synthesized proteins translocated to the lumen of the ER, enhances protein-

folding capacity and secretion potential of the ER to facilitate transport and

degradation of ER-localized proteins 151. Grp78/BiP release from ATF6 leads to the

translocation of the latter from the ER to the Golgi apparatus, where it is cleaved and

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 41

activated. This stimulates transcription of genes encoding chaperones that refold

misfolded proteins 153. This also leads to decreased protein synthesis and increased

production of ER chaperones and activation of ERAD, which shunts misfolded

proteins to the proteasome. Grp78/BiP thus monitors the folding status of proteins in

the ER and regulates the UPR response to stress 154,155.

I.1.4. Drawbacks of EAE as a model of MS

Since the first experiments by Rivers 156, in which autoimmune

neuroinflammation and demyelination were observed, many animal models have been

used to study MS. MBP, proteolipid protein (PLP), MAG, MOG and S100 proteins

are the major known CNS antigens that elicit an immune response and behavioural

phenotype resembling MS in mice. EAE is said to be a good model for acute

disseminated encephalomyelitis (ADEM), where there is acute monophasic illness

compared to chronic relapsing disease course in MS 157. Among other features, the

disease pathology in EAE is aggravated when MOG is injected along with anti-MOG

antibodies, suggesting a role for the humoral response 158. CD8+ T cells rather than

CD4+ T cells characterize the immune response to MOG in certain mice strains; CD4+

T cells that dominate the perivascular regions, however, characterize EAE induced by

MBP and PLP. It has also been contended that in MS, CD8+ T cells and macrophages

are the predominant cells, while CD4+ T cells are infrequent. Thus, a collective

agreement regarding the predominant T cells in MS pathogenesis seems to be lacking.

CD8+ T cells are found in close association with myelin membranes suggesting a

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 42

possible role in tissue damage, but though these MOG-reactive CD8+ T cells can

induce EAE, their role is only now being investigated intensively in MS 99.

The traditional ‘outside to inside’ hypothesis of myelin-specific T cell

trafficking to the CNS to induce pathology as seen in EAE was challenged by studies

demonstrating death of oligodendrocytes before inflammation in the brain of MS

patients, suggesting that inflammatory responses in MS brain may be an

epiphenomenon or a response to factors in the brain that initiate oligodendrocyte

death 159. Though a decrease in cell numbers was demonstrated, immunotherapy

+ directed against CD4 T cells has also failed. Therapeutic modulation of TH1 cells to

TH2 cell phenotype must be cautioned as TH2 cells are found in aggressive lesions.

Multiple therapeutic drugs have successfully been implemented in

ameliorating EAE in mice (Table 1), with no subsequent therapeutic benefits for MS

patients. Further, several approaches have also been established for promoting

remyelination, ranging from stem cells 160 to immunoglobulins 161. The dynamics of

disease progression in MS, the ability of the CNS to remyelinate itself and the failure

of several established therapeutics suggest that we should examine the disease

without the constraints of EAE, particularly focusing on genetics, epidemiological

and imaging studies in humans and examine issues outside the autoimmune

hypothesis 157.

I.1.5. Genetics of MS

Epidemiological studies have provided evidence for environmental factors

in the disease process. MS relapses are frequently associated with common viral

43

Table 1: Therapeutics employed in EAE (From Sriram and Steiner, 2006)

Compound Target Antibodies to T-cell surface antigens CD3, CD4, T-cell receptor, CD2, IL-2R, CD24, CD40L, CD28 Antibodies to APCs MHC class II, CD40, B7-1, B7-2, Fc receptor blockage Antibodies to NK cells Anti-NK cell antibody, α-Gal ceramide Antibodies to adhesion molecules VLA-4, ICAM-1, LFA-1 Antibodies to cytokines IL-2, IL-6, IL-12, IL-15, TNF-α, IL-1, IL-23 Antibodies to chemokines Anti-MIP-1-α, RANTES Anti-inflammatory cytokines IL-4, IL-10, TGF-β, IFN-β, IFN-α, IFN-γ Antagonists of signaling molecules Tyrphostins (JAK-STAT inhibitors), lysofyline, MAPkinase inhibitors, inhibitors of NF-κB and iNOS activation, amsamysin, cholera toxin, AMPA antagonists, glutamate antagonists, IL-1 receptor antagonists Activation of nuclear receptors PPAR-γ, retinoic acid Hormones Estrogen, progesterone, Vitamin D, DHEA, leptin antagonists Antibiotics Minocycline, rapamycin Anti-metabolites and immunosuppressants FK-506, cyclosporine, dyspergualin, corticosteroids, azathioprine, cyclophosphamide, mycophenolate, bone- marrow transplantation Gene therapies Targeted delivery of IL-4 and IL-10 Enzyme inhibitors HOMG co-reductase inhibitors (statins), COX-2 inhibitors Peptides/proteins Oral myelin proteins, omega-3 fatty acid, curcumin, padma-28, fish oil Small organic molecules Linomide, silica, sodium phenyl acetate, copper chelators (N- acetylcysteine amide), laaquinamod, piperazylbutroxide, uric acid, dermatan sulphate, aminoguanidine, cuprizone, roliprim, H-2 receptor antagonists, indoleamine 2-3 deoxygenase, FTY- 270, pentoxyfyline Miscellaneous Incomplete Freund’s adjuvant, BCG vaccination, Helminthic infections , Glatiramer acetate Chapter 1: INTRODUCTION AND LITERATURE REVIEW 44

infections and migration from low-to-high risk areas exacerbates the risk of

developing MS. A viral infection may initiate MS, presumably an autoimmune

disease 162. Thus, both genetic and environmental factors seem to contribute to

development and progression of the disease 163. MS exhibits several characteristics

that are common to autoimmune diseases including polygenic inheritance, evidence

of environmental exposure, increased frequency in women, and partial susceptibility

conferred by a HLA-associated gene 164. Despite substantial evidence for polygenic

inheritance, the MHC-containing HLA is the only region that has clearly and

consistently demonstrated linkage and association in MS genetic studies.

Susceptibility to MS involves a significant number of different genes, each with a

relatively small contribution. Particularly, MHC genes, namely HLA-DRB1 and

HLA-DQB1 showed strong associations with MS in Canadian and Finnish cohorts

165. The strongest linkage result is on chromosome 1q44 with an increase in the

multipoint Logarithm of the Odds (LOD) score and also to other autoimmune

diseases such as systemic lupus erythematosus and rheumatoid arthritis 166. In

addition, there is a potential presence of an MS susceptibility locus on chromosome

7q21-22, delineated by the markers D7S3126 and D7S554. Interestingly, within this

region, there are 2 putative genes, protachykinin-1 precursor gene (TAC1, pos.

37,224–41,031 in Genbank entry AC004140) and Thymosin-β4, which constitutes a

multigene family of IFN-inducible proteins (Genbank entry M17733). TAC1 encodes

four products of the tachykinin peptide hormone family, Substance P and neurokinin

A, as well as the related peptides, neuropeptide -K and -γ 167. An etiological role for

Substance P in CNS inflammation, such as seen in MS, is likely since Substance P is

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 45

an inflammatory mediator. Astrocytes express the high-affinity NK-1 Substance P

receptor 168. In astrocytes, Substance P induces secretion of IL-8 through the NF-κB

activation pathway 169, and of IL-6 through the p38 MAPK pathway 170. In the

context of my thesis, however, the importance of chromosome 7q21-22 lies in the

fact that it also contains the ancestral proviral copy of HERV-W7q, which

encodes Syncytin-1 7.

I.1.6. Environmental factors in MS

I.1.6.1. Infectious agents in MS pathogenesis

Application of sophisticated molecular tools has led to identification of

several viruses that show association with MS. No pathogen has nonetheless been

accepted as the causal agent of MS. Studies related to virus infection in MS are

serological and are demonstrated by antibody titers against a particular virus, though

it has not been elucidated whether these antibodies are elevated in response to the

etiological agent or if it’s an epiphenomenon. It can be said that inflammatory

mediators and/or demyelination of axons block impulse conduction leading to clinical

manifestations of exacerbations. These inflammatory mediators may include IFN-γ

and IL-12, cytokines that are induced in response to active infection 171. A

prospective study identified that there was worsened neurological damage during

exacerbations with onset around the time of clinical infection than that of non-

infection. Infections contributing to increased clinical activity were supposedly of

viral origin in the upper-respiratory tract, self-limiting and mild, but may also include

pathogens such as Chlamydia pneumoniae and Mycoplasma pneumoniae, that mimic

viral pathogens 172 173.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 46

I.1.6.2. Demyelinating viruses

Following infection and a long period of latency, a virus can be

reactivated and affect oligodendrocytes as in PML caused by JC virus infection. A

virus can also initiate an acute or sub-acute demyelinating immunopathology, as in

the case of Theiler’s murine encephalitis virus (TMEV) model of demyelination. The

mechanisms by which viruses induce autoimmunity include molecular mimicry,

epitope spreading and bystander activation. Molecular mimicry involves the de novo

activation of autoreactive T cells due to cross reactivity between self and viral

epitopes during a virus infection. The recognition of self-antigens at moderate affinity

levels can lead to positive selection and export to the periphery. However, when these

potentially self-reactive T cells cross-react with foreign antigens, they can be

activated and recruited into the nervous system. Based on structural studies, for

molecular mimicry to take place, minimum contact motifs between MHC and TCR is

sufficient.5 This was further clarified by studies in which viral and bacterial peptides

sharing contact motifs led to activation of MBP-specific T-cell clones in MS patients

174. Examples of such mimicking antigens include TMEV and the myelin component,

galactocerebroside 175. Also, a non-pathogenic strain of TMEV mimics the

immunodominant epitope of the myelin protein, PLP139-151, with subsequent

demyelination in the absence of adjuvants such as complete Freud’s adjuvant (CFA),

which imposes an artificial inflammatory environment 176.

Epitope spreading occurs when the immune response from an initiating

antigenic epitope widens to different epitopes on the same molecule (intramolecular

spreading) or to epitopes on a different antigenic molecule (intermolecular

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 47

spreading). For example, cells responding to PLP139-152 isolated from lymph nodes of

TMEV-infected mice have the ability to demyelinate organotypic spinal cord cultures

177. The third mechanism, bystander activation, is the non-specific activation of T

cells resulting from direct inflammatory and/or necrotic effects of virus infection of

tissue in the target organ. Destruction of affected tissues would release sequestered

antigens with increased local inflammation followed by recruitment of activated

lymphocytes, which are not responsible for the initial insult or reactive to the viral

antigens 178.

Myelin-specific CD4+ T cells can potentially cross-react with foreign

antigens such as viral peptides, a process called molecular mimicry, in which self-

reactive T cells become activated by infectious agents to mediate an autoimmune

process. Chronic inflammatory demyelinating disease induced by TMEV is an

example of an acquired acute or persistent infection of neural cells that could result in

release of tissue antigens, provoking a disease relevant autoimmune response. Mouse

hepatitis virus (MHV)-infected mice exhibit chronic demyelinating encephalomyelitis

characterized by mononuclear cell infiltrates. CD8+ and CD4+ T cells are crucial for

controlling the virus while B cells and antibodies are involved to a lesser extent 179.

I.1.6.3. Retroviruses and MS pathogenesis

A retroviral cause for MS was postulated when 70% of MS patients had

cross-reactive antibodies to Human T lymphotropic virus (HTLV-1/2) and Human

Immunodeficiency virus (HIV) antigens 180, although subsequently disproven 181.

Furthermore, there are unconfirmed reports of isolation of HTLV-1-related nucleotide

sequences in cells from the CSF of some MS patients 182. Among retroviruses to be

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 48

associated with MS, probably MSRV is the most intensely studied. MSRV was

initially called LM7 virus, an oncovirus, isolated from the leptomeningeal LM7 cells

obtained from the CSF of a patient with MS 183. However, there is substantial

controversy as to whether MSRV is an endogenous or exogenous virus 184. MSRV

belongs to the ERV9 family of endogenous retroviruses (ERVs), and partial

molecular characterization revealed that MSRV has 75% homology to ERV9 185. In

fact, the MSRV envelope shares about 87% amino acid identity with the human

endogenous retrovirus (HERV)-W7q envelope protein, Syncytin-1

(www.ncbi.nlm.nih.gov). Using MSRV probes obtained from virion-associated RNA,

a novel HERV family was identified that is different from ERV9 and genetically

related to MSRV sequences and was named HERV-W 186,187. The pathogenicity of

MSRV retroviral particles has been evaluated in severe combined immunodeficiency

(SCID) mice grafted with human lymphocytes and injected intraperitoneally with

MSRV virion. MSRV-injected mice suffered acute neurological symptoms and died

within 5 to 10 days post injection. In ill animals, reverse transcription-polymerase

chain reaction (RT-PCR) analyses showed circulating MSRV RNA in serum, with

overexpression of TNF-α and IFN-γ in spleen 188. This in vivo study suggests that

MSRV retroviral particles from MS cultures have potent immunopathogenic

properties mediated by T cells corroborating previously reported superantigen activity

of HERVs in vitro, which appear to be mediated by overexpression of

proinflammatory cytokines 189.

The in vitro production of retrovirus particles (RVP) in cell cultures from

MS patients but not healthy controls may be enhanced or activated by infectious

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 49

triggers such as Herpesviruses (e.g. herpes simplex virus (HSV), EBV). Independent

molecular analysis of retroviral RNA associated with RVP revealed two different

genetic families of endogenous retroviral elements: MSRV/HERV-W and

RGH/HERV-H. Retroviral particles of the latter were found to be transmitted to

mitogen-stimulated lymphocytes from healthy donors 190,191. Though this study was

not confirmed, it suggests that one or more retroviruses may be associated with MS.

Production of pathogenic molecules has been associated with retroviral

expression 192, especially the envelope protein 193. It has been postulated that there

may exist a pathogenic 'chain-reaction' in MS involving several step-specific

pathogens interacting with particular genetic elements leading to enhanced retroviral

expression 194. However, there is no conclusive proof that any human endogenous

retroviruses (HERVs) play a role in human disease, although investigators have

postulated several possible pathogenetic mechanisms. HERVs, for example, could

enhance the transcription of cellular genes downstream of HERV-long terminal

repeats (LTR) that contains the promoter elements. HERVs have been postulated to

encode superantigens that result in enhanced inflammatory responses, or mimic self-

antigens leading to autoimmune pathologies such as lupus or MS 195. Increased

expression of the HERV-W env-encoded glycoprotein, Syncytin-1, in astrocytes was

found to induce ROS that mediated oligodendrocyte toxicity 196. Moreover, Syncytin-

1 expression was enhanced by HSV-1 infection 197. Yet expression of these

endogenous retroviruses are seen in several neuroinflammatory diseases including

MS, with no specific disease pathology 198, suggesting that HERVs found in MS are a

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 50

by-product of the inflammatory component of MS, diluting the contention of some

researchers that HERVs are a causative pathogen of MS 195.

Due to its partial immune-restricted environment, the CNS is particularly

vulnerable to immune mediated damage. In this context, leukocyte infiltration into the

CNS is a double-edged sword with excessive responses leading to irreparable

secondary damage while a hypoimmune response fails to eliminate the pathogen.

Demyelination and neurodegeneration are some of the immune-mediated outcomes

that occur while inflammation-induced apoptosis and activation of neural cells to

stimulate neurotrophic factor production enhances brain repair. The latter

phenomenon is collectively called protective autoimmunity and can be defined as a

physiological response elicited by a threatening situation in the CNS, which is

beneficial but, if impaired, can lead to autoimmune disease. Tolerance to self is not a

state of non-responsiveness but, rather, is an ability to tolerate an autoimmune

response to self-antigens without developing an autoimmune disease 199. Thus,

inflammation can lead to acceleration of brain repair process that ensures longevity of

intrinsic cells in the CNS cells. In the light of several recent studies 45,99,179, more

important than ever, there is a need for better mouse models of MS that actually

reflects the pathogenesis, so that rational therapies can be designed. I have tried to

address this issue by developing another mouse model for studying MS, wherein a

HERV protein is transgenically expressed in the brain. The rationale being that

though HERVs do not cause MS, they participate in the pathogenesis and blocking

HERV function through specific therapies may aid in slowing down the progression

of MS. Having reviewed various components of the neuroinflammatory pathway, I

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 51

will now discuss the relevance of these cascades in the context of MS and how

retroviral pathogenesis has modified our understanding of neuroinflammation.

1.2. Retroviruses: Introduction

Global health is threatened by the emergence of new infectious agents that

have the potential to cause neurological infections 200. Microbial evolution due to

adaptive changes in host or ecological niches further contributes to disease, which

becomes unmanageable by drugs. Among these infectious agents, the Human

Immunodeficiency virus (HIV) retrovirus that causes acquired immunodeficiency

syndrome (AIDS) has devastated economies and threatened livelihoods globally.

Several strategies to eradicate HIV have proven to be futile due to inherent properties

of the virus. Indeed, retroviruses have attracted considerable attention over the past

century with the recognition of their contribution to cancer, infectious diseases and

other facets of virology. Thus far, 3 Nobel Prizes have been awarded for studies of

retroviruses including Peyton Rous (1966), David Baltimore, Rennato Dulbecco and

Howard Martin Temin (1975) and Michael Bishop and Harold Varmus (1989). The

immense impact of retroviruses on our understanding of health and disease makes

these the most pathogenic infectious agents known.

Retroviruses possess a unique replication cycle, which directly or

indirectly leads to formation of diverse strains with different pathogenic effects. Host-

virus interactions result in benign infections to fatal consequences. Retroviruses are

unique in that they acquire and alter the structure of host-encoded sequences leading

to oncogenesis in some cases. Upon insertion into the host genome, retroviruses

behave as transposable elements, thus affecting evolutionary processes. In addition to

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 52

being investigated for diseases, scientists have harnessed the ability of retroviruses to

serve as vectors of foreign genes for therapeutic purposes 201. There is an increasing

trend in using viral vectors for gene therapy in the brain. Retrovirus vectors make a

good choice for gene transfer since they integrate into the target cell genome to

promote stable gene transfer and integration is precise, resulting in unrearranged

transfer of the target genes. A key element in retroviral vector design is the viral

envelope protein, as it is this protein that binds to specific cell-surface proteins and is

the primary determinant of the range of cells that can be transduced by the vector.

The virus from which the envelope protein is derived is known as the pseudotype of

the vector 202. Lentiviral vectors can transduce (gene transfer and expression mediated

by replication-incompetent vectors) both dividing and non-dividing cells. Further,

retrovirus vectors can be designed to eliminate specific viral protein coding regions

without affecting gene transfer rates and can be made in the absence of replication-

competent virus by using retrovirus packaging cell lines which supply all of the viral

proteins required for vector transmission 202.

I.2.1. Genomic and structural organization

Retroviruses are single-stranded RNA viruses that replicate through a

double-stranded DNA intermediate 203. Retrovirus particles consist of the viral protein

core consisting of the reverse transcriptase and integrase enzymes necessary for

replication. Surrounding the core is the viral envelope, made up of a cellular

membrane-derived lipid bilayer and viral-encoded envelope glycoproteins. The

retrovirus genome is approximately 10 kb and is contained within the structural core,

in association with the nucleocapsid protein, surrounded by the capsid protein outside

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 53

of which is the matrix protein. The genome consists of two positive sense identical

RNA molecules that are contained within the viral particle (Fig. 4). The 5’ end of the

genomic RNA begins with the “r” (for repeat) and “u5” (for unique 5’ region)

segment followed by the viral genes gag, pol, and env. The 3’ end of the genomic

RNA terminates with the u3 (for unique 3’ region) and r (identical to the 5’ R region)

regions and a polyA tail. Each end of the proviral genome consists of regions called

LTRs, which contain the U3, R and U5 regions. This rearrangement at the termini

enables appropriate expression of viral genes. The U3 region of the 5’-LTR contains

the viral promoter and enhancer at the 5’-U3/R from where the transcription of the

viral genome is initiated. In a simple retrovirus, the gag and pol genes are expressed

from an unspliced transcript while the env gene is expressed from a spliced transcript.

The splice donor (SD) is situated between the 3’ end of the 5’-LTR and the 5’ end of

the gag gene, while the splice acceptor (SA) is located at the 5’ end of the env gene.

The transcription terminal polyadenylation signal is situated in the 3’-LTR. The gag

gene encodes viral core structural proteins: matrix, capsid and nucleocapsid. The pol

gene encodes the viral replication enzymes: protease, reverse transcriptase and

integrase. The env gene encodes the envelope glycoprotein, which is processed into

the transmembrane (TM) and surface (SU) subunits 201 (Fig. 5).

I.2.2. Classification

Classification of retroviruses is based on sequence identity and functional

properties or genomic organization in simple and complex structure. There are 7

genera (Fig. 6) 204. The exogenous horizontally or vertically transmitted viruses

including feline (FeLV) and murine leukemia viruses (MuLV), HTLV-1/2, HIV-1/2

Chapter 1: Introduction and Literature Review

Nucleocapsid Surface unit (SU)

Capsid

Transmembrane Protein (TM)

Reverse transcriptase Matrix

Envelope

Diploid ss (+) RNA genome Integrase

Fig. 4: Schematic of a retrovirus

54 Chapter 1: Introduction and Literature Review

gag env

pol SU TM 5’ LTR 3’ LTR PR RT IN U3 R U5 U3 R U5

Fig. 5: Typical proviral structure with identical 5’ and 3’ LTRs. Genes illustrated are shared by all replication-competent retroviruses. The horizontal arrow marks the start site of transcription. The arrowhead denotes the site of 3’-end processing and polyadenylation in the RNA transcript. All viruses synthesize full- length genomic RNA. In simple retroviruses, only a single splice donor or, occasionally, two splice acceptors are used. In complex viruses, multiple spliced products are found.

55 Chapter 1: Introduction and Literature Review

Epsilonvirus:WDSV CAEV Spumavirus: (HSRV) VMV : MuLV EIAV FIV lentiviruses HIV -1 SIVagm HIV-2 primate non- primate SIVmac : MMTV, HERV

Class I: HERV-W

Alpharetrovirus: RSV Class II: HERV-K :HTLV Class III: HERV-L

• 1. -eg. Avian leucosis virus • 2. Betaretrovirus- eg. Mouse Mammary Tumor virus • 3. Gammaretrovirus-eg. Murine and , • 4. Deltaretrovirus-eg. , Human lymphotropic virus • 5. -eg. Walleye dermal sarcoma virus • 6. -eg. HIV, SIV, FIV • 7. Spumavirus- eg. Chimpanzee foamy virus

Fig. 6: Schematic of Retroviral Classification (Adapted from Power C; 2001) 56 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 57

etc. are pathogenic while most endogenous retroviruses are not 201. Early

classification of HERVs was based on the tRNA specificity of the primer binding site

by adding the one-letter code for the specific amino acid to HERV 205 but due to

redundancy in usage of tRNA, this system has been abandoned. Current classification

is based on sequence identity to known exogenous retroviruses and also to copy

numbers (Table 2) 206,207.

I.2.3. Retroviral Biology

Retrovirus infection is initiated by the binding of the viral envelope

glycoprotein to host cell surface molecules, which act as viral receptors. Binding of

envelope protein to a cell surface receptor is the first obstacle for the establishment of

an infection. Thus, envelope/receptor binding in part determines viral tropism.

However, various postbinding events and cell factors, independent of cell surface

receptors, influence the outcome of completion of the retroviral replication cycle and

thus impact on tropism. The envelope glycoprotein exists as an oligomeric complex,

usually a trimer. Binding of the SU protein to specific cell surface receptors initiates

conformational changes in the SU protein that result in the exposure of a fusogenic

domain in the TM protein. The fusion domain is responsible for the fusion of the

virus and host cell membranes, followed by release of viral genetic material into the

host cell (Fig. 7).

After the viral genome is uncoated, the viral reverse transcriptase initiates

minus strand synthesis using the RNA genome as the template. During reverse

transcription, the u3 and u5 regions are duplicated such that the 5’ and 3’ ends of the

proviral genome differ in structure from the ends of the RNA genome. A cellular

58

Table 2: Classification of HERVs in the genome (From Gifford and Tristem, 2003)

HERV Family Primer Copy number Class I HERV.Z69907 ND 30 HERV.ADP tRNA Thr 60 HERV.E tRNA Glu 85 HERV-F tRNA Phe 15 HERV-F (Type B) tRNA Phe 30 HERV-FRD tRNA His 15 HERV-H TRNAHis 660 HERV-H49C23 NO LTRs 70 HERV-I tRNA Ile 85 RRHERV-I TRNAIle 15 ERV-9 tRNA Arg 70 HERV-F (Type C) tRNA Phe ND HERV-P tRNA Pro 70 HERV-R tRNA Arg 15 HERV-R (Type B) tRNA Arg 15 HERV-T tRNA Thr 15 HERV-W tRNA Thr 115 HERV-XA tRNA Trp 15 Class II HERV-K.HML1-4 tRNA Lys 170 HERV-K.HML5 tRNA Ile 45 HERV-K.HML6 tRNA Lys 70 HERV-K.HML9 ND ND Class III HERV-L tRNA Leu 575 HERV-S tRNA Ser 70 HERV-U2 ND ND HERV-U3 ND ND Chapter 1: Introduction and Literature Review

Exogenous retrovirus

INFECTIOUS VIRUS

BUDDING INFECTION TRANSCRIPTION TRANSLATION AA mRNA GAG, POL PROVIRUS

cDNA REVERSE INTEGRATION TRANSCRIPTION

TRANSCRIPTION AA REVERSE mRNA TRANSCRIPTION TRANSLATION cDNA RE-INTEGRATION GAG, POL BUDDING

Endogenous retrovirus ?

59 INFECTIOUS VIRUS

Fig. 7: Life cycle of endogenous and exogenous retroviruses (Adapted from Lower, Lower and Kurth, 1996) Chapter 1: INTRODUCTION AND LITERATURE REVIEW 60

tRNA is used as a primer, which is bound to the primer binding site located

downstream of the 5’-LTR. Upon strand transfer of the reverse transcriptase, a

nascent DNA strand is formed for synthesis to continue. DNA synthesis is initiated by

utilization of the polypurine tract (ppt) at the 3’ end of the genome. The reverse

transcriptase transfers to the second identical RNA genome, but not in all cases and

completes the reverse transcription step with LTRs at either end of the full-length

double stranded DNA proviral copy.

Following reverse transcription of the RNA genome into a double-

stranded cDNA molecule, the cDNA form of the viral genome is integrated into the

host cell chromosomal DNA as the provirus. The provirus is integrated into host

genome by a processes known as 3’-processing and strand transfer 208 and is mediated

by the viral integrase enzyme, which is encoded by the pol gene. During 3’-

processing, which takes place in the cytoplasm within the pre-integration complex

(PIC), integrase removes a pGT dinucleotide at the 3’ end of each LTR that is

adjacent to a highly conserved CA dinucleotide. PIC contains linear viral DNA and

several viral proteins including matrix, reverse transcriptase, integrase and

nucleocapsid 208. Strand transfer occurs in the nucleus following the nuclear import of

the PIC. Integrase mediates a nucleophilic attack by the 3’-OH residues of the viral

DNA on phosphodiester bridges located on either side of the major groove in the host

DNA. In a single transesterification reaction, the processed CA-3’-OH viral DNA

ends are ligated to the 5’-O-phosphate ends of the target DNA, forming a gapped

intermediate product in which the 5’-phosphate ends of the viral DNA are not

attached to the 3’-OH ends of the host DNA, as the 3’ ends of the target DNA are not

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 61

joined after strand transfer. The integration process is completed by cleavage of the

unpaired dinucleotides from the 5’ ends of the viral DNA and repair of the single-

stranded gaps created between the viral and target DNA by host-cell DNA-repair

enzymes (Fig. 7). Transcription during the late phase produces the full-length viral

RNA, which is capped at the 5’ end and polyadenylated at the 3’ end by the cellular

machinery and transported to the ribosome. Translation of the unspliced RNA

produces the viral gag and occasional read-through, gag-pol proteins, which assemble

and bud to form immature particles. The actual site of initial genome recognition and

binding is unknown. Proteolytic cleavage of gag by the viral protease produces the

matrix, capsid and nucleocapsid proteins, which rearrange to form the mature particle

209. Retroviruses thus exist as RNA-containing infectious virions and as DNA

proviruses, which may be active or silent 201.

I.2.4. Retroviral Pathogenesis in the Nervous System

Retroviruses represent a group of highly pathogenic infectious agents that

are associated with the development of neurological disease depending on the

individual virus and its host 193. Indeed, human exogenous retroviruses, including

HIV-1/-2 and HTLV-1/-2 viruses, cause neurological disorders involving both the

CNS and peripheral nervous system (PNS) 210-214. Retroviral infections frequently

lead to nervous system disease but HIV-1 infection exhibits the broadest range of

associated neurological phenotypes, which are associated with immunosuppression

and increased HIV-1 molecular diversity within the host. The underlying mechanisms

by which infection and disease occur during exogenous retrovirus infections of the

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 62

nervous system have consistently implicated the amino acid sequence and level of

expression of retrovirus-encoded envelope proteins 215.

Along with host neurosusceptibility, neuropathogenic mechanisms

underlying retroviral infections of the nervous system can be defined by

neuroinvasion, neurotropism and neurovirulence of virus 216. Age, species, immune

status and genetic background determine the host’s vulnerability to HIV-induced

neurological disease. Genetic variation in CCR5, TNF-α and APOE genes, extremes

of age and immunodeficiency characterized by progressive loss of CD4+ cells are

factors that lead to neurosusceptibility to HAD 217,218. The ability of retroviruses to

infect neural cells is neurotropism, which is in part cell-type specific and is dependent

on expression of receptors that mediate viral entry and the viral strain. In the CNS,

HIV infection predominantly occurs within macrophages and microglia. HIV-1

macrophage tropism is correlated with CCR5 while the chemokine receptor CXCR4

suggests T-lymphocyte tropism, though dual-tropic HIV-1 capable of using both

these chemokine receptors have also been identified. Neurovirulence is the capacity

of the virus to cause disease within the nervous system and this involves both host-

and virus-specific factors. Among properties of viruses, variation at the nucleotide

and protein sequences in certain viral genes determines lentivirus-induced disease.

Mediators of host immune response such as cytokines, chemokines, cellular and

humoral immunity that comprise the innate and adaptive immune systems as well as

soluble factors contribute significantly to neuropathogenesis 216.

Retroviral infections might lead to cell death through several ways:

immune system-mediated killing of infected cells, direct toxicity of a virus gene

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 63

product, extensive replication of the virus causing break-down in cell survival and

indirect effects of virus gene products on cell-cell communication 201. Retroviral env

genes encoding structural proteins necessary for cell-surface binding and cell-entry

are also implicated in direct neuronal killing or indirectly through activation of glial

cells that release neurotoxins. Both the adaptive and innate arms of the immune

system can be activated by neurotropic retroviruses; the former consisting of T and B-

lymphocytes and the latter, macrophage/microglia and astrocytes. In the next section,

I will discuss how retroviruses are specifically involved in 2 pathways of

neurodegeneration that are of relevance to my thesis.

I.2.4.1. Retrovirus and Nitric oxide

NO has a dichotomous role in HIV infection; low levels of NO can inhibit

HIV-1 while high levels can induce viral replication and also induce neuronal injury

through apoptosis of neuronal cells, oxidative damage and immune depletion of

lymphocytes and dendritic cells (DCs) leading to immunosuppression 219. Studies

have implicated NO as a key mediator in numerous pathophysiological and

neurodegenerative diseases including HIV-associated dementia (HAD) 220. NO is

vital in inducing innate immunity against virus directly or indirectly (through IFN-γ)

and can inhibit replication of the retroviruses, Friend (F-

MuLV) and HIV-1 221. It does so by deamination of viral DNA 221, nitrosylation of

viral cysteine proteases, essential for viral virulence and replication and also by

inhibiting reverse transcriptase by modulating catalytic activity of cysteine residues.

Also, NO reacts with iron-containing proteins and interferes with the function of

several ribonucleotide reductases that induce cytostatic properties 221. HIV replication

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 64

in astrocytes is not productive because NO binds to NFκB, preventing its binding to

HIV-LTR 222.

Another retrovirus, PVC-211, a variant of leukemia-inducing F-MuLV,

primarily targets brain capillary endothelial cells (BCEC), which are resistant to F-

MuLV infection, but not reactive astrocytes and degenerating neurons. Toll-like

receptor (TLR)-4 expressed on microglia and BCECs could interact with viral

envelope to activate NFκB and induce NO. At the early stages, NO may be protective

by inhibiting viral replication 223. NO generated by iNOS has been shown to inhibit

leukocyte adhesion and also proliferation of T cells in the CNS, and, as a result,

MuLV-induced degeneration is not associated with inflammation or infiltration of

leukocytes 223.

I.2.4.2. Retrovirus-mediated ER stress

Viral infection of mammalian cells elicits cellular responses, such as ER

stress and IFN responses 224. Viruses have evolved mechanisms to challenge these

responses that limit/inhibit viral replication. The ER is an essential organelle for viral

replication and maturation and in the course of a productive infection, many viral

proteins are synthesized in infected cells, where unfolded or misfolded proteins

activate the ER stress response 224. Several viral proteins trigger Grp78/BiP

expression during infection, which in turn associates transiently with folding

intermediates of viral glycoproteins. This binding facilitates folding or assembly of

viral proteins along the maturation process 224. Hepatitis C replication stimulates the

ATF6 pathway, but suppresses the IRE1-XBP1 pathway 225. This favours translation

of viral proteins. There is an increased level of Grp78/BiP, which may be stimulated

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 65

by viral replication. Infection by cytomegalovirus transiently induces Grp78/BiP at

early stages but returns to basal levels at the later stage. This coincides with

expression of other markers of UPR. Increased Grp78/BiP at the early stages inhibits

the stress response by interacting with PERK, ATF6 and IRE1. Thus the virus seems

to be inducing Grp78/BiP in order to control ER stress 224.

Caspase-12 is an initiator caspase required for transduction of the death

signal from the ER in infected cells and is activated by several viruses and the onset

occurs before activation of caspase-8 and -3 152. Several viruses induce apoptosis

mediated by ER stress through the activation of GADD153/CHOP. Virus infection

activates the p38 MAPK pathway, which then acts on GADD153/CHOP to initiate

apoptosis in infected cells. Through the mediation of as-yet unidentified proteins,

several viruses including African swine fever virus, block the expression of

GADD153/CHOP 224. It is unclear however, why some viruses promote ER-mediated

apoptosis. Among retroviruses, a temperature sensitive mutant of Moloney murine

leukemia virus (MoMuLV-ts1) has a single point mutation in the env gene that

confers the ability to destroy T cells and motor neurons, causing a progressive

spongiform encephalopathy 226. Neurodegeneration is probably due to loss of glial

support and release of TNF-α, IL-1 and NO from adjacent ts1-infected glial cells 226.

Neurons display apoptosis, vacuolization and inclusion bodies. Elucidation of the

pathogenic mechanisms has revealed a role for ER-stress in neuronal death. TNF-α or

NO induces the ER to release Ca2+ leading to activation of ER-stress signaling

pathways and cell death through apoptosis.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 66

MoMuLV infection is correlated with phosphorylation of PERK and

eIF2α. During late stages, levels of Grp78/Bip may be insufficient to protect neurons

against ER-stress associated with the retroviruses, FrCasBrE and ts1 infection.

Neurons, but not astrocytes, show increased levels of ER-stress associated genes in

infected mice, which result in activation of ER-associated caspase-12 and subsequent

2+ cleavage of caspase-3. Stressed neurons also showed increase in [Ca ]i-mediated

phosphorylation of calmodulin (CaM) kinase IIα. Since ts1 infects glial cells, but not

neurons, an indirect mechanism of neuronal death, perhaps glutamate excitotoxicity

associated with NMDA receptor activation due to neurotoxins from infected glia, may

lead to neurodegeneration 226. The envelope protein of an avirulent strain, F43, binds

to Grp78/BiP and is processed through the normal secretory pathway. In contrast, the

envelope protein of FrCasBrE bound to Grp78/BiP for a prolonged period is retained

in the ER and diverted to the proteasome for degradation 154. ts1-infected astrocytes

demonstrate expression of GADD153/CHOP in neurons but not astrocytes 226,227.

GADD153/CHOP and Grp78/BiP as well as iNOS and apoptosis are induced in p53-/-

microglial cells when treated with lipopolysaccharide (LPS) or IFN-γ, suggesting

p53-independent NO-induced apoptotic mechanism in microglia 228. Both FrCasBrE

and ts1 strains induce a protein misfolding disease since Grp78/BiP binds to

hydrophobic residues and attempts to prevent protein aggregation of misfolded

proteins and activate an UPR. Recently, the Power lab has shown that replication-

competent feline immunodeficiency virus (FIV) is essential for inducing an ER stress

response and a vigorous neurotoxic and immune response in feline macrophages and

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 67

in vivo. However, mere exposure and/or expression of the envelope protein of FIV

was insufficient to induce an ER stress response 229.

Recent studies indicate that ER stress can be linked to inflammation,

specifically the systemic inflammatory component of innate immunity, called acute

phase response (APR) 230. Members of the membrane-bound transcription factor

family with homology to ATF6 include CREBH, Luman and OASIS (old astrocyte

specifically induced substance)231. CREBH is a hepatocyte-specific bZip transcription

factor belonging to the cyclic AMP response element binding protein transcription

factor (CREB/ATF) family, which requires proteolytic cleavage for its activation.

Interestingly, proinflammatory cytokines induce and cleave CREBH, which regulates

C-reactive protein (CRP) and serum amyloid P-component (SAP), which are

implicated in several pathologies 230 including MS, where levels of these proteins are

augmented in the serum 232. CRP can bind to and activate monocyte-macrophages 233,

whereas SAP plays important roles in leukocyte adhesion 234. These results are highly

significant as their relevance to pathologies in the CNS is immense. In the next

section, I will discuss retroviral infections of the nervous system, focusing on the

common pathogenic mechanisms of both the exogenous and endogenous retroviruses.

1.2.5. Exogenous retroviral pathogenesis

1.2.5.1. Type C Retroviruses

Type C or oncogenic retroviruses are associated with neurovirulence in a

range of host species, including cats, rodents, birds and humans with or without

concurrent immunological disease. Genomes consist of three structural genes and 2

LTRs and perhaps an accessory gene. Neurotropic type C retroviruses include MuLV,

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 68

and HTLV-1/-2. Herein, I will discuss only MuLV and HTLV-1 because they are best

understood in terms of neuropathogenesis.

1.2.5.1.1. Murine leukemia virus (MuLV)

Infection with MuLV, a type C neurotropic retrovirus, is associated with

tremor, hindlimb paralysis, and lack of coordination. Several different strains of

MuLV exist, each with varying cell-tropism, usually infecting astrocytes,

oligodendrocytes and microglia. MuLV enters the CNS via leukocyte trafficking in

the nervous system or by infection of the brain endothelial cells, and are known to use

the cationic amino acid transporter (mCAT-1) 193. Spongiform changes or loss of

CNS parenchyma in brain stem and spinal cord or hemorrhagic stroke-like pathology

are the pathological changes associated with different strains of MuLV 235.

Inflammation-induced breach in the BBB leading to its increased permeability and

glutamate excitotoxicity resulting in neuronal death, are associated with the

pathology. TNF-α production in the vicinity of the neurons increases glutamate

concentration, in turn activating the NMDA receptor and causing Ca2+ influx.

Glutamate concentrations in the extracellular fluid of the striatum of MuLV infected

mice are increased, leading to neurodegenerative changes and contributing to

cognitive deficits 236. Altered intracellular processing and expression of MuLV env-

encoded proteins and levels of viral replication in the brain contribute to

neurovirulence. Though proinflammatory cytokines have been identified, pathogenic

host response to MuLV infections of the nervous system remains undefined. A

variant of the oncogenic retrovirus, MuLV, called WB91-GV induces lymphomas in

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 69

a high percentage of adult mice. WB91-GV was highly selective for white matter

infection, especially replicating in oligodendrocytes 237.

Resistance to MoMuLV-induced neurological disease has been localized

to several host genes including Fv1, Fv4 (gag) and Akvr-1 (Env) 238. Akvr-1 prevents

neurovirulence by blocking the putative viral receptor, mCAT-1, while Fv4 gag

inhibits a post-entry step 193. The env gene that contains the principal determinants of

neurovirulence, encodes the SU and the TM involved in receptor binding and fusion.

In CNS tissues of MoMuLVts1-infected mice, astrocytes, microglia,

oligodendrocytes and endothelial cells are infected but neurons are not. Astrocyte

death observed in infected mice occurs through apoptosis, mediated by ER and

mitochondrial stress 239. In addition, infected astrocytes also undergo oxidative stress

wherein cellular glutathione and cysteine are depleted, ROS are increased and

importantly, the basic leucine zipper transcription factor, Nrf-2 is induced which

translocates to the nucleus to drive expression of genes associated with oxidative

stress 240.

1.2.5.1.2. HTLV

HTLV-1 infection currently persists in 10-20 million people worldwide

but is a particular problem in the Caribbean, Japan, South America, Africa and

amongst high-risk groups in the United States. A subset of HTLV-1/-2-infected

patients develop a progressive myelopathy, HTLV-1/-2-associated

myelopathy/Tropical spastic paraparesis (HAM/TSP), characterized by inflammation,

particularly in the thoracic region of the spinal cord 193. In addition, HTLV-1-infected

patients may also develop adult T cell leukemia (ATL). Prednisolone, IFN-α,

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 70

immunoglobulin, and plasma exchange are reported to improve disability, but most

studies were performed on small numbers of patients with a short-term follow-up 241.

HTLV-1 belongs to the Deltaretrovirus genus of family

and a member of the Oncovirinae type C subfamily whose diploid plus strand RNA is

9032 nucleotides long. HTLV-1 envelope uses the glucose transporter GLUT1 242 as a

receptor. This receptor also mediates infection by HTLV-1 envelope pseudotyped

virus and abrogates HTLV-1 envelope viral interference to superinfection 243. GLUT1

appears to be concentrated in viral synapses 243 and in this context, HTLV-1 subverts

the immunological synapse to transfer viral genomes directly from cell to cell 244.

Although the CD4+ T cells comprise a dominant viral reservoir,

maintaining HTLV-1 infection during the lifespan of an infected individual, other

hematopoietic cells (non-CD4+ T cell subsets, B lymphocytes, monocytes and

macrophages, DCs and megakaryocytes) as well as glial cells (astrocytes and

microglial cells) with limited infection, are also part of HTLV-1 in vivo tropism. Viral

proteins and genomes have been detected in astrocytes and lymphocytes within spinal

cord lesions 193. This is of particular importance in HTLV infections, due to the stable

integration of the viral genome in infected T cells and astrocytes 245, whose in vivo

lifespan may exceed a decade.

In addition to gag, pol and env genes, HTLV-1 also encodes regulatory

proteins, Tax and Rex, derived from the pX open reading frame (ORF) in the 3’

portion of the viral genome. Rex regulates viral gene expression post-transcriptionally

by allowing cytoplasmic and intracellular transport of incompletely spliced viral

mRNA 246. Tax acts in trans to activate transcription initiating from the viral

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 71

promoter in the U3-LTR and also regulates transcription of the host gene, CD25, by

binding to the promoter region. The consequence of CD25 activation by Tax, is

induction of IL-2 in CD4+ T cells where the virus is chiefly found, though CD8+ T

cells also carry the virus. Further, Tax stimulates NF-κB activity via activation of the

signaling cascade controlling IκB phosphorylation specifically through interaction

with IKKγ subunit of the IκB kinase complex 247. Since NF-κB regulates immune and

growth regulatory gene transcription, these events culminate in dysregulation of T

cell function leading to CD4+ T cell proliferation that precedes ATL. Additionally,

cytokines and MMPs induced by the Tax protein may alter BBB permeability,

allowing infiltration of inflammatory cells into the CNS 193. Activated T lymphocytes

directly kill infected cells expressing Tax protein and indirectly injure neurons

through a bystander effect and lead to astrocyte proliferation 248, causing the release

of inflammatory cytokines, or killing CNS cells that express a cross-reactive cellular

determinant 249.

The pathology of HAM/TSP autopsied tissue indicates that affected areas

in the CNS include the myelinated regions of the lateral funiculi of the spinal cord

where inflammatory cell infiltrates are seen. This eventually leads to loss of myelin

and axons; the tissue is replaced by glial proliferation and fibrillary astrocytosis. Loss

of myelin and axons is associated with perivascular and parenchymal lymphocytic

proliferation, foamy macrophages, proliferation of astrocytes and fibrillary gliosis.

Demyelination and axonal loss observed in spinal cord lesions may be due to

bystander activation from activation of T cells and expression of cytokines 250. In

another study, since HTLV-1 provirus DNA was amplified only in infiltrating

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 72

lymphocytes, but not detected in neurons and oligodendrocytes, the demyelination

observed in the spinal cord is unlikely to be due to viral infection of the astrocytes,

oligodendrocytes and/or neurons 251.

Tax expression is generally low in HTLV-1 infected individuals.

Interestingly, HTLV-1 gene expression is regulated by a protein tyrosine phosphatase,

probably SHP-1 252. Infection of CD4+ T lymphocytes results in their activation and

ability to cross the BBB. Interaction of these activated lymphocytes with the CNS

results in cytokine expression, adhesion molecule and receptor expression and

secretion of MMPs leading to disruption of the BBB, allowing cytotoxic T cells and

antibodies against Tax to enter the CNS. HAM/TSP patients exhibit an autoimmune

response to the heterogeneous nuclear ribonuclear protein-A1 (hnRNP-A1), which is

critical for the transport of nuclear mRNA to the cytoplasm. Autoantibodies to

hnRNP-A1 enter cells, bind the protein and decrease neuronal firing, enhancing

myelopathy 253.

1.2.5.2. Lentiviruses

Lentiviral infections of the nervous system have attracted increasing

attention due to the growing HIV/AIDS pandemic and the concomitant high rates of

neurological disease among HIV-infected individuals 254. Despite extensive genetic

diversity among lentiviruses, they share several biological properties, including

structural and genomic organizations, Mg2+-dependent reverse transcriptase activity

and broad cellular tropisms involving both proliferating and non-proliferating cells,

within and outside the nervous system. On the basis of pathogenicity, lentiviruses are

subdivided into immunodeficiency viruses including human (HIV), simian (SIV),

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 73

feline (FIV) and bovine (BIV) immunodeficiency virus. Further, lentiviruses that

activate the immune system include caprine arthritis encephalitis virus (CAEV),

maedi-visna virus (MVV) and equine infectious anemia virus (EIAV). Of interest,

MVV served as a model of MS in the past 255. Visna is a slow infection of sheep

caused by MVV, which infects the oligodendrocytes 256 and demyelination might be

due to host immune response 257. Though astrocytes are not considered to be major

targets of visna virus, infection of astrocytes leads to sustained activation of MAPK,

triggering reactive astrogliosis 258.

Lentiviruses exhibit a disease pattern in which a primary infection

induces an acute disease and an intense immune response, followed by a lengthy

period of subclinical infection and a terminal phase resulting in death. The intense

host immune response is diminished over time, increasing the individual’s

susceptibility to opportunistic infections. Lentiviruses are distinguished by their

ability to replicate in non-dividing cells such as macrophages. Viral tropism involves

cells of bone-marrow lineage, such as microglia and macrophages. Lentiviruses are

assumed to enter the CNS through infected macrophages crossing the BBB and

subsequent microglial infection. The primary receptors utilized by HIV (CD4)259, FIV

(CD134)260 and SIV (CD4)261, and chemokine co-receptors, CXCR4 and CCR5 are

expressed in the nervous system. The viruses of topical interest in our research are

HIV, MVV and FIV, which will be described below.

The high error rate of HIV-1/-2 reverse transcriptase, high replication

rates, frequent recombination and increased selection make HIV-1 the fastest

evolving organism. Point mutations, deletions, insertions and recombination events

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 74

within a viral population or quasi-species leading to synonymous and non-

synonymous changes and immunosuppression contribute to viral molecular diversity.

The benefit of increased molecular diversity is that HIV-1 has an enhanced ability to

evade the immune system with concurrent depletion of immunological defenses and

gain or loss of (co) receptor binding or replication competence as well as gain drug

resistance. Increased viral diversity at onset of infection may predict a higher viral

load and accelerated disease progression. The interaction of a genetically large and

outbred human population with a highly diverse and dynamic viral population has the

potential for emergence of novel and potentially virulent strains of HIV-1 262.

Lentivirus-associated neurovirulence has been demonstrated by

neurotoxicity caused by viral proteins from HIV, SIV, MVV and FIV, directly or

indirectly. SIV gp41, FIV gp95 and hypervariable regions within HIV gp120

comprise the viral determinants associated with envelope-mediated neurovirulence.

HIV enters the CNS early in the course of infection, and the virus resides primarily in

microglia and macrophages, contributing to productive infection and subsequent

neurodegeneration by forming viral reservoirs. Although HIV-1 infection of

astrocytes has been reported, this appears to be largely restricted to the expression of

mRNA transcripts and protein of the HIV-1 early expressed genes tat, nef and rev.

Brain-derived progenitor cells are permissive to infection but thus far, no convincing

evidence of neuronal infection by HIV-1 has been demonstrated, but are highly

susceptible to injury by direct or indirect/bystander effects 214,263.

Neuropathological features of HIV-1 infection of the brain include HIV-

encephalitis, characterized by multinucleated giant cells, astrogliosis, microglial

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 75

nodules, macrophage infiltration, white matter pallor, reduced synaptic density and

neuronal loss in the cortex and basal ganglia. At the time of seroconversion, about

50% of patients infected with HIV experience acute demyelinating neuropathy 264 and

these characteristics are associated with a subcortical clinical disorder (HAD),

defined by cognitive, behavioural and motor dysfunction 265. HIV infection within the

CNS can be detected by measurements of viral RNA in CSF. There may exist a

positive correlation between CSF viral load and viral load in the brain and the degree

of cognitive dysfunction in patients with HAD. Vacuolar myelopathy is caused due to

HIV infection in the spinal cord and is predominantly found in patients with low

CD4+ T cell count. The pathogenesis remains unclear 266 but intralamellar vacuolation

is observed in the spinal white matter with extensive macrophage activation and

production of cytokines, particularly TNF-α 254.

γ-aminobutyric acid (GABA)-ergic and pyramidal neurons within the

cortex are prone to death in HIV-1-infected brain. Though neurons are not infected by

HIV-1, the primary cause of neuronal injury remains unknown but both apoptotic and

necrotic death are observed. Two theories prevail and are described as ‘direct injury’

and ‘indirect or bystander effect’. HIV proteins can injure neurons directly without

intermediate cells such as microglia and astrocytes. HIV-gp120 interacts with

chemokine and NMDA receptors (by engaging the glycine-binding site) to induce

neuronal apoptosis 267. Neuronal injury can also occur when HIV-Tat is internalized

by receptor-mediated mechanisms into neuronal cells and HIV-Vpr allows formation

of cation-selective ion channels in lipid bilayers 268. Apoptotic neurons do not co-

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 76

localize with infected microglia suggesting that neurons die when HIV-infected

microglia-macrophages release soluble factors 214.

Activated circulating monocytes expressing CD16 and CD69 adhere to

normal endothelium of the brain microvasculature, transmigrate and trigger

deleterious processes during HIV-1 infection. HIV Tat expression can induce

astrocytes to produce CCL2/MCP-1 which is chemotactic for monocytes/microglia

and whose recruitment into the CNS microenvironment leads to potentiation of

toxicity. Chemokines (CCL2/MCP-1) and cytokines (TNF-α) produced by microglia

and astrocytes regulate migration (involving adhesion molecules, vascular cell

adhesion molecule [VCAM-1]) of peripheral blood mononuclear cells (PBMCs)

through the BBB. Neurotoxic factors secreted by brain resident macrophages or

microglia in response to HIV-1 infection and stimulation by viral proteins or immune

activation can induce neuronal death. Among the potential neurotoxins, eicosanoids,

pro-inflammatory cytokines, free radicals and neurotoxic amines are elevated in brain

mononuclear phagocytes infected with HIV-1. TNF-α acts in an autocrine and

paracrine role through its TNF-RI (p55) receptor to activate microglia. HIV-

associated neuronal injury is induced by HIV (gp120 or Tat) through the action of IL-

1α/β, IL-6, TNF-α, chemokines 121 and glutamate excitotoxicity 269. Other microglial

factors released are quinolinic acid (which induces neurotoxicity through NMDA

receptors), cysteine and low molecular weight amine, Ntox 270. These factors induce a

vicious cycle of immune dysregulation and BBB dysfunction leading to neuronal

injury 263.

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 77

Detection of apoptotic astrocytes is more common in HIV-associated

dementia (HAD) patients compared to non-demented HIV/AIDS patients and in the

gp120 transgenic mouse, implying a role for astrocyte cell loss in the

neuropathogenesis of HAD 271. Astrocytes positive for activated caspase-3 in

autopsied subcortical brain tissue from HAD brain suggest ongoing injury to

astrocytes. During HIV infection in the brain, the normal functions of astrocytes are

impaired and they are unable to re-uptake glutamate 272. Astrocytes perform trophic

roles in the CNS and their death could impact on glutamate levels, production of

neurotrophic factors and maintenance of the BBB 214. Cell-specific

compartmentalization of viral strains exists within the CNS as evidenced by

astrocyte-specific env sequences of HIV 265. The mechanism of viral entry into

astrocytes is unclear, because they do not express detectable levels of CD4+ or the

HIV co-receptors 265. Apart from mediating neuronal death in HIV (gp120)-CXCR4

interaction 273, TNF-α produced by macrophages during HIV infection in the brain

blocks glutamate uptake by astrocytes contributing to neuronal injury 274. Stimulation

2+ of metabotropic glutamate receptors (mGluRs) on astrocytes leads to increased [Ca ]i

and further release of glutamate. Cytokines and viral proteins also promote iNOS

induction within astrocytes. NO, thus released reacts with superoxide anions to form

neurotoxic peroxynitrite 263. Thus, astrocytosis induced by viral proteins and

macrophage products might play a role in loss of astrocyte function and brain

homeostasis, leading to neuropathogenesis 214.

Although there is no evidence of oligodendrocyte infection in vivo, the

binding of gp120 to galactosylceramide or other proteoglycans can reduce myelin

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 78

2+ 275 synthesis and increase [Ca ]i levels and apoptotic cell numbers . Further, NMDA

receptors present on myelin can lead to oligodendrocyte damage through bystander

effect 89.

Unlike primate and human immunodeficiency viruses, Maedi visna virus

(MVV) does not cause immunodeficiency in its infected host, primarily due to its

inability to productively infect CD4+ T lymphocytes 276. MVV infects mainly

monocyte/macrophages, dendritic cells and does not require CD4+ nor CXCR4 for

infection, but are required for syncytium formation 277. In the CNS, MVV causes

meningitis, periventricular inflammation and infiltration of the choroid plexus and is

known to induce apoptosis in sheep brain cells 278.

FIV-induced brain disease occurs in 20-50% of infected animals, usually

concurrent with diminished CD4+ T-lymphocyte levels 279. Sharing structural and

biochemical properties with HIV-1, FIV-infected cats have been used as an animal

model that recapitulates many salient aspects of HIV infection including

immunosuppression with depletion of CD4+ and CD8+ T lymphocytes in blood and

high viral burdens 280. Neonatal infection by HIV, SIV and FIV causes neuro-

developmental delay and frank encephalopathy in 50% of those infected, signifying a

worsened overall survival prognosis. Studies have shown that lentivirus envelope

proteins derived from HIV-1 and FIV are highly cytotoxic to neural cells 281. Growth

hormone was shown to improve both neurological and immunological outcomes in a

FIV-infected model by up-regulating IGF-1 in cortical neurons 282. Analogous to the

cell tropisms of HIV and SIV, FIV infects feline macrophages, CD4+ T lymphocytes,

neural cells including microglia and astrocytes, but not neurons 283, by means of the

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 79

chemokine receptors CXCR4 or CCR5 120. However, FIV’s cell tropism has a broader

range infecting both CD8+ T and B lymphocytes 284. Both in vivo and in vitro studies

indicate that FIV is a neurovirulent virus causing neurological complications

manifested as FIV encephalopathy and peripheral neuropathy, similar to the clinical

findings observed during HIV-1 infection 285.

1.2.6. Endogenous retroviral pathogenesis

I.2.6.1. Characteristics of endogenous retroviruses

Human DNA contains ~3,100,000,000 bp comprising ~20,000-25,000

genes, with many transcripts that are non-protein encoding and of no known function

286. All humans carry HERV sequences as an integral part of their genomes,

comprising almost 8% of the human genome and are likely vestiges of retroviral

infections during primate evolution. At some point during evolution, exogenous

progenitors of HERVs inserted themselves into the cells of the germ line, where they

have been replicated along with the host’s cellular genes following a Mendelian

pattern 287,288. Integration of endogenous retroviruses into the germ line is thought to

have occurred 2 to about 70 million years ago and were introduced by mechanisms

involving reverse transcription. HERVs were discovered as a result of approaches

such as low-stringency screening of human genomic libraries 289, PCR by

oligonucleotide homology to viral primer binding sites 290 and during analyses of

human gene loci 291. They are identified as retroviruses because of their provirus-like

structure containing LTRs flanked by short direct repeats and primer binding site

flanking internal coding regions. Other retroelements that make up the genome

include LINES (Long Interspersed Repeat Sequences) that move due to inherent

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 80

reverse transcription activity and processed pseudogenes whose motion is dictated by

reverse transcription not encoded by the pseudogenes 292. Of the retroelements present

in the human genome, HERVs are significant in terms of both physiology and

pathogenesis. HERVs are divided into two groups based upon the presence or absence

of LTRs. Those HERVs with LTRs can be further divided based on infectivity.

Infectious elements with LTRs are retroviruses, while noninfectious elements with

LTRs are retrotransposons while those lacking LTRs are termed retroposons 293.

HERVs represent approximately integrated 1500–2000 proviruses,

together with at least 20000–40000 copies of solitary HERV LTRs per genome (Fig.

8). They have been amplified during evolution by repeated reintegration of reverse-

transcribed mRNA into the DNA of germ line cells 293-296. By virtue of their ability to

integrate randomly into the host genome, HERVs are considered to be an important

class of insertional mutagens 297,298 and exert pathogenic effects through several

possible mechanisms (Fig. 9). Endogenous retroviruses have a life cycle that is quite

different in many aspects. Following reverse transcription, they may re-integrate as

demonstrated for HERV-K 299 or the gene product (envelope) can combine with other

available retroviral proteins such as gag or pol and form a recombinant virus which

may or may not bud out to form an infectious particle (Fig. 7). Exogenous

retroviruses occasionally infect germline cells and become fixed in the host germline.

Intracellular retrotransposition and new integrations in germ cells lead to

amplification of proviral copies. The amplification will decline over time due to

mutations, deletions or recombination between viral LTRs. It is probable that

retrotranposons may give rise to infectious retroviruses by recombination events 300.

Chapter 1: Introduction and Literature Review

Provirus

AAA Genomic RNA

AAA Full length retrosequence

AAA Truncated retrosequence

AAA

Solitary LTR

Fig.8: Various forms of HERVs in the genome (Adapted from Costas J, 2002).

81 Chapter 1: Introduction and Literature Review

Activation of HERVs

Transactivation of cellular genes

LTR (gag) (pol) (env) LTR DNA

cellular gene cytoplasm nucleus mRNA (A)6 enhancer/ insertional promoter mutagenesis insertion

Retroviral proteins Integration events Molecular mimicry Superantigens

Figure 9: Pathogenic potential of HERVs (Adapted from http://www3.gsf.de/imv/gif/lmhome1.gif

82 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 83

The consequences of reverse transcription and integration are insertional mutagenesis,

presence of multiple homologous genomic regions, insertion of heterologous reverse

transcripts and non-transposed reverse transcripts 301.

To date, 31 HERV families have been identified and are named according

to the transfer RNA used to prime reverse transcription 302 303 (Table 2). However,

based on sequence homology, HERVs can be grouped into 80 distinct families 300.

Several members of the HERV-W and -H families have been shown to encode intact

envelope proteins and currently 18 full-length HERV env sequences have been

defined 186,303-305. HERV gene expression is principally regulated by their individual

LTRs. Varied levels of expression and cell-type specificity of isolated HERV LTRs in

human cell lines suggest that HERV LTRs may be a valuable source of

transcriptional regulatory elements for the construction of targeted retroviral

expression vectors 306.

1.2.6.2. HERVs and evolutionary advantages

There is evidence for the ability of HERVs to mediate genomic

rearrangements during primate evolution 307. Examples of the nonfunctional viral

sequences that may have arisen through several duplications, recombination events,

and mutations acquired during primate evolution are HERV-H, HERV-F and HERV-

K. Comparison of sequences, particularly in the LTRs of certain HERV families

provide insight into the length of time a particular genetic sequence may have been

present in the genome. In addition to its evolutionary advantages in mediating

necessary recombination events in the human genome, expressed ERV envelope

genes may have beneficial functions to the host. A possible advantage of expressing

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 84

an ERV envelope protein is to resist exogenous retroviral infections 308. ERVs found

in the genomes of birds, mice and cats protect these animals from certain exogenous

retroviral infections. For example, ERV envelope (ev3 and ev6) in chickens protect

against infection by Avian leucosis virus (ALV) 309; an endogenous feline leukemia

virus protects cats against feline leukemia virus-B infection and the Fv-4 locus in

mice confers resistance to infection by Friend leukemia virus 310. In this context, it is

also worth considering if HIV is on its way to becoming endogenized into the human

genome as it may have occurred in the past for HERVs. However, for this to happen,

HIV must infect germline cells.

The most abundant expression of different HERVs is observed in placenta

and embryonic tissues, and also in reproductive tissues or cells such as testis and

oocytes. The broad expression of HERVs in embryonic tissues may be sufficient for

induction of immunological tolerance towards HERV-encoded proteins. HERV

proteins could influence foeto-maternal immunosuppression 311 during pregnancy

when expressed in the placenta 312, fuse trophoblast cells in the placenta 313 and also

provide protection against exogenous retroviruses through receptor interference 308.

Interestingly, no other human gene has been discovered that encodes these functions.

Lower Syncytin-1 expression in human placentas has been correlated with placental

dysfunction, including pre-eclampsia and hemolysis, elevated liver enzymes and low

platelets syndrome 311.

1.2.6.3. Murine endogenous retroviruses

Mice have two types of retroviral elements, the intracisternal particles

(IAPs) and early transposon element (Etn) families which are responsible for dozens

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 85

of documented germ line and somatic mutations, present in both wild-type and inbred

mice 314. Mouse IAPs are endogenous retroviruses showing sequence homologies to

B/D- and avian type C retroviruses and a gene expression strategy similar to that of

type D retroviruses. These particles are competent for allowing retrotransposition

within the virus-producing cell. IAPs are abundant in many murine tumours and cell

lines. F-MuLV is another endogenous, defective leukemia virus. Cells expressing the

envelope protein of the defective F-MuLV virus are resistant to infection by ecotropic

MuLVs 315. F-MuLV infection during EAE showed many inflammatory cells in the

gray matter including the frontal lobe, whereas almost no inflammatory cells were

found in rats with EAE alone, with pathological features resembling HAM/TSP,

suggesting that retroviral infections can modify the pattern of EAE to a certain extent

316. Two previously uncharacterized retroviral envelope proteins have been identified

in the mouse, namely Syncytin-A and -B. They are present at a single copy and are

phylogenetically unrelated to Syncytin-1 and -2. Northern blot analysis revealed

expression of Syncytin-A and -B in the placenta and mediates syncytia formation 317.

Retrovirus replication cycle does not require the infected cell to be

harmed. HERVs potentially influence progression of human diseases through

insertional mutagenesis, molecular mimicry or induction of aberrant cellular

responses, mediated through viral receptors 193. The increased expression of HERV

genes may be important in modulating host innate and adaptive immune responses

with ensuing disease effects although definitive proof of specific HERV-related

pathogenic effects is lacking. Several human and animal exogenous retrovirus

proteins, particularly the env-encoded proteins, demonstrate a tendency for causing

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 86

neuropathogenic effects 193. However, the precise functions of endogenous retrovirus

proteins in the nervous system are uncertain despite being abundantly expressed in

many species including rodents, cats and non-human primates 193,318,319 (Fig. 10).

HERVs are expressed in healthy tissues such as placenta and interestingly they are

also up-regulated in neoplasic, autoimmune and inflammatory conditions 320. Apart

from controversies regarding the involvement of HERVs in insulin-dependent

diabetes mellitus 321, most reports of involvement of HERVs in disease have been

speculative. Anti-HERV antibodies and retroviral nucleic acid are detected frequently

in several autoimmune diseases such as systemic lupus, MS and schizophrenia. This

however, does not prove cause or consequence of HERV expression but the probable

mechanisms may include break-down of tolerance to HERV proteins, transactivation

of cellular genes by HERV proteins, expression of superantigens, molecular mimicry

etc (Fig. 9). Retroviral involvement in tumorigenesis is well established through

several mechanisms: acute transformation of cells by insertional mutagenesis and

transactivation involving cellular proteins but there is no evidence that human cancer

is caused by a HERV. Development of cancer could be a multi-step process wherein

HERV contribution may be restricted to perhaps insertional mutagenesis 322. In this

regard, there are implications for retrotransposition involving HERV-K which may be

active in the human population 299. Other modes include retroviral superantigen

activation that reacts with the immune system in an unusual way. Normal antigens

stimulate an immune response through presentation by MHC molecules on B cells to

the T cells whose TCRs are arranged in a specific fashion. The stimulated T cells

divide and release cytokines. Superantigens, in contrast, bind simultaneously to MHC

Chapter 1: Introduction and Literature Review

HERV-W HERV-E ERV-FRD (Primate) HIV HERV-H MuLV (Mouse) FeLV (Cat) ZFERV (Fish) HTLV-1 HERV-K 0.5

Fig. 10: A phylogenetic tree describing the evolutionary relationships between different endogenous (ERV-FRD, MuLV, ZFERV, HERV-W, -E, -H, -K) and exogenous (HIV, HTLV-1) retrovirus envelope genes found in mice, fish, cats, non-human primates and humans. Amino acid sequences were obtained from GenBank. The DNA analysis software, MEGA3 (Available from the Lab of Masatoshi Nei, Penn State University) was used to draw the phylogenetic tree.

87 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 88

class II molecules and to all TCR molecules expressing a particular class of Vβ

chains. This accounts for about 5% of the T cells to be stimulated at one time, leading

to a very large immune response following infection of newborn mouse with mouse

mammary tumor virus (MMTV) 323.

Induction of several HERVs is reported in different cell types derived

from patients with various diseases. The HERV-K gag protein and RNA transcript

have been reported to be highly expressed in teratocarcinoma and breast cancer cell

lines 324,325, HERV-E in prostate carcinoma 326, HERV-H in leukemia cell lines 327

and HERV-W in brain tissue and CSF from MS 186 and schizophrenic patients 328.

Induction of HERV-H/RGH, HERV-W and ERV-9 expression was reported after

specific cell types (mainly B cells) from MS patients were cultivated in vitro 329. Viral

RNA from these HERVs has been detected by RT-PCR in blood and brain tissues

from MS patients, although not exclusively from this patient group 329. HERVs could

play several roles in MS pathogenesis, acting predominantly as a component of the

inflammatory cytokine-mediated cascade of events. Thus HERVs may act as auto-,

super- or neoantigens (upon expression of oncogenic viruses) with the potential to

enhance inflammatory responses or induce autoimmune reactions 329.

I.2.6.4. Syncytin-1: Characteristics of protein

The multi-copy (30 known copies of full length env gene) HERV-W

family contains a unique proviral locus, located on chromosome 7q21-q22. The

HERV-W7q envelope gene encodes a 538-amino acid envelope (Syncytin-1) but with

presence of inactivating mutations in the HERV-W7q gag and pol genes 186,330. The

deduced 538-amino acid envelope protein contains an N-terminal leader peptide and a

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 89

putative C-terminal hydrophobic membrane anchoring sequence. Northern blot

analysis detects 8.0, 3.1 and 1.3 kb Syncytin-1 transcripts in the placenta 186 (Fig. 11).

The Syncytin-1 envelope glycoprotein is synthesized as a gPr73 precursor, which is

specifically cleaved at a consensus furin cleavage site to give rise to two mature

subunits, gp50 SU and gp24 TM unit. The non-glycosylated Syncytin-1 molecule is

53 kDa, corresponding to gPr73 lacking seven N-glycans. Presence of the leucine

zipper-like motif and the CX6CC motif suggests that the envelope protein can

oligomerize and the SU and TM subunits are covalently linked. The retroviral

protease cleavage site and the C-terminal 16 residue (R peptide) region are missing

from the 69-amino acid intracytoplasmic tail of Syncytin-1. Fusion competence does

not depend on the R-like peptide cleavage but on the first 16 residues of the

intracytoplasmic tail. The Syncytin-1 sequence shows a 4 amino acid deletion in the

intracytoplasmic tail, located after the conserved 16 residues. This deletion modifies

the potential retroviral protease cleavage site allowing fusogenic activity. This finding

suggests that Syncytin-1 evolved from an infectious ancestor with an original R

peptide fusion inhibition function 331.

Computer analysis (ExPASy Proteomics Tool, available free online,

(http://ca.expasy.org/tools/) of the Syncytin-1 protein revealed a glycosylphosphatidyl

inositol (GPI) linkage on serine 524, through which Syncytin-1 is tethered to the

membrane. There were, in addition, 13 serine, 8 threonine and 9 tyrosine predicted

phosphorylation sites and 189 proteasomal potential cleavage sites identified in the

Syncytin-1 protein.

Chapter 1: Introduction and Literature Review

Fig.11: Northern Blot analysis of Syncytin-1 expression. Poly (A)+ RNAs from placental tissues with the U5(g) (nt 115 to 717 of cl.6A2) and U5(e) (nt 1 to 491 of cl.24.4) probe, gag (Pgag-LB19), pro (Ppro-E), pol (Ppol- MSRV), and env (Penv-C15) probes and the U3(e) (nt 732 to 1116 of cl.C4C5) probe (From Blond et al., 1999). For permission to use this figure, please see Appendix F.

90 Chapter 1: INTRODUCTION AND LITERATURE REVIEW 91

I.2.6.5. Syncytin-1: Interaction with receptors

Syncytin-1 303,332 is functional in that it can induce cell-cell fusion when

expressed in cells expressing the feline endogenous virus (RD114)/simian type D

retrovirus (RDR) receptor 333,334 and as infectious pseudotypes generated with HIV-1

virions 332,333. A widely dispersed group of interfering retroviruses, which includes

the feline endogenous virus (RD114), baboon endogenous virus (BaEV), HERV-W,

and type D primate retrovirus, uses the human sodium-dependent neutral amino acid

transporters type 1 and 2 (hASCT1; gene name, SLC1A4 and hASCT2; gene name,

SLC1A5) as common cell surface receptors 335. Syncytin-1 engages these two highly

divergent (54% amino acid identity) ASCT members, as receptors, which also

transports polar neutral amino acids including, Ala, Ser, Cys and Thr 333. It also uses

the mouse ASCT transporter orthologues of these receptors if the N-linked

oligosaccharides in their extracellular loop 2 regions are eliminated 333. ASCT1 and -

2 transport an overlapping but non-identical set of neutral amino acids, with an

important difference being the transport of glutamine only by ASCT2. The carboxyl

terminal regions of extracellular loop 2 in both ASCT1 and -2 play critical roles in

controlling retroviral reception. ASCT1 and -2 are localized on both neurons and glia

in the brain 336,337 and are essential for maintaining cellular integrity through the

transport of amino acids with potentially neurotrophic 338 or neurotoxic 339 effects.

Conclusion, Statement of Hypothesis and Objectives

Understanding the roles of HERVs in health and disease is of immense

importance considering the diversity and abundance of these retroelements in the

human genome. The lack of reagents and appropriate animal models has restricted

Chapter 1: INTRODUCTION AND LITERATURE REVIEW 92

studies on HERVs. Availability of the human genome sequence and establishment of

several antibodies and vectors has revitalized research on HERVs. We have taken a

step further by establishing a transgenic animal model for MS as well as elucidating a

potentially pathogenic role for Syncytin-1.

Based on previous work by others and our own preliminary studies, I developed the

working hypothesis:

Inflammation in the brain enhances expression of Syncytin-1 in astrocytes,

leading to cellular dysfunction. Through Syncytin-1-mediated induction of ER

stress, free radicals and receptor modulation, astrocytes can affect

oligodendrocyte viability and myelination.

To systematically address this hypothesis, we developed the following objectives:

Objective 1: Determine which HERV envelope gene exhibits up-regulation in

neuroinflammation and the mechanism of oligodendrocyte injury.

Hypothesis 1: Syncytin-1 (HERV-W env) is up-regulated in astrocytes and

microglia in MS.

Objective 2: Define the precise Syncytin-1 abundance in clinical samples and

correlation with disease pathogenesis.

Hypothesis 2: Syncytin-1-mRNA and DNA copy numbers are increased in

tissues from MS patients.

Objective 3: Investigate the mechanisms leading to Syncytin-1-induced ER stress

and the consequences of ER stress on oligodendrocyte viability.

Hypothesis 3: Syncytin-1 induces ER stress in astrocytes resulting in cellular

dysfunction subsequently affecting oligodendrocyte survival.

Chapter 2: MATERIALS AND METHODS 93

CHAPTER 2

MATERIALS AND METHODS

Chapter 2: MATERIALS AND METHODS 94

II.1. Cell culture

II.1.1. Primary cells

Oligodendrocytes were derived from adult Sprague-Dawley rat brains. After

isolating brains from rats in sterile conditions, they were rinsed in PBS and minced with

sterile scalpels in a plastic dish. The contents were pipetted into a 100 ml flask. Two ml

of DNase I (1 mg/ml stock solution; Sigma) and 10 ml of 2.5% trypsin (final concentration 0.25%; Invitrogen) were added to the contents and stirred at 37oC for 45 min. Later, 3 ml of heat-inactivated FBS (Invitrogen) was added to the contents to inhibit trypsin activity. The cell suspension was then forced through a 132 μm mesh into a sterile glass bottle using a plunger. The contents were washed liberally with PBS and centrifuged at 1200 rpm for 10 min at room temperature and low brakes. The supernatant was discarded and the pellets were pooled in PBS up to 21 ml and then 9 ml of Percoll

(Sigma) was added to the contents. The mix was then added to sterile polycarbonate tubes and centrifuged at 15000 rpm for 30 min at 4oC without brakes. The myelin layer

was aspirated off and the cell layer was removed and pooled in 50 ml tubes and topped

up with PBS. The contents were washed by centrifugation at 2000 rpm for 10 min at

room temperature and low brakes. The resulting cells were incubated in RBC lysis buffer

(ammonium chloride, 0.15 M; potassium carbonate, 10 mM; EDTA, 0.1 mM in distilled

water) for 5 min followed by a final wash in PBS at 1200 rpm under similar conditions

and resuspended in feeding medium (500 ml MEM (Invitrogen), 0.2 mM glutamine

(Invitrogen), 0.1 % Dextrose (BDH, Canada), 2.5 ml penicillin-streptomycin (Invitrogen)

and 10% FBS). The cells were counted, and after adjusting the cell density to 3 x 106/ml, flasks were seeded and incubated overnight at 37oC. Floating cells were harvested by

Chapter 2: MATERIALS AND METHODS 95

centrifugation. Oligodendrocytes thus obtained were seeded onto poly-ornithine coated

chamber slides at a density of 50,000/well.

Human fetal astrocytes (HFA) cultures were a gift from Dr. VW Yong

(University of Calgary). 12-14 week old fetal brain tissue was minced and 3 ml of 2.5%

trypsin and 6-8 ml of DNase-I was added to the tissue. Cells were passed through a 125

μm mesh and washed with PBS. Cells were centrifuged at 1200 rpm for 10 min and resuspended in culture medium comprising MEM supplemented with 10% FBS, 1%

glutamine and antibiotics (penicillin-streptomycin). Cells were counted and seeded on

poly-ornithine coated plates at a density of 3-5 x106 cells/ml. Human fetal neurons (HFN)

culture were a gift from Dr. VW Yong (University of Calgary). Neurons were purified

from the mixed cultures obtained above by treating cells with 25 μM cytosine B-D-

arabinofuranoside (Ara-C; Sigma), which selectively blocks DNA synthesis. Adult human oligodendrocytes (HOLs), also a gift from Dr. VW Yong (University of Calgary), were derived from brain biopsy specimens from patients who underwent surgical

resection to ameliorate drug-intractable epilepsy. Preparation procedures followed are as

described above for adult rat oligodendrocytes.

Bone-marrow-derived macrophages (BMDM) were obtained from 8-10 week old

mice. Mononuclear phagocyte progenitor cells derived from femoral and tibial bone

marrow were propagated in the presence of M-CSF. This macrophage growth factor is

secreted by L929 cells and is used in the form of L929 cell conditioned medium. The

progenitor cells proliferate and differentiate through monoblast, promonocyte and

monocyte stages before maturing to macrophages. At this time, the cells adhere firmly to the culture vessel. After sacrificing animals by carbon dioxide CO2 overdose and a

Chapter 2: MATERIALS AND METHODS 96

thorough wash with 70% alcohol, the skin is removed from the lower part of the body.

The tissue from legs is removed and the limb is dissected out. In order to prevent

contamination of the marrow preparation with extraneous cells that could potentially

overgrow the macrophages, the remaining tissue from the pelvic and femoral bones are

removed and then separated at the knee joint. Each end of bone is cut with a scalpel and

using a 25-gauge needle and a 12 cc syringe filled with bone marrow medium (DMEM

supplemented with antibiotics (penicillin-streptomycin) and glutamine [29.2 mg/1000

ml]), the bone marrow is expelled from both ends of the bone with a jet of medium

directed into a 50 ml screw top Falcon tube. The cells are washed by centrifugation at

1000 rpm for 10 minutes and resuspended in 10 ml bone marrow medium. The cell

aggregates are broken up by gentle aspiration using an 18-gauge needle attached to a 12

cc syringe. After bringing the sample volume to 40 ml with bone marrow medium, the

cells are counted and suspended to a concentration of 11.0 × 106 cells/12 ml and

dispensed into culture dishes and incubated for 5 to 7 days at 37° under 10% (v/v) CO2.

On day 5, half of the media is replaced followed by a complete change on day 6.

To obtain and culture human monocytes-derived macrophages (MDMs) and peripheral blood lymphocytes (PBLs), blood from healthy donors was diluted at 50% in

RPMI (Invitrogen) medium. 25 ml of diluted blood was overlaid on 15 ml Histopaque

(Sigma) and centrifuged for 20 min at 1800 rpm with low brakes. The buffy coat was removed and washed with RPMI by centrifugation at 1200 rpm for 10 min followed by a second wash at 800 rpm for 10 min. Cells were resuspended in 12% RPMI supplemented with antibiotics (penicillin-streptomycin). Cells were counted and seeded onto poly- ornithine-coated tissue culture plates at a density 10 times more than the final number of

Chapter 2: MATERIALS AND METHODS 97

MDM required per well. After allowing cells to adhere for 3h, the medium was changed and 20% (10% FBS and 10% human serum or L-929 (macrophage-colony stimulating factor rich) medium) RPMI was added. MDMs were used for experiments after 7 days in culture. Human PBLs were obtained from the cultures described above for MDM. Cells that did not attach to the poly-ornithine coated plates were identified as peripheral blood leukocytes (PBLs). All primary cultures were grown and maintained in MEM with 10% glucose.

II.1.2. Cell lines

BHK-21 cells (ATCC, Manassas, VA) were cultured in MEM with 1% sodium pyruvate. U373 astrocytoma, HeLa 340 and HEK293T cells (ATCC, Manassas, VA), were cultured in DMEM. U937 monocytoid cells (ATCC, Manassas, VA) and MYA-1 feline cells 341 were grown in RPMI. Cells were cultured in media with 10% FBS unless otherwise mentioned. Human LAN-2 neuronal cells were proliferated in 10% MEM with

1% N2 supplement (Invitrogen) and differentiated in L-15 medium (Invitrogen) with 50

μg/ml gentamicin (Invitrogen) and cyclic adenine monophosphate (AMP) (1 mM;

Sigma). Cell lines were used when they were between passage numbers 10-25.

II.2. Syncytin-1 constructs

II.2.1. Construction of SINrep5-Syncytin-1 plasmid

The Sindbis virus-based (SIN) vector system used in this study has been described previously 342. The constructs pSINrep5-EGFP and pSINrep5-Syncytin-1 were obtained by cloning the EGFP and HERV-W env (Syncytin-1) genes, respectively, into the multiple cloning site present in the pSINrep5 expression vector as described 342. Briefly, to construct pSINrep5-EGFP, pEGFP-N1 (Clontech, Palo Alto, CA) was digested with

Chapter 2: MATERIALS AND METHODS 98

SmaI and HpaI, and the resulting 862 bp DNA fragment, containing the EGFP ORF, was

isolated from an agarose gel using the Concert Rapid Gel extraction kit (Life

Technologies Inc., Burlington, ON) and inserted into the StuI restriction site of the

pSINrep5 multiple cloning site. The Syncytin-1 envelope insert was obtained by

digesting the expression vector phCMVenvpH74 334 with EcoR1 and Xba1 and subsequent isolation from an agarose gel. The 1.832 kbp fragment was inserted into the

EcoR1 and Xba1 restriction sites of pBluescript SK+. The resulting construct, pBS-

Syncytin-1, was digested with EcoRV and Xba1 to generate a fragment containing the

Syncytin-1 ORF, which was subsequently cloned into the StuI site of the multiple cloning site of pSINrep5 (Appendix A). For all constructs, correct insertion of the inserts was determined by restriction enzyme digest analysis and sequencing. All restriction and other enzymes were obtained from New England Biolabs (Mississauga, ON) or Life

Technologies (Burlington, ON) and used according to their specifications.

II.2.2. Preparation and titration of viral stocks

Constructs pSINrep5-EGFP and pSINrep5-Syncytin-1 and helper virus construct

pDH-BB 343,344 were linearized with XhoI. The linearized plasmids were used as

templates for the generation of capped RNA transcripts by in vitro run-off transcription

using the SP6 mMESSAGE mMACHINE kit (Ambion Inc, Austin, TX). 10 μg of DH-

BB RNA transcript and 10 μg SINrep5-EGFP or SINrep5-Syncytin-1 RNA transcript

were transfected into 3-5 x 106 BHK-21 cells/ml in diethyl pyrocarbonate (DEPC)-treated

PBS by electroporation according to a previously published protocol 343. Briefly, the cells

were subjected to two pulses at 850V, 50 μF and infinity (∞) resistance (Ω), using a Gene

Chapter 2: MATERIALS AND METHODS 99

Pulser II (BioRad Laboratories Canada Ltd., Missisauga, ON). Cells were plated in

culture media and incubated at 37°C/5% CO2. The media containing recombinant

SINrep5 was harvested 20-24 hr after transfection. When high titers were needed, the

SINrep5-Syncytin-1 viral stock was concentrated by centrifugation at 120,000 g at 4°C for 4 hr, and the virus pellet dissolved in culture media. The recombinant SINrep5 stocks

(Fig. 12) were titered on BHK cells. For SINrep5-EGFP, the titer was determined by counting the number of EGFP-positive cells using fluorescent microscopy 24 hr post infection. To determine the titer of SINrep5-Syncytin-1, cells were immunostained using monoclonal antibody 6A2B2 334 directed against the Syncytin-1 envelope protein and the

number of Syncytin-1-positive cells were counted. The average of the numbers of

Syncytin-1 immunopositive BHK cells in each well in chamber slide at dilutions of 1:100

and 1:1000 were counted. Viral titer = Average x dilution (100 or 1000) x 1000/100 per

mL. On average, 5 x 106 to 3 x 107 infectious virus particles per ml were obtained for

SINrep5-EGFP and 5 x 106 particles per ml for SINrep5-Syncytin-1.

II.2.3. Construction of pFGH-Syncytin-1 envelope plasmid

(5’-AAGGAATAAAGCGGCCGCATGGCCCTCCCTTATCATATCTTTC-3’) and (5’-

AAAAGGAAAAGCGGCCGCCTAACTGCTTCCTGCTG-3’) oligonucleotides with

Not1 tags (underlined) in both sequences and a silent mismatch (C, italicized) in the sense

sequence were designed. The Not1 restriction sites allowed PCR amplification and

cloning into suitable vectors, whereas the silent mismatch prevented five ‘T’s in the

sequence. PCR was performed with phCMVph74 334 as the template, resulting in a 1.8 kb

product. The pFGH vector containing the GFAP promoter (kindly provided by Iain

Chapter 2: Materials and Methods

DHBB SINrep5- Syncytin-1 Syncytin-1

REPLICASE EGFP

REPLICASE REPLICASE

Syncytin-1 Structural proteins (Capsid/envelope proteins)

Translation of Syncytin-1 Specific assembly of SINrep5 genome in virus particle

SINrep5-Syncytin-1 virus

Fig. 12: Sindbis virus vector expressing Syncytin-1 (Adapted from Schlesinger, S (2000)

100 Chapter 2: MATERIALS AND METHODS 101

Campbell, University of Sydney) and the PCR product were digested with Not1

restriction enzyme. The PCR product was cloned into pFGH to obtain the construct,

pFGH-Syncytin-1.

II.2.4. Construction of pseudotyped virus

Pseudotyped virions expressing Syncytin-1 were generated by co-transfecting

293T cells with plasmids expressing firefly luciferase within an envelope-inactivated

HIV-1 clone (pNL-Luc-E-R-) 340 and the expression vector pCMV containing the full-

length Syncytin-1 sequence (pCMV-Syncytin-1) 334 or pCDNA 3.1 (+) (Clontech, CA,

USA). Transduction of target cells by pseudotyped virus led to expression of luciferase,

which was quantified in cells, lysed 48 h following infection, using the Luciferase Assay

Kit (Pharmingen, Canada). For experimental purposes, equal numbers of target cells were treated with 100 μl per well (96 well plate) of supernatant containing pseudotyped virus.

II.2.5. Soluble Syncytin-1 protein expression

Five μg of pVGW427 vector (a gift from Dr. Francoiş Mallet) expressing soluble

Syncytin-1 was transfected into HEK 293T cells using Lipofectamine-2000 (4 μl;

Invitrogen) and 100 μg of soluble Syncytin-1 protein was used for various experiments or

stored at –80oC.

II.3. Transfections

Constructs (5 μg) expressing various ER stress proteins were transfected into

astrocytes using Transfectin Lipid reagent (5 μl; Biorad) following the manufacturer’s

protocol. CHOP was obtained from David Ron (NYU School of Medicine); OASIS was a

kind gift from Kazunori Imaizumi (University of Miyazaki).

Chapter 2: MATERIALS AND METHODS 102

II.4. Extraction of protein from CSF and plasma

Protein from CSF, plasma and cell culture supernatants was precipitated by

adding 9 volumes of sample to 4 volumes of buffer (10% trichloroacetic acid, 0.07% β- mercaptoethanol in acetone) as described previously 345. Following centrifugation at

15000 g for 30 min at 4oC, 4 volumes of acetone containing 0.07% β-mercaptoethanol

were added to the pellet and incubated for 1 h at -20oC followed by centrifugation as

above. The pellet was air-dried and incubated for 90 min at room temperature in 2x

Laemmli buffer that was pre-heated to 95-100oC.

II.5. Antioxidant activity

The antioxidant activity of the test drugs was measured using a

spectrophotometric DPPH assay as described previously 346. NCX-2216 and ferulic acid

were dissolved in 95% ethanol after which serial dilutions of each drug were made in

95% ethanol. An aliquot (20 µl) of each drug, at various dilutions, was added to the wells

of a 96-well plate. A stable free radical [1,1-diphenyl-2-picrylhydrazyl (DPPH)], also

dissolved in 95% ethanol (180 µl), was then added to each well, and absorbance at 540

nM was monitored over a 15-min period. DPPH is a purple-colored substance, which is

converted to a colourless substance in the presence of antioxidants. The concentration of

each test compound that reduced absorbance to 50% was then calculated (EC50). Ascorbic

acid was included in this analysis as a positive control. All test compounds were assessed

at concentrations ranging from 1 to 300 µM.

Chapter 2: MATERIALS AND METHODS 103

II.6. Infection and treatment of cells

II.6.1. Infection of cells

All SINrep5 infections were performed with the same number of virus particles

(multiplicity of infection (MOI) of 1.0). HFA and MDM (5×104/well) were seeded in 16-

well chamber slides and infected with SINrep5-Syncytin-1 or SINrep5-EGFP (MOI 1.0

each) or mock conditioned medium (CM).

II.6.2. Treatment with cytokines

Cells were treated with human recombinant TNF-α, IL-1β, IL-10 (10 ng/ml, R &

D Systems, MN, USA) and IFN-β (100 U/ml, Serrono, USA).

II.6.3. Treatment with drugs

To assess the effect of astrocyte and monocyte stimulation on HERV expression,

U373, PBL and U937 cells were treated with 50 ng/ml of PMA (phorbol-12-myristate-

13-acetate) (Sigma, Canada) for 4-72 hours prior to harvest and RNA isolation as

described previously 198. Cells were treated with minocycline (10 μM, Sigma), sodium nitroprusside, an NO donor (100 nM, Axxora Life Science Inc., San Diego, CA, USA),

NBQX (30 µM; Sigma), MK-801 (30 µM; Sigma), glatiramer acetate (25 μg/ml; Teva,

Israel), IFN-β (100 U/ml; Serrono, Oakville, ON, Canada), ferulic acid (0.005, 0.5, 5, 50 and 250 μM; Sigma), NCX-2216 (60 nM, 600 nM and 6 μM; generously provided by

NicOx S.A., Nice, France), L-NIL (N6- (1-iminoethyl)-lysine, hydrochloride) (0.5 μM,

Sigma), L-NAME (NG-nitro-L-arginine methyl ester, a non-specific NOS inhibitor) (5.0

μM, Sigma), IGF-1 (3 ng/mL, R&D Systems, MN, USA) and Growth Hormone (100

μg/mL; GH, Serrono, USA); Thapsigargin and Tunicamycin (5 ng/ml; Sigma) were gifts from Dr. M. Marek (University of Alberta, Canada).

Chapter 2: MATERIALS AND METHODS 104

II.7. Animals and in vivo procedures

II.7.1. Stereotaxic implantation with SINrep5-Syncytin-1 virus

SINrep5-EGFP or SINrep5-Syncytin-1 virus (0.5 x 106 particles/ml in 3 μl) was stereotaxically implanted bilaterally into the corpus callosum of 10-week old CD1 mice

(n=6 for each treatment). The corpus callosum was chosen since this region of the brain is predominantly white matter with myelin-producing oligodendrocytes. Control mice (n=6) were implanted with conditioned medium from mock-infected cultures (1 mm anterior,

2.0 mm lateral and 1.5 mm deep relative to bregma). In addition, the right striatum of 10- week old CD1 male mice (n = 4 for each treatment) was stereotaxically implanted with

SINrep5-EGFP and SINrep5-Syncytin-1 (0.5 x 106 particles/ml). The striatum was chosen since this region of the brain is predominantly rich in neurons. The coordinates for the striatal implant were 3 mm posterior, 2.5 mm lateral, and 3 mm deep relative to the bregma (Fig. 13).

II.7.2. Syncytin-1 transgenic mouse

The construct pFGH-Syncytin-1 was digested with EcoR1 to produce a fragment of 5 kb that was used for pronuclear microinjection. We produced three independently- derived transgenic (Tg) lines on a CD1 background, bearing the Syncytin-1 transgene.

For genotyping, we isolated genomic DNA from tail biopsies by lysis in 0.25 mg/ml proteinase K (Sigma) in 1% SDS, 100 mM NaCl, 100 mM EDTA, 50 mM Tris, pH 8 at

55°C overnight followed by salt extraction and ethanol precipitation. We determined the presence of the transgene using PCR and the Syncytin-1 primers described in Table 3.

Genotyping was further confirmed by a second set of Syncytin-1 primers (5'-

ACCCATACCTCAAACCTCACCTG-3' and 5'-CTTTTGTTGCGGGGCTTAGATA-3').

Chapter 2: Materials and Methods

Fig. 13: Stereotaxic implantation of SINrep-Syncytin-1/EGFP/mock into corpus callosum (arrows) and striatum (arrowheads).

105 106

Table 3: List of Primers used in the study

Primer Sequence Genbank ID Primer Location HERV-W env 5’-TGCCCCATCGTATAGGAGTCT-3’ and NM_014590.3 1187-1207 Syncytin-1 5’-CATGTACCCGGGTGAGTTGG-3’ 1464-1445 HERV-W env 5'-ACCCATACCTCAAACCTCACCTG-3' and NG_004112 8870-8893 Syncytin-1* 5'-CTTTTGTTGCGGGGCTTAGATA-3' 9240-9219 HERV-H env 5’-TTACCCCATCATCAGTCCCCATTAC-3’ and AJ289709 870-895 5’-GAGCTCTTCGGTCCCATTTG-3’ 1125-1105 HERV-E env 5’- TCCCCTGTCCTCCTGCTCTTT -3’ and HEU34991 25-46 5’- AGGGTTGTCTGGGCTTGGTCT -3’ 491-511 HERV-K 5’- CCTGCAGTCCAAAATTGGTT -3’ and X82272 955-975 (HML2) env 5’- GGGGCAAGTTTTCCCTTTAG -3’ 1120-1100 HERV-R env 5’-CCATGGGAAGCAAGGGAACT-3’ and NM_001007253.1 793-812 5’-CTTTCCCCAGCGAGCAATAC-3’ 933-914 HERV-FRD 5’-GCCTGCAAATAGTCTTCTTT-3’ and BC068585.1 631-650 env 5’-ATAGGGGCTATTCCCATTAG-3’ 744-725 (Syncytin-2) HERV-T env 5’-CCAGGATTTGATGTTGGG-3’ and AJ862655.1 640-657 5’-GGGGTGAGGTTAAGGAGATGG-3’ 884-865 IL-10 5’-CCTCTCACCGTCTTGCTTTC-3’ and DQ217938.1 4457-4476 5’-GCAGAGGTTGCTTGTTCTCC-3’ 4710-4691 IL-1β 5’-CCAAAGAAGAAGATGGAAAAGCG-3’and NM_000576.2 706-728 5’-GGTGCTGATGTACCAGTTGGG-3’ 807-787 iNOS 5’- CAAAGGCTGTGAGTCCTGCAC -3’ and NM_000625.3 2418-2398 5’- ACTTTGATCAGAAGCTGTCCC -3’ 2203-2186 nNOS 5’- TCAGTCTCCCAGGCTAATGG-3’ and NM_008712.1 609-628 5’- CTGTCCACCTGGATTCCTGT-3’ 808-789 Alu 5’-GTGGATCACCTGAGGTCAGGAGTTTC-3’ Universal Alu NCBI Primer Handbook Syncytin-1- 5’-GGCAAAGACAGGAGGTAAAGAAAT-3’ and AF520550.1 2103-2126 LTR 5’-TGAAAACAGCTCCCATACAAAG-3’ 293-272 PLP 5′-CTTCCCTGGTGGCCACTGGATTGT-3′ and NM_011123.2 128-151 5′-CCGCAGATGGTGGTCTTGTAGTCG-3 407-384 MOG 5′-CCTCTCCCTTCTCCTCCTTC-3′ and NM_010814.1 39-58 5′-AGAGTCAGCACACCGGGGTT-3′ 470-451 CNPase 5′-CTACCCTCCACGAGTGCAAGACGCT-3′ and BC021904.1 147-171 5′-AGTCTAGTCGCCACGCTGTCTTGGG-3’ 485-461 OASIS 5’-CAACGCACCCCACTCACAGACACC-3’ and AB063321.1 1975-1998 5’-GGAGCAGCAAAGCCCGCACTAACT-3’ 2171-2148 GADD153 5’-AACCAGCAGAGGTCACAAGC-3’ and S40706.1 377-396 5’-AGCCGTTCATTCTCTTCAGC-3’ 593-574 CGT 5’-TTATCGGAAATTCACAAGGAT-3’ and X92122.1 1710-1730 5’-TGGCGAAGAATGTAGTCTATC-3’ 1786-1766 IFN-α 5’-GTGATCTCCCTGAGACCCAC-3’ and NM_006900.2 101-120 5’- GGTAGAGTTCGGTGCAGAAT-3’ 409-390 TNF-α 5’-ATTCAGGAATGTGTGGCCTGC-3’ and NM_000594.2 1041-1061 5’-GTTTGAATTCTTAGTGGTTGCCAG-3’ 1096-1073 107

PERK 5’-AAGTAGATGACTGCAATTACGCTATCAA-3’ NM_004836.3 2005-2032 and 2090-2070 5’-TTTAACTTCCCGCATTACCTTCTC-3’

ERp57 5’-TCAAGGGTTTTCCTACCATCTACTTC-3’ and NM_005313.4 1493-1513 5’-TTAATTCACGGCCACCTTCAT-3’ 1568-1548 BiP 5’-TCATCGGACGCACTTGGAA-3’ and NM_022310.2 523-541 5’-CAACCACCTTGAATGGCAAGA-3’ 591-571 ASCT1 (H) 5’-TCCCCATAGGCACTGAGATAGAAG-3’ and BC026216.1 819-842 5’-CAAGGAACATGATGCCCACAGGTA-3’ 1013-990 ASCT2 (H) 5’-CCTGCTGGGGGTGCTCTTTGGACA-3’ and NM_005628.1 2240-2263 5’-TTGAGTTGGGGACATGAGTGAGAA-3’ 2448-2425 ASCT-1 (M) 5’-CCTGGCTTGATGATGAACGC-3’ and BC043483.1 624-605 5’-CTGGTGCTGCTCACCGTGTC-3’ 348-329 ASCT-2 (M) 5’-CCATCGGCGCCACGGTCAACAT-3’ and NM_009201.1 1669-1690 5’-GTGGCGAGGGGCAGTGGATTCAGA-3’ 2071-2048 Egr1 5’-AGCAGCACCTTCAACCCTCA-3’ and BC073983.1 509-528 5’-CAGCACCTTCTCGTTGTTCAGA-3’ 610-589 Egr3 5'-TTGGGAAAGTTCGCCTTCG-3 and NM_004430.2 614-632 5’-ATGATGTTGTCCTGGCACCA-3’ 666-647 Egr4 5'-CCCCGCTGGATGCCCCTTTTC-3' and NM_001965.1 537-557 5'-ACTCTCCGCCGTCGCCGCTACTCC-3' 1073-1050 EAAT1 5’- GCGGGCCTGGTCACTATGGT -3’ and NM_004172.3 1991-1968 5’- AGAAGGGAGGAAAGGGAAGATGAC -3’ 1580-1599 GAPDH** 5’-AGCCTTCTCCATGGTGGTGAAGAC-3’ AND AY841947.1 214-191 (R) 5’-CGGAGTCAACGGATTTGGTCG-3’ BC002547.1 70-90 (H)

* Oligonucleotide primers used for genotyping Syncytin-1 transgenic mice ** Oligonucleotide primers designed to amplify GAPDH from a variety of species H: Human R: Rat M: Mouse Chapter 2: MATERIALS AND METHODS 108

The PCR mix contained approximately 100 ng genomic DNA, 200 μM dNTPs, 100 nM

each primer, 2 units Taq DNA polymerase I (5 units/μl; Invitrogen) in 1x reaction buffer

supplemented with 1.5 mM MgCl2 (Invitrogen). A 267 bp fragment was amplified under

the following PCR conditions: 94°C for 1 min, 56°C for 2 min, 72°C for 1 min for 40

cycles, then 72°C for 10 min. For stereotaxic implantations, 10-week transgenic and wild

type (WT) littermate controls were stereotaxically implanted with TNF-α or PBS

bilaterally into the corpus callosum (n=6 for each treatment) (1 mm anterior, 2.0 mm

lateral and 1.5 mm deep relative to bregma). All studies and procedures were carried out following University of Calgary Animal Care Committee guidelines.

II.7.3. Behaviour studies

Behavioural tests were conducted in mice that were injected with the SINrep5-

Syncytin-1, SINrep5-EGFP virus and the mock-infected conditioned medium on days 3,

7, 10 and 14 days post implantation. The horizontal bar test involved a test of co-

ordination and forelimb strength using a horizontal bar that was 0.2 cm thick, 38 cm long,

held 49 cm above a bench 347. The mouse was positioned to hold the bar using its

forelimbs and the time taken to either fall off the bar or reach the ends of the bar was

noted. The static rod test involved a test of co-ordination using five rods each 60 cm long

and of varying thickness (diameter) (Rod 1: 35 mm; Rod 2: 28 mm; Rod 3: 22 mm; Rod

4: 15 mm and Rod 5: 9 mm). These rods were bolted to the edge of a bench such that the

rods horizontally protruded their full 60 cm length into space. A mouse was placed at the

exposed end of the widest rod and the time taken to orient 180 degrees from the starting

position and the time taken to travel to the other end were noted 347. A test of strength and

seeking behavior was performed using the inverted screen test. The invertible screen was

Chapter 2: MATERIALS AND METHODS 109

a 43 cm2 area of wire mesh consisting of 12 mm squares of 1 mm diameter wire and

surrounded by a 4 cm deep wooden beading 347. We modified the inverted screen test by

placing the mouse at a point that was equidistant from the edges of the screen and the

stopclock was started once the screen was inverted and the time taken to reach the edge

of the inverted screen was determined, as a measure of curiosity and seeking behaviour.

Rotary behaviour of the animals implanted in the striatum was analyzed at 3, 7, 10 and 14

days after implantation, as described previously 342. Briefly, total and ipsiversive percent

rotation was assessed over 10 minutes after intraperitoneal injection of amphetamine (1 mg/kg, Sigma) at days 3, 7, 10 and 14 following intrastriatal injection.

II.7.4. Oral drug treatment

For treatment with ferulic acid, mice (n=6) were implanted with SINrep5-

Syncytin-1 followed by daily oral gavage with ferulic acid (20 mg/kg on a daily basis) for

14 days. Animals were sacrificed at day 14 and intracardially perfused with saline followed by 4% paraformaldehyde (PFA).

II.8. PCR

II.8.1. Isolation of DNA and RNA and cDNA preparation

Approximately 100 mg brain tissue or 1 x 106 cells were homogenized in Trizol

reagent (Invitrogen) followed by phase separation in Trizol-chloroform. The resulting

aqueous phase was used for RNA extraction while the inter- and -phenol phases were

used for DNA extraction. DNA was precipitated from the inter- and phenol phases by

ethanol, washed in a solution containing 0.1 M sodium citrate in 10% ethanol and the

DNA pellet was finally dissolved in 8 mM NaOH. Quality and quantity of DNA thus

obtained was measured spectrophotometrically (Appendix B). PBMC-derived DNA from

Chapter 2: MATERIALS AND METHODS 110

a cohort of Canadian MS patients and non-MS controls was obtained from Dr. George

Ebers (University of Oxford). Ten ng per reaction of DNA was used for PCR. Total RNA

and cDNA were prepared from brain tissue or cells. RNA was isolated using Trizol

reagent (Invitrogen) following the manufacturer’s protocol. Total RNA (encapsidated and non-encapsidated) was extracted from CSF and plasma of control and MS patients by

Trizol LS reagent (Invitrogen) following the manufacturer’s instructions. Briefly, after chloroform extraction, the resulting aqueous phase was treated with isopropanol and the

RNA was precipitated. The RNA pellet thus obtained was washed with 75% ethanol in

DEPC-treated water and finally suspended in DEPC water for cDNA preparation with the following conditions. Approximately 3 μg of RNA was dissolved in 8 μl of DEPC water and was subjected to RNase-free DNase-1 (Promega, Madison, WI) treatment for 45 min

o o at 37 C and heat inactivated at 65 C for 10 min. After addition of dNTP and random N6

oligomers to prime cDNA synthesis (Invitrogen, Burlington, ON), the reaction was

performed at 70oC for 5 min and reverse-transcribed at 37oC for 1 hour using

SuperscriptII (Invitrogen) to a final volume of 50 μl. Enzymes were heat-inactivated at

90oC for 5 min and the cDNA thus obtained was diluted 3-4 times in water before PCR

amplification.

II.8.2. Relative quantification real time RT-PCR

Relative quantification real time PCR analysis of cDNA or genomic DNA levels

in brain was performed. This was achieved by monitoring in real-time, the increase of

fluorescence of SYBR-green dye on a Biorad iCycler. A threshold cycle (Ct) value for

each gene of interest was calculated by determining the point at which the fluorescence

exceeded a threshold limit (12-fold increase above the standard deviation of the initial

Chapter 2: MATERIALS AND METHODS 111

baseline). To confirm single-band production, a melt-curve analysis was performed and

subsequently confirmed by electrophoresis and ethidium bromide staining. The 2-∇∇Ct method was used to quantify relative gene expression 348. The calculation assumes that

the amplification efficiencies of target and reference genes are equal. The change in

expression of the target gene was normalized to that of housekeeping genes such as β-

actin or GAPDH and expressed as mRNA relative fold change (RFC). Triplicate samples

were collected from the real-time RT-PCR experiment. The data were analyzed using the

-∇∇Ct equation 2 , where -∇∇Ct = (Ct (target)-Ct (housekeeping gene) CONTROL- Average (Ct (target)-Ct

(housekeeping gene) TREATMENT). Total RNA from CSF and plasma samples was subjected to

DNase treatment and a combined RT-PCR was done with Syncytin-1 specific primers

(Table 3) generating a 278 bp PCR product. PCR analysis of brain tissue was performed

using the primer pair 5'-TGCCCCATCGTATAGAGTCT-3' and 5'

CATGTACCCGGGTGAGTTGG-3'. The product was sequenced and confirmed to be

that of Syncytin-1.

II.8.3. Quantitative real time PCR

The construct, pBS-Syncytin-1, contained the 1.832 kbp Syncytin-1 env gene cloned into the EcoR1 and Xba1 sites downstream of the T3 promoter in pBluescript SK

II (+) (See section II.2.2.). Ten-fold serial dilutions of this clone were used to obtain the

DNA standard used for real time PCR to determine DNA copy numbers. Serial dilutions

of U937 cells were performed prior to extraction of genomic DNA for developing a

GAPDH standard. To obtain in vitro RNA transcripts to develop the RNA standard curve,

pBS-Syncytin-1 plasmid was linearized with XhoI, treated with proteinase-K (0.2 mg/ml,

0.5% SDS, 50°C for 30 minutes) and purified by phenol-chloroform extraction and

Chapter 2: MATERIALS AND METHODS 112

ethanol precipitation. About 2-3 μg of linearized DNA was used as template to generate in vitro run-off RNA transcripts using a T3 in vitro RNA transcription Kit (Ambion,

Austin, TX). RNA transcripts were purified by phenol-chloroform extraction and

isopropanol precipitation. Genomic DNA and/or total RNA were measured by

spectrophotometry (Appendix B). For quantitation of Syncytin-1 DNA or RNA levels, a

standard curve was generated with serial 10-fold dilutions (1011 copy/8μg to 1 copy/8μg)

of the DNA plasmid (pBS-Syncytin-1) or cDNA derived from the in vitro transcribed

RNA, respectively. Subsequently, the in vitro RNA transcripts were treated with RNase-

free DNase (Promega, Madison, WI) and reverse-transcribed using SuperscriptII

(Invitrogen) and random N6 oligomers to prime cDNA synthesis (Invitrogen, Burlington,

ON). The cDNA was diluted 1:1 with sterile water and 5 μl were used per PCR reaction.

Cycle threshold values from relative quantification RT-PCR experiments were analyzed

for levels of expression of several genes using equal concentrations of cDNA as templates

for the PCR. A real-time PCR protocol, using primers that amplified Syncytin-1 gene,

described in Table 3, was developed to determine the number of copies of Syncytin-1

encoding DNA and RNA sequences/μg of brain or RNA/μL of plasma or CSF.

II.8.4. Sequencing of virus-cell junctions

Primers were designed to PCR amplify circular, single and/or double LTR

sequences. Virus-host cell sequence junctions that included HERV-W7q LTR (U5) and

the universal Alu primer sequence were also designed (Table 3). PCR amplification of

either 780 or ~1500 bp fragments indicating single or double LTRs (Fig. 14) respectively

was performed and shown as Circ (+) if a PCR product was amplified and Circ (-), if not

amplified. First round PCR assay was performed using Syncytin-1-sense and Alu primers

Chapter 2: Materials and Methods

Preintegration complex 2-LTR circle

1-LTR circle

Target DNA Terminal cleavage, strand transfer

Integration intermediate

Repair Provirus

Fig. 14: Schematic of retroviral LTR circles and formation of provirus

113 Chapter 2: MATERIALS AND METHODS 114

(Table 3). A second round PCR was performed using 5 μl of the first round reaction with

HERV-W7q LTR (U5)-sense and Alu primers (Fig. 15). Results were expressed as percent detection of PCR amplified products.

II.9. In vitro assays

II.9.1. Toxicity of astrocyte conditioned medium

Adult rat oligodendrocytes and LAN-2 neurons were treated with conditioned

medium (diluted 1:1 with AIM-V medium). Conditioned media for toxicity assays were

obtained from HFA and MDM infected with SINrep5-Syncytin-1 or SINrep5-EGFP.

Cells were examined for cytotoxicity by trypan-blue dye exclusion for up to 5 days.

Culture medium in 96 well plates or chamber slide (Nunc) was carefully removed by

pipetting, followed by addition of trypan-blue dye (50 μl per well). After incubating cells

for 5 min, the trypan-blue dye was carefully removed and cells were washed twice with

PBS. Cells were counted on a Zeiss inverted microscope using the 10X objective. All

experiments were repeated at least three times. For measurements of cellular injury,

oligodendrocytes were immunostained with antibody to Gal-C, CNPase and MBP, and

cells with and without processes were counted. In addition, oligodendrocyte injury was

also measured by in-cell western analysis using quantitative immunofluorescence

(section II.11).

II.9.2. Analysis of supernatant for protein carbonyls and 4-HNE

The level of protein oxidation was determined by an oxidized protein detection kit

(OxyblotTM, ONCOR) 349. Samples were incubated for 20 min with 12% SDS and 2,4- dinitrophenylhydrazine (DNPH) in 10% trifluoroacetic acid, vortexing every 5 min, and

then neutralized with OxyblotTM Neutralization solution. 600 ng of protein was blotted

LTR-F ltr gag pol env LTR-F

Alu LTR-R LTR-R

Fig. 15: Location of oligonucleotide primers for PCR amplification of virus-host cell junctions and quantification of Syncytin-1 DNA copy numbers

115 Chapter 2: MATERIALS AND METHODS 116

onto nitrocellulose paper by the slot blotting technique. Membranes were incubated with

blocking buffer for 30 min at room temperature, exposed to rabbit anti-DNPH protein

antibody (1:150) for 90 min, followed by anti-rabbit IgG coupled to alkaline phosphatase

(1:15,000) for 2 h at room temperature. Following washing and development with

SigmaFastTM chromogen, blots were analyzed by computer-assisted imaging software,

Scion Imaging. Samples for HNE detection were similarly analyzed by slot blotting

technique except that rabbit-anti-HNE antibody (1:4,000; Calbiochem) was used as a

primary antibody as described 349. Results were expressed as relative fold change (RFC)

over levels in AIM-V medium.

II.10. Microarray Analysis

U373 astrocytes were infected with Sindbis virus-derived clones (0.5 x 106) or mock-infected for 24 h in AIM-V medium. Total cellular RNA from infected astrocytes

was purified and subjected to hybridisation using Affymetrix Human Genome U133 Plus

2 arrays. Following purification of RNA, discrete 28S rRNA and 18S rRNA peaks were

observed. RNA concentrations were determined spectrophotometrically and all samples

had an OD260/OD280 ratio of 1.7-1.9. RNA amplification was followed by hybridization,

washing, staining and scanning as described in the Affymetrix GeneChip Expression

Analysis Technical Manual. Relevant sections are provided in Appendix C.

Hybridizations were carried out at 45°C for 16 h. Following hybridization, the arrays

were washed and stained with streptavidin–phycoerythrin in the Affymetrix Fluidics

Station 400, using the standard antibody amplification protocol. Arrays were scanned

with the Affymetrix GeneArray Scanner at 488 nm and 3 μm per pixel. Relative expression values were calculated using GeneChip Operating Software (Affymetrix). The

Chapter 2: MATERIALS AND METHODS 117

experimental approach and data acquisition were performed in accordance with MIAME

requirements.

II.11. Quantitative Immunofluorescence

Astrocytes were plated in a 96-well plate and grown to confluence overnight

before being treated with soluble Syncytin-1 protein or supernatant from empty vector

transfected HEK293T cells. Treatment period ranged from 24-96 hours after which cells

were processed for analysis. Cells were permeabilized by five washes of 5 minutes each

in PBS containing 0.1% Triton X-100. Cells were blocked by a 90-minute incubation in

Li-Cor Odyssey blocking buffer at room temperature with gentle shaking. The primary

antibodies (anti-ASCT1, anti-CNPase, anti-MBP, anti-Egr1) were added at a

concentration of 1:500 and incubated overnight at 4°C with gentle shaking. Cells were washed 5 × 5 minutes each in PBS containing 0.1% Tween-20. The secondary antibodies

(1:800, room temperature, IRDye800-conjugated or Alexafluor-conjugated, Rockland,

Gilbertsville, PA, USA) were added at a concentration of 1:500 and incubated 1 hour at room temperature with gentle shaking, after which cells were washed 5 × 5 min in PBS containing 0.1% Tween. The plate was then scanned using the Odyssey Infrared Imaging

System (700 nm and 800 nm wavelength; 169 μm resolution, 2 mm offset, intensity setting of 5 for both channels). Label intensity was measured by densitometric analysis of the wells.

II.12. Detection of proteins in brain homogenate and cell cultures by western blot

Protein extracts were prepared from various brain tissue and cell samples with

lysis buffer (10 mM Tris, pH 7.4, 10 mM NaCl, 3 mM MgCl2, 0.5 % NP-40) and

concentrations were determined by Bradford assay (Biorad, Mississauga, ON, Canada).

Chapter 2: MATERIALS AND METHODS 118

Equal amounts of protein from each sample (100-200 μg) were separated by 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE, under reducing conditions), transferred to nitrocellulose membranes and blocked with 10% skimmed milk in TBST (25 mM Tris-buffered saline and 0.05% Tween-20). Primary antibodies were diluted in TBST containing 5% skimmed milk and incubated with membranes for at least 2 h at room temperature. Membranes were washed and incubated for at least 2 h at room temperature with horseradish peroxidase (HRP)-conjugated goat-anti-mouse (or rabbit anti-goat) IgG (Jackson ImmunoResearch Lab Inc. Westgrove, PA, USA) diluted in 5% milk-TBST. Immunoreactive proteins were detected by chemiluminescence

(Roche) and protein abundance measured by densitometry using Scion Image (Scion

Corp. MD, USA). Blots were stripped in stripping buffer (100 mM β-mercaptoethanol in

2% SDS-containing TBS) for 2 h at 55oC and probed with either other primary antibodies or against β-actin. When needed, densitometric analysis was performed and the average of the results was expressed as the RFC in the density of the band in treatment versus controls.

II.13. Human tissue samples

Brain tissue (frontal white matter) was collected at autopsy. Age-matched control subjects included 18 patients (mean age 56 ± 16.4 years) who were diagnosed with

Alzheimer’s Disease (AD) (n=6), HIV-infection (encephalitis (n=4); gliosis (n=4)), cerebral arteriosclerosis (n=2), anoxic encephalopathy (n=1) or normal brain pathology

(n=1). MS patients included 20 patients (age 63.3 ± 13.4 years) who had been classified as primary progressive (n=6), secondary progressive (n=10), and relapsing-remitting

(n=4) who had Expanded Disability Status Scale score (EDSS) ranging from 7 to 9 prior

Chapter 2: MATERIALS AND METHODS 119

to death. Frozen brain tissue from MS patients was obtained from the Multiple Sclerosis

Patient Care and Research Clinic, Edmonton, AB, Canada. (See Appendix D for list of

patients and their disease conditions). In addition, one case of an acute MS lesion was obtained from Dr. Arthur Clarke (University of Calgary, Canada) and tissue sections of acute lesions from MS patients was also obtained from Dr. Robert Hammond (University of Western Ontario, Canada). Matched plasma and CSF samples from controls (n=40) and MS (n=38) patients with relapsing-remitting disease were also obtained with consent through the MS clinic at the University of Alberta, through Dr. K.G. Warren and centrifuged to remove cellular debris. In addition, resting PBMC subsets (B and T cells, monocytes) from a previously reported cohort of acute RR-MS patients and healthy age- and sex-matched controls at the Montreal Neurological Institute, McGill University,

Montreal QC, were used 350.

II.14. Immunohistochemistry and immunocytochemistry

II.14.1. Detection of proteins in culture, human and mouse brain tissue sections

Paraffin-embedded sections were immunostained with antibodies to ionized

calcium binding adaptor protein (Iba-1) 351 (macrophages and microglia), GFAP

(astrocytes) (DAKO), iNOS (Santa Cruz Biotechnology, Santa Cruz, CA, USA), rabbit

anti-mouse GSTYp (oligodendrocytes) (Biotrin), APC (Ab-7) (oligodendrocytes)

(Oncogene Research Products, CA, USA), Syncytin-1 (6A2B2) 334, SLC1A4 (ASCT1),

SLC1A5 (ASCT2) (US Biologicals), CD45 (macrophage/microglial cells) (Zymed

Laboratories, San Francisco, CA), mouse anti-myelin basic protein (MBP Sternberger

Monoclonals Inc.), CNPase (Chemicon, USA), (Iba-1 1.0 μg/ml, GFAP 1:2000, iNOS

1:500, GSTYp 1:600, APC 1:500, Syncytin-1 1:1000, ASCT1 1:40, ASCT2 1:40, CD45

Chapter 2: MATERIALS AND METHODS 120

1:1000, MBP 1:1000, GSTYp 1:600, CNPase 1:100). ER stress proteins

GADD153/CHOP, Grp78/BiP, Grp58/ERp57 and PERK were detected using antibodies sc-575, sc-1050, sc-18619 and sc-13073 respectively (1:100, Santa Cruz Biotechnology,

CA, USA). Cultured oligodendrocytes derived from adult human and rat brains were stained with a mAb (O1, 1:1, a kind gift from Dr. V.W. Yong, University of Calgary) that recognizes galactocerebroside (Gal-C), a marker for mature oligodendrocytes 352, CNPase

or MBP.

II.14.2. Double label immunohistochemistry and microscopy

To double label both microglia and astrocytes with cell type-specific markers and

Syncytin-1, sections were initially probed with antibodies to Iba-1 and GFAP

respectively. This was followed by incubation with biotinylated goat anti-rabbit Ig (H+L)

(1:500, Vector Laboratories Inc. Burlingame, CA, USA) and subsequently with Elite

ABC reagent (Vector Laboratories Inc. Burlingame, CA, USA) or peroxidase-conjugated

affinipure goat anti-rabbit IgG (H+L) (1:500, Jackson Immunoresearch Laboratories, PA,

USA) and later developed by DAB treatment for peroxidase or BCIP/NBT for alkaline

phosphatase activity. Slides were examined on a Zeiss Axioskop2 microscope.

Immunofluorescence studies on brain sections from mouse models were performed when

required using secondary antibodies conjugated to Alexa488 (Molecular Probes) or Cy3

(1:200, Jackson Immunoresearch Laboratories, PA, USA). Images were captured on an

Olympus FV300 confocal laser-scanning microscope at the University of Calgary or at

the University of Alberta using a LSM510 META (Carl Zeiss MicroImaging, Inc.) laser-

scanning microscope. Images were analyzed by Scion Image software (Spot Diagnostic

Instruments, MD, USA).

Chapter 2: MATERIALS AND METHODS 121

II.14.3. Luxol fast blue staining for myelin

Sections were stained for myelin using Luxol fast blue (Solvent blue 38; Sigma).

Briefly, sections were deparaffinized, incubated in the solvent blue solution for 3 hr at

60°C, destained with 0.05% lithium carbonate, and counterstained with

hematoxylin/eosin 353.

II.14.4. Syncytia formation in astrocytes

HFA infected with SINrep5-Syncytin-1 or SINrep5-EGFP or mock-infected

astrocytes were immunostained for expression of Syncytin-1 using the monoclonal

antibody 6A2B2 334. Cells were double labeled with anti-GFAP antibody to determine

GFAP-positive astrocytes overexpressing Syncytin-1.

II.15. Quantification of cell numbers in vivo and in vitro

Fluorescently (Alexa488, Molecular Probes)-labeled cells were counted (10

fields) from a total area of 23760 μm2 using a 10X objective on a Zeiss Axioskop

microscope and expressed as a percentage. In addition, cells were also counted by a

stereological method 354. APC (Ab-7)-positive oligodendrocytes as well as GFAP-

positive astrocytes in the vicinity of the implantation site were counted per well (5 fields

at 400 x; expressed as total number of GSTYp-positive cells in an area of 2376 μm2).

Three serial sections (5 μm) from 6 animals per treatment group were used for

quantitative analysis of numbers of GFAP-positive astrocytes and GSTYp-positive

oligodendrocytes. Briefly, a systematic random sample of 3 sections that span the corpus

callosum was selected for analysis. Sections were selected at equal intervals in the series

comprising a known fraction of the section series (ssf). The labeled astrocytes and

oligodendrocytes were counted (ΣQ) under a known fraction of the section area (asf). The

Chapter 2: MATERIALS AND METHODS 122 height of the optical dissectors (h) positioned in the central part of the section thickness

(t) was used to determine the ratio h/t (tsf). The total number (N) of astrocytes or oligodendrocytes were estimated as N= ΣQ. 1/ssf. 1/asf. 1/tsf. The coefficient of error was determined by the formula CE (ΣQ)= SEM/Mean.

II.16. Statistical analyses

Statistical tests were performed using GraphPad InStat version 3.01 software

(GraphPad Software, San Diego California USA). Syncytin-1 RNA and DNA copy numbers were shown as the median viral copy numbers with 95% confidence intervals.

Contingency tables were analyzed using Chi-square and Fisher’s Exact Tests. The Mann-

Whitney U or unpaired t tests were used to analyze data and when multiple treatments were used, a one-way Analysis of variance (ANOVA) with Tukey-Kramer Multiple

Comparisons Test was also performed. When data from treatments were normalized to the control values, Dunnet’s post-hoc test was applied. When distribution of our data was unknown and non-parametric tests were used, data were analyzed with the Kruskal-

Wallis test.

Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 123

CHAPTER 3

SYNCYTIN-1 INDUCES NEUROINFLAMMATION

Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 124

III.1. Introduction

As much as 8% of the human genome is derived from retrovirus-like elements,

presumably remnants of retroviral infections during primate evolution 287,307,355. Many

HERVs have retained functional promoter, enhancer, and polyadenylation signals, and

these regulatory sequences have the potential to modify the expression of adjacent genes

318,319 . Despite most HERVs being replication defective because of mutations within

structural retrovirus genes, specific ORFs corresponding to HERV genes encode

detectable proteins 356. The increased expression of HERV genes may be important in

modulating host innate and adaptive immune responses with ensuing disease effects

although definitive proof of specific HERV-related pathogenic effects is lacking.

Earlier studies suggested that HERV expression in human brain was augmented in

circumstances of neuroinflammation 198. MS is a prototypic neuroinflammatory disease,

characterized by infiltration of inflammatory cells, damage and death of oligodendrocytes

with demyelination, resulting in physical and cognitive disabilities. Indeed, cytokines,

arachidonic acid metabolites and redox reactants including NO are major determinants of

pathogenicity in MS 138,357,358, which is also influenced by an individual’s genetic susceptibility. The role of both exogenous and endogenous infectious pathogens in MS pathogenesis is uncertain but several viruses and bacteria have been implicated through specific mechanisms including transactivation of aberrant immune responses and molecular mimicry. Herein, we examined the effects of Syncytin-1 expression on neural

cell function and survival, focusing in particular on the pathogenic effects.

III.2. RESULTS

III.2.1. Syncytin-1 is inducible and up-regulated in MS lesions.

Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 125

To investigate gene expression of phylogenetically related HERVs (Fig. 10) in

neuroinflammatory diseases, we examined the abundance of different HERV env mRNAs

in brains from patients with MS and other neurological diseases as controls. We analyzed

a broad range of endogenous retroviral envelope genes, whose sequences have since

become available (www.ncbi.nlm.nih.gov). The recently described Syncytin-2 (HERV-

FRD)303, HERV-R and -T env expression was analyzed by real-time RT-PCR, in addition

to HERV-H, -K (HML-2), -E and Syncytin-1. Syncytin-1 mRNA expression was

selectively up-regulated in brain tissue from MS patients relative to control patients,

while other HERV env mRNAs were not elevated in MS patients (Fig. 16). Sequencing

of the resulting PCR products confirmed that Syncytin-1 was overexpressed in the brain

samples, based on comparisons with HERV-W and other HERV env sequences located in

Genbank (www.ncbi.nlm.nih.gov/Genbank). Western blotting showed that HERV-W

env-encoded Syncytin-1 (75 kDa) immunoreactivity was detected in brain tissue from

MS patients but exhibited limited detection in control patients (Fig. 17A). Comparison of

Syncytin-1 immunoreactivity in brain, showed a ~3.0-fold increase among MS patients

relative to controls (Fig. 17B). Lesions from MS patients containing active demyelination

[D] in acute (Fig. 18A) tissue sections with numerous lipid/myelin filled macrophages and hypertrophied astrocytes demonstrated Syncytin-1 immunoreactivity. Syncytin-1 expression was observed in acute (Fig. 18C) lesions in cells resembling activated glia, which also exhibited iNOS immunoreactivity (Fig. 18C, inset), but not in controls (Fig.

18 D) that showed normal myelination (Fig. 18 B). Lipid vacuole filled Syncytin-1- immunopositive cells resembling phagocytic macrophages, were evident at the margin of the lesion (Fig. 18 G) as well as within the core of the lesion (Fig. 18H) in both acute and

Chapter 3: Syncytin-1 induces neuroinflammation

7 Brain Control ** 6 MS 5

4

mRNA RFC mRNA 3

2

1

0 -R -FRD -E - W -H -T -K

HERV

Fig. 16: A significant increase (3-fold) in the mRNA of HERV-W env (Syncytin-1) in MS patients relative to other HERVs and disease control patients was observed by real-time RT-PCR analysis. Statistical comparisons were made between controls and MS and also among different HERVs (Tukey’s Multiple Comparison Test was used to compare all columns; ** p<0.01).

126 Chapter 3: Syncytin-1 induces neuroinflammation

A MS Controls KDa

Syncytin-1 75 β-actin 42

B 4 MS (n=16) * 3 Controls (n=18) 2 1 Protein RFC 0 MS Controls

Fig. 17: Representative western blot of brain tissue lysate from MS patients (MS) that exhibited increased Syncytin-1 immunoreactivity compared to controls (A). Quantitation of western blots showed increased Syncytin-1 immunoreactivity in MS brains compared to controls (B) (Dunnet’s test was used to compare patient groups; * p<0.05).

127 Chapter 3: Syncytin-1 induces neuroinflammation

D AB

CD

EF

D *

G H

Fig. 18: (A) An active demyelinating (D) lesion from frontal lobe sections of a MS patient brain shows myelin debris in macrophages, minimal Luxol fast blue (LFB), hematoxylin and eosin staining, compared to normal myelin (B). (C) Serial sections from the same active lesion show increased Syncytin-1 expression in an area of active demyelination (D) that was absent in control sections (D). iNOS immunoreactivity (dark blue) was co-localized with Syncytin-1 (brown) in glia (C, inset). Double-label immunohistochemistry of acute lesions in MS brain shows Syncytin-1 expression in activated astrocytes colocalized with GFAP immunoreactivity (E), (Syncytin-1-blue and GFAP-brown, arrows) (E; inset figure shows only GFAP- immunoreactive astrocyte) or microglia and macrophages colocalized with Iba-1 immunoreactivity (Iba-1-blue and Syncytin-brown, arrowheads) (F; inset figure shows only Iba-1 immunoreactive microglia). Syncytin-1 immunoreactivity (G) is detected at the edge (asterix) of an acute demyelinated (D) lesion. (H) Phagocytic macrophages containing lipid vacuoles, representing myelin debris (arrow), are evident at the edge of the lesion (G, box) in the vicinity of Syncytin-1-positive hypertrophied astrocytes or macrophages (arrowheads). 128 Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 129

chronic demyelinating lesions (Table 4). As astrocytes 359 and microglia 6 are important

modulators of neuroinflammation, we determined if Syncytin-1 expression was

selectively up-regulated in these immunologically active cells. Double-label

immunohistochemistry revealed enhanced expression of Syncytin-1 in astrocytes (Fig.

18E) and microglia (Fig. 18F) of acute brain lesions (frontal white matter) from MS

patients, but not in other neural cells including neurons and myelin-forming

oligodendrocytes.

To examine the relative expression of different HERV env mRNAs in specific cell

types implicated in neuroinflammation, we studied human cell lines with and without cellular activation. Peak mean HERV transcript abundance was detected at 4 h in PMA- treated monocytoid (U937) cells with a significant increase in Syncytin-1 mRNA level

compared to other HERVs (Fig. 19A). HERV-H env mRNA was the most highly

expressed HERV in PMA-treated astrocytic (U373) cells at 24 h together with a

significant increase in Syncytin-1 (Fig. 19B). In contrast, we were unable to induce

HERV env expression in lymphocytes (PBLs) with PMA stimulation although IL-1β

expression was significantly elevated (Fig. 19C), similar to monocytoid and astrocytic cells. In fact, we observed suppression of HERV env mRNA levels when PBLs were stimulated with PMA (Fig. 19C). Thus, these studies indicated up-regulation of Syncytin-

1 occurred among MS patients in active demyelinating lesions with selective expression in cells mediating neuroinflammation.

130

Table 4: Clinical and neuropathological features of multiple sclerosis patients exhibiting Syncytin-1 immunoreactivity in [Acute (A), sub-acute (SA) and chronic (C)] demyelinating lesions Section ID 1 2 3 Age at Death 65 71 38 Gender F F M MS Type1 SP SP RR Duration of MS >10 years >10 years 8 years Pattern of Syncytin-1 immunoreactivity2 Lesion type SA (n=1), SA (n=2), C (n=3) A (n=3), SA (n=3), C (n=3) C (n=2) Lesion type SA C C SA SA C C C SA SA SA A A A C C C Lesion edge 3+ 1+ 0 2+ 1+ 2+ 1+ 0 3+ 4+ 3+ 4+ 4+ 4+ 3+ 2+ 3+ Lesion core 4+ 2+ 1+ 4+ 3+ 3+ 2+ 1+ 2+ 2+ 2+ 4+ 4+ 0 1+ 1+ 0 Periplaque white matter 1+ 0 0 1+ 1+ 1+ 1+ 0 0 1+ 2+ 0 1+ 0 0 1+ 0 Normal appearing white matter 1+ 0 0 1+ 1+ 1+ 1+ 0 0 0 0 0 0 1+ 0 0 0

1 Clinical course: Secondary Progressive (SP), relapsing-remitting (RR) 2 Scoring pattern: 0, no cells; 1+, 1-10 cells/hpf; 2+, 10-50 cells/hpf; 3+, 50-100 cells/hpf; 4+, 100-500 cells/hpf (hpf = X 200) Chapter 3: Syncytin-1 induces neuroinflammation

ABC 60 25 Control ** Control *** Control PMA PMA 250 20 PMA ** 150 40 15 50 5 ** ** ** 10 4 20 * 3 mRNA RFC mRNA * 5 2 1 0 0 HERVHERV-KHERVHERV-EIL-1 HERV-EHERV-KHERV-HHERV-W HERV HERV HERV-HHERV β -H -W -E -K -W

Monocytoid cells Astrocytes Lymphocytes (U937) (U373) (PBL)

Fig. 19. Monocytoid cells (U937) (A) and astrocytic cells (U373) (B) show an increase in individual HERV mRNA relative fold change (RFC) when stimulated in the presence of 50 ng/ml of PMA as detected by real-time RT-PCR analysis using HERV envelope specific primers. All HERVs’ mRNA levels were suppressed in peripheral blood lymphocytes (PBL) (C) upon PMA stimulation despite marked induction of IL-1β mRNA levels. Statistical comparisons were made between controls and PMA and also among different HERVs (Dunnet’s Test was used to compare all PMA treatments to untreated controls; ***p<0.001, **p<0.01, *p<0.05).

131 Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 132

III.2.2. Syncytin-1 activates pro-inflammatory molecules in glial cells

As Syncytin-1 was abundantly expressed in vivo in MS brain tissue (Fig. 18), we

constructed a SINrep5-based vector (Fig. 12) that efficiently expressed the HERV-W-env

ORF encoding Syncytin-1 (Fig. 20A) and also mediated syncytia formation in BHK cells

(Fig. 20B). The SINrep5-Syncytin-1 virus infected HFA (Fig. 20C) that expressed GFAP

(Fig. 20E) compared to controls (Fig. 20 D&F). Comparison of cellular localization of

Syncytin-1 in BHK and HFA revealed distinct patterns of localization, with localized

staining in BHK (Fig. 20G) compared to a more dispersed pattern in HFA (Fig. 20H)

relative to SINrep5-EGFP expressing controls that did not display any Syncytin-1

immunoreactivity (Fig. 20 I&J). Further, RT-PCR analysis revealed comparable levels

of Syncytin-1 expression in fetal astrocytes and brain tissue from MS patients (Fig. 20K).

Syncytin-1-mediated induction of genes related to neuroinflammation was

examined 24 h post infection (p.i), revealing that the pro-inflammatory cytokine, IL-1β,

was significantly increased in both HFA (Fig. 21A) and MDMs (Fig. 21B) infected with

SINrep-5-Syncytin-1 compared to the controls including SINrep5-EGFP and mock-

infected cells. Moreover, mean iNOS expression was also enhanced in astrocytes upon infection with SINrep5-Syncytin-1 (Fig. 21A) but not in MDMs (Fig. 21B). However,

the anti-inflammatory cytokine, IL-10, was not induced in either cell type, suggesting that

Syncytin-1 expression selectively induced pro-inflammatory responses. A marked increase in mean protein carbonyl levels was also observed in the conditioned medium from SINrep5-Syncytin-1-infected HFA compared to controls (Fig. 21A), but not in

MDM-derived conditioned medium (Fig. 21B). Conversely, there was no significant difference in the mean levels of malondialdehyde (a product of lipid peroxidation) 349 in

Chapter 3: Syncytin-1 induces neuroinflammation

A B BHK EGFP Syncytin-1Mock 75 KDa C DC GFAP

EF Syncytin-1 GHH Syncytin-1 I J EGFP

K BHK HFA 7 ** 6 * 5 4 3

mRNA RFC mRNA 2 1 0 -1 MS Non-MS FP n G E ncyti y (n=13) (n=11) (n=5) S (n=6) Fig. 20: Western blot analysis of BHK cells transfected with SINrep5-Syncytin-1 RNA transcript. Syncytin-1 immunoreactivity on western blot (A) and syncytia formation (B) could be detected in transfected cells but not in mock-transfected or cells transfected with SINrep5-EGFP. Confocal microscopy of uninfected human fetal astrocytes (HFA) immunostained for GFAP (C) and Syncytin-1 (D) and astrocytes infected with SINrep5-Syncytin-1 (multiplicity of infection (MOI) of 1.0) immunostained for GFAP (E) and Syncytin-1 (F). Localization pattern of Syncytin-1 in BHK cells (G) and HFA (H) expressing Syncytin-1 reveal distinct patterns of immunoreactivity. Syncytin-1 expression is not observed in BHK (I) and HFA (J) infected with SINrep5-EGFP. Comparable levels of expression of 133 Syncytin-1 was observed in Syncytin-1 expressing brain tissue from MS patients and astrocytes (K). Levels of Syncytin-1 was compared between brain tissue of MS patients and SINrep5-Syncytin-1 infected astrocytes (Tukey’s Multiple Comparison Test was used to compare all columns**p<0.01, *p<0.05). Chapter 3: Syncytin-1 induces neuroinflammation

A

20 Carbonyls (%) ** Mock HFA EGFP * 250 Protein 15 Syncytin 200 10 150

mRNA RFC mRNA 100 *** 5 50 0 0 IL-1β IL-10 iNOS Protein B carbonyl Mock 100 *** MDM Carbonyls (%) EGFP 180 80 Syncytin Protein 140 60 100 40 mRNA RFC mRNA 60 20 20 0 IL-1β IL-10 iNOS Protein carbonyl C 1000 800 600 400 4-HNE (RFC) 200 0 ck -CM -CM o M EGFP DM M HFA SINrep5-Syncytin-1 Fig. 21: SINrep5-Syncytin-1-infected HFA show a significant increase in mRNA expression of IL-1β and iNOS together with increased protein carbonyl levels relative to SINrep5-EGFP and mock controls while IL-10 mRNA expression was similar between treatments (A). IL-1β expression was increased in MDMs relative to controls while IL-10 and iNOS mRNA expression and levels of protein carbonyls did not differ between viruses (B). Levels of 4-HNE, a product of lipid peroxidation was not significantly different in the conditioned medium from astrocytes (HFA) and macrophages (MDM) infected with SINrep5- Syncytin-1 compared to controls. (C) Lipid peroxidation was not significantly different between samples. Samples were analyzed by slot blotting technique 134 that used a rabbit-anti-HNE antibody (1:4,000; Calbiochem) as a primary antibody (Dunnet’s Test was used to compare all PMA treatments to untreated controls; *p<0.05 , **p< 0.01, *** p< 0.001; compared to values obtained in Mock or SINrep5-EGFP infected astrocytes) Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 135

conditioned medium from SINrep5-Syncytin-1 infected HFA or MDMs compared to

controls (Fig. 21C). These observations indicated that Syncytin-1 induced a

proinflammatory molecular profile in astrocytes that included increased oxidation of

cellular proteins.

III.2.3. Syncytin-1 causes oligodendrocyte damage and death

Since oligodendrocytes are the principal cell types susceptible to injury associated

with neuroinflammation and demyelination, their morphology and survival was examined

after treatment with conditioned medium from HFAs and MDMs infected with SINrep5-

Syncytin-1. Conditioned medium from HFA infected with SINrep5-Syncytin-1 was

highly cytotoxic to human oligodendrocytes compared to SINrep5-EGFP- and mock-

infected HFA-derived supernatants (Fig. 22A). Importantly, conditioned medium from

HFA infected with SINrep5-Syncytin-1 also induced higher mean oligodendrocyte death

than that from SINrep5-Syncytin-1-infected MDM (Fig. 22B). Additionally, human

oligodendrocytes treated with media from HFA infected with SINrep5-Syncytin-1 also

displayed significant retraction of cellular processes (p<0.001) (Fig. 22C), compared to

the controls.

These results were confirmed using rat oligodendrocytes in the same cytotoxicity

protocol, which showed that supernatants from SINrep5-Syncytin-1-infected HFA

similarly caused significantly higher mean levels of cell death than that from SINrep5-

Syncytin-1 infected MDMs (Fig. 23A). As a control, the envelope protein from HIV-

JRFL, another neurotropic retrovirus from a patient with HIV-associated dementia, expressed in astrocytes using the same vector (SINrep5) did not cause cytotoxicity of oligodendrocytes (Fig. 23A). Aside from cell loss, there was also marked process

Chapter 3: Syncytin-1 induces neuroinflammation

A

Mock Syncytin-1 EGFP

BC

10 * HFA-CM *** 80 HFA-CM 8 MDM-CM 6 60 4 40 2 20 Cytotoxicity (%) Cytotoxicity 0 0 Oligodendrocytes with Oligodendrocytes Mock Syncytin-1 EGFP retracted processes ( %) Mock Syncytin-1 EGFP

Fig.22: Syncytin-1 causes oligodendrocyte damage and death. (A) Human oligodendrocytes (H-OL) treated with conditioned medium (CM) from SINrep5-Syncytin-1- infected HFA exhibited retracted cellular processes (arrowhead) with cell loss compared to SINrep5-EGFP and mock controls which showed abundant cell numbers and intact processes (arrowhead). (B) H-OLs treated with CM from SINrep5- Syncytin-1- infected HFA for 24 h showed significantly higher cytotoxicity than the controls. CM from SINrep5-Syncytin-1-infected MDMs was not toxic to H-OLs. (C) H-OLs treated with CM from SINrep5-Syncytin-infected HFA for 24 h showed significantly more cells with retracted processes compared to controls. Statistical comparisons 136 were made between Syncytin-1 and EGFP relative to mock controls (Tukey’s Multiple Comparison Test was used to compare all columns *p< 0.05, *** p< 0.001). Chapter 3: Syncytin-1 induces neuroinflammation

A

30 MDM-CM *** ROL 25 HFA-CM 20 15 10

Cytotoxicity (%) Cytotoxicity 5

FL R Mock EGFP HIV-J velope Syncytin-1 n e B C

Syncytin EGFP

Fig. 23: (A) Conditioned medium (CM) from SINrep5-Syncytin-1- infected HFA induced significant cytotoxicity in rat oligodendrocytes but not CM from SINrep5-HIV-envelope. CM from HFA infected with SINrep5-Syncytin-1 caused rat OL cytotoxicity and process retraction (B) compared to conditioned media from SINrep5-EGFP-infected HFAs (C). Cells were stained for Gal-C using the monoclonal antibody O1 (1:50 dilution). OLs were immunostained for p53 to determine if cytotoxicity is mediated through the p53 pathway. Indeed, p53 immunopositive OLs (B, inset) were observed when OLs were treated with conditioned medium from Syncytin-1 expressing astrocytes. Statistical comparisons were made between Syncytin-1 and EGFP and HIV-JRFL relative to mock controls (Dunnet’s Test was used to compare all PMA treatments to untreated controls; *** p< 0.001). 137 Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 138

retraction in rat oligodendrocytes treated with conditioned medium from SINrep5-

Syncytin-1 infected HFA (Fig. 23B) compared to controls (Fig. 23C). Examination of rat

oligodendrocytes for apoptotic markers revealed enhanced immunostaining for p53 (Fig.

23B, inset) in Gal-C-positive cells treated with conditioned medium from Syncytin-1

expressing astrocytes (Fig. 23B).

Conditioned medium from SINrep5-Syncytin-1 infected HFA and MDM was not

cytotoxic to human neurons under similar conditions unlike the envelope protein from

HIV-JRFL, which was highly cytotoxic to neurons (Fig. 24). Thus, soluble factors released from astrocytes selectively induced by Syncytin-1 caused cellular injury and death in oligodendrocytes.

III.2.4. Anti-oxidants prevent Syncytin-1-induced oligodendrocyte injury

Since protein carbonyl formation is mediated by redox reactants such as NO and

its metabolite peroxynitrite 349, we hypothesized that compounds that scavenge redox

reactants might reduce death of oligodendrocytes. We examined the free radical

scavenging properties of a polyphenolic anti-oxidant, ferulic acid and a non-steroidal

anti-inflammatory-based anti-oxidant, NCX-2216 by the DPPH assay. Ferulic acid (Fig.

25B) efficiently scavenged free radicals whereas NCX-2216 was not efficient at doing so

(Fig. 25C) compared to the positive control, ascorbic acid (Fig. 25A). SINrep5-Syncytin-

1 infected HFA were treated with ferulic acid 360, NCX-2216 and the NOS inhibitors, L-

NIL (0.5 μM) and L-NAME (5.0 μM). Oligodendrocytes treated with the conditioned

media from these astrocytes showed a marked reduction in both mean oligodendrocyte cytotoxicity and protein carbonyl levels, compared to oligodendrocytes not treated with

either drug (Fig. 26A). Indeed, protection of oligodendrocytes by ferulic acid (Fig. 26B)

Chapter 3: Syncytin-1 induces neuroinflammation

A 90 *** 80 70 60 50 40 30

Cytotoxicity (%) Cytotoxicity 20 10

HIV Mock EGFP velope n Syncytin e

Fig. 24. SINrep5-Syncytin-1 expression in astrocytes and conditioned medium, therein, is not neurotoxic. Conditioned media from Sinrep5- Syncytin-1 and SINrep5-EGFP infected astrocytes were not toxic to neurons but control conditioned media from HFA infected with SINrep5-HIV-JRFL envelope was highly cytotoxic. Statistical comparisons were made between Syncytin-1 and EGFP and HIV-JRFL relative to mock controls (Dunnet’s Test was used to compare all PMA treatments to untreated controls; *** p< 0.001).

139 Chapter 3: Syncytin-1 induces neuroinflammation

A B

C

Fig. 25: Antioxidant activity of various drugs were examined. Ascorbic acid was used as the positive control, showing a potent antioxidant

activity with an EC50 of 40.06 μM(A). Ferulic acid had an EC50 of 83.18 μM (B) while NCX2216 had an EC50 of >100 μM (C) and was not effective as an antioxidant. 140 Chapter 3: Syncytin-1 induces neuroinflammation

A (%)Protein carbonyl 30 300 *** Cell death 25 * Protein carb. 20 200 15 10 100

Cytotoxicity (%) Cytotoxicity 5 0 0 16 2 Mock S + FA S +L-NIL S + L-NAME Syncytin (S) S+ NCX-2 B C 25 20

20 16 ** 15 * 12

10 8 ***

Cytotoxicity ( %) Cytotoxicity *** ( %) Cytotoxicity 5 4

0 0 M M M M μ μ μ 5 0 μ 0 nM nM Mock .5 5 6 0 0 0 Mock 6 6 Syncytin Syncytin Ferulic acid NCX-2216

Fig. 26: (A) Both Syncytin-1-induced OL cytotoxicity and protein carbonyl abundance in the CM of HFA were reduced when HFA were treated with the antioxidants, ferulic acid (50 μM) and NCX- 2216 (6 μM), L-NIL (N6- (1-iminoethyl)-lysine, hydrochloride) (0.5 μM) and L-NAME (N-nitroωarginine methyl ester) (5.0 μM). Syncytin-mediated rat oligodendrocyte toxicity was prevented by the anti-oxidants, ferulic acid (B) and NCX-2216 (C) in a dose- dependent manner. 50 μM of ferulic acid and 6.0 μM of NCX-2216 were the optimum doses required to block oligodendrocyte cytotoxicity mediated by free radicals. Statistical comparisons (ANOVA) were made between treatments relative to mock controls 141 Values are Mean ± SEM; (Dunnet’s Test was used to compare all PMA treatments to untreated controls *p < 0.05 ***p< 0.001). Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 142

and NCX-2216 (Fig. 26C) against Syncytin-1-mediated toxicity was found to be dose-

dependent. Treatment with ferulic acid did not affect infection or expression by SINrep5-

Syncytin-1. Conversely, treatment with other established neuroprotectants including

MK801 (NMDA-receptor antagonist), NBQX [α-amino-3-hydroxy-5-methyl-4-

isoxazolepropionate- (AMPA) receptor antagonist], IFN-β and glatiramer acetate failed

to reduce Syncytin-1-related oligodendrocyte toxicity. In fact, oligodendrocyte toxicity

increased in the presence of NMDA/AMPA receptor antagonists (Fig. 27A). Moreover, pre-treatment of rat oligodendrocytes with IGF-1 (3 ng/mL) and growth hormone (GH,

100 μg/mL) did not protect cells from cytotoxicity induced by conditioned medium from fetal astrocytes overexpressing Syncytin-1 (Fig. 27B). Hence, Syncytin-1-induced

oligodendrocyte cytotoxicity was likely mediated by the pathogenic effect of redox

reactants in this in vitro system.

III.2.5. Syncytin-1-induced neuroinflammation and neurobehavioral abnormalities are inhibited by ferulic acid

Given that Syncytin-1 caused a significant increase in cytotoxicity of

oligodendrocytes, the in vivo effects of Syncytin-1 were examined following stereotaxic

implantation of the SINrep5-Syncytin-1 virus into the corpus callosum of CD1 mice. This

brain region was selected because it is abundant in myelin, oligodendrocytes and

astrocytes and is specifically injured in demyelinating diseases such as MS, resulting in

motor and cognitive abnormalities. Following implantation with SINrep5-Syncytin-1,

Syncytin-1 was detected near the implantation site (Fig. 28c) and Syncytin-1-expressing

astrocytes were visible in the corpus callosum (Fig. 28c, inset). Serial tissue sections

displayed increased numbers of hypertrophied astrocytes (Fig. 28f) and microglia (Fig.

Chapter 3: Syncytin-1 induces neuroinflammation

AB 10 25 *** CNPase *** *** 8 20 *** ***

15 6 * 4 10 Cytotoxicity (%) (%) Cytotoxicity 5 Fluorescence (AU) 2

0 0 1 1 1 P 1 β trol GH rol GA GH t -801 IGF- IGF- Mock EGF NBQX IFN- Con Con MK Syncytin- Syncytin- Syncytin-1 Syncytin-1

Fig. 27: (A) Treatment of astrocytes with NMDA and AMPA receptor agonists (MK-801 and NBQX respectively; 30 μM each), interferon-β (IFN-β; 100 U/ml) or glatiramer acetate (GA; 25 μg/ml) prior to infection with SINrep5-Syncytin-1 did not block oligodendrocyte cytotoxicity relative to untreated cells and in some instances appeared to exacerbate Syncytin-1induced cytotoxicity. (B) Pre-treatment of oligodendrocytes with IGF-1 (3 ng/mL) and Growth Hormone (GH, 100 μg/mL) did not protect cells from toxicity mediated by conditioned medium from astrocytes overexpressing Syncytin-1 This assay was performed by quantitative immunofluorescence for CNPase immunoreactivity (LI-COR, Odyssey) and values are expressed as arbitrary units of fluorescence. Statistical comparisons (ANOVA) were made between treatments relative to mock controls. Values are Mean ± SEM; (Dunnet’s Test was used to compare all PMA treatments to untreated controls; *p < 0.05, **p < 0.001).

143 Chapter 3: Syncytin-1 induces neuroinflammation

Mock SINrep5-EGFP SINrep5-syncytin-1 abc Syncytin-1

def GFAP

ghi MBP

jkl GSTYp

mon Iba-1

Fig. 28: Syncytin-1 induces neuroinflammation in mice. Syncytin-1 immunoreactivity was detected in the corpus callosum up to 14 days after infection with SINrep5-Syncytin-1 (c) but not in mock-CM- (a) and SINrep5- EGFP- (b) implanted animals. Astrogliosis indicated by intense GFAP immunoreactivity was observed in the corpus callosum of mice injected with SINrep5-Syncytin-1 (f) while normal astrocytes are observed in mock CM- (d) and SINrep5-EGFP- (e) implanted animals. White matter of mice injected with SINrep5-Syncytin-1 showed a vacuolar appearance (i) but in contrast, healthy myelin is seen in mock CM- (g) and SINrep5-EGFP- (h) implanted animals. GSTYp-immunopositive OLs were decreased in the white matter of mice implanted with SINrep5-Syncytin-1 (l) compared to the controls (j and k). Syncytin induces microgliosis in mice brain. Mock (m) and SINrep5-EGFP (n)- implanted mice brains showed fewer activated microglia compared to those implanted with SINrep5-Syncytin-1 (o), 144 suggesting microglial activation by Syncytin-1(White Bar represents 0.05 mm; Original magnification X400 (a-c); inset X1000); X200 (d-f); X200 (g-i); X600 (j-l); X200 (m-o). Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 145

28o) in the corpus callosum of SINrep5-Syncytin-1 implanted mice compared to

SINrep5-EGFP (Fig. 28 e&n) and the mock-implanted control (Fig. 28 d&m). In addition, myelin in the corpus callosum of mice implanted with SINrep5-Syncytin-1 showed a vacuolar appearance after immunostaining for MBP (Fig. 28i), compared to the controls (Fig. 28 g&h). Immunoreactive oligodendrocytes (GSTYp-positive) were substantially fewer in the animals receiving the Syncytin-1-expressing virus (Fig. 28 l), compared to controls (Fig. 28 j&k). To verify the latter observation, stereological cell counts of immunoreactive oligodendrocytes were performed revealing a significant reduction in mean cell numbers in the SINrep5-Syncytin-1 implanted animals compared to controls (Table 5). In contrast, astrocyte counts in the same animals revealed a significant increase in activated astrocytes. However, the decrease in oligodendrocyte numbers and increase in hypertrophied astrocytes caused by Syncytin-1 expression were reversed by daily ferulic acid treatment (Table 5).

The above neuropathological findings were confirmed by neurobehavioural testing of animals receiving SINrep5-EGFP, mock-implanted conditioned medium and

SINrep5-Syncytin-1 with and without concurrent ferulic acid treatments. At days 3 and 7, there were no differences in performance among the different groups (Fig. 29 (A-C)).

However, at days 10 and 14 post-implantation, SINrep5-Syncytin-1 implanted mice grasped the horizontal rod significantly less time compared to the controls (p<0.05) (Fig.

29D). In addition, mice implanted with SINrep5-Syncytin-1 were not sufficiently impaired that they could not hold onto the screen, but were slower to reach the screen edge (Fig. 29E), while mice implanted with SINrep5-EGFP or control conditioned medium were more curious and reached the edges of the inverted screen more quickly

146

Table 5: Cell counts of immunoreactive astrocytes and oligodendrocytes in corpus callosum1

Mock SINrep5- SINrep5- SINrep5- EGFP Syncytin-1 Syncytin-1 +Ferulic acid Oligodendrocytes 653.97 (0.05) 760.53 (0.06) 356.32 (0.07) ‡ 731.99 (0.04)

Astrocytes 130.50 (0.12) 198.98 (0.14) 402.49 (0.09) ‡ 177.08 (0.17)

1 Cell counts were performed using stereological methods and expressed as mean and coefficient of error (CE). GST-Yp (oligodendrocytes) and GFAP (astrocytes) immunopositive cells were counted in the corpus callosum of animals implanted with SINrep5-Syncytin-1, SINrep5-Syncytin-1 and ferulic acid, SINrep5-EGFP or mock- infected. Tukey’s Multiple Comparison Test was used for post-hoc analysis (‡, P<0.0001) Chapter 3: Syncytin-1 induces neuroinflammation

50 Control A B Control C Control EGFP 200 EGFP 300 EGFP 40 Syncytin Syncytin Syncytin 250 160 30 200 120

Seconds 150 Seconds

20 Seconds 80 100 10 40 50 0 0 0 Day 3 Day 7 Day 3 Day 7 Day 3 Day 7

35 Day 10 D Day 10 E Day 10 ** F 60 300 Day 14 Day 14 Day 14 250 *** 50 ** 25 40 200 30 150 15 Seconds Seconds

Seconds 20 100 10 * 50 5

l -1 l -1 -1 FP n -1 + n -1 + l n -1 + n FP n G G n FP E E G Contro ncyti ncyti E ncyti y ncyti FA Contro y ncyti FA Contro y ncyti S y S y S y FA S S S

Horizontal bar test Modified Screen test Beam test

Fig. 29: Syncytin-1 induces neurobehavioral deficits in mice. At days 3 and 7, behavior of mice implanted with SINrep5-Syncytin-1 did not differ significantly among various treatment groups in the Horizontal Rod test (A), Modified screen test (B) and the Beam test (C). (D) SINrep5-Syncytin-1-implanted mice showed significantly reduced ability to grasp a horizontal rod (Horizontal Bar test), compared to SINrep5-EGFP and control CM implanted controls (n=6 per group). (E) SINrep5-Syncytin-1-implanted mice exhibited significantly diminished ability to grasp and escape from an inverted screen (Modified Screen test) compared to controls (n=6 per group). (F) SINrep5-Syncytin-1-implanted mice exhibited delays in time taken to cross a cantilevered beam (Beam test) compared to controls (n=6 per group). Ferulic acid (20 mg/kg administered daily by oral gavage) treatment increased the animal’s ability to grasp the bar (D), improved ability to grasp and escape from the inverted screen (E) and cross the cantilevered beam in less time (F) compared to SINrep5-Syncytin-1, SINrep5- EGFP and mock-CM implanted mice. Syncytin-1 induced neurobehavioral changes in mice. Mice were subjected to the following tests after implantation 147 with SINrep5-Syncytin-1, SINrep5-EGFP or mock conditioned medium. Statistical comparisons (ANOVA) were made between treatments relative to mock controls Values are Mean ± SEM; (Tukey’s Multiple Comparison Test was used to compare all columns; *p < 0.05, **p < 0.005, ***p < 0.0001). Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 148

(p<0.0001). Finally, mice implanted with SINrep5-Syncytin-1 exhibited mean delays in

the time taken to cross a cantilevered beam, compared to mice implanted with SINrep5-

EGFP or control conditioned medium (p<0.005) (Fig. 29F), suggesting that the SINrep5-

Syncytin-1 implanted mice showed diminished motor activity and exploratory behaviour.

When SINrep5-Syncytin-1 implanted mice were treated with ferulic acid for 14 d, the neurobehavioural outcomes in the Horizontal Bar test (Fig. 29D), the Modified Screen test (Fig. 29E) and the Beam test (Fig. 29F) were significantly improved. Further, behavioral studies on mice implanted with SINrep5-Syncytin-1 or EGFP in the striatum were examined up to Day 14 of the experiment. Both total (Fig. 30A) and ipsiversive

(Fig. 30B) rotations did not reveal significant differences between groups. These in vivo

studies indicated that Syncytin-1 induced neuroinflammation, oligodendrocyte and

myelin damage together with neurobehavioural abnormalities, which were abrogated by

the anti-oxidant ferulic acid, similar to our in vitro observations.

Chapter 3: Syncytin-1 induces neuroinflammation

A B 100 120 CM EGFP Syncytin-1 CM EGFPEGFPSyncytin-1 100 Mock 80 Syncytin 80 60 60 40 40 20 20 Total rotations/10 min 0 0 Day 3 Day 7 Day 10 Day 14 Ipsiversive rotations/10 min Day 3 Day 7 Day 10 Day 14

Fig. 30: The striatum of 8-10-week-old CD1 mice were stereotaxically implanted with SINrep5-Syncytin-1, SINrep5-EGFP or BHK conditioned media. To assess neuronal damage as a result of expression of Syncytin-1, the rotational behavior of the mice was analyzed 3, 7, 10 and 14 days after striatal implantation. The total number of rotations (A) and ipsiversive rotations (B) over time of the animals following implantation with SINrep5-Syncytin-1, SINrep5- EGFP or mock-infected-CM animals were not significantly different among groups, suggesting Syncytin-1 expression does not cause striatal injury 149 Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 150

III.3. Discussion

Herein, we report both mRNA and protein (Syncytin-1) encoded by the HERV-W

env gene showed increased expression in the brains of MS patients and in specific cell

types involved in neuroinflammation within demyelinating and demyelinated lesions.

Furthermore, in vitro, Syncytin-1 mediated the production of pro-inflammatory

molecules such as iNOS, IL-1β and redox reactants that at high levels are damaging to

the brain 349,357,361. An explicit function for Syncytin-1 in the brain is unknown in contrast

to the placenta where it seems important for placental development 186,313,334,362.

Nonetheless, our findings are consistent with the neuroinflammatory profile in astrocytes associated with MS including elevated levels of iNOS and redox reactants 363-365. In

addition to the pro-inflammatory effects of Syncytin-1 in glial cells, soluble factors derived from Syncytin-1-expressing astrocytes were toxic to oligodendrocytes but not to neurons implying a select mechanism of killing and/or targeting of individual cell-types, which is congruent with earlier studies showing astrocytes may influence oligodendrocyte survival 93,366,367. Since pre-treatment of oligodendrocytes with IGF-1

and GH did not protect cells from death and injury, withdrawal of these trophic factors

due to astrocyte dysfunction may not be the cause of oligodendrocyte injury.

Interestingly, the increase in oligodendrocyte toxicity associated with NMDA/AMPA-

receptor antagonist treatment might be due to the decrease in neurotrophic factors, BDNF

and NGF as suggested previously 368.

Syncytin-1 was not detected in supernatants from astrocytes infected by SINrep5-

Syncytin-1, precluding a direct interaction between Syncytin-1 and the target cells that

led to cytotoxicity. Nonetheless, Syncytin-1 expression in astrocytes resulted in cellular

Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 151

stress, manifested by induction of IL-1β, iNOS and redox reactants. Importantly,

Syncytin-1 expression was evident in glia at both the margin and core of acute

demyelination, emphasizing its role in cellular stress. Of interest, polymorphisms in

retrovirus-encoded envelope proteins from HIV 342 and MuLV that alter intracellular

envelope expression in glial cells, have been associated previously with pathogenic

effects in the nervous system 369. These effects may occur through misfolding of the envelope protein or protein accumulation in the endoplasmic reticulum, which results in a stress response by the cell and the subsequent release of neurotoxic molecules including redox reactants 226,370. Given that iNOS was induced in astrocytes, likely redox stress

products include NO, reactive nitrogen-oxygen species, peroxynitrite and superoxide

anions, which are capable of damaging target tissues, particularly the brain 349,363 and are also toxic to oligodendrocytes 371. In contrast, in vitro generation of lipid peroxidation

products was not increased in the present study, in agreement with earlier reports of MS

pathogenesis 372. The precise pathway by which oxidation of a protein released by

astrocytes mediates oligodendrocyte toxicity remains to be determined but a potential mechanism might occur through oxidation of a released MMP 373.

We also demonstrated that by inducing pro-inflammatory molecules and redox reactants, ultimately resulting in oligodendrocyte death, Syncytin-1 modulates neurobehavioural changes in a mouse model displaying pathology similar to other MS animal models. The present neurobehavioural changes are also reminiscent of those seen among individuals with MS, including weakness, gait unsteadiness and altered executive functions 374,375. In addition, our studies showed that the plant-derived phenolcarboxylic

acid, ferulic acid, which acts as an anti-oxidant, 360,376 ameliorates death of

Chapter 3: SYNCYTIN-1 INDUCES NEUROINFLAMMATION 152

oligodendrocytes in vitro and in vivo and significantly improves the neurobehavioural

outcomes. Thus, the present study indicates that Syncytin-1 maybe involved in the

pathogenesis of active demyelination, principally by evoking redox reactant-mediated

cellular damage in the brain. Alternatively, given its persistent expression in lesion cores,

the Syncytin-1-mediated glial stress reaction might antagonize remyelination. In this

regard, ferulic acid or equivalent compounds might be considered for trials designed to

evaluate efficacy for reducing demyelination or enhancing remyelination, similar to its

protective effects in neurons 360. Moreover, because HERVs represent a substantial proportion of the human genome and in some cases express proteins, the potential pathogenic (or reparative) effects of other HERV proteins expressed in the nervous system warrant further investigation.

Chapter 4: QUANTIFICATION OF SYNCYTIN-1 153

CHAPTER 4

QUANTIFICATION OF SYNCYTIN-1

Chapter 4: QUANTIFICATION OF SYNCYTIN-1 154

IV.1. Introduction

HERVs are comprised of approximately 1500–2000 integrated proviruses, which

may have been amplified during evolution by repeated reintegration events in germ line cells, although most HERVs are not replication competent per se 296. Although 18

HERVs possess a coding retroviral envelope gene 304, expression of only 5 HERVs has

been detected in healthy human brain tissue to date 303. Importantly, the HERV-W family

has received extensive attention in the literature 196,331, in part because its envelope (env)-

encoded protein has retained the ability to interact with a receptor for the D-type

retroviruses 333,334. The multi-copy HERV-W family contains a unique proviral locus,

located on chromosome 7q, flanked by two intact LTRs and encodes a 538-amino acid

envelope (Syncytin-1) together with presence of inactivating mutations in the gag and pol

genes 186,330.

In addition to expression of HERVs in healthy tissues such as placenta, they are also up-regulated in neoplasic, autoimmune and inflammatory conditions 186,198,377.

Induction of several HERVs is reported in different cell types derived from patients with

MS 329. Relative quantification analyses from Chapter #3 indicated that Syncytin-1 mRNA and protein levels are increased in the brains of MS patients, predominantly in

glial cells 196. In this Chapter, we have quantitatively analyzed Syncytin-1 expression in

brain, CSF and plasma, together with relevant leukocytes. These studies revealed that

Syncytin-1 RNA was selectively up-regulated in the brains of MS patients. Increase in

Syncytin-1-encoding DNA sequences in the brain occurred without evidence of new

integration events.

Chapter 4: QUANTIFICATION OF SYNCYTIN-1 155

IV.2. Results

IV.2.1. Syncytin-1 expression is cell-type and tissue specific

Given that Syncytin-1 RNA levels were increased in brains of MS patients, we

also investigated its expression in blood-derived leukocyte populations because these

cells are pivotal in MS pathogenesis. Analysis of different HERVs in PBMC subsets

showed that env RNA abundance for HERV-H, -K (HML-2), -E, and Syncytin-1 in T

cells (Fig. 31A), monocytes (Fig. 31B) and B cells (Fig. 31C) did not differ between MS and non-MS controls. Using matched cDNA quantities, the average cycle threshold (Ct ±

standard error) values were 26.1 ± 0.16, 27.3 ± 0.3 and 26.6 ± 0.5 for HERV-H env in

monocytes, T and B cells, respectively. Similarly, Ct values for Syncytin-1 and HERV-K

(HML-2) env did not differ between PBMC subsets. However, expression of HERV-E

env was significantly lower in monocytes (32.7 ± 0.8) and T cells (32.5 ± 0.6) compared

to B cells (28 ± 0.3) (Fig. 32A). Thus, there were cell-type-specific differences in

expression of HERVs within PBMC subsets, though differences between clinical groups

were not significant. To determine if Syncytin-1 was in fact inducible in leukocytes, we

stimulated PBLs, monocytoid (MDM) and astrocyte (U373) cells with PMA. PMA

treatment induced Syncytin-1 mRNA expression in monocytoid (46 fold) and astrocytic

(10 fold) cell lines (p<0.001) (Fig. 32B). However, PMA suppressed Syncytin-1 mRNA

expression in PBLs from healthy individuals (p<0.05) although IL-1β was inducible in

these same cells (Fig. 32B).

Chapter 4: Quantification of Syncytin-1

A

6 Control T 5 MS 4 3 2 mRNA RFC mRNA 1

-W -H -E -K HERV BC 2.5 4 B Control M Control MS MS 2 3 1.5 2 1 mRNA RFC mRNA mRNA RFC mRNA 1 0.5

-W -H -E -K -W -H -E -K HERV HERV

Fig. 31: Expression of HERV env mRNA in resting T cells (T) (A), monocytes (M) (B) and B cells (C) did not differ between age- matched non-MS controls (n=9) and RR-MS patients (n=9). Data were not considered significant.

156 Chapter 4: Quantification of Syncytin-1 A 40 Monocytes 35 T-cells 30 B-cells * 25 Ct 20 15 10 5 0 WEKH

B *** 1 50

60 ***

40 mRNA RFC mRNA 20 ** ** * * 0 PMA -+-+-+ -+-+-+ PBL MDM Ast PBL MDM Ast Syn-1 IL-1β

Fig. 32: Detection of HERV env transcript abundance by real time RT- PCR suggests comparable levels of HERV-W (Syncytin-1), -H and -K, but abundance of HERV-E was lower in B cells (A). PMA stimulated Syncytin-1 (Syn-1) expression in monocytoid (MDM) cells and astrocytic (Astro) cells (U373). Syncytin-1 mRNA levels were suppressed in PMA-stimulated peripheral blood lymphocytes (PBL) despite marked induction of IL-1β mRNA levels (B). Statistical comparisons (ANOVA) were made between treatments relative to controls (Dunnet’s Test was used to compare all treatments to untreated controls; *** p<0.001, ** p <0.01, * p<0.05). 157 Chapter 4: QUANTIFICATION OF SYNCYTIN-1 158

IV.2.2. Minocycline inhibits Syncytin-1 expression

Since inflammation drives Syncytin-1 expression, we wished to determine

whether Syncytin-1 expression is regulated by minocycline, an anti-inflammatory drug.

PMA-induced Syncytin-1 transcript abundance was significantly suppressed by

minocycline (10 μM) (Fig. 33).

IV.2.3. Syncytin-1-RNA and DNA copy numbers are significantly enhanced in brain

of MS patients relative to controls

As relative quantification analysis disclosed increased Syncytin-1 expression in

the brains of MS patients, we focused on determining the actual copy numbers of

Syncytin-1-encoding RNA and DNA-encoding sequences in different tissues relevant to

neuroinflammation. Syncytin-1 RNA copy numbers were measured using a quantitative

real time RT-PCR, which were derived as copy numbers from a standard curve plotted

with RNA transcribed from pBS-Syncytin-1 (Fig. 34A). Median Syncytin-1 RNA copy numbers were significantly increased in MS brains (5.04 log 10) compared to non-MS

brains (4.57 log 10) per μg RNA (p<0.05) (Fig. 34B). The mean PCR cycle threshold

differences between GAPDH cDNA from the same MS (20.52) and non-MS (20.24)

brain tissues did not differ significantly. Total RNA extracted from CSF and plasma of

MS and non-MS patients showed that Syncytin-1 RNA was detected in samples from all

patients examined. However, there were no significant differences in Syncytin-1 RNA

copy numbers between groups in both CSF (MS: 3.8 log 10/ml and non-MS: 5.0 log 10/ml)

(Fig. 34C) and plasma (MS: 2.9 log 10/ml and non-MS: 5.033 log 10/ml) (Fig. 34D).

The median Syncytin-1 DNA copy number was increased (79 fold) in the brains

of MS patients (9.8 log 10/μg DNA), compared to that of the non-MS brains (7.9 log 10/μg

Chapter 4: Quantification of Syncytin-1

60 *** Control 50 PMA PMA + Minocycline 40

30

mRNA RFC mRNA 20 ** ** 10 * 0 Syn-1 IL-1β Syn-1 IL-1β

U937 U373

Fig. 33: Minocycline suppresses PMA-induced Syncytin-1 expression Monocytoid cells (U937) and astrocytic cells (U373) show an increase in Syncytin-1 mRNA relative fold change (RFC) when stimulated in the presence of 50 ng/ml of PMA as detected by real-time RT-PCR analysis. Minocycline (10 μM) treatment significantly suppressed expression of the envelope gene. Statistical comparisons (ANOVA) were made between treatments relative to controls (Dunnet’s Test was used to compare all treatments to untreated controls; *** p<0.001, ** p <0.01, * p<0.05).

159 Chapter 4: Quantification of Syncytin-1

AB

30 8 * 25 g μ 6

t 20 C 15 4 copies/ 10 RNA

10 2 5 RNA Brain log 0 024681012 MS Control Log copy number

C D L L ns μ ns μ 6 6 4 4 copies/ copies/ copies/

2 10

10 2 log log 0 CSF 0 Plasma MS Control MS Control

Fig. 34: Syncytin-1 RNA copy numbers are increased in brain of MS patients. A standard curve for Syncytin-1 RNA copy number (correlation coefficient: 0.978; slope: -1.857; intercept: 32.632; Y= -1.857 X + 32.632) was derived (A). Absolute numbers of Syncytin-1 RNA copies were increased in brains of MS patients

(5.04 log10) relative to non-MS controls (4.57 log10) (B). Quantitative analysis of Syncytin-1 RNA abundance in CSF (C) and plasma (D) did not reveal statistically significant differences between clinical groups. Statistical comparisons (Mann-Whitney 2-tailed test) were made between MS and non-MS controls (* p<0.05).

160 Chapter 4: QUANTIFICATION OF SYNCYTIN-1 161

DNA) (p<0.001) (Fig. 35C), based on a standard curve derived for Syncytin-1-encoding

DNA (Fig. 35A). To normalize for the quantity of DNA used as PCR template, a

GAPDH standard curve was developed (Fig. 35B), revealing that the Syncytin-1- encoding DNA copy numbers, when expressed as a ratio of the absolute copy numbers of

Syncytin-1 relative to the copies of GAPDH per μg DNA, displayed an increased median ratio for MS patients (3.1) compared to that of the control patients (1.6) (p<0.001) (Fig.

35D). We also investigated PBMC-derived DNA from non-MS and MS patients from two cohorts of patients. The ratio of Syncytin-1 DNA to GAPDH DNA copy numbers did not differ between MS patients and unaffected family members from an Iranian cohort

(Fig. 35E). Similarly, another cohort of patients from Canada did not reveal any differences in the ratio of Syncytin-1 DNA to GAPDH DNA copies between non-MS controls and MS patients (Fig. 35F).

IV.2.4. Increased Syncytin-1 DNA copies reflect un-integrated cDNA

To determine if the increased brain levels of Syncytin-1 DNA in MS patients reflected unintegrated linear/episomal cDNA or newly integrated proviral DNA, we developed PCR assays to detect HERV-W7q circular, single and/or double LTR sequences and integration events using oligonucleotide primers targeting Syncytin-1,

HERV-W7q LTR U3-U5 regions and Alu sequences (Fig. 15). Comparisons of the relative frequency of detection of circular Syncytin-1-LTR encoding DNA (Fig. 36A) and integrated LTR-U5-Alu sequences (Fig. 36B) did not reveal statistically significant differences between MS and non-MS brain-derived DNA samples.

Chapter 4: Quantification of Syncytin-1 A B DNA STANDARD GAPDH STANDARD 30 30 25 25

20 t 20 C t

C 15 15 10 10 5 5

0 246810 01234567 C Log copy number D Log copy number

12 *** 5 *** 10 4 g DNA μ 8 3 6 DNA 2 4 copies/ 1

10 2 Brain Brain MS Control Syncytin-1/GAPDH MS Control log

EF Iran-PBMC Canada-PBMC 4 7 6 3 5 4 DNA 2 DNA 3 1 2

Syncytin-1/GAPDH Syncytin-1/GAPDH 1 0 Syncytin-1/GAPDH MS CONTROL MS CONTROL (n=28) (n=27) (n=22) (n=24)

Fig. 35: Standard curves for Syncytin-1 DNA copies (A) ( Correlation Coefficient: 0.979; Slope: -1.365; Intercept: 29.435; Y= -1.365 X + 29.435; PCR efficiency: 440.6%) and GAPDH (Correlation Coefficient: 0.965; Slope: -2.276; Intercept: 29.371; Y= -2.276 X + 29.371; PCR efficiency: 175.0%) were obtained (B). Syncytin-1 DNA was quantified by real time PCR in brain tissue (white matter) of MS patients (n=20) and control patients (n=15) that included HIV (n=6), Alzheimer’s (n=6) and neuroinflammatory disease controls (n=3). Absolute number of viral DNA copies indicates a significant increase in viral DNA copy number in brain of MS patients relative to control (C). Ratio of Syncytin-1 copy number to GAPDH copy number also shows a significant increase in Syncytin-1 DNA copy number in brain of MS patients relative to control (D). Ratio of copy numbers of Syncytin-1 to GAPDH did not differ between MS and unaffected family members from PBMC DNA of an Iranian cohort (E). Similarly, PBMC from a 162 Canadian cohort of MS patients and non-MS controls did not reveal differences

between clinical groups (F). Data shown are log 10 (median copy number) (Mann- Whitney 2-tailed test; *** p<0.001; p<0.01). Chapter 4: Quantification of Syncytin-1

A B 100 70 MS ns MS Control ns 80 Control 50 ns 60 30 40 ns Detection (%) Detection

Detection (%) 20 10 0 Circ+ Circ- LTR-Alu+ LTR-Alu-

Fig. 36. Double LTR sequences from brain tissue of non-MS controls and MS patients did not reveal detection differences between clinical groups. Circ (+) refers to the numbers of DNA samples where the PCR product was detected, while Circ (-) refers to the numbers of DNA samples where the PCR product was not detected (A). Detection of PCR products from amplification of HERV-W7q-cell junctions (LTR-Alu) showed no detection differences between clinical groups. Detection of PCR products was denoted as LTR-Alu+ and absence as LTR-Alu - (B). Chi-square and Fisher’s exact tests were used to analyze the contingency tables obtained in this assay.

163 Chapter 4: QUANTIFICATION OF SYNCYTIN-1 164

IV.3. Discussion

The present study represents the first quantitative PCR analysis of a human

endogenous retrovirus expression in a range of tissues. Syncytin-1 mRNA copy number

was consistently higher in brain tissue from MS patients relative to non-MS controls.

Further, DNA copy numbers were also higher, though without evidence of new

integration events or viral replication. Syncytin-1 was inducible by the mitogen, PMA, in

astrocytic and monocytoid cells but not in lymphocytes. Expression level of Syncytin-1

in different leukocyte populations did not differ between clinical groups. These

observations highlight the perturbed expression of Syncytin-1 in neuroinflammation,

providing new insights into the pathogenesis of MS and other neuroinflammatory

disorders.

Though Syncytin-1 expression was increased in brains of MS patients relative to

non-MS controls, it was detected in both MS and non-MS patient brains and in

leukocytes from each group in the current study. Thus, it is improbable that HERV-W is

a conventional infectious agent causing MS, thereby supporting the notion that Syncytin-

1 may act as an immunomodulator in the brain, as proposed for endogenous retroviruses

in other organs 378,379. The specific cellular milieu is an important determinant of HERV transcription since it is regulated by chemokines, inflammatory molecules infectious

agents or cytokines 379. However, it is clear that HERV expression is differentially

regulated depending on the cell types involved in MS pathogenesis, as leukocytes from

MS patients did not differ from non-MS controls (Fig. 32 A-C), whereas differences were observed in brain-derived tissue. Further, mitogen stimulation of healthy PBLs did

Chapter 4: QUANTIFICATION OF SYNCYTIN-1 165

not induce Syncytin-1 expression. Using env-specific PCR primers for a PCR-based quantification assay for Syncytin-1, copy numbers of Syncytin-1 mRNA were

significantly higher in brain tissue of MS patients extending an earlier study 196 by showing selective up-regulation of Syncytin-1 only in brains of patients with MS.

However, levels of Syncytin-1 RNA copies in CSF and plasma did not reflect the enhanced levels in brains of MS patients relative to non-MS controls. These findings are in contrast to low HIV-1 copy numbers in brains of patients with another neurological disorder, HIV-associated dementia, but simultaneously exhibit elevated levels of HIV-1 viral copies in CSF 193. High frequency of detection of another HERV-W family member,

MSRV, in lymphocytes (B cells) and sera of MS patients has led to speculation regarding

its involvement in the etiopathogenesis of MS 380-383. In the present study, we used

Syncytin-1 primers, which did not recognize MSRV env, thus solely focusing on the role

of Syncytin-1 in MS neuropathogenesis. Nevertheless, there are molecular differences

(13.2% at the amino acid level, www.ncbi.nlm.nih.gov) between the two members of the

HERV-W family 319, including the absence of a 4-amino acid gap in MSRV env, a unique signature of Syncytin-1 384 and MSRV is postulated to be an exogenous retrovirus,385 rather than a HERV.

Increased Syncytin-1 DNA copy numbers were present in the brains of MS patients, which did not reflect increased viral re-infection or integration as the number of single/double DNA LTRs and integrated genomes did not differ between groups. Due to the detection of predominantly ~800 bp single LTR PCR fragments suggestive of aborted replication, the present study suggests that Syncytin-1 is reverse transcribed but not integrated. These results underscore the widely-held supposition that HERV-W is a non-

Chapter 4: QUANTIFICATION OF SYNCYTIN-1 166

replicating agent 330. In fact, this may indicate accumulation of un-integrated extra-

chromosomal viral cDNA, similar to reports of HIV-1 infection of the brain 386,387, which

is found to be associated with S35 domains in the interchromosomal space 388. The precise mechanism by which HERVs are induced or suppressed is likely dependent on the individual HERV, cell type and other host determinants but may be an important aspect to investigate further, given the interest in HERVs as biomarkers for disease.

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 167

CHAPTER 5

SYNCYTIN-1 INDUCES ER STRESS

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 168

V.1. Introduction

Multiple Sclerosis is a progressive demyelinating disease of the CNS defined by

inflammatory destruction of the myelin sheath and ensuing axonal damage. The

neuropathological hallmarks of acute MS lesions are demyelination and inflammation, involving T and B cells and activated macrophage/microglia, which result in tissue damage characterized by loss of oligodendrocytes, astrocytes, and axons 389. Although largely assumed to be a T cell-mediated disease, increasing evidence indicates involvement of other immune cells including microglia/macrophages 5 and astrocytes 389 in MS pathogenesis. The latter group of cells can efficiently present MOG peptides and act as a secondary line of APCs in the CNS, simultaneously acting to down-modulate proinflammatory T cell-driven inflammation 390. Up-regulation of ER stress molecules including ATF-4 has been demonstrated in MS lesions 391 and accumulation of proteins

such as MHC class I in the ER 150 or induction of ER stress by IL-1β or nitric oxide 392 can affect reparative abilities of oligodendrocytes 393.

In Chapter #3, we have demonstrated up-regulation of Syncytin-1 in the brains of

MS patients and its overexpression in areas of acute demyelination 196. Syncytin-1

appears to bind to at least two receptors (ASCT1 and 2) including sodium-dependent

transporters of polar neutral amino acids, Ala, Ser, Cys, Thr 333. ASCT1 and 2 are

localized on both neurons and glia in the brain 337 and are essential for maintaining

cellular integrity through the transport of amino acids with potentially neurotrophic 338 or neurotoxic 339 effects.

Astrocytes are more than ‘brain glue’; in addition to their roles in modulating

synaptic plasticity 64, they provide trophic support to oligodendrocytes and neurons 394,

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 169

while also protecting neurons from damage such as excitotoxicity 395. Extrinsic and

intrinsic factors can lead to astrocyte dysfunction, including accumulation of misfolded

proteins contributing to disruption of ER function, resulting in ER stress 396. Specific

signaling pathways including the UPR are triggered during ER stress to enable cells

survive the perturbed condition of the ER 396. During the pre-stress period, nascent

folding-competent polypeptides are maintained in a soluble form in interaction with ER

luminal chaperones, chief among which is BiP. BiP is a cytoprotective protein 397 that associates with 3 proximal sensors of the UPR namely, PERK, ATF6 and the inositol- requiring enzyme 1 (IRE1). Upon ER stress, BiP disassociates from the UPR transducers to permit their signaling 392. Recently, a basic leucine zipper (bZIP) transcription factor,

old astrocyte specifically induced substance (OASIS), which has a structure similar to

that of ATF6, was found to be activated by cleavage in response to ER stress. The

cleaved fragment, p50OASIS, activates the BiP promoter and thus protects astrocytes

from ER stress 398, but its other functions remain unknown.

Several lines of evidence implicate astrocytes in retrovirus-induced ER stress and

neuropathogenesis 370. Indeed, astrocyte activation and death are closely correlated with

the severity of HIV-associated dementia 399. Infection of astrocytes with MoMuLV-ts1 induces an ER stress response, in particular inducing the expression of the redox- sensitive transcription factor NF-E2-related factor 2 (Nrf2) 400, which is also required for

cell survival during ER stress 401. Importantly, xCT, a cystine transporter 402 and also a

viral receptor 403 was induced by Nrf2 370 and reactive oxygen species 404. These findings

prompted us to ask whether Syncytin-1 induced an ER stress response, modulated its

receptor expression and affected oligodendrocyte viability. Although Syncytin-1 blocks

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 170

infection by an exogenous retrovirus 308, how it affects glial physiology or if it mediates

interference with its cognate receptors, ASCT1 or 2 is not known. Since retroviral

envelopes are known to interfere with their respective receptor’s function 405, we hypothesized that enhanced Syncytin-1 might influence expression of its receptor through

ER stress induction, contributing to MS pathogenesis. Earlier studies showed that

Syncytin-1 protein and transcript expression in glial cells including astrocytes and macrophage/microglia were substantially greater in the brains of MS patients compared to non-MS controls 196,406. In addition, iNOS expression is enhanced in demyelinating regions of MS brains relative to controls 138. Moreover, increased expression of iNOS in

Syncytin-1-expressing astrocytes in vitro as well as in acute MS lesions was observed 196.

The present studies revealed that Syncytin-1 induced several ER stress molecules including OASIS. Using a new model of MS, we describe a novel pathway for oligodendrocyte injury in which Syncytin-1 induced OASIS expression in astrocytes accompanied by increased production of nitric oxide and the transcription factor, early growth response (Egr1), leading to diminished ASCT1 expression and oligodendrocyte loss.

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 171

V.2. Results

V.2.1. Syncytin-1 induces ER stress in astrocytes

Since retroviral proteins cause neuropathogenic effects through protein misfolding

235, we investigated whether enhanced levels of Syncytin-1 in the brains of MS patients

196 contributed to ER stress. To examine the effects of Syncytin-1 up-regulation in ER

stress, we overexpressed Syncytin-1 in human astrocytes by transduction of a Syncytin-1 expressing neurotropic Sindbis virus 196, which revealed syncytia formation in astrocytes

(Fig. 37A), confirming its functional role in GFAP-positive (Fig. 37A, inset) astrocytes.

Levels of GADD153, BiP, PERK, ERp57 and OASIS transcripts were significantly

increased (Fig. 37B) in astrocytes overexpressing Syncytin-1 but not in astrocytes

transduced by an EGFP-expressing virus (control) (p<0.05). However, ER stress genes

were not induced in monocyte-derived macrophages (MDMs) overexpressing Syncytin-1

relative to EGFP-expressing MDMs (Fig. 37C). To determine the comparative transcript

levels of ER stress genes in Syncytin-1 expressing astrocytes and brains of MS patients,

we examined ER stress gene induction in brain white matter from MS (n=15) and non-

MS controls (n=12). The expression profile of ER stress genes observed in Syncytin-1

expressing astrocytes closely resembled transcript abundance in brain white matter tissue

of MS patients relative to non-MS controls in areas of acute demyelination (Fig. 38A). In

particular, the induction of OASIS was highly significant (p<0.001) in MS brains

compared to non-MS controls. To determine the cell-types expressing ER stress proteins,

we analyzed tissue sections from MS and non-MS control brain sections. Demyelinated

(D) lesions from MS patient brains (B i) stained less intensely with luxol fast blue (LFB),

hematoxylin and eosin compared to a non-MS control brain section (B ii). Serial sections

Chapter 5: Syncytin-1 induces ER stress

A

SYNCYTIN-1 CONTROL B

30 * Control 25 Syncytin-1 (HFA) 20 15

mRNA RFC mRNA 10 * * * 5 * * 0 7 -1 BiP RK p5 N D153 E R Y D P E OASIS S GA

C Control 7 6 Syncytin-1 5 (MDM) 4 3 mRNA RFC 2 1

7 RK p5 BiP D153 E R D P E GA

Fig.37: Syncytin-1 expressing human fetal astrocytes (HFA) but not controls exhibit syncytia formation (A) (inset: GFAP-positive astrocytes) with concurrent increase in mRNA levels of Syncytin-1 and ER stress genes, BiP, GADD153, PERK, ERp57 and OASIS compared to controls (B). SINrep5-Syncytin-1 infected monocyte- derived macrophages (MDMs) showed no significant differences in the level of induction of ER stress genes, BiP, GADD153, PERK and ERp57 compared to controls (C). Statistical comparisons (t test with Dunnet’s test for post-hoc analysis) were made between Syncytin-1 relative to controls (* p<0.05) 172 Chapter 5: Syncytin-1 induces ER stress

A B MS Non-MS i ii

500 LFB D 400 Non-MS MS *** 300 iiii iv 200 50 * 40 * mRNA RFC mRNA 30 * 20 SYNCYTIN-1 10 * * 0 7 -1 v vi BiP RK p5 N D153 E R Y D P E OASIS S GA GADD153

vii viii BiP

ix x ERp57

Fig. 38: Increased transcript levels of Syncytin-1, BiP, GADD153, PERK, ERp57, and OASIS were evident in the white matter tissue of MS (n=12) brains relative to non-MS controls (n=11) (A). Demyelinated (D) lesions from MS patient brains (B i) stained less intensely with luxol fast blue (LFB), hematoxylin and eosin compared to a non-MS control (B ii). MS brains showed marked increase in Syncytin-1 immunoreactivity (B iii), particularly in GFAP-positive astrocytes (B iv), GADD153 (B v) and BiP (B vii) expression compared to non-MS control brain (B vi & viii). ERp57 immunoreactivity was observed in both MS (B, ix) and non-MS control brain (B, x) in and around blood vessels (Original magnification X400; 173 (inset, X1000). Statistical comparisons (t test with Dunnet’s test for post-hoc analysis) were made between treatments relative to controls (*** p<0.001, * p<0.05). Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 174

of demyelinated regions in the brains of MS patients demonstrated marked increase in

Syncytin-1 immunoreactivity (B iii), particularly in astrocytes expressing GFAP (B iv)

(Syncytin-1-blue and astrocytes-brown) (inset, figure shows GFAP-positive astrocyte).

Sections from the same lesions show increased GADD153 (B v) and BiP (B vii) expression compared to non-MS control brain (B vi & viii; arrow shows blood vessel).

GADD153 expression was increased in several cell-types but most prominently in

astrocytes, co-localized with GFAP immunoreactivity (B v, inset) (GADD153-blue and

GFAP-brown, arrowhead) in brains of MS patients. BiP expression was found in several

cell types including astrocytes (arrow) and macrophages (arrowhead) in the white matter

(B vii) of demyelinating lesions (arrow shows blood vessel). ERp57 was minimally expressed with no differences between clinical groups (B, ix & x). Thus, multiple ER stress genes including OASIS were overexpressed in brain glia of MS patients, suggesting a role for ER stress in the pathogenesis of MS.

V.2.2. OASIS down-regulates ASCT1 expression in astrocytes

As several retroviral proteins including Syncytin-1 mediate receptor interference

308 and induce free radicals in astrocytes 196, we hypothesized that Syncytin-1 might

influence expression and function of its putative receptors, ASCT1 and -2 in the brain 131, perhaps through regulation by free radical production and removal, similar to the related transporter, xCT 402,407. Hence, we examined the expression of ASCT1 and 2 in brain

white matter from MS and non-MS brains. Intense ASCT1 immunoreactivity was

observed in the white matter of non-MS brain sections (Fig. 39A) on glial cells including

both astrocytic (Fig. 39A, inset) and CD45-positive monocytoid cells (Fig. 39B, inset)

but was reduced in MS sections (Fig. 39B). Expression of ASCT2, however, revealed no

Chapter 5: Syncytin-1 induces ER stress

Non-MS MS

A B ASCT1

C D ASCT2

EF Non-MS (n=15) 35 30 * Non-MSMS kDa 25 MS (n-12) 50 * 20 25 42 15 10 15 mRNA RFC mRNA 5 OASIS/ACTIN 0 ND 5 WM CTX WM CTX Non-MS MS G ASCT1 ASCT2 H 2.5 Control 2 SYN-1 Vec OASIS kDa OASIS 1.5 iNOS 130 β-Actin 42 1

mRNA RFC mRNA 0.5 ** 0 ** ASCT1 ASCT2 Fig. 39: ASCT1 expression was detected in non-MS brains (A), particularly in astrocytes (A, inset) and CD45-positive leukocytes (B, inset) compared to minimal immunoreactivity in MS white matter (B). ASCT2 expression did not differ between MS (D) and non-MS controls (C). MS brains showed down-regulation of ASCT1 mRNA in white matter (WM) but not in the frontal cortex (CTX) compared to non-MS controls. ASCT2 mRNA was undetectable in the CTX and did not differ between groups in the WM (E). OASIS immunoreactivity (50 kDa) was increased in MS brains compared to non-MS controls (F, inset). Graphic analysis of the OASIS immunoreactivity in brains relative to actin revealed a significant increase in MS brains (F). OASIS expressing astrocytes revealed increased iNOS expression (G). 175 Astrocytes overexpressing OASIS or Syncytin-1 down-regulated ASCT1, but not ASCT2 compared to empty vector (pCDNA 3.1) and mock controls (H) (Original magnification X400 (C); (inset, X1000) (Dunnet’s test for post-hoc analysis ** p< 0.01, * p<0.05). Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 176

differences in immunoreactivity between non-MS (Fig. 39C) and MS patients’ white

matter (Fig. 39D). Interestingly, ASCT2 expression was predominantly expressed on

activated microglia (Fig. 39D). To examine the effects of Syncytin-1 expression in

astrocytes, we performed microarray analysis of astrocytic cells transduced with the

Sindbis virus-derived vector expressing Syncytin-1. Investigation of gene expression profile (Table 6) revealed several genes, particularly an MS lesion-specific transcript and

a disintegrin and metalloproteinase (ADAM)-10 in addition to multiple genes involved in

ER stress, myelin synthesis and immune response (Table 6), which were also modulated

in acute lesions from brains of MS patients 408 (Table 7). Importantly, ASCT1 and 2 were

down-regulated in astrocytes overexpressing Syncytin-1 (Table 6). Corroborating these

findings was the observation that ASCT1 transcripts were significantly diminished in MS brain white matter relative to non-MS brains. Conversely, ASCT2 transcript levels did

not differ in white matter between clinical groups. ASCT1 transcript levels did not differ

in (frontal) cortex between non-MS and MS patients while ASCT2 was not detected in

cortex (Fig. 39E).

Corresponding to increased transcript levels of OASIS in MS brain lesions (Fig.

38A), western blot analysis (Fig. 39F, inset) of non-MS (n=6) and MS (n=6) patient

brain lysates revealed a significant increase in OASIS protein levels in the brain (Fig.

39F). Since OASIS activates the transcription of target genes via acting on the ER stress

responsive element (ERSE) and cyclic AMP responsive element (CRE) 409, we hypothesized that OASIS might trigger iNOS through CRE on the iNOS promoter 410.

Indeed, increased expression of iNOS in OASIS transfected astrocytes was evident compared to empty vector (Fig. 39G). Based on these results, we transfected astrocytes

177

Table 6: Gene profile in Syncytin-1 expressing astrocytes showing up-regulation and down-regulation of immune response, myelin-related and ER stress genes.

Gene Function Fold GenBank ID Name change STCH stress 70 protein chaperone, microsome-associated, 60kDa 8.3 AI718418 HSPH1 heat shock 105kDa/110kDa protein 1 5.29 NM_006644 LONP peroxisomal lon protease 3.32 AV700132 FLJ23560 hypothetical protein FLJ23560 2.56 NM_024685 ER stress FLJ14281 hypothetical protein FLJ14281 2.38 NM_024920 response HSPD1 heat shock 60kDa protein 1 (chaperonin) 2.24 NM_002156 SERP1 stress-associated endoplasmic reticulum protein 1 2.26 AL136807 homocysteine-inducible, endoplasmic reticulum stress- HERPUD1 inducible, ubiquitin-like domain member 1 0.74 BC015447 heat shock 70kDa protein 5 (glucose-regulated protein, HSPA5BP1 78kDa) binding protein 1 0.71 AB046803 GADD45B growth arrest and DNA-damage-inducible, beta 0.70 AV658684 Syncytin-1 HERV-W envelope glycoprotein 3.11 AC000064 solute carrier family 1 (glutamate/neutral amino acid SLC1A4 transporter), member 4 0.68 W72527 solute carrier family 1 (neutral amino acid transporter), SLC1A5 member 5 0.69 AF105230 ADAM10 a disintegrin and metalloproteinase domain 10 8.25 N51370 MS- MS-lesion MS lesion transcript 6.82 N73682 related Charcot-Marie-Tooth neuropathy 4B2 (autosomal recessive, CMT4B2 with myelin outfolding) 4.48 AK022478 MBP myelin basic protein 0.73 NM_002385 MAG myelin associated glycoprotein 0.69 X98405 SOCS5 suppressor of cytokine signaling 5 5.38 NM_014011 Immune STAT1 signal transducer and activator of transcription 1, 91kDa 2.12 NM_007315 response TLR4 toll-like receptor 4 2.05 NM_003266 TLR9 toll-like receptor 9 0.73 AB045180 IL23A interleukin 23, alpha subunit p19 0.72 NM_016584 OLIG1 oligodendrocyte transcription factor 1 0.72 AL355743 TLR8 toll-like receptor 8 0.61 NM_016610 OAS1 2',5'-oligoadenylate synthetase 1, 40/46kDa 0.41 NM_002534

178

Table 7: Genes exhibiting increased and decreased transcript levels in Syncytin-1 expressing astrocytes, showing a similar profile in lesions from MS patients (Lock et al., 2002) Gene Function Fold GenBank ID Name change CEBPG CCAAT/enhancer binding protein (C/EBP), gamma 2.75 BE622659 TNFRSF6 tumor necrosis factor receptor superfamily, member 6 2.43 NM_000043 RCN1 reticulocalbin 1, EF-hand calcium binding domain 1.61 NM_002901 GAD2 glutamate decarboxylase 2 (pancreatic islets and brain, 65kDa) 0.96 BQ128302 SPOCK2 sparc/osteonectin, cwcv and kazal-like domains proteoglycan (testican) 2 0.96 NM_014767 SST somatostatin 0.91 NM_001048 NEUROD2 neurogenic differentiation 2 0.88 AB021742 PVALB parvalbumin 0.87 NM_002854 CCNI cyclin I 0.85 BG530368 SCD stearoyl-CoA desaturase (delta-9- desaturase) 0.84 BC005807 RPS4Y1 ribosomal protein S4, Y-linked 1 0.83 NM_001008 RAB5B RAB5B, member RAS oncogene family 0.82 AF267863 PEG3 paternally expressed 3 0.82 AF208967 HTR7 5-hydroxytryptamine (serotonin) receptor 7 (adenylate cyclase-coupled) 0.71 NM_019859 ACAT2 acetyl-Coenzyme A acetyltransferase 2 (acetoacetyl Coenzyme A thiolase) 0.70 BC000408 HSPA6 heat shock 70kDa protein 6 (HSP70B') 0.70 NM_002155 AQP11 aquaporin 11 0.68 AI886656 MAP1D methionine aminopeptidase 1D 0.68 AA779679 KIF5A kinesin family member 5A 0.67 AF063608 CNP 2',3'-cyclic nucleotide 3' phosphodiesterase 0.62 AK098048 EXTL3 Exostoses (multiple)-like3 0.62 BC006363 Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 179

with an OASIS expression vector, which resulted in down-regulation of Syncytin-1 receptor, ASCT1, but not ASCT2 with the latter gene demonstrating an increase in expression (Fig. 39H). These results were supported by the finding of down-regulation of

ASCT1, but not ASCT2, in Syncytin-1 expressing astrocytes (Fig. 39H). Thus, Syncytin-

1-induced OASIS expression and resulting ER stress was associated with down- regulation of ASCT1 expression, but not ASCT2, which recapitulated the present observations in MS brain lesions.

V.2.3. Syncytin-1 diminishes oligodendrocyte viability

Oligodendrocyte death may represent the earliest neuropathological feature of

MS, in advance of infiltrating inflammatory cells 159 but is also a cardinal feature of

established MS lesions 411. Indeed, we have previously demonstrated that free radicals

induced by Syncytin-1 contribute to oligodendrocyte damage 196. To determine whether

Syncytin-1 expression in astrocytes affected oligodendrocyte viability, we treated

oligodendrocytes with supernatants from astrocytes overexpressing Syncytin-1 and

examined oligodendrocyte viability by measuring the levels of two markers of

myelination, CNPase (2′,3′-cyclic nucleotide 3′-phosphodiesterase) and MBP.

Supernatant from Syncytin-1-transfected astrocytes reduced the number of CNPase-

positive oligodendrocytes (Fig. 40Aii) compared to mock-treated cells (Fig. 40Ai).

Counts of CNPase-positive cells revealed that indeed Syncytin-1-transfected astrocytes significantly reduced the number of CNPase-positive oligodendrocytes compared to mock-treated cells (Fig. 40B). Immunofluorescence levels of CNPase-expressing

oligodendrocytes (Fig. 40C) were reduced by Syncytin-1-mediated toxicity but there was

no effect of Syncytin-1 on MBP-immunofluorescence (Fig. 40C). Since our observations

Chapter 5: Syncytin-1 induces ER stress

A B MOCK SYN-1 250 iii 200 150

CNPase 100 *** 50 CNPase (+)OLs 0 -1 N Y C D MOCK S

140 CNPase MBP 120 Control BS 14 100 ** 12 ** + 80 10 MBP 60 + 8 ** ♣ ♣ 40 6

Rat OL 4 20 Fluorescence (%) 2 0 Casp.3 Act. 0 00.11.0 1 N- Benzylserine (mM) Y S MOCK

Benzylserine

Fig. 40: Oligodendrocytes treated with supernatants from mock, Syncytin-1 overexpressing human fetal astrocytes (A) or supernatants from astrocytes treated with 1 mM benzylserine (C) revealed that the supernatant from Syncytin-1 overexpressing astrocytes (A) and supernatants from astrocytes treated with the ASCT inhibitor, benzylserine (C) were cytotoxic to oligodendrocytes compared to mock. (B) Quantitative analysis of immunofluorescence depicted as a percent of CNPase-immunoreactivity was decreased by both treatments (C). Immunoreactivity of MBP was not affected by various treatments (C). Oligodendrocytes were immunostained for MBP (red) and activated caspase-3 (green). Number of activated caspase-3 and MBP-positive oligodendrocytes were counted per field and expressed as a ratio of positive cells in benzylserine containing supernatant to that of DMSO containing supernatant from astrocytes. Results indicate a significant increase in oligodendrocyte damage and injury with benzylserine treatment. Original magnification 400x 180 (A). Statistical comparisons (ANOVA) were made between treatments relative to mock controls (Dunnet’s test for post-hoc analysis; ♣ p<0.001, ** p<0.01). Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 181

indicated a reduction in expression of ASCT1 in MS brain white matter (Fig. 39E) and in astrocytes overexpressing Syncytin-1 (Fig. 39G), we hypothesized that inhibition of

ASCT transporters may adversely affect amino acid flux, thus contributing to astrocyte

dysfunction. Inhibitors of ASCT transporters include benzylserine and benzylcysteine 412.

Supernatants from astrocytes treated with benzylserine were found to reduce immunofluorescence levels of both CNPase and MBP, more so than astrocytes expressing Syncytin-1 (p<0.01) (Fig. 40C). Further, supernatant from astrocytes treated with benzylserine induced caspase-3 in rat oligodendrocytes in a dose-dependent manner

(Fig. 40D). Thus, Syncytin-1 expression and inhibition of ASCT transporter function exerted cytotoxic effects on oligodendrocytes.

V.2.4. iNOS and Egr1 suppress ASCT1 in astrocytes

Increased expression of iNOS 138,196 and Egr1 391, an established transcriptional repressor of TNF-α 413 and ASCT1 414 are consistent observations in the brains of MS

patients. Given that OASIS (Fig. 38 & 39) was induced in brains of MS patients, we

examined the contributions of iNOS, Egr1 and OASIS to the expression of ASCT1.

Transfection of astrocytes with a pCDNA vector expressing Syncytin-1 revealed

significant induction of Egr1 but not Egr3 and Egr4 (Fig. 41A). Treatment of astrocytes

with the NO donor, sodium nitroprusside (SNP) reduced ASCT1 mRNA (p<0.05) (Fig.

41B). Expression of Egr1 was also significantly increased by SNP in keeping with earlier

studies showing that NO enhanced Egr1 expression 415 (Fig. 41B). Our observations were

confirmed by quantitative immunofluorescence analysis of Egr1 expression, which was

increased in astrocytes after SNP or Syncytin-1 treatment (Fig. 42A). SNP treatment

decreased ASCT1 expression in a dose-dependent manner in astrocytes (Fig. 42B),

Chapter 5: Syncytin-1 induces ER stress

A B

30 40 ** * Control ASCT1 25 30 Vector Egr1 20 20 Syncytin-1 10 15 5 10 4 mRNA RFC mRNA

mRNA RFC mRNA 3 5 2 1 * 0 0 Egr1 Egr3 Egr4 -SNP +SNP

Fig. 41: Expression of Syncytin-1 in astrocytes induced the expression of the transcription factor, Egr1 but not Egr3 and Egr4 (A). Treatment of astrocytes with sodium nitroprusside (SNP) significantly decreased ASCT1 expression, concurrently increasing the expression of Egr1 (B). Statistical comparisons (ANOVA) were made between treatments relative to controls (Dunnet’s test for post-hoc analysis ; ** p<0.01, * p<0.05).

182 Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 183

confirming our PCR results (Fig. 41B). Enhanced expression of Egr1 was also observed in MS brain white matter lesions (Fig. 42D) compared to non-MS controls (Fig. 42C).

Egr1 was localized predominantly in the cytoplasm of astrocytes in the white matter

(GFAP-positive astrocytes, blue; Egr1-brown) (Fig. 42D). Thus, these observations

indicated that Syncytin-1 expression in astrocytes diminished ASCT1 expression, which

involved induction of the transcription factors, OASIS and Egr1 by Syncytin-1.

V.2.5. Soluble Syncytin-1 down-regulates ASCT1 in astrocytes

As soluble retroviral envelope proteins are capable of modulating their cognate

receptors’ expression 416, directly inducing immune responses in different cell types 417 and down-regulating amino acid transporters 418, we hypothesized that secreted Syncytin-

1 might also affect astrocyte function. Since Syncytin-1 is highly expressed in brain

tissue 196, its expression was investigated in plasma and CSF of MS patients. Protein was

extracted as described previously 345 followed by SDS-PAGE analysis. Syncytin-1 was

detected in plasma and CSF and to a lesser extent in supernatant from mitogen (PMA)-

stimulated monocytoid cells (Fig. 43A). To determine if secreted Syncytin-1 engaged its

putative receptor on glial cells, we constructed pseudotyped virions expressing Syncytin-

1. HERVs can be assembled into virions through transcomplementation with virion

proteins encoded by different retroviruses 256 and thus, we determined whether Syncytin-

1 could act as a functional envelope glycoprotein permitting neural cell infection. By

pseudotyping an envelope-defective HIV-1 clone containing a luciferase reporter gene

(pNL-Luc-E-R-) 264 with Syncytin-1, I observed that supernatants from cells transfected with pCMVenv-Syncytin-1 alone, or co-transfected with pNL-Luc-E-R-, exhibited

Syncytin-1 immunoreactivity suggesting that Syncytin-1 and pseudotyped virions were

Chapter 5: Syncytin-1 induces ER stress

A B

(%) 140 120

* (%) 120 * 100 100 80 * 80 60 60 ASCT1 * 40

Egr1/Tubulin 40 20 20

P 100 10 1 N-1 N Con Y S Con pcDNA S pcDNA SNP (nM)

Non-MS MS C D Egr1

Fig. 42: Quantitative immunofluorescence of Egr1 showed significant induction in astrocytes treated with soluble Syncytin-1 or SNP relative to supernatant from pCDNA (empty) vector transfected HEK293T cells (A). SNP dose-dependently suppressed ASCT1 in astrocytes as determined by quantitative immunofluorescence analysis (B). MS brains (D) revealed increased expression of Egr1 in astrocytes in the white matter compared to non-MS controls (C). Statistical comparisons (t test) were made between treatments relative to controls (Original magnification 400x) (E & F), (inset F, X1000) (Dunnet’s test for post-hoc analysis; * p<0.05)

184 Chapter 5: Syncytin-1 induces ER stress

B A Syn-1 Syn-1+Luc Cont MA S φ 70 KDa C -ND A-M -CSF M 4 MS PLA HIV P MS ASTRO MDM *** 75 kDa Syn-1 25 Neurons 20 1 N- 15 D Y Vector S 10 *** 75 kDa 5 Relative Light Units x 10– x Units Light Relative Syn-1 Luc Syn-1+Luc

Fig. 43: Soluble Syncytin-1 was detectable in plasma and CSF of MS patients and supernatant of monocytoid (U937) cells (MΦ) stimulated with PMA but not in brain tissue from HIV patients without dementia (HIV-ND) (A). Syncytin-1 was detectable in supernatants from HEK293T transfected with a Syncytin-1 vector, which was enhanced by pseudotyping with the HIV vector, pNL- Luc-E-R-+ (Luc) (B). MDMs and HFAs but not neurons were permissive to pseudotyped virus, based on cellular luciferase activity. Supernatants from cells transfected with pNL-Luc-E-R-+ (Luc) failed to show any luciferase activity (C). Soluble Syncytin-1 protein from the vector pVGW427 exhibited an immunoreactive band at 70 kDa (D). Statistical comparisons (ANOVA) were made between pseudotyped virus relative to Syncytin-1 alone (Dunnet’s test for post-hoc analysis; ***p<0.001).

185 Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 186

released into the culture medium. Importantly, Syncytin-1 immunoreactivity was

increased in supernatant from the co-transfected cells (Fig. 43B). Examination of the cell-

type specificity associated with pseudotyped virus infection revealed that human

macrophages and astrocytes were permissive to infection by pseudotyped viruses while human neurons (LAN-2) were not infected based on luciferase expression (Fig. 43C).

Indeed, HeLa cells and a feline lymphocyte line (MYA-1) were also not permissive to the

pseudotyped virus, underscoring the selective cell tropism mediated by Syncytin-1,

similar to the neural cell tropism of exogenous retroviruses 419. We then produced soluble

Syncytin-1 that was secreted by pVGW427-transfected HEK293T cells (Fig. 43D). A

distinct immunoreactive band at 75 kDa was observed by western blot analysis using a

mAb to Syncytin-1 (Fig. 43D).

Following treatment of astrocytes with soluble Syncytin-1, its temporal effects on receptor expression was examined. Suppression of ASCT1 levels was observed in astrocytes after Syncytin-1 treatment for 96 h (Fig. 44A). Conversely, β-tubulin and

ASCT2 levels in similarly treated cells revealed no differences in protein expression at all time points examined (Fig. 44A). ASCT1 suppression was independent of regulation by

cytokines, as both IL-10 and IL-1β significantly increased the transcription of ASCT1

(Fig. 44B). Interestingly, soluble Syncytin-1 also induced OASIS transcription in

astrocytes, which was inhibited by the mAb to Syncytin-1 (Fig. 44C), corroborating

previous observations (Fig. 37B). The suppression of ASCT1 transcripts observed in

Syncytin-1-overexpressing astrocytes was significantly reversed by treatment of cells

with a NOS inhibitor, L-NAME [5 μM] (Fig. 45A) and minocycline treatment, which

also inhibited PMA-induced iNOS expression (Fig. 45B). Thus, ASCT1 expression in

Chapter 5: Syncytin-1 induces ER stress

A B 180 Tubulin ASCT1 8 U373 7 * 140 ASCT2 U937 6 * * 5 100 4 60 3 2 Fluorescence (%) Fluorescence 20 1 ASCT1 mRNA RFC mRNA ASCT1 0 Cont IL-10 IL-1β TNF-α 24 48 72 96 Control Hours

C 100 U373 80

60

40

20 OASIS mRNA RFC 0 Vector Syn-1 Syn1+mAb

Fig. 44: Chronic treatment of astrocytes with soluble Syncytin-1 resulted in diminished ASCT1 expression immunoreactivity although β-tubulin and ASCT2 were not altered (A). Astrocytes (U373) and monocytoid cells (U937) were treated with IL-10, IL-1β and TNF-α (10 ng/ml) for 24 h and expression of ASCT1 was examined by real time RT-PCR. Expression of ASCT1 was examined following cytokine treatment, which revealed a significant increase with IL-10 and IL-1β, but not with TNF-α treatment, particularly in astrocytes (U373) but not monocytoid cells (U937) (B). Treatment of human fetal astrocytes with soluble Syncytin-1 significantly induced mRNA levels of OASIS, which was inhibited when Syncytin-1 was blocked with the anti- Syncytin-1 mAb (6A2B2) (C). Statistical comparisons (ANOVA) 187 were made between treatments relative to controls (Dunnet’s test for post-hoc analysis; * p<0.05). Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 188

astrocytes could also be diminished by secreted Syncytin-1, suggesting that ASCT1

expression may be regulated by both extracellular and intracellular Syncytin-1 exposure.

V.2.6. Syncytin-1 transgenic mice exhibit neuroinflammation

We have previously reported up-regulation of Syncytin-1 in the brains of MS

patients relative to age-matched controls 196. To elucidate the mechanism by which

Syncytin-1-mediates oligodendrocyte injury and to confirm in vitro data obtained above, we generated three independent lines of transgenic (Tg) mice expressing Syncytin-1, driven by the GFAP promoter 420 (Fig. 46A). Transgene detection was performed by PCR assay using tail-derived genomic DNA (Fig. 46B). Lines of Tg mice were obtained by serial back-crossing onto C57/BL6 mice as the latter are susceptible to EAE (Fig. 46C).

As Syncytin-1 Tg mice displayed no obvious disease phenotype, we utilized a model of

MS in which PBS or murine recombinant TNF-α was stereotaxically implanted into the corpus callosum 196, an anatomical region usually exhibiting demyelination in MS

patients 421. In fact, TNF-α is a highly potent inducer of Syncytin-1 expression in

astrocytes (Fig. 47A). Since Syncytin-1 Tg and wild type (WT) littermates stereotaxically

implanted with PBS did not demonstrate Syncytin-1 expression (Fig. 47B), subsequent comparisons were made between Tg and WT animals implanted with TNF-α, which significantly induced Syncytin-1 mRNA (Fig. 48A) and protein (Fig. 48B) expression in brains of Tg mice. To determine whether Syncytin-1 expression in the brain provoked an inflammatory response in mice, we examined TNF-α-implanted Syncytin-1-Tg and WT littermates, which showed induction of the pro-inflammatory genes, TNF-α and IFN-α

(Fig. 49A). Interestingly, expression of CNPase and ceramide galactosyltransferase

(CGT), indicators of oligodendrocyte viability/myelination, was significantly reduced in

Chapter 5: Syncytin-1 induces ER stress

A B 2.5 ASCT1 * 65 *** iNOS 2 ASCT1 45 *** 1.5 25

1 7.5 mRNA RFC mRNA mRNA RFC mRNA * * 0.5 5.0 2.5 0 Control Syn-1 Syn-1 CPP+MCPP+M + L-NAME Astro MDM

Fig. 45: Suppression of ASCT1 transcripts observed in Syncytin-1- overexpressing astrocytes was significantly reversed by treatment of cells with an iNOS inhibitor, L-NAME (A). Minocycline restored ASCT1 in astrocytes and monocytoid cells and simultaneously suppressed PMA-induced iNOS (B). Statistical comparisons (ANOVA) were made between treatments relative to controls (Dunnet’s test for post-hoc analysis; *** p<0.001, * p<0.05).

189 Chapter 5: Syncytin-1 induces ER stress A B

Syn-1 1 kb GFAP hGH promoter Not1 Syncytin-1 ATG poly A EcoR1 EcoR1

SV40 1.6 Kb 267 bp SD/SA

C

Ear Tag No. 1 2 3 N 5 6 7 N N N N N 13 N 15 Founder Mice

95 5 11 F1 112 149 137

F2 19

122 F3 124

Fig. 46: The Syncytin-1 gene was cloned into pFGH vector containing the GFAP promoter (A). Transgene integration was confirmed by genotyping revealing a 267 bp product (Syn-1) (B). Map showing lineage of transgenic mice

190 Chapter 5: Syncytin-1 induces ER stress

A B

*** 100 Syncytin-1 U373 75 PBS 50 Tg- Tg WT 25 TNF-α 1 2 3 1 2 3 kDa 5 Syncytin-1 75 4 mRNA RFC mRNA 3 β-Actin 42 2 1

l α -β A 5 10 15 20 5 10 15 20 N N M F IF P α ControI β -1 IL TNF-

Fig. 47: Syncytin-1 expression in U373 astrocytes was determined by real time RT-PCR analysis after treatment with IFN-α and IFN-β (100 U/ml), PMA (50 ng/ml) and varying doses of IL-1β and TNF-α. Syncytin-1 expression was significantly induced in astrocytes treated with 15 ng/ml of TNFα compared to untreated control (A). Implantation of PBS into the corpus callosum of Syncytin-1 transgenic mice did not induce Syncytin-1 expression compared to implantation with TNF-α, where Syncytin-1 immunoreactive band is seen in Lane 1. Tg (n=3) and WT (n=3) mice implanted with PBS did not reveal any Syncytin-1 immunoreactive bands when SDS-PAGE followed by Western blot analysis was performed (B). Statistical comparisons (ANOVA) were made between treatments relative to controls (Dunnet’s test for post-hoc analysis; *** p< 0.001).

191 Chapter 5: Syncytin-1 induces ER stress

A B

2.5 Syncytin-1 * TNF-α 2 Tg Wt 1.5 1 2 3 1 2 3 + kDa 1 Syncytin-1

mRNA RFC mRNA 75 0.5 β-Actin 42 Tg-PBS Tg-TNFα

Fig. 48: TNF-α-implantation induced Syncytin-1 in Tg mice but not in Tg mice implanted with PBS (A). TNF-α-implantation induced Syncytin-1 immunoreactivity on western blot in Tg mice brains but not in WT littermates (B). Statistical comparisons (t test) were made between treatments relative to controls (Dunnet’s test for post-hoc analysis; * p<0.05).

192 Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 193

Tg mice (Fig. 49A). The transcript levels of MOG and proteolipid protein (PLP) were not

affected in the Syncytin-Tg mice relative to WT animals (Fig. 49B). The brains of TNF-

α-implanted Syncytin-1 Tg mice revealed astrocytosis, demonstrated by increased GFAP immunoreactivity (Fig. 49C i&ii). Expression of Iba-1-positive microglia/macrophages was enhanced in the brains of Tg mice compared to WT littermates (Fig. 49C iii&iv).

Brains of Tg mice displayed increased numbers of infiltrating CD3-positive T cells relative to controls (Fig. 49C v&vi). Importantly, Syncytin-1-positive astrocytes (Fig.

49C viii, inset) in brains of Tg mice also demonstrated enhanced iNOS immunoreactivity

(Fig. 49C viii) relative to WT littermates (Fig. 49C vii), which supported the notion that iNOS and ER stress induction are closely coupled 228. Analysis of CNPase-

immunoreactivity in the corpus callosum demonstrated a marked reduction in Syncytin-1

Tg mice (Fig. 49C x) compared to WT littermates implanted with TNF-α (Fig. 49C ix),

confirming earlier observations 196. Interestingly, there was no difference in MBP immunoreactivity between Tg (Fig. 49C xii) and WT littermates (Fig. 49C xi) implanted with TNF-α.

V.2.7. Syncytin-1 Tg animals show ER stress

Brain tissue from TNF-α-implanted Tg mice also revealed significant induction

of ER stress gene transcripts, ERp57, OASIS, and GADD153 compared to WT

littermates (Fig. 50A), while both BiP and PERK showed a trend towards increased

transcript levels. Of particular interest, GADD153 was observed in astrocytes in brains of

Syncytin-1 Tg mice (Fig. 50C). BiP was up-regulated in brains of Syncytin-1 Tg mice

(Fig. 50A), but expression was predominantly in cortical neurons (Fig. 50C).

Interestingly, OASIS, which is induced during astrocytic ER stress 398 and in acute

Chapter 5: Syncytin-1 induces ER stress

A B

1500 TNFα *** 3 Wt-TNFα (n=6) 1200 IFNα 900 Tg-TNFα (n=6) CGT 600 2 300 CNPase 3 * mRNA RFC mRNA

2 RFC mRNA 1

1 * * 0 0 Wt-TNFα Tg-TNFα MOG PLP

C GFAP Iba-1 CD3 iNOS CNPase MBP i iii v vii ix xi

cc

ii iv vi viii x xii

cc Tg-TNF Wt-TNF

Fig. 49: TNF-α implantation in corpus callosum of Syncytin-1 Tg mice enhanced levels of pro-inflammatory genes, IFN-α and TNF-α and decreased levels of the oligodendrocyte markers, CGT and CNPase in Syncytin-1 Tg mice relative to WT littermates (A). Transcript levels of MOG and PLP did not differ between groups (B). Syncytin-1 Tg mice implanted with TNF-α revealed astrocytosis (GFAP immunoreactivity) (A ii inset), microgliosis (Iba-1- immunoreactivity) (A iv), infiltrating CD3-positive T cells (A vi) and iNOS immunoreactivity in Syncytin-1-positive cells (A viii, inset) compared to WT littermates (A i, iii, v & vii). CNPase expression was reduced in the corpus callosum of Syncytin-1Tg mice implanted with TNF-α (A x) compared to WT littermates (A ix). Syncytin-1 Tg (A xi) mice did not show any difference in the expression of myelin basic protein (MBP) in the corpus callosum (CC) after TNF-α implantation compared to WT littermates (A xii) (Original 194 magnification 50x A, xi&xii) (400x, inset 1000x). Statistical comparisons (ANOVA) were made between expression levels in Tg mice relative to Wt littermates (Dunnet’s test for post-hoc analysis; *** p<0.001, * p<0.05). Chapter 5: Syncytin-1 induces ER stress AB

1000 Wt-TNFα (n=6) Tg-TNFα (n=6) ** 100 Tg-TNFα Wt-TNFα kDa * GADD153 30 mRNA RFC mRNA 10 * BiP 78 1 β-Actin 42 7 RK BiP D153 p5 E R D P E OASIS GA

Wt-TNFα Tg-TNF α Tg-PBS C i alsmCortex callosum Corpus BiP

ii BiP

iii callosum Corpus GADD153

Fig. 50: TNF-α implantation significantly enhanced levels of ER stress genes, particularly GADD153, ERp57 and OASIS in the brains of TNF-α- implanted Syncytin-1 Tg mice relative to WT controls (A). These results were also confirmed by western blot analysis, which revealed induction of GADD153 and BiP in Syncytin-1 Tg mice brains (C) Immunohistochemical analyses of expression of ER stress proteins, BiP and GADD153 in Syncytin-1 Tg mice revealed BiP immunostaining in glial cells in the corpus callosum (i) and also in neurons in the cortex (ii). Expression of GADD153 was mostly observed in astrocytes in the corpus callosum of Syncytin-1 Tg mice implanted with TNF-α and to a lesser extent in Tg mice implanted with PBS (iii) (Original magnification 630x). Statistical comparisons ( t test) were 195 made between treatments relative to controls (Dunnet’s test for post-hoc analysis; ** p<0.01, * p<0.05). Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 196 lesions of MS patients (Fig. 38) was also increased in the brain of Syncytin-1 Tg mice

(Fig. 50A).

Complementing these findings, Syncytin-1 Tg mice implanted with TNF-α displayed significantly lower ASCT1 transcript levels but ASCT2 did not differ between groups. This finding corresponded to higher iNOS mRNA levels in the Tg mice (Fig.

51A), emphasizing our immunohistochemical analysis. Induction of pro-inflammatory molecules was specific to the brain, since treatment of macrophages from Syncytin-1-Tg mice with TNF-α did not induce iNOS expression differentially compared with WT- derived macrophages (Fig. 51B). Further, comparison of nNOS transcripts did not differ between groups (Fig. 51C). The number of ASCT1-positive cells was reduced in

Syncytin-Tg mice implanted with TNF-α compared to WT littermates (Fig. 51D).

Stereotaxic implantation of TNF-α into the corpus callosum of Syncytin-1 Tg animals resulted in significant neurobehavioral deficits compared to TNF-α implanted WT littermates at days 3 and 6 post-implantation (Fig. 51E). The mean deficit scores were obtained from a combination of three behavioral tests described previously 347. TNF-α- implanted Tg mice grasped a horizontal rod for significantly less time compared to the

WT littermates. In addition, Tg mice were sufficiently impaired that they could not hold on to an inverted screen and reach the screen edge, while WT littermates reached the edges of the inverted screen more quickly. Lastly, Tg mice implanted with TNF-α exhibited mean delays in the time taken to cross a cantilevered beam, compared to WT littermates, suggesting that the Tg mice showed diminished motor activity and exploratory behavior. Thus, in vivo Syncytin-1 overexpression caused oligodendrocyte

Chapter 5: Syncytin-1 induces ER stress

A B C Wt-TNFα (n=6) WT-TNFα Tg-TNFα (n=6) 1.4 Tg-TNFα 250 1.2 BMDM-iNOS *** 150 1.0 50 1 5 0.8 4 0.6 0.6

3 0.4 RFC mRNA mRNA RFC mRNA mRNA RFC mRNA 2 1 0.2 0.2 0 * ASCT1 ASCT2 iNOS Wt-TNFα Tg-TNFα nNOS

D E Wt-TNFα (n=6) 60 3.5 Tg-TNFα (n=6) * 2.5 40 * * cells (%) + MDS 1.5 20 0.5 ASCT1 0 Wt-TNFα Tg-TNFα Day 3 Day 6

Fig. 51: TNF-α-implantation induced iNOS but reduced ASCT1 expression, but not ASCT2, in Syncytin-1 Tg mice relative to WT littermates (A). Bone marrow-derived macrophages (BMDM) from Syncytin-1 Tg mice and WT littermate controls were treated with TNFα. Expression of iNOS did not differ between WT and Tg groups (B). Transcript levels of nNOS in brains of Tg mice did not differ from that of the WT littermates (C) Analysis of ASCT1-immunopositive cells in the corpus callosum (D, inset) revealed significantly lower numbers in TNF- α-implanted brains of Syncytin-1 Tg mice compared to WT littermate controls (D). Mean deficit scores (MDS) revealed significantly high MDS scores for Syncytin-1 Tg mice implanted with TNF-α compared to WT littermates (E) (Original magnification: D, X400). Statistical comparisons (ANOVA) were made between treatments relative to controls (Tukey’s Multiple Comparison Test was performed; *** p<0.001, * p<0.05). 197 Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 198 loss and neurobehavioral abnormalities, which were accompanied by ER stress and down-regulation of Syncytin-1’s receptor, ASCT1.

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 199

V.3. Discussion

In this Chapter, we have demonstrated enhanced expression of ER stress proteins,

GADD153, ERp57, BiP, PERK and OASIS, in demyelinating brain lesions of MS

patients. We have also identified a novel role for Syncytin-1 whereby through the

induction of ER stress molecules, and in particular OASIS, this retroviral protein mediates down-regulation of its receptor, ASCT1 with ensuing adverse effects on oligodendrocyte survival. Using a newly developed mouse model of MS, Syncytin-1 was

found to induce neuroinflammation, myelin injury and neurobehavioral deficits that were

accompanied by ER stress in astrocytes, recapitulating our observations in MS patients.

Retrovirus receptor interference and down-regulation proceeds from direct

interactions between the virus and the receptor or through indirect (intracellular)

mechanisms through redox regulation of the putative receptor 422. Excessive or long-term

ER stresses result in apoptotic cell death, and a balance between apoptotic and ER stress

signals determines cell fate after ER stress 398. Since inflammation can drive an ER stress

response 230, induction of ER stress genes in the brains of Syncytin-1 Tg mice may be the

result of enhanced inflammation in conjunction with Syncytin-1 expression observed in

the brain of these animals. We have previously demonstrated that Syncytin-1 expression

in astrocytes increases pro-inflammatory cytokine expression 196. Herein, we demonstrate

that Syncytin-1-induced inflammation enhances ER stress in astrocytes and in particular,

the expression of OASIS. The regulation of this recently discovered molecule 398 has not

been elucidated in great detail. Nevertheless, for the first time, we have shown that

OASIS is induced in MS brain tissue, astrocytes overexpressing Syncytin-1 and

following chronic exposure to Syncytin-1, and in each case with an ensuing reduction in

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 200

ASCT1. Suppression of ASCT1 might be brought about by OASIS-mediated iNOS

expression through the production of Egr1. Moreover, our present observations imply an

interaction of Syncytin-1 with ASCT1, leading to diminished oligodendrocyte.

The receptors for Syncytin-1, ASCT1 and 2, are widely expressed on most cell

types 333, but their expression in the human brain has not been previously characterized.

Expression of ASCT2 is low in the adult brain 423 and our observations of ASCT2

immunostaining in human brains represent the first report of its expression predominantly

in microglial cells. In the mouse brain, ASCT1 is chiefly expressed in GFAP-positive

astrocytes in the cerebral cortex and corpus callosum, but not in neurons,

oligodendrocytes or activated/resting microglia 424, similar to our observations in the

white matter of human brains. Interestingly, weak ASCT1 labeling was observed around

ER cisterns, revealing its intracellular trafficking pathway or ASCT1 may have a

functional role in exchange of ions between cytoplasm and ER lumen 424. In our study,

we provide evidence for the down-regulation of ASCT1 in MS brain white matter.

ASCT1 was also found to be significantly down-regulated in glial cells treated with 7-

ketocholestrol, a by-product of myelin, damaged by oxidative stress, a key feature of MS

425, which supports the present findings. Reduced expression of ASCT1 in brain

astrocytes of MS patients appears to have adverse consequences for oligodendrocytes and

perhaps other proximate cells’ health and function. ASCT1 is the principal transport

system involved in the secretion of L-serine 426, a potent astrocyte-derived neurotrophic

factor 424, essential for myelination 427 and neuronal survival 338. Conversely, ASCT1 is also responsible for mediating intracellular transport of the excitotoxic amino acid, cysteine 338, preventing its extracellular accumulation. Interestingly, a reciprocal change

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 201

of Syncytin-1 and its putative receptor, along with diminished amino acid influx has been

observed in the placenta 428. Down-regulation of ASCT1 expression in mouse brain

capillaries has been observed during the second postnatal week and is speculated to be due to lowered demand for small neutral amino acids from circulation or their increased

synthesis in local glial cells 424. Our results suggest a highly specific role for ASCT1

down-regulation, since another astrocytic, amino acid transporter, EAAT1 429, was unaffected by Syncytin-1. Indeed, this finding supports our earlier studies showing that

Syncytin-1-mediated effects on oligodendrocytes were not dependent on the glutamate receptors NMDA-R and AMPA-R 172.

To investigate the mechanism of ASCT1 down-regulation in astrocytes, we

hypothesized that NO might play a role as NO donors are known to modulate ASCT2

expression 430. Further, our rationale for using NO donors was that Syncytin-1 mediates

NO production and formation of peroxynitrites 196, both of which are known to induce

ER stress 431. iNOS expression and overproduction of NO in astrocytes of MS

demyelinating lesions also contributes to inflammation and tissue injury 138,196. Indeed,

SNP, an NO donor, diminished ASCT1 expression in astrocytes, but also induced Egr1,

an established repressor protein of ASCT1 in neural cells 414,432. Of note, Egr1 also

suppresses TNF-α 413, which may have pathogenic consequences in MS due to the

protective nature of this pro-inflammatory cytokine 433. Indeed, Bcl-2-induced Egr1 DNA

binding activity has been correlated to oligodendrocyte death 434. Since iNOS and Egr1

are significantly enhanced in brain lesions of MS patients 391 with concurrent down-

regulation of ASCT1, we might have also identified a potential role for Egr1 in MS

neuropathogenesis.

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 202

Syncytin-1 is a complex endogenous retroviral protein that is largely beneficial to

the host in terms of enhancing placental development 334. However, in the brains of MS

patients, it becomes a part of the inflammatory cascade when its expression is increased

by various factors including viruses 197. Syncytin-1 overexpression in astrocytes is detrimental 196 and also results in syncytia formation, which corroborates recent

observations in an MS neuropathology wherein syncytium formation was reported 435.

Cytokines are known to enhance expression of HERV env genes 379 and in the present

study we found that TNF-α increased Syncytin-1 expression in the Tg mice, likely

through activation of the GFAP promoter. Enhanced inflammatory responses in the brain

of Syncytin-1 Tg mice as well as recruitment of CD3-positive T cells are highly

significant in our model of MS since the effect of T cell-mediated pathology as well as

the potential role of Syncytin-1 to chemoattract T cells into the brain can be investigated.

The significance of my study lies in the pathogenic consequences of Syncytin-1, whose

expression in astrocytes has negative consequences for oligodendrocyte viability.

CNPase, an early marker of myelination and oligodendrocyte viability, was suppressed in

our transgenic model compared to MBP, a late marker. Supporting our observation is the finding that CNPase, but not MBP, is regulated by redox reactants 436. A major

implication of our findings, therefore, is that compromised expression of ASCT1 in the

brain, particularly in astrocytes, can contribute to altered concentrations of small neutral

amino acids, which might potentiate neurotoxicity by excessive stimulation of the

NMDA receptor 437. In the light of recent evidence for NMDA receptor expression on

oligodendrocytes 89, the effect of neutral amino acids on viability of oligodendrocytes

warrants further investigation. Availability of specific antagonists/inhibitors for ASCT1

Chapter 5: SYNCYTIN-1 INDUCES ER STRESS 203

will also be useful in dissecting the molecular mechanisms of ASCT1 suppression in

mediating neuropathogenesis. We speculate that the supernatant from astrocytes

expressing Syncytin-1 might be containing lower amounts of neutral amino acids,

particularly, L-serine than the controls. If so, this might contribute to the demyelination

seen in our transgenic mouse model as well as in vitro experiments.

Thus, we have identified a novel pathway in the pathogenesis of MS, wherein

Syncytin-1 induces ER stress in astrocytes. The prototypic ER stress molecule examined in this study, OASIS, and Syncytin-1-mediated NO production suppressed ASCT1 in astrocytes, which compromised oligodendrocyte viability (Fig. 52). These observations highlight the perturbed expression of Syncytin-1 and its receptor, ASCT1, in neuroinflammation providing new insights into the pathogenesis of MS and other neuroinflammatory disorders. Based on our results, this new mouse model of MS expressing a HERV glycoprotein in astrocytes can be used to dissect the pathogenic mechanisms of MS, without the constraints reported for other MS models 157.

Activated astrocyte Syncytin-1 Quiescent astrocytes OASIS Syncytin-1- OASIS Egr1 NO P ASCT1 ASCT1 induced ER stress Syncytin-1 Egr1

Neuroinflammation ASCT1

Healthy Oligodendrocyte toxins oligodendrocyte Demyelination and axonal injury Axon Damaged Myelin ↓ oligodendrocyte

204 Fig. 52: Syncytin-1-mediated neuropathogenesis GENERAL DISCUSSION AND CONCLUSIONS 205

CHAPTER 6

GENERAL DISCUSSION AND CONCLUSIONS

GENERAL DISCUSSION AND CONCLUSIONS 206

VI. General Discussion and Conclusions VI.1. Overview

Since the completion of sequencing of the human genome, scientific focus has

shifted from studying genes, to analyzing the proteins encoded by them. Among these

genes, the endogenous retroviral protein, Syncytin-1, has been characterized as an

essential component for placental development 235. However, as many other proteins do,

expression of Syncytin-1 in cell types other than trophoblasts in the placenta may not

exert beneficial effects. To the best of our knowledge, the present studies represent the

first direct evidence of Syncytin-1 being neuropathogenic, particularly inducing ER stress

and free radicals that are responsible for demyelination, damage and death of oligodendrocytes.

VI.2. Syncytin-1 vs MSRV

It is speculated that Syncytin-1 behaves as a complex host gene that has evolved

from a retrovirus and might not exist as an infectious virus, though evidence supporting

this contention is still lacking. Among infectious agents, several different viruses have been posited to have an association with MS 438. Studies by others in the field of HERV-

induced inflammation have focused primarily on MSRV. MSRV is a component of

previously characterized extracellular virions derived from MS choroid plexus cell

cultures 183 and is a prototype member from which the whole HERV-W family was later

characterized 185. Syncytin-1 is another member of this family and maybe an endogenous counterpart of the exogenous retrovirus, MSRV. Another HERV implicated in MS is

HERV-H, which has also been demonstrated to be transmissible 167 but convincing

evidence is lacking. Nevertheless, the complex reality is that pathogenicity of one

member might be amplified by unavoidable interactions with other family members.

GENERAL DISCUSSION AND CONCLUSIONS 207

Indeed, superinfection with exogenous retroviruses that are closely related appear to

escalate the disease course 439. In fact, other non-RNA viruses have the ability to drive

expression of Syncytin-1 in the brain 197, further corroborating our contention that MS is not caused by Syncytin-1, but rather is an essential component of the inflammatory

cascade that comprises MS demyelinating pathogenesis.

VI.3. Syncytin-1-associated neuroinflammation

In Chapter #3, we have shown that Syncytin-1 expression is increased in the

brain. As mentioned earlier, Syncytin-1 expression is enhanced by other viruses notable

among them being HSV 197. In placental cells, Syncytin-1 expression is decreased under

hypoxic conditions, resulting in decreased cell fusion and placental abnormalities in pre- eclampsia, a common clinical problem in pregnant women 440. The transcription factors

CBF (CCAAT binding factor), Oct-1 (octamer protein-1), AP-1 and Sp1 are known to bind to the Syncytin-1 gene promoter. CBP and Oct-1 sites are critical for transcriptional regulation of the gene in trophoblast cells 441, suggesting that one or more of these transcription factors may be involved in the induction of Syncytin-1 in MS. CBP induces the expression of granulocyte/macrophage colony-stimulating factor (GM-CSF) 442, a hematopoietic growth factor which is increased in association with demyelination in MS and whose deficiency in mice results in resistance to EAE 443. Other transcription factors,

Sp1 and Oct-1 that bind to Syncytin-1 promoter are also known to up-regulate osteopontin in MS 444. Mice lacking osteopontin are resistant to demyelination in the EAE model 445. The pathways upstream of these transcription factors in astrocytes are unknown, but might include cytokines induced in lymphocytes and macrophages early in the MS disease process.

GENERAL DISCUSSION AND CONCLUSIONS 208

VI.3.1. Syncytin-1 mediates inflammation through nitric oxide intermediates

We have shown that Syncytin-1 can cause the production of proinflammatory cytokines and reactive oxygen species in astrocytes. Viral envelope glycoproteins are known to affect immune responses, and Syncytin-1 has amino acid sequences that would be predicted to affect the activation of lymphocytes and macrophages 313. Thus far, a single nucleotide polymorphism (SNP) in Syncytin-1 gene has been reported in the transmembrane domain of Syncytin-1 (www.ncbi.nlm.nih.gov), whose gene is carried by all people in the general population and indeed expresses the protein. However, no studies exist to explain if this polymorphism predisposes certain individuals to MS.

HIV-1 coat protein gp120 can be cytotoxic to neurons and glial cells by a

mechanism involving membrane-associated oxidative stress and up-regulation of IL-1β

and other oxidative stress genes 446, suggesting similarities in the pathogenic actions of

Syncytin-1 and gp120. IL-1β can also increase ROS by enhancing NO production in astrocytes and possibly the activation and cytokine production by MS lesion-associated T cells and macrophages. However, we do know that ROS and intermediate compounds are responsible for damaging oligodendrocytes but additional cytolytic mediators may also be generated by Syncytin-1 expression since neuronal cells were not damaged in our model. It could be because of constitutively low levels of antioxidants in oligodendrocytes that may make them prone to oxidative stress 131. In particular, the

levels of glutathione has been determined to be lower in oligodendrocytes than in neurons

and this might make the former cells vulnerable to oxidative stress. It might perhaps be

interesting to determine whether Syncytin-1 can induce neuropathological changes in

iNOS deficient mice. We have demonstrated a modest but significant increase in protein

GENERAL DISCUSSION AND CONCLUSIONS 209 carbonyls that can kill oligodendrocytes. In fact, a 30% increase in protein carbonyls is also seen in other models of oxidative stress mediated neurodegeneration 447. Moreover, amyloid β protein can induce 60% increase in protein carbonyls, which corresponds to

20-40% neuronal death. Thus there is no direct correlation between amount of protein carbonyls and cell death 447.

Previously it has been demonstrated that T cells reactive to MSRV were specifically and clonally selected for expansion 189. Since the contribution of T cells to

MS pathogenesis is well established and cannot be ignored in the current model, it is important to determine whether T cells derived from MS patients are capable of mounting an immune response to Syncytin-1, which is up-regulated in activated glial cells of MS patients. It may be interesting to demonstrate if EAE is worsened in a

Syncytin-1 transgenic mouse, which is part of an ongoing study in the laboratory. If tissue injury or trauma releases the otherwise sequestered Syncytin-1, a normal immune response comprising autoantibodies and autoreactive T-lymphocytes might be generated which can modulate pathogenesis in MS patients. It is also imperative to understand the immunological tolerance to Syncytin-1 during development.

VI.4. Quantification of Syncytin-1 copy numbers in tissues

VI.4.1. Syncytin-1 RNA copy numbers in the brain

Chapter#4 deals with using HERV copy number as an important variable for understanding retroviral dynamics. Using simple RNA and DNA extraction and performing real time PCR analyses, we have demonstrated that Syncytin-1 RNA and

DNA copy numbers are increased in brain tissue of MS patients, but not from the PBMC.

This study has several implications, notably that it cannot be used as a biomarker, unless

GENERAL DISCUSSION AND CONCLUSIONS 210 of course brain biopsies will be performed regularly to monitor disease status. Indeed,

Syncytin-1 RNA copy numbers were quantified, revealing increased levels in the brains of MS patients. This was not surprising since we have shown this by relative quantification in Chapter#3. However, the copy numbers that we obtained in this study may not reflect actual copy numbers for several reasons. Firstly, the assay may not be the most accurate. We have compared the copy numbers in the brain RNA sample to RNA transcribed in vitro from a Syncytin-1 expressing construct. RNA extracted from tissues exhibit less efficient amplification than from in vitro RNA. It may be necessary to make standard curves including non-human tissue extracts to overcome this problem. Secondly, there is tremendous variability in the stability of RNA in post mortem tissue resulting in

RNA levels that differ extensively between clinical samples as well as stability of

GAPDH mRNA which may be quite different from that of Syncytin-1 mRNA. This may explain why the difference in quantitative analyses (p<0.05) of Syncytin-1 mRNA is less significant than that of the relative quantification analysis (p<0.01). The 2.5 fold change in RNA copy numbers in MS patients over controls seen in this study could be low either due to difference in patterns of death among different patients and removal of necropsy samples or it could be high due to insensitivity of the assay for the above mentioned reasons. However, in our analysis, the mean PCR threshold cycle differences between cDNA from MS (20.52) and control (20.24) brain tissue for the housekeeping gene,

GAPDH, was not significant, suggesting that the differences observed in RNA copy numbers were not due to differences in RNA and cDNA quality between groups.

Moreover, the 0.5log10 difference (2 fold change) seen with RNA copy numbers has translated into a 3-fold change in Syncytin-1 protein expression in the brain by western

GENERAL DISCUSSION AND CONCLUSIONS 211

blot analysis. Relative quantification analysis (2.74 fold) of Syncytin-1 RNA in the brain

of MS patients relative to controls agreed with our quantitative analysis (2.95 fold)

highlighting the utility of our assays, as well as indicating that the variations in RNA

extracted from autopsy specimen and in vitro transcription reactions may be minimal.

VI.4.2. Syncytin-1 RNA copy numbers in CSF and plasma

One concern with quantification of HERV is that cellular RNA may be released

due to cell lysis in samples after blood collection or CSF obtained by lumbar puncture

during sample treatment and storage. Previously, quantification of MSRV virion was

complex since it could not be differentiated from cellular RNA released from dead cells.

This problem was overcome by incorporating centrifugation, filtration, RNase and DNase

digestion steps to ensure that only particle-associated MSRV RNA was extracted and to

remove any non-encapsidated RNA 184,448 and thus prevented contamination of irrelevant

HERV sequences from cellular DNA or RNA in extracellular fluids. However, in our study, we have examined the total RNA extracted from cells and other body fluids in an effort to understand copy numbers of Syncytin-1 as one might see in the clinical setting, comprising both particle-associated and non-encapsidated RNA. The biological relevance of Syncytin-1 in CSF and plasma, amplified in my study is yet to be fully understood,

though extracellular Syncytin-1 protein might be involved in regulation of its cognate

receptor expression (Chapter #5).

Other issues that we pondered over but did not address in my thesis are whether

our PCR primers cross react with other overexpressed loci since competition in a PCR

reaction will favour the higher amount of mRNA. Syncytin-1 mRNA is derived from a single specific locus (7q21q2), whereas there is no specific locus for MSRV and it was

GENERAL DISCUSSION AND CONCLUSIONS 212

also revealed that it was the Syncytin-1 locus that had a role in placental morphogenesis

due to a conserved ORF as opposed to mutated gag, pol pseudogenes of the same locus

334. Indeed, we sequenced the PCR product, which revealed that it is Syncytin-1 locus

(7q) that is overexpressed. Studies suggest that of 30 env sequences, only one on

chromosome 7q is expressed 449. Northern blot analysis revealed the presence of genomic, sub-genomic and large spliced mRNAs 186. Transcripts from defective

Syncytin-1-like sequences or due to alternate splicing might influence the results of

Syncytin-1 quantitative PCR assay developed in this study, although the latter is

improbable because retrovirus envelopes do not usually contain splice variants. cDNA

synthesis performed in this study employed random primers, which can bind to mRNA

transcripts of varied length. The real time RT-PCR experiments could be performed with

oligodT primers that bind only to polyadenylated mature mRNA.

VI.4.3. Syncytin-1 DNA copy numbers and integration events

Analysis of DNA copy numbers by PCR has proven to be an interesting

challenge. Measurement of MSRV pol gene DNA copy numbers by fluorescent in situ

hybridization (FISH) revealed an increased copies of the gene in MS patient samples 450.

Similarly rigorous PCR quantification procedures applied in my thesis for Syncytin-1 revealed massive increases in Syncytin-1-encoding DNA copy numbers in the brains of

MS patients relative to controls (Fig. 35C). Whether the increase in DNA copy numbers implied new viral DNA insertions due to ongoing reinfection by replication competent

HERV was investigated. We looked for evidence of newly acquired proviruses by cloning virus-cell junctions and determining new integration junctions. Since our PCR assays did not detect increased numbers of HERV-W-LTR (U5)-Alu PCR fragments, we

GENERAL DISCUSSION AND CONCLUSIONS 213

speculated that the increase in DNA copy numbers can be attributed to accumulation of

unintegrated linear retroviral cDNA sequences 301, which were assessed by PCR of 1 and

2 LTR circles. Also Southern blotting of supernatant DNA, a technique named Hirt assay could be performed to detect LTR circles or episomal DNA 451, although this was not performed in my studies. We have demonstrated that the env sequences are amplified but not the LTR sequences perhaps by a mechanism similar to that of the dihydrofolate reductase (DHFR) gene under methotrexate selection 452. Accumulation of abundant

unintegrated retroviral DNA post reverse transcription, suggests that integration may be

aborted. This also suggests that superinfection of replication-competent retrovirus may be occurring, overcoming cellular receptor interference, which was also not investigated in our study.

Appendix E shows the calculations performed to study the retroviral copy

numbers. We acknowledge that there may be several issues here. Firstly, the

measurement of DNA may not be accurate since the samples may contain, in addition to

genomic DNA, contaminating RNA and ribonucleotides that could influence the results

despite rigorous attempts to generate pure DNA. To control for this, GAPDH was used to

normalize the data though the PCR efficiency for detection of Syncytin-1 and GAPDH

are not similar. Secondly, the human genome consists of 654 LTRs of HERV-W 292 of

which we examined one in our assay. This may not reflect precisely the Syncytin-1 locus

unless extensive sequencing of various PCR fragments was carried out. Thirdly, use of

appropriate controls and methodology is essential here. As mentioned before, Syncytin-1

is not unique to MS and it is assumed that Syncytin-1 copy number is fairly constant

among a control population. However, we have observed that the non-MS controls from

GENERAL DISCUSSION AND CONCLUSIONS 214

brain and PBMC differ by more than 1000 fold in copy numbers, which we attribute to

different methods of DNA purification from 2 different cohorts of MS patients and from

2 different tissues. Fourthly, our method of normalization may have inherent problems.

The ratio of Syncytin-1 to GAPDH is in the 1-4 range, which means Syncytin-1 copy

number is 2-8 per cell (since the observed 100 fold difference in Syncytin-1 DNA/μg of

total DNA is reduced to 2 fold when normalized to GAPDH) but sometimes this ratio is

less than 1 which suggests that Syncytin-1 copies are less than 2 per cell, a number that is

lower than the existing 30 copies of full-length env in the diploid genome 330. This anomaly can be attributed to normalizing the data to GAPDH and the real time PCR quantification methodology. To normalize the data between 2 genes, it is absolutely critical to have similar PCR efficiencies, which is difficult to achieve with the quality of autopsy material used in this study. Using Taqman probes to perform real time PCR assays might overcome this problem.

The issue of using appropriate clinical samples also needs to be addressed. Our disease controls were obtained from autopsied specimens of other neurological disorders.

Patients suffering from AD, HIV, Cancer etc. may die from a variety of different causes

(Appendix D). Our tissue bank consists of samples that were not standardized in terms of autopsy time. Further, for the purpose of our study, we did not take into consideration differences in copy numbers between patients’ brain tissue by matching for age and sex.

MS is disproportionately distributed with younger females affected whereas AD is more common among older people. Further, comparisons were made between different cohorts of MS patients.

GENERAL DISCUSSION AND CONCLUSIONS 215

Nevertheless, we have made a preliminary attempt to quantify Syncytin-1 DNA

copy numbers in clinical samples, which has not been undertaken before. It must be

acknowledged that syncytia formation in brain of MS patients is relatively unknown

except for a single report 435. So the relevance of Syncytin-1 as a fusion protein in MS

pathogenesis needs further investigation. It could be that Syncytin-1 does not reach the

cellular membrane when overexpressed, but rather is trapped in the ER where it manifests

its pathogenicity and thus no fusion is seen in brain of MS patients or perhaps it may be a temporal effect and there may no way of knowing it since our analyses are performed on autopsy tissue. No systematic studies have ever been carried out to understand if HERVs can be used as a biomarker. We now know that Syncytin-1 expression is increased in MS

brains. However, its expression is also increased relative to healthy control brains in other neuroinflammatory disorders, including HIV-dementia and Alzheimer’s disease, though

its expression was higher in MS.

VI.5. Syncytin-1 induces ER stress in astrocytes

Chapter#5 deals with the mechanism of Syncytin-1-induced neurodegeneration.

An observation that we made was that prolonged overexpression (up to 5 days) of

Syncytin-1 in astrocytes led to cell death. Though apoptosis/necrosis of astrocytes can be

mediated through several pathways, several studies indicate a strong link between

retroviral infections and the induction of ER stress proteins. The UPR is an evolutionarily

conserved response, triggered by accumulation of unfolded proteins in the ER. Viral

infections trigger the UPR and this could represent an ancient evolutionary adaptation

that links ER stress to apoptosis in order to avoid spread of viruses 453. It is possible that

the cell death observed among astrocytes expressing Syncytin-1 over a prolonged period

GENERAL DISCUSSION AND CONCLUSIONS 216 of time is an ER-linked process. Since recent reports suggest oligodendrocyte death in the absence of T cell infiltration and inflammation 159, the culprits may be activated astrocytes in the vicinity of oligodendrocytes 131.

Induction of the UPR permits cells to adapt to the changing microenvironment and establish normal ER function. The adaptive mechanism involves several processes that integrate into restoring cellular normalcy. Translation of new mRNAs is inhibited, thereby reducing the amount and number of proteins destined for the ER for folding and allowing UPR proteins to be synthesized. Transcription of genes that increase the protein folding capacity and ER-associated degradation (ERAD) for removal of misfolded proteins is enhanced. However, due to the nature of the protein that accumulates in the

ER and the overwhelming taxation on the cellular machinery, the adaptive mechanisms may fail, leading cells to initiate an alarm pathway involving NF-κB, a transcription factor involved in host defense response. However, prolonged and excessive ER stress triggers apoptosis, which perhaps is the phenomenon occurring in astrocytes herein.

VI.6. Syncytin-1 causes astrocyte dysfunction

We chose to focus on the dysfunction of astrocytes in our study, rather than apoptosis because frank apoptosis induced by Syncytin-1 was a highly contrived circumstance resulting from very efficient transduction of astrocytes. There are several studies that describe death of astrocytes in MS 389 454 455 456 and this may be reversed by

IFN-β treatment 455. However, this is not a well-established clinical pathology, perhaps because astrocytes are proliferating in MS 457. Astrocyte dysfunction resulting in reduced trophic support to oligodendrocytes may lead to oligodendrogliopathy observed in MS lesions 458. Rat pups injected with astrocytes demonstrated oligodendrocyte remyelination

GENERAL DISCUSSION AND CONCLUSIONS 217 in an ethidium bromide model of demyelination, perhaps by secreting platelet-derived growth factor (PDGF) 459. An astroglial response called isomorphic gliosis is associated with improved recovery from tissue damaging insults. Soluble trophic and growth factors produced by these astrocytes enhance the survival of adjacent neurons and glia as well as contribute to tissue remodeling 111. Oligodendrocyte precursors proliferate in response to

FGF-2 induced by CNTF-secreting astrocytes 108. Thus, astrocytes contribute substantially to oligodendrocyte viability and myelination, which has implication for design of therapeutics in MS.

VI.7. Syncytin-1 indirectly regulates its receptor expression

In Chapter #3, we brought to attention the enhanced expression of Syncytin-1 in

MS brains. During investigation of the consequence of Syncytin-1 overexpression in astrocytes, we discovered that expression of the receptor for Syncytin-1, a transporter molecule named ASCT1, was diminished. Since ASCT1 transports alanine, serine, cysteine and threonine, accumulation or withdrawal of these neutral amino acids in the astrocyte culture supernatant, might harm adjacent oligodendrocytes. The amount of L- serine and other neutral amino acids in the culture media can be determined by high performance liquid chromatography (HPLC) analysis and we intend to complete this experiment in due course. To investigate the mechanism of Syncytin-1-induced neuroinflammation and whether MS pathogenesis is enhanced by an ER stress response that has not subsided, we developed a transgenic mouse model of MS that expresses

Syncytin-1 in astrocytes under the GFAP promoter. Indeed, the Syncytin-1 transgenic mouse exhibited clinical features resembling MS. In fact, we also observed T cell infiltration into the corpus callosum of transgenic mice upon TNF-α implantation,

GENERAL DISCUSSION AND CONCLUSIONS 218

suggesting that the adaptive immune component of neuroinflammation is activated in this

model. The role of T cells in Syncytin-1-mediated neuropathogenesis can be investigated

in our novel Syncytin-1 transgenic mouse model of MS.

Based on the data obtained from analysis of CSF and plasma of MS patients,

wherein we detected Syncytin-1 by Western blotting, we wish to caution from deriving

conclusions from this observation, as this may not necessarily mean presence of soluble

and secreted Syncytin-1. Rather, we may have detected Syncytin-1 from lysed cells that

released host proteins into the CSF and plasma. Soluble and secreted Syncytin-1 may

interact with its putative receptors and may enter astrocytes via pinocytic mechanisms.

The implication of the recent identification of two mouse orthologues of

Syncytin-1 and Syncytin-2, named Syncytin-A and Syncytin-B 317 respectively is important to understand in the context of the Syncytin-1 transgenic mouse model of MS developed in this study. Experiments must be carried out to determine whether the transcript and protein levels of Syncytin-A are modulated in the Syncytin-1 transgenic mouse model in various treatment paradigms.

GENERAL DISCUSSION AND CONCLUSIONS 219

VI. 8. Conclusions and Future Perspectives

The goals of this project were to identify and characterize the function of a HERV glycoprotein encoded by the human genome and its relationship to neuropathogenesis.

The knowledge gained from these studies might be used to identify and evaluate novel targets for intervention of neuroinflammation in MS. Though our studies imply that MS is not caused by Syncytin-1 per se, the results indicate a strong relationship between induction of Syncytin-1 in astrocytes and diminished oligodendrocyte viability mediated by ER stress and free radical production. They also highlight the role of astrocytes in the pathogenesis of MS. Future research in this direction may help identify novel therapeutic compounds that modulate the behaviour of astrocytes during neuroinflammation.

APPENDICES 220

APPENDICES

221

APPENDIX A: SINrep5 VECTOR USED IN THIS THESIS

222

Appendix B: Quality and quantity of DNA extracted from brain tissue Patient ID Concentration (ng/μl) OD (26/280) 1 63.9 1.82 2 652.7 1.74 3 364.4 1.82 4 201.5 1.72 5 312.8 1.66 6 560.3 1.68 7 195.6 1.79 8 109.8 1.71 9 214.3 1.75 10 375.7 1.74 11 862.9 1.68 12 394.6 1.87 13 257.5 1.74 14 131.2 1.61 15 131.2 1.77 16 449.7 1.68 17 176.8 1.71 18 144.9 1.94 19 294.6 1.66 20 719.8 1.61 21 244.5 1.94 22 348.3 1.79 23 1656.2 1.89 24 774.4 1.79 25 625.6 1.76 26 820.8 1.9 27 1757.9 1.9 28 799.5 1.69 29 1016.6 1.7 30 469.7 1.77 31 222.5 1.71 32 312.4 1.59 33 502.8 1.7 34 749.7 1.61 35 278.8 1.76 36 306.9 1.73 37 1537.8 1.81 38 407.2 1.85 39 317.1 1.81 40 329 1.86

223

APPENDIX C: AFFYMETRIX GENECHIP EXPRESSION ARRAY 224

APPENDIX D: AUTOPSIED SAMPLES FROM PATIENTS

Patient ID Disease 1- LA MS 2- MF MS 3- EH MS 4- SH MS 5- MH MS 6- WC MS 7- JZ MS 8- MF MS 9-GD MS 10-RW MS 11-RS MS 12-JMK MS 13-JK MS 14-LG (F) MS 15- LA (F) MS 16-MJH MS 17-LJ MS 18-M230-98 MS 19-RS MS 20-SM MS 21-AD1 AD 22-AD2 AD 23-AD3-cerebellum AD 24-AD4 AD 25-AD5 AD 26-AD6 AD 27-A93-125 Control (cerebral arteriolosclerosis, multiple old cerebral infarcts) 28-A93-111 Control (Normal brain) 29-A93-120 Control (Normal brain, drug overdose) 30-A93-119 Control (cerebral arteriolosclerosis, minute foci of old cerebral infarcts) 31-A94-75 Control (Candida endocarditis with septic emboli) 32-A95-117 Control 33-A96-28 Control (Nocardiosis with meningitis and cerebral abscess) 34-A94-82 HIV (MGN encephalitis consistent with CMV) 35-A95-89 HIV (CMV optic neuritis, CMV encephalitis, HSV encephalitis) 36-A93-121 HIV (HIV encephalitis) 37-A94-66 HIV (acute aroxic/ischemic encephalopathy) 38-A95-91 HIV (florid CNS toxo with multiple necrotizing microabscesses) 39-A93-109 HIV (subacute encephalitis) 40-A95-121 HIV 41-A94-97 HIV (microglial nodules, microinfarcts; MNL) 225

APPENDIX E: Calculation of Syncytin-1 DNA copy number in the genome

Total size of human genome (haploid): 3x 109 bases Diploid genome: 6 x 109 bases

1000 bp = 9.1 x 1011 molecules = 1 μg DNA

Therefore, 1 molecule = 1/9.1 x 1011 = 10-12 μg DNA 1000 bp = 10-12 μg DNA 1 bp = 10-15 μg DNA

Therefore, 6 x 109 bases = 6 x 109 x 10-15 μg DNA = 6 pg DNA/cell

6 pg DNA= 30 copies of Syncytin-1 100 ng DNA = 5 x 105 copies of Syncytin-1 226

APPENDIX F: COPYRIGHTS OBTAINED FROM PUBLISHERS REFERENCES 227

REFERENCES

REFERENCES 228

1. Beck, C.A., Metz, L.M., Svenson, L.W. & Patten, S.B., Regional variation of multiple sclerosis prevalence in Canada, Mult Scler, 11, 516, 2005. 2. Steinman, L., Martin, R., Bernard, C., Conlon, P. & Oksenberg, J.R., Multiple sclerosis: deeper understanding of its pathogenesis reveals new targets for therapy, Annu Rev Neurosci, 25, 491, 2002. 3. Lucchinetti, C.F., Brueck, W., Rodriguez, M. & Lassmann, H., Multiple sclerosis: lessons from neuropathology, Semin Neurol, 18, 337, 1998. 4. Yong, V.W. et al., The promise of minocycline in neurology, Lancet Neurol, 3, 744, 2004. 5. Sospedra, M. & Martin, R., Immunology of multiple sclerosis, Annu Rev Immunol, 23, 683, 2005. 6. Carson, M.J., Microglia as liaisons between the immune and central nervous systems: functional implications for multiple sclerosis, Glia, 40, 218, 2002. 7. Villoslada, P., Barcellos, L.F. & Oksenberg, J.R., Chromosome 7q21-22 and multiple sclerosis, J Neuroimmunol, 150, 1, 2004. 8. Mirsattari, S.M. et al., Aboriginals with multiple sclerosis: HLA types and predominance of neuromyelitis optica, Neurology, 56, 317, 2001. 9. Prat, A. & Antel, J., Pathogenesis of multiple sclerosis, Curr Opin Neurol, 18, 225, 2005. 10. Kanki, P.J. et al., Human immunodeficiency virus type 1 subtypes differ in disease progression, J Infect Dis, 179, 68, 1999. 11. Frohman, E.M., Racke, M.K. & Raine, C.S., Multiple sclerosis--the plaque and its pathogenesis, N Engl J Med, 354, 942, 2006. 12. Grigoriadis, N. & Hadjigeorgiou, G.M., Virus-mediated autoimmunity in Multiple Sclerosis, J Autoimmune Dis, 3, 1, 2006. 13. Trapp, B.D., Bo, L., Mork, S. & Chang, A., Pathogenesis of tissue injury in MS lesions, J Neuroimmunol, 98, 49, 1999. 14. Bruck, W., Inflammatory demyelination is not central to the pathogenesis of multiple sclerosis, J Neurol, 252 Suppl 5, v10, 2005. 15. Merkler, D., Ernsting, T., Kerschensteiner, M., Bruck, W. & Stadelmann, C., A new focal EAE model of cortical demyelination: multiple sclerosis-like lesions with rapid resolution of inflammation and extensive remyelination, Brain, 2006. 16. Lassmann, H., Bruck, W. & Lucchinetti, C., Heterogeneity of multiple sclerosis pathogenesis: implications for diagnosis and therapy, Trends Mol Med, 7, 115, 2001. 17. Lucchinetti, C.F., Bruck, W. & Lassmann, H., Evidence for pathogenic heterogeneity in multiple sclerosis, Ann Neurol, 56, 308, 2004. 18. Cifelli, A. et al., Thalamic neurodegeneration in multiple sclerosis, Ann Neurol, 52, 650, 2002. 19. Giuliani, F., Goodyer, C.G., Antel, J.P. & Yong, V.W., Vulnerability of human neurons to T cell-mediated cytotoxicity, J Immunol, 171, 368, 2003. 20. Rao, S.M., Leo, G.J., Bernardin, L. & Unverzagt, F., Cognitive dysfunction in multiple sclerosis. I. Frequency, patterns, and prediction, Neurology, 41, 685, 1991.

REFERENCES 229

21. Ferguson, B., Matyszak, M.K., Esiri, M.M. & Perry, V.H., Axonal damage in acute multiple sclerosis lesions, Brain, 120 ( Pt 3), 393, 1997. 22. Gehrmann, J., Banati, R.B., Cuzner, M.L., Kreutzberg, G.W. & Newcombe, J., Amyloid precursor protein (APP) expression in multiple sclerosis lesions, Glia, 15, 141, 1995. 23. Trapp, B.D. et al., Axonal transection in the lesions of multiple sclerosis, N Engl J Med, 338, 278, 1998. 24. Snell, R.S. Clinical Neuroanatomy for Medical Students, 653 (Little, Brown and Company, Boston, MA, USA, 1992). 25. Williams, R.W. & Herrup, K., The control of neuron number, Annu Rev Neurosci, 11, 423, 1988. 26. Kaur, C., Hao, A.J., Wu, C.H. & Ling, E.A., Origin of microglia, Microsc Res Tech, 54, 2, 2001. 27. Neumann, H., Control of glial immune function by neurons, Glia, 36, 191, 2001. 28. Maehlen, J., Schroder, H.D., Klareskog, L., Olsson, T. & Kristensson, K., Axotomy induces MHC class I antigen expression on rat nerve cells, Neurosci Lett, 92, 8, 1988. 29. Neumann, H., Schmidt, H., Cavalie, A., Jenne, D. & Wekerle, H., Major histocompatibility complex (MHC) class I gene expression in single neurons of the central nervous system: differential regulation by interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha, J Exp Med, 185, 305, 1997. 30. Johnson, R.T. Viral infections of the nervous system, 265 (Lippincott-Raven Publishers, Philadelphia, 1998). 31. Lowenstein, P.R., Immunology of viral-vector-mediated gene transfer into the brain: an evolutionary and developmental perspective, Trends Immunol, 23, 23, 2002. 32. Steinman, L., Elaborate interactions between the immune and nervous systems, Nature Immunology, 5, 575, 2004. 33. Kosugi, I., Kawasaki, H., Arai, Y. & Tsutsui, Y., Innate immune responses to cytomegalovirus infection in the developing mouse brain and their evasion by virus-infected neurons, Am J Pathol, 161, 919, 2002. 34. Brocke, S., Piercy, C., Steinman, L., Weissman, I.L. & Veromaa, T., Antibodies to CD44 and integrin alpha4, but not L-selectin, prevent central nervous system inflammation and experimental encephalomyelitis by blocking secondary leukocyte recruitment, Proc Natl Acad Sci U S A, 96, 6896, 1999. 35. Griffin, D.E., Immune responses to RNA-virus infections of the CNS, Nat Rev Immunol, 3, 493, 2003. 36. Callahan, M.K. & Ransohoff, R.M., Analysis of leukocyte extravasation across the blood-brain barrier: conceptual and technical aspects, Curr Allergy Asthma Rep, 4, 65, 2004. 37. Ransohoff, R.M., Kivisakk, P. & Kidd, G., Three or more routes for leukocyte migration into the central nervous system, Nat Rev Immunol, 3, 569, 2003. 38. Kerfoot, S.M. & Kubes, P., Overlapping roles of P-selectin and alpha 4 integrin to recruit leukocytes to the central nervous system in experimental autoimmune encephalomyelitis, J Immunol, 169, 1000, 2002.

REFERENCES 230

39. Yednock, T.A. et al., Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin, Nature, 356, 63, 1992. 40. Ropper, A.H., Selective treatment of multiple sclerosis, N Engl J Med, 354, 965, 2006. 41. Langer-Gould, A., Atlas, S.W., Green, A.J., Bollen, A.W. & Pelletier, D., Progressive multifocal leukoencephalopathy in a patient treated with natalizumab, N Engl J Med, 353, 375, 2005. 42. Yousry, T.A. et al., Evaluation of patients treated with natalizumab for progressive multifocal leukoencephalopathy, N Engl J Med, 354, 924, 2006. 43. Chavarria, A. & Alcocer-Varela, J., Is damage in central nervous system due to inflammation?, Autoimmun Rev, 3, 251, 2004. 44. Tran, E.H., Hoekstra, K., van Rooijen, N., Dijkstra, C.D. & Owens, T., Immune invasion of the central nervous system parenchyma and experimental allergic encephalomyelitis, but not leukocyte extravasation from blood, are prevented in macrophage-depleted mice, J Immunol, 161, 3767, 1998. 45. Huseby, E.S. et al., A pathogenic role for myelin-specific CD8(+) T cells in a model for multiple sclerosis, J Exp Med, 194, 669, 2001. 46. Monson, N.L. et al., Receptor revision and atypical mutational characteristics in clonally expanded B cells from the cerebrospinal fluid of recently diagnosed multiple sclerosis patients, J Neuroimmunol, 158, 170, 2005. 47. Dines, K.C. & Powell, H.C., Mast cell interactions with the nervous system: relationship to mechanisms of disease, J Neuropathol Exp Neurol, 56, 627, 1997. 48. Orr, E.L. & Stanley, N.C., Brain and spinal cord levels of histamine in Lewis rats with acute experimental autoimmune encephalomyelitis, J Neurochem, 53, 111, 1989. 49. Secor, V.H., Secor, W.E., Gutekunst, C.A. & Brown, M.A., Mast cells are essential for early onset and severe disease in a murine model of multiple sclerosis, J Exp Med, 191, 813, 2000. 50. Rozniecki, J.J., Dimitriadou, V., Lambracht-Hall, M., Pang, X. & Theoharides, T.C., Morphological and functional demonstration of rat dura mater mast cell- neuron interactions in vitro and in vivo, Brain Res, 849, 1, 1999. 51. Tuomisto, L., Kilpelainen, H. & Riekkinen, P., Histamine and histamine-N- methyltransferase in the CSF of patients with multiple sclerosis, Agents Actions, 13, 255, 1983. 52. Zappulla, J.P., Arock, M., Mars, L.T. & Liblau, R.S., Mast cells: new targets for multiple sclerosis therapy?, J Neuroimmunol, 131, 5, 2002. 53. Mukaida, N., Interleukin-8: an expanding universe beyond neutrophil chemotaxis and activation, Int J Hematol, 72, 391, 2000. 54. Lucchinetti, C.F. et al., A role for humoral mechanisms in the pathogenesis of Devic's neuromyelitis optica, Brain, 125, 1450, 2002. 55. Lund, B.T. et al., Increased CXCL8 (IL-8) expression in Multiple Sclerosis, J Neuroimmunol, 155, 161, 2004. 56. Pashenkov, M., Teleshova, N. & Link, H., Inflammation in the central nervous system: the role for dendritic cells, Brain Pathol, 13, 23, 2003. 57. Tailor, P., Tamura, T. & Ozato, K., IRF family proteins and type I interferon induction in dendritic cells, Cell Res, 16, 134, 2006.

REFERENCES 231

58. Wakimoto, H., Johnson, P.R., Knipe, D.M. & Chiocca, E.A., Effects of innate immunity on herpes simplex virus and its ability to kill tumor cells, Gene Ther, 10, 983, 2003. 59. Serafini, B. et al., Dendritic cells in multiple sclerosis lesions: maturation stage, myelin uptake, and interaction with proliferating T cells, J Neuropathol Exp Neurol, 65, 124, 2006. 60. Dejaco, C., Duftner, C., Grubeck-Loebenstein, B. & Schirmer, M., Imbalance of regulatory T cells in human autoimmune diseases, Immunology, 117, 289, 2006. 61. Hong, J., Zang, Y.C., Nie, H. & Zhang, J.Z., CD4+ regulatory T cell responses induced by T cell vaccination in patients with multiple sclerosis, Proc Natl Acad Sci U S A, 103, 5024, 2006. 62. van der Vliet, H.J. et al., Circulating V(alpha24+) Vbeta11+ NKT cell numbers are decreased in a wide variety of diseases that are characterized by autoreactive tissue damage, Clin Immunol, 100, 144, 2001. 63. Marin-Padilla, M., Prenatal development of fibrous (white matter), protoplasmic (gray matter), and layer I astrocytes in the human cerebral cortex: a Golgi study, J Comp Neurol, 357, 554, 1995. 64. Volterra, A. & Meldolesi, J., Astrocytes, from brain glue to communication elements: the revolution continues, Nat Rev Neurosci, 6, 626, 2005. 65. Li, R. et al., Glial fibrillary acidic protein mutations in infantile, juvenile, and adult forms of Alexander disease, Ann Neurol, 57, 310, 2005. 66. Liedtke, W. et al., GFAP is necessary for the integrity of CNS white matter architecture and long-term maintenance of myelination, Neuron, 17, 607, 1996. 67. Bartlett, W.P., Knapp, P.E. & Skoff, R.P., Glial conditioned medium enables jimpy oligodendrocytes to express properties of normal oligodendrocytes: production of myelin antigens and membranes, Glia, 1, 253, 1988. 68. Gay, F.W., Early cellular events in multiple sclerosis. Intimations of an extrinsic myelinolytic antigen, Clin Neurol Neurosurg, 108, 234, 2006. 69. Matute, C., Domercq, M. & Sanchez-Gomez, M.V., Glutamate-mediated glial injury: mechanisms and clinical importance, Glia, 53, 212, 2006. 70. Ambrosini, E. et al., Astrocytes produce dendritic cell-attracting chemokines in vitro and in multiple sclerosis lesions, J Neuropathol Exp Neurol, 64, 706, 2005. 71. Seifert, G., Schilling, K. & Steinhauser, C., Astrocyte dysfunction in neurological disorders: a molecular perspective, Nat Rev Neurosci, 7, 194, 2006. 72. Bush, T.G. et al., Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice, Neuron, 23, 297, 1999. 73. Neufeld, A.H. & Liu, B., Glaucomatous optic neuropathy: when glia misbehave, Neuroscientist, 9, 485, 2003. 74. Sofroniew, M.V., Reactive astrocytes in neural repair and protection, Neuroscientist, 11, 400, 2005. 75. Miller, R.H., Regulation of oligodendrocyte development in the vertebrate CNS, Prog Neurobiol, 67, 451, 2002. 76. Cohen, R.I., Exploring oligodendrocyte guidance: 'to boldly go where no cell has gone before', Cell Mol Life Sci, 62, 505, 2005.

REFERENCES 232

77. Lappe-Siefke, C. et al., Disruption of Cnp1 uncouples oligodendroglial functions in axonal support and myelination, Nat Genet, 33, 366, 2003. 78. Chang, A., Nishiyama, A., Peterson, J., Prineas, J. & Trapp, B.D., NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions, J Neurosci, 20, 6404, 2000. 79. Lledo, P.M., Alonso, M. & Grubb, M.S., Adult neurogenesis and functional plasticity in neuronal circuits, Nat Rev Neurosci, 7, 179, 2006. 80. Peters, A. & Palay, S.L., The morphology of synapses, J Neurocytol, 25, 687, 1996. 81. Hoek, R.M. et al., Down-regulation of the macrophage lineage through interaction with OX2 (CD200), Science, 290, 1768, 2000. 82. Nelson, P.T., Soma, L.A. & Lavi, E., Microglia in diseases of the central nervous system, Ann Med, 34, 491, 2002. 83. Gimsa, U., Peter, S.V., Lehmann, K., Bechmann, I. & Nitsch, R., Axonal damage induced by invading T cells in organotypic central nervous system tissue in vitro: involvement of microglial cells, Brain Pathol, 10, 365, 2000. 84. Hill, K.E., Zollinger, L.V., Watt, H.E., Carlson, N.G. & Rose, J.W., Inducible nitric oxide synthase in chronic active multiple sclerosis plaques: distribution, cellular expression and association with myelin damage, J Neuroimmunol, 151, 171, 2004. 85. Huitinga, I., van Rooijen, N., de Groot, C.J., Uitdehaag, B.M. & Dijkstra, C.D., Suppression of experimental allergic encephalomyelitis in Lewis rats after elimination of macrophages, J Exp Med, 172, 1025, 1990. 86. Scolding, N.J., Jones, J., Compston, D.A. & Morgan, B.P., Oligodendrocyte susceptibility to injury by T-cell perforin, Immunology, 70, 6, 1990. 87. Russell, J.H. & Ley, T.J., Lymphocyte-mediated cytotoxicity, Annu Rev Immunol, 20, 323, 2002. 88. Antel, J.P., Williams, K., Blain, M., McRea, E. & McLaurin, J., Oligodendrocyte lysis by CD4+ T cells independent of tumor necrosis factor, Ann Neurol, 35, 341, 1994. 89. Micu, I. et al., NMDA receptors mediate calcium accumulation in myelin during chemical ischaemia, Nature, 2005. 90. Sun, D. et al., Myelin antigen-specific CD8+ T cells are encephalitogenic and produce severe disease in C57BL/6 mice, J Immunol, 166, 7579, 2001. 91. Lassmann, H. & Ransohoff, R.M., The CD4-Th1 model for multiple sclerosis: a crucial re-appraisal, Trends Immunol, 25, 132, 2004. 92. de Rosbo, N.K. & Ben-Nun, A., T-cell responses to myelin antigens in multiple sclerosis; relevance of the predominant autoimmune reactivity to myelin oligodendrocyte glycoprotein, J Autoimmun, 11, 287, 1998. 93. Takahashi, J.L., Giuliani, F., Power, C., Imai, Y. & Yong, V.W., Interleukin- 1beta promotes oligodendrocyte death through glutamate excitotoxicity, Ann Neurol, 53, 588, 2003. 94. Casaccia-Bonnefil, P., Carter, B.D., Dobrowsky, R.T. & Chao, M.V., Death of oligodendrocytes mediated by the interaction of nerve growth factor with its receptor p75, Nature, 383, 716, 1996.

REFERENCES 233

95. Jurewicz, A., Matysiak, M., Andrzejak, S. & Selmaj, K., TRAIL-induced death of human adult oligodendrocytes is mediated by JNK pathway, Glia, 53, 158, 2006. 96. Matysiak, M., Jurewicz, A., Jaskolski, D. & Selmaj, K., TRAIL induces death of human oligodendrocytes isolated from adult brain, Brain, 125, 2469, 2002. 97. Venters, H.D., Dantzer, R. & Kelley, K.W., Tumor necrosis factor-alpha induces neuronal death by silencing survival signals generated by the type I insulin-like growth factor receptor, Ann N Y Acad Sci, 917, 210, 2000. 98. Pouly, S., Becher, B., Blain, M. & Antel, J.P., Interferon-gamma modulates human oligodendrocyte susceptibility to Fas-mediated apoptosis, J Neuropathol Exp Neurol, 59, 280, 2000. 99. Lassmann, H. & Ransohoff, R.M., The CD4-Th1 model for multiple sclerosis: a critical [correction of crucial] re-appraisal, Trends Immunol, 25, 132, 2004. 100. van Oosten, B.W. et al., Increased MRI activity and immune activation in two multiple sclerosis patients treated with the monoclonal anti-tumor necrosis factor antibody cA2, Neurology, 47, 1531, 1996. 101. Nguyen, M.D., Julien, J.P. & Rivest, S., Innate immunity: the missing link in neuroprotection and neurodegeneration?, Nat Rev Neurosci, 3, 216, 2002. 102. Ono, S.J. et al., Chemokines: roles in leukocyte development, trafficking, and effector function, J Allergy Clin Immunol, 111, 1185, 2003. 103. Gerard, C. & Rollins, B.J., Chemokines and disease, Nat Immunol, 2, 108, 2001. 104. Martino, G., How the brain repairs itself: new therapeutic strategies in inflammatory and degenerative CNS disorders, Lancet Neurol, 3, 372, 2004. 105. Pavelko, K.D. et al., Interleukin-6 protects anterior horn neurons from lethal virus-induced injury, J Neurosci, 23, 481, 2003. 106. Butzkueven, H. et al., LIF receptor signaling limits immune-mediated demyelination by enhancing oligodendrocyte survival, Nat Med, 8, 613, 2002. 107. Lee, J.W. & Juliano, R., Mitogenic signal transduction by integrin- and growth factor receptor-mediated pathways, Mol Cells, 17, 188, 2004. 108. Albrecht, P.J. et al., Astrocytes produce CNTF during the remyelination phase of viral-induced spinal cord demyelination to stimulate FGF-2 production, Neurobiol Dis, 13, 89, 2003. 109. Brenneman, D.E. & Gozes, I., A femtomolar-acting neuroprotective peptide, J Clin Invest, 97, 2299, 1996. 110. Gozes, I. et al., A novel signaling molecule for neuropeptide action: activity- dependent neuroprotective protein, Ann N Y Acad Sci, 897, 125, 1999. 111. Liberto, C.M., Albrecht, P.J., Herx, L.M., Yong, V.W. & Levison, S.W., Pro- regenerative properties of cytokine-activated astrocytes, J Neurochem, 89, 1092, 2004. 112. Picard-Riera, N. et al., Experimental autoimmune encephalomyelitis mobilizes neural progenitors from the subventricular zone to undergo oligodendrogenesis in adult mice, Proc Natl Acad Sci U S A, 99, 13211, 2002. 113. Nakanishi, H., Microglial functions and proteases, Mol Neurobiol, 27, 163, 2003. 114. Burster, T. et al., Cathepsin G, and not the asparagine-specific endoprotease, controls the processing of myelin basic protein in lysosomes from human B lymphocytes, J Immunol, 172, 5495, 2004.

REFERENCES 234

115. Pozio, E. & Morales, M.A., The impact of HIV-protease inhibitors on opportunistic parasites, Trends Parasitol, 21, 58, 2005. 116. Yong, V.W., Metalloproteinases: mediators of pathology and regeneration in the CNS, Nat Rev Neurosci, 6, 931, 2005. 117. Larsen, P.H., Wells, J.E., Stallcup, W.B., Opdenakker, G. & Yong, V.W., Matrix metalloproteinase-9 facilitates remyelination in part by processing the inhibitory NG2 proteoglycan, J Neurosci, 23, 11127, 2003. 118. Larsen, P.H. & Yong, V.W., The expression of matrix metalloproteinase-12 by oligodendrocytes regulates their maturation and morphological differentiation, J Neurosci, 24, 7597, 2004. 119. Vos, C.M. et al., Cytotoxicity by matrix metalloprotease-1 in organotypic spinal cord and dissociated neuronal cultures, Exp Neurol, 163, 324, 2000. 120. Johnston, J.B., Olson, M.E., Rud, E.W. & Power, C., Xenoinfection of nonhuman primates by feline immunodeficiency virus, Curr Biol, 11, 1109, 2001. 121. Zhang, K. et al., HIV-induced metalloproteinase processing of the chemokine stromal cell derived factor-1 causes neurodegeneration, Nat Neurosci, 6, 1064, 2003. 122. Giancotti, F.G. & Ruoslahti, E., Integrin signaling, Science, 285, 1028, 1999. 123. Scarisbrick, I.A. et al., Activity of a newly identified serine protease in CNS demyelination, Brain, 125, 1283, 2002. 124. Noorbakhsh, F. et al., Proteinase-activated receptor-2 induction by neuroinflammation prevents neuronal death during HIV infection, J Immunol, 174, 7320, 2005. 125. Noorbakhsh, F. et al., Proteinase-activated receptor 2 modulates neuroinflammation in experimental autoimmune encephalomyelitis and multiple sclerosis, J Exp Med, 203, 425, 2006. 126. Vollgraf, U., Wegner, M. & Richter-Landsberg, C., Activation of AP-1 and nuclear factor-kappaB transcription factors is involved in hydrogen peroxide- induced apoptotic cell death of oligodendrocytes, J Neurochem, 73, 2501, 1999. 127. Keller, J.N. et al., Impairment of glucose and glutamate transport and induction of mitochondrial oxidative stress and dysfunction in synaptosomes by amyloid beta- peptide: role of the lipid peroxidation product 4-hydroxynonenal, J Neurochem, 69, 273, 1997. 128. Chiou, C.C. et al., Urinary 8-hydroxydeoxyguanosine and its analogs as DNA marker of oxidative stress: development of an ELISA and measurement in both bladder and prostate cancers, Clin Chim Acta, 334, 87, 2003. 129. Drake, J., Sultana, R., Aksenova, M., Calabrese, V. & Butterfield, D.A., Elevation of mitochondrial glutathione by gamma-glutamylcysteine ethyl ester protects mitochondria against peroxynitrite-induced oxidative stress, J Neurosci Res, 74, 917, 2003. 130. Barnham, K.J., Masters, C.L. & Bush, A.I., Neurodegenerative diseases and oxidative stress, Nat Rev Drug Discov, 3, 205, 2004. 131. Mattson, M.P. & Taub, D.D., Ancient viral protein enrages astrocytes in multiple sclerosis, Nat Neurosci, 7, 1021, 2004.

REFERENCES 235

132. Merrill, J.E., Ignarro, L.J., Sherman, M.P., Melinek, J. & Lane, T.E., Microglial cell cytotoxicity of oligodendrocytes is mediated through nitric oxide, J Immunol, 151, 2132, 1993. 133. Ulvestad, E. et al., Fc receptors for IgG on cultured human microglia mediate cytotoxicity and phagocytosis of antibody-coated targets, J Neuropathol Exp Neurol, 53, 27, 1994. 134. Mackenzie-Graham, A.J., Mitrovic, B., Smoll, A. & Merrill, J.E., Differential sensitivity to nitric oxide in immortalized, cloned murine oligodendrocyte cell lines, Dev Neurosci, 16, 162, 1994. 135. Alderton, W.K., Cooper, C.E. & Knowles, R.G., Nitric oxide synthases: structure, function and inhibition, Biochem J, 357, 593, 2001. 136. Aktan, F., iNOS-mediated nitric oxide production and its regulation, Life Sci, 75, 639, 2004. 137. Nelson, E.J., Connolly, J. & McArthur, P., Nitric oxide and S-nitrosylation: excitotoxic and cell signaling mechanism, Biol Cell, 95, 3, 2003. 138. Bo, L. et al., Induction of nitric oxide synthase in demyelinating regions of multiple sclerosis brains, Ann Neurol, 36, 778, 1994. 139. Bredt, D.S., Endogenous nitric oxide synthesis: biological functions and pathophysiology, Free Radic Res, 31, 577, 1999. 140. Broholm, H. et al., Nitric oxide synthase expression and enzymatic activity in multiple sclerosis, Acta Neurol Scand, 109, 261, 2004. 141. Cha, C.I. et al., Reactive astrocytes express nitric oxide synthase in the spinal cord of transgenic mice expressing a human Cu/Zn SOD mutation, Neuroreport, 9, 1503, 1998. 142. Catania, M.V., Aronica, E., Yankaya, B. & Troost, D., Increased expression of neuronal nitric oxide synthase spliced variants in reactive astrocytes of amyotrophic lateral sclerosis human spinal cord, J Neurosci, 21, RC148, 2001. 143. Tieu, K., Ischiropoulos, H. & Przedborski, S., Nitric oxide and reactive oxygen species in Parkinson's disease, IUBMB Life, 55, 329, 2003. 144. Lipton, S.A. et al., Cysteine regulation of protein function--as exemplified by NMDA-receptor modulation, Trends Neurosci, 25, 474, 2002. 145. Gow, A.J., Farkouh, C.R., Munson, D.A., Posencheg, M.A. & Ischiropoulos, H., Biological significance of nitric oxide-mediated protein modifications, Am J Physiol Lung Cell Mol Physiol, 287, L262, 2004. 146. Bizzozero, O.A., DeJesus, G., Bixler, H.A. & Pastuszyn, A., Evidence of nitrosative damage in the brain white matter of patients with multiple sclerosis, Neurochem Res, 30, 139, 2005. 147. Eiserich, J.P. et al., Microtubule dysfunction by posttranslational nitrotyrosination of alpha-tubulin: a nitric oxide-dependent mechanism of cellular injury, Proc Natl Acad Sci U S A, 96, 6365, 1999. 148. Mihm, M.J. et al., Free 3-nitrotyrosine causes striatal neurodegeneration in vivo, J Neurosci, 21, RC149, 2001. 149. Credle, J.J., Finer-Moore, J.S., Papa, F.R., Stroud, R.M. & Walter, P., On the mechanism of sensing unfolded protein in the endoplasmic reticulum, Proc Natl Acad Sci U S A, 102, 18773, 2005.

REFERENCES 236

150. Lin, W. et al., Interferon-{gamma} inhibits central nervous system remyelination through a process modulated by endoplasmic reticulum stress, Brain, 2006. 151. Rao, R.V., Ellerby, H.M. & Bredesen, D.E., Coupling endoplasmic reticulum stress to the cell death program, Cell Death Differ, 11, 372, 2004. 152. Liu, N. et al., ATM deficiency induces oxidative stress and endoplasmic reticulum stress in astrocytes, Lab Invest, 85, 1471, 2005. 153. Oyadomari, S. & Mori, M., Roles of CHOP/GADD153 in endoplasmic reticulum stress, Cell Death Differ, 11, 381, 2004. 154. Dimcheff, D.E., Faasse, M.A., McAtee, F.J. & Portis, J.L., Endoplasmic reticulum (ER) stress induced by a neurovirulent mouse retrovirus is associated with prolonged BiP binding and retention of a viral protein in the ER, J Biol Chem, 279, 33782, 2004. 155. Kaufman, R.J., Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls, Genes Dev, 13, 1211, 1999. 156. Rivers TM, S.F., Encephalomyelitis accompanied by myelin destruction experimentally produced in monkeys, journal of experimental medicine, 61, 689, 1935. 157. Sriram, S. & Steiner, I., Experimental allergic encephalomyelitis: a misleading model of multiple sclerosis, Ann Neurol, 58, 939, 2005. 158. Genain, C.P. et al., Late complications of immune deviation therapy in a nonhuman primate, Science, 274, 2054, 1996. 159. Barnett, M.H. & Prineas, J.W., Relapsing and remitting multiple sclerosis: pathology of the newly forming lesion, Ann Neurol, 55, 458, 2004. 160. Pluchino, S. & Martino, G., The therapeutic use of stem cells for myelin repair in autoimmune demyelinating disorders, J Neurol Sci, 233, 117, 2005. 161. Trebst, C. & Stangel, M., Promotion of remyelination by immunoglobulins: implications for the treatment of multiple sclerosis, Curr Pharm Des, 12, 241, 2006. 162. Olson, J.K. & Miller, S.D., Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs, J Immunol, 173, 3916, 2004. 163. Noseworthy, J.H., Progress in determining the causes and treatment of multiple sclerosis, Nature, 399, A40, 1999. 164. Haines, J.L. et al., Multiple susceptibility loci for multiple sclerosis, Hum Mol Genet, 11, 2251, 2002. 165. Lincoln, M.R. et al., A predominant role for the HLA class II region in the association of the MHC region with multiple sclerosis, Nat Genet, 37, 1108, 2005. 166. Kenealy, S.J. et al., Examination of seven candidate regions for multiple sclerosis: strong evidence of linkage to chromosome 1q44, Genes Immun, 7, 73, 2006. 167. Vandenbroeck, K. et al., Chromosome 7q21-22 and multiple sclerosis: evidence for a genetic susceptibility effect in vicinity to the protachykinin-1 gene, J Neuroimmunol, 125, 141, 2002. 168. Palma, C. et al., Functional characterization of substance P receptors on cultured human spinal cord astrocytes: synergism of substance P with cytokines in inducing interleukin-6 and prostaglandin E2 production, Glia, 21, 183, 1997.

REFERENCES 237

169. Lieb, K., Fiebich, B.L., Berger, M., Bauer, J. & Schulze-Osthoff, K., The neuropeptide substance P activates transcription factor NF-kappa B and kappa B- dependent gene expression in human astrocytoma cells, J Immunol, 159, 4952, 1997. 170. Fiebich, B.L., Schleicher, S., Butcher, R.D., Craig, A. & Lieb, K., The neuropeptide substance P activates p38 mitogen-activated protein kinase resulting in IL-6 expression independently from NF-kappa B, J Immunol, 165, 5606, 2000. 171. van Boxel-Dezaire, A.H. et al., Decreased interleukin-10 and increased interleukin-12p40 mRNA are associated with disease activity and characterize different disease stages in multiple sclerosis, Ann Neurol, 45, 695, 1999. 172. M Mayne, J.J. Latent and activated brain flore: human herpes virus, endogenous retroviruses,coronaviruses and Chlamydia and their role in neurological disease. in Emerging Neurological Infections (ed. Johnson, C.P.a.R.T.) 363 (Taylor and Francis, 2005). 173. Buljevac, D. et al., Prospective study on the relationship between infections and multiple sclerosis exacerbations, Brain, 125, 952, 2002. 174. Wucherpfennig, K.W. & Strominger, J.L., Molecular mimicry in T cell-mediated autoimmunity: viral peptides activate human T cell clones specific for myelin basic protein, Cell, 80, 695, 1995. 175. Tsunoda, I. & Fujinami, R.S., Two models for multiple sclerosis: experimental allergic encephalomyelitis and Theiler's murine encephalomyelitis virus, J Neuropathol Exp Neurol, 55, 673, 1996. 176. Olson, J.K., Croxford, J.L., Calenoff, M.A., Dal Canto, M.C. & Miller, S.D., A virus-induced molecular mimicry model of multiple sclerosis, J Clin Invest, 108, 311, 2001. 177. Dal Canto, M.C., Calenoff, M.A., Miller, S.D. & Vanderlugt, C.L., Lymphocytes from mice chronically infected with Theiler's murine encephalomyelitis virus produce demyelination of organotypic cultures after stimulation with the major encephalitogenic epitope of myelin proteolipid protein. Epitope spreading in TMEV infection has functional activity, J Neuroimmunol, 104, 79, 2000. 178. Horwitz, M.S. & Sarvetnick, N., Viruses, host responses, and autoimmunity, Immunol Rev, 169, 241, 1999. 179. Hemmer, B., Archelos, J.J. & Hartung, H.P., New concepts in the immunopathogenesis of multiple sclerosis, Nat Rev Neurosci, 3, 291, 2002. 180. Azoulay-Cayla, A., [Is multiple sclerosis a disease of viral origin?], Pathol Biol (Paris), 48, 4, 2000. 181. Rice, G.P., Armstrong, H.A., Bulman, D.E., Paty, D.W. & Ebers, G.C., Absence of antibody to HTLV I and III in sera of Canadian patients with multiple sclerosis and chronic myelopathy, Ann Neurol, 20, 533, 1986. 182. Christensen, T., Moller-Larsen, A. & Haahr, S., A retroviral implication in multiple sclerosis?, Trends Microbiol, 2, 332, 1994. 183. Perron, H. et al., Leptomeningeal cell line from multiple sclerosis with reverse transcriptase activity and viral particles, Res Virol, 140, 551, 1989. 184. Garson, J.A., Tuke, P.W., Giraud, P., Paranhos-Baccala, G. & Perron, H., Detection of virion-associated MSRV-RNA in serum of patients with multiple sclerosis, Lancet, 351, 33., 1998.

REFERENCES 238

185. Perron, H. et al., Molecular identification of a novel retrovirus repeatedly isolated from patients with multiple sclerosis. The Collaborative Research Group on Multiple Sclerosis, Proc Natl Acad Sci U S A, 94, 7583, 1997. 186. Blond, J.L. et al., Molecular characterization and placental expression of HERV- W, a new human endogenous retrovirus family, J Virol, 73, 1175, 1999. 187. Komurian-Pradel, F. et al., Molecular cloning and characterization of MSRV- related sequences associated with retrovirus-like particles, Virology, 260, 1, 1999. 188. Firouzi, R. et al., Multiple sclerosis-associated retrovirus particles cause T lymphocyte- dependent death with brain hemorrhage in humanized SCID mice model, J Neurovirol, 9, 79, 2003. 189. Perron, H. et al., Multiple sclerosis retrovirus particles and recombinant envelope trigger an abnormal immune response in vitro, by inducing polyclonal Vbeta16 T- lymphocyte activation, Virology, 287, 321, 2001. 190. Christensen, T., Pedersen, L., Sorensen, P.D. & Moller-Larsen, A., A transmissible human endogenous retrovirus, AIDS Res Hum Retroviruses, 18, 861, 2002. 191. Christensen, T. et al., Molecular characterization of HERV-H variants associated with multiple sclerosis, Acta Neurol Scand, 101, 229, 2000. 192. Menard, A. et al., Detection of a gliotoxic activity in the cerebrospinal fluid from multiple sclerosis patients, Neurosci Lett, 245, 49, 1998. 193. Power, C., Retroviral diseases of the nervous system: pathogenic host response or viral gene-mediated neurovirulence?, Trends Neurosci, 24, 162, 2001. 194. Perron, H., Perin, J.P., Rieger, F. & Alliel, P.M., Particle-associated retroviral RNA and tandem RGH/HERV-W copies on human chromosome 7q: possible components of a 'chain-reaction' triggered by infectious agents in multiple sclerosis?, J Neurovirol, 6 Suppl 2, S67, 2000. 195. Kolson, D.L. & Gonzalez-Scarano, F., Endogenous retroviruses and multiple sclerosis, Ann Neurol, 50, 429, 2001. 196. Antony, J.M. et al., Human endogenous retrovirus glycoprotein-mediated induction of redox reactants causes oligodendrocyte death and demyelination, Nat Neurosci, 7, 1088, 2004. 197. Ruprecht, K. et al., Regulation of human endogenous retrovirus W protein expression by herpes simplex virus type 1: Implications for multiple sclerosis, J Neurovirol, 12, 65, 2006. 198. Johnston, J.B. et al., Monocyte activation and differentiation augment human endogenous retrovirus expression: implications for inflammatory brain diseases, Ann Neurol, 50, 434, 2001. 199. Schwartz, M. & Kipnis, J., Protective autoimmunity: regulation and prospects for vaccination after brain and spinal cord injuries, Trends Mol Med, 7, 252, 2001. 200. Domingo, E. Microbial Evolution and emerging diseases. in Emerging Neurological Infections (ed. RT, C.P.a.J.) (Taylor and Francis, 2005). 201. Coffin, J.M. Retroviridae: The viruses and their replication. in Fields Virology, Vol. 2 (ed. Fields BN, K.D., Howley PM, Chanock RM, Melnick JL, Monath TP, Roizman B and Straus SE) 1767 (Lippincott-Raven, Philadelphia, New York, 1996).

REFERENCES 239

202. Miller, A.D., Cell-surface receptors for retroviruses and implications for gene transfer, Proc Natl Acad Sci U S A, 93, 11407, 1996. 203. Goff, S.P., Integration of retroviral DNA into the genome of the infected cell, Cancer Cells, 2, 172, 1990. 204. van Regenmortel MHV, F., CM, Bishop DHL, Carsten EB, Estes MK, Lemon SM, Maniloff J, Mayo MA, McGeoch DH, Pringle CR and Wickner RB. Virus Taxonomy, (Academic Press, San Diego, 2000). 205. Larsson, E., Kato, N. & Cohen, M., Human endogenous proviruses, Curr Top Microbiol Immunol, 148, 115, 1989. 206. Andersson, M.L. et al., Diversity of human endogenous retrovirus class II-like sequences, J Gen Virol, 80 ( Pt 1), 255, 1999. 207. Medstrand, P. & Blomberg, J., Characterization of novel reverse transcriptase encoding human endogenous retroviral sequences similar to type A and type B retroviruses: differential transcription in normal human tissues, J Virol, 67, 6778, 1993. 208. Van Maele, B., Busschots, K., Vandekerckhove, L., Christ, F. & Debyser, Z., Cellular co-factors of HIV-1 integration, Trends Biochem Sci, 31, 98, 2006. 209. D'Souza, V. & Summers, M.F., How retroviruses select their genomes, Nat Rev Microbiol, 3, 643, 2005. 210. Jacobson, S., Immunopathogenesis of human T cell lymphotropic virus type I- associated neurologic disease, J Infect Dis, 186 Suppl 2, S187, 2002. 211. Jones, G. et al., Peripheral nerve-derived HIV-1 is predominantly CCR5- dependent and causes neuronal degeneration and neuroinflammation, Virology, 334, 178, 2005. 212. Zehender, G. et al., Increased risk of developing peripheral neuropathy in patients coinfected with HIV-1 and HTLV-2, J Acquir Immune Defic Syndr, 31, 440, 2002. 213. Barmak, K., Harhaj, E.W. & Wigdahl, B., Mediators of central nervous system damage during the progression of human T-cell leukemia type I-associated myelopathy/tropical spastic paraparesis, J Neurovirol, 9, 522, 2003. 214. Gonzalez-Scarano, F. & Martin-Garcia, J., The neuropathogenesis of AIDS, Nat Rev Immunol, 5, 69, 2005. 215. Zhou, N. et al., The N-terminal domain of APJ, a CNS-based coreceptor for HIV- 1, is essential for its receptor function and coreceptor activity, Virology, 317, 84, 2003. 216. Patrick, M.K., Johnston, J.B. & Power, C., Lentiviral neuropathogenesis: comparative neuroinvasion, neurotropism, neurovirulence, and host neurosusceptibility, J Virol, 76, 7923, 2002. 217. Wang, C. et al., Cytokine and chemokine gene polymorphisms among ethnically diverse North Americans with HIV-1 infection, J Acquir Immune Defic Syndr, 35, 446, 2004. 218. Knight, J.C., Regulatory polymorphisms underlying complex disease traits, J Mol Med, 83, 97, 2005. 219. Torre, D., Pugliese, A. & Speranza, F., Role of nitric oxide in HIV-1 infection: friend or foe?, Lancet Infect Dis, 2, 273, 2002.

REFERENCES 240

220. Carreras, M.C., Franco, M.C., Peralta, J.G. & Poderoso, J.J., Nitric oxide, complex I, and the modulation of mitochondrial reactive species in biology and disease, Mol Aspects Med, 25, 125, 2004. 221. Akarid, K., Sinet, M., Desforges, B. & Gougerot-Pocidalo, M.A., Inhibitory effect of nitric oxide on the replication of a murine retrovirus in vitro and in vivo, J Virol, 69, 7001, 1995. 222. Colasanti, M. & Persichini, T., Nitric oxide: an inhibitor of NF-kappaB/Rel system in glial cells, Brain Res Bull, 52, 155, 2000. 223. Jinno-Oue, A. et al., Expression of inducible nitric oxide synthase and elevation of tyrosine nitration of a 32-kilodalton cellular protein in brain capillary endothelial cells from rats infected with a neuropathogenic murine leukemia virus, J Virol, 77, 5145, 2003. 224. He, B., Viruses, endoplasmic reticulum stress, and interferon responses, Cell Death Differ, 2006. 225. Tardif, K.D., Mori, K., Kaufman, R.J. & Siddiqui, A., Hepatitis C virus suppresses the IRE1-XBP1 pathway of the unfolded protein response, J Biol Chem, 279, 17158, 2004. 226. Kim, H.T. et al., Activation of endoplasmic reticulum stress signaling pathway is associated with neuronal degeneration in MoMuLV-ts1-induced spongiform encephalomyelopathy, Lab Invest, 84, 816, 2004. 227. Liu, N. et al., Interaction between endoplasmic reticulum stress and caspase 8 activation in retrovirus MoMuLV-ts1-infected astrocytes, Virology, 348, 398, 2006. 228. Kawahara, K. et al., Induction of CHOP and apoptosis by nitric oxide in p53- deficient microglial cells, FEBS Lett, 506, 135, 2001. 229. Noorbakhsh, F. et al., Lentivirus envelope protein exerts differential neuropathogenic effects depending on the site of expression and target cell, Virology, 2006. 230. Zhang, K. et al., Endoplasmic reticulum stress activates cleavage of CREBH to induce a systemic inflammatory response, Cell, 124, 587, 2006. 231. Omori, Y. et al., CREB-H: a novel mammalian transcription factor belonging to the CREB/ATF family and functioning via the box-B element with a liver- specific expression, Nucleic Acids Res, 29, 2154, 2001. 232. Boylan, M.T. et al., Interferon-beta1a administration results in a transient increase of serum amyloid A protein and C-reactive protein: comparison with other markers of inflammation, Immunol Lett, 75, 191, 2001. 233. Crowell, R.E., Du Clos, T.W., Montoya, G., Heaphy, E. & Mold, C., C-reactive protein receptors on the human monocytic cell line U-937. Evidence for additional binding to Fc gamma RI, J Immunol, 147, 3445, 1991. 234. Kim, J.K., Scott, E.A. & Elbert, D.L., Proteomic analysis of protein adsorption: serum amyloid P adsorbs to materials and promotes leukocyte adhesion, J Biomed Mater Res A, 75, 199, 2005. 235. Dimcheff, D.E., Askovic, S., Baker, A.H., Johnson-Fowler, C. & Portis, J.L., Endoplasmic reticulum stress is a determinant of retrovirus-induced spongiform neurodegeneration, J Virol, 77, 12617, 2003.

REFERENCES 241

236. Sei, Y. et al., The encephalopathy associated with murine acquired immunodeficiency syndrome, Ann N Y Acad Sci, 840, 822, 1998. 237. Simonian, N.A. et al., Specific infection of central nervous system white matter by a variant of gross murine leukemia virus, Virology, 177, 384, 1990. 238. Gardner, M., Dandekar, S. & Cardiff, R., Molecular mechanism of an ecotropic MuLV restriction gene Akvr-1/FV-4 in California wild mice, Curr Top Microbiol Immunol, 127, 338, 1986. 239. Liu, N. et al., Possible involvement of both endoplasmic reticulum- and mitochondria-dependent pathways in MoMuLV-ts1-induced apoptosis in astrocytes, J Neurovirol, 10, 189, 2004. 240. Qiang, W. et al., Activation of transcription factor Nrf-2 and its downstream targets in response to moloney murine leukemia virus ts1-induced thiol depletion and oxidative stress in astrocytes, J Virol, 78, 11926, 2004. 241. Khan, R.B., Bertorini, T.E. & Levin, M.C., HTLV-1 and its neurological complications, Neurologist, 7, 271, 2001. 242. Manel, N. et al., The ubiquitous glucose transporter GLUT-1 is a receptor for HTLV, Cell, 115, 449, 2003. 243. Manel, N., Battini, J.L., Taylor, N. & Sitbon, M., HTLV-1 tropism and envelope receptor, Oncogene, 24, 6016, 2005. 244. Dustin, M., Viral spread through protoplasmic kiss, Nat Cell Biol, 5, 271, 2003. 245. Levin, M.C., Rosenblum, M.K., Fox, C.H. & Jacobson, S., Localization of retrovirus in the central nervous system of a patient co-infected with HTLV-1 and HIV with HAM/TSP and HIV-associated dementia, J Neurovirol, 7, 61, 2001. 246. Feuer, G. & Green, P.L., Comparative biology of human T-cell lymphotropic virus type 1 (HTLV-1) and HTLV-2, Oncogene, 24, 5996, 2005. 247. Xiao, G. & Sun, S.C., Activation of IKKalpha and IKKbeta through their fusion with HTLV-I tax protein, Oncogene, 19, 5198, 2000. 248. Benveniste, E.N. & Merrill, J.E., Stimulation of oligodendroglial proliferation and maturation by interleukin-2, Nature, 321, 610, 1986. 249. Araujo, A. & Hall, W.W., Human T-lymphotropic virus type II and neurological disease, Ann Neurol, 56, 10, 2004. 250. Umehara, F. et al., Cytokine expression in the spinal cord lesions in HTLV-I- associated myelopathy, J Neuropathol Exp Neurol, 53, 72, 1994. 251. Casseb, J. & Penalva-de-Oliveira, A.C., The pathogenesis of tropical spastic paraparesis/human T-cell leukemia type I-associated myelopathy, Braz J Med Biol Res, 33, 1395, 2000. 252. Langlois, M. et al., Activation of HTLV-I gene transcription by protein tyrosine phosphatase inhibitors, Virology, 329, 395, 2004. 253. Levin, M.C. et al., Autoimmunity due to molecular mimicry as a cause of neurological disease, Nat Med, 8, 509, 2002. 254. McArthur, J.C., Brew, B.J. & Nath, A., Neurological complications of HIV infection, Lancet Neurol, 4, 543, 2005. 255. Lewin, R., Promising animal model for MS, Science, 221, 1364, 1983. 256. Stowring, L. et al., Detection of visna virus antigens and RNA in glial cells in foci of demyelination, Virology, 141, 311, 1985.

REFERENCES 242

257. Chebloune, Y. et al., Neuroinvasion by ovine lentivirus in infected sheep mediated by inflammatory cells associated with experimental allergic encephalomyelitis, J Neurovirol, 4, 38, 1998. 258. Barber, S.A. et al., Visna virus-induced activation of MAPK is required for virus replication and correlates with virus-induced neuropathology, J Virol, 76, 817, 2002. 259. Zaitseva, M., Peden, K. & Golding, H., HIV coreceptors: role of structure, posttranslational modifications, and internalization in viral-cell fusion and as targets for entry inhibitors, Biochim Biophys Acta, 1614, 51, 2003. 260. Shimojima, M. et al., Use of CD134 as a primary receptor by the feline immunodeficiency virus, Science, 303, 1192, 2004. 261. Marx, P.A. & Chen, Z., The function of simian chemokine receptors in the replication of SIV, Semin Immunol, 10, 215, 1998. 262. van Marle, G. & Power, C., Human immunodeficiency virus type 1 genetic diversity in the nervous system: evolutionary epiphenomenon or disease determinant?, J Neurovirol, 11, 107, 2005. 263. Kaul, M., Garden, G.A. & Lipton, S.A., Pathways to neuronal injury and apoptosis in HIV-associated dementia, Nature, 410, 988, 2001. 264. Hollander, H. & Stringari, S., Human immunodeficiency virus-associated meningitis. Clinical course and correlations, Am J Med, 83, 813, 1987. 265. Jones, G. & Power, C., Regulation of neural cell survival by HIV-1 infection, Neurobiol Dis, 21, 1, 2006. 266. Anneken, K., Fischera, M., Evers, S., Kloska, S. & Husstedt, I.W., Recurrent vacuolar myelopathy in HIV infection, J Infect, 2005. 267. Xin, K.Q. et al., Evidence of HIV type 1 glycoprotein 120 binding to recombinant N-methyl-D-aspartate receptor subunits expressed in a baculovirus system, AIDS Res Hum Retroviruses, 15, 1461, 1999. 268. Piller, S.C., Ewart, G.D., Jans, D.A., Gage, P.W. & Cox, G.B., The amino- terminal region of Vpr from human immunodeficiency virus type 1 forms ion channels and kills neurons, J Virol, 73, 4230, 1999. 269. Tomonaga, K., Virus-induced neurobehavioral disorders: mechanisms and implications, Trends Mol Med, 10, 71, 2004. 270. Giulian, D., Vaca, K. & Noonan, C.A., Secretion of neurotoxins by mononuclear phagocytes infected with HIV-1, Science, 250, 1593, 1990. 271. Petito, C.K. & Roberts, B., Evidence of apoptotic cell death in HIV encephalitis, Am J Pathol, 146, 1121, 1995. 272. Garden, G.A., Microglia in human immunodeficiency virus-associated neurodegeneration, Glia, 40, 240, 2002. 273. Corasaniti, M.T. et al., Exploitation of the HIV-1 coat glycoprotein, gp120, in neurodegenerative studies in vivo, J Neurochem, 79, 1, 2001. 274. Fine, S.M. et al., Tumor necrosis factor alpha inhibits glutamate uptake by primary human astrocytes. Implications for pathogenesis of HIV-1 dementia, J Biol Chem, 271, 15303, 1996. 275. Yoshioka, M. et al., Role of immune activation and cytokine expression in HIV-1- associated neurologic diseases, Adv Neuroimmunol, 5, 335, 1995.

REFERENCES 243

276. Villet, S. et al., Maedi-visna virus and caprine arthritis encephalitis virus genomes encode a Vpr-like but no Tat protein, J Virol, 77, 9632, 2003. 277. Hovden, A.O. & Sommerfelt, M.A., The influence of CD4 and CXCR4 on maedi- visna virus-induced syncytium formation, Apmis, 110, 697, 2002. 278. Duval, R., Bellet, V., Delebassee, S. & Bosgiraud, C., Implication of caspases during maedi-visna virus-induced apoptosis, J Gen Virol, 83, 3153, 2002. 279. Podell, M. et al., AIDS-associated encephalopathy with experimental feline immunodeficiency virus infection, J Acquir Immune Defic Syndr, 6, 758, 1993. 280. Podell, M., Hayes, K., Oglesbee, M. & Mathes, L., Progressive encephalopathy associated with CD4/CD8 inversion in adult FIV-infected cats, J Acquir Immune Defic Syndr Hum Retrovirol, 15, 332, 1997. 281. Power, C. et al., Neurovirulence in feline immunodeficiency virus-infected neonatal cats is viral strain specific and dependent on systemic immune suppression, J Virol, 72, 9109, 1998. 282. van Marle, G., Antony, J.M., Silva, C., Sullivan, A. & Power, C., Aberrant cortical neurogenesis in a pediatric neuroAIDS model: neurotrophic effects of growth hormone, Aids, 19, 1781, 2005. 283. Dow, S.W., Poss, M.L. & Hoover, E.A., Feline immunodeficiency virus: a neurotropic lentivirus, J Acquir Immune Defic Syndr, 3, 658, 1990. 284. Dean, G.A., Reubel, G.H., Moore, P.F. & Pedersen, N.C., Proviral burden and infection kinetics of feline immunodeficiency virus in lymphocyte subsets of blood and lymph node, J Virol, 70, 5165, 1996. 285. Kennedy, J.M. et al., Peripheral neuropathy in lentivirus infection: evidence of inflammation and axonal injury, Aids, 18, 1241, 2004. 286. Little, P.F., Structure and function of the human genome, Genome Res, 15, 1759, 2005. 287. Costas, J., Characterization of the intragenomic spread of the human endogenous retrovirus family HERV-W, Mol Biol Evol, 19, 526, 2002. 288. Urnovitz, H.B. & Murphy, W.H., Human endogenous retroviruses: nature, occurrence, and clinical implications in human disease, Clin Microbiol Rev, 9, 72, 1996. 289. Ono, M., Kawakami, M. & Takezawa, T., A novel human nonviral retroposon derived from an endogenous retrovirus, Nucleic Acids Res, 15, 8725, 1987. 290. Harada, F., Tsukada, N. & Kato, N., Isolation of three kinds of human endogenous retrovirus-like sequences using tRNA(Pro) as a probe, Nucleic Acids Res, 15, 9153, 1987. 291. La Mantia, G. et al., Identification of regulatory elements within the minimal promoter region of the human endogenous ERV9 proviruses: accurate transcription initiation is controlled by an Inr-like element, Nucleic Acids Res, 20, 4129, 1992. 292. Pavlicek, A., Paces, J., Elleder, D. & Hejnar, J., Processed pseudogenes of human endogenous retroviruses generated by LINEs: their integration, stability, and distribution, Genome Res, 12, 391, 2002. 293. Lower, R., Lower, J. & Kurth, R., The viruses in all of us: characteristics and biological significance of human endogenous retrovirus sequences, Proc Natl Acad Sci U S A, 93, 5177, 1996.

REFERENCES 244

294. Jurka, J., Repeats in genomic DNA: mining and meaning, Curr Opin Struct Biol, 8, 333, 1998. 295. Leib-Mosch, C. & Seifarth, W., Evolution and biological significance of human retroelements, Virus Genes, 11, 133, 1995. 296. Patience, C., Wilkinson, D.A. & Weiss, R.A., Our retroviral heritage, Trends Genet, 13, 116, 1997. 297. Di Cristofano, A., Strazzullo, M., Parisi, T. & La Mantia, G., Mobilization of an ERV9 human endogenous retroviral element during primate evolution, Virology, 213, 271, 1995. 298. Di Cristofano, A., Strazullo, M., Longo, L. & La Mantia, G., Characterization and genomic mapping of the ZNF80 locus: expression of this zinc-finger gene is driven by a solitary LTR of ERV9 endogenous retroviral family, Nucleic Acids Res, 23, 2823, 1995. 299. Belshaw, R. et al., Long-term reinfection of the human genome by endogenous retroviruses, Proc Natl Acad Sci U S A, 101, 4894, 2004. 300. de Parseval, N. & Heidmann, T., Human endogenous retroviruses: from infectious elements to human genes, Cytogenet Genome Res, 110, 318, 2005. 301. Mourier, T., Reverse transcription in genome evolution, Cytogenet Genome Res, 110, 56, 2005. 302. Katzourakis, A., Rambaut, A. & Pybus, O.G., The evolutionary dynamics of endogenous retroviruses, Trends Microbiol, 13, 463, 2005. 303. Blaise, S., de Parseval, N., Benit, L. & Heidmann, T., Genomewide screening for fusogenic human endogenous retrovirus envelopes identifies syncytin 2, a gene conserved on primate evolution, Proc Natl Acad Sci U S A, 100, 13013, 2003. 304. Dewannieux, M., Blaise, S. & Heidmann, T., Identification of a functional envelope protein from the HERV-K family of human endogenous retroviruses, J Virol, 79, 15573, 2005. 305. Lindeskog, M., Mager, D.L. & Blomberg, J., Isolation of a human endogenous retroviral HERV-H element with an open env reading frame, Virology, 258, 441, 1999. 306. Schon, U. et al., Cell type-specific expression and promoter activity of human endogenous retroviral long terminal repeats, Virology, 279, 280, 2001. 307. Hughes, J.F. & Coffin, J.M., Evidence for genomic rearrangements mediated by human endogenous retroviruses during primate evolution, Nat Genet, 29, 487, 2001. 308. Ponferrada, V.G., Mauck, B.S. & Wooley, D.P., The envelope glycoprotein of human endogenous retrovirus HERV-W induces cellular resistance to spleen necrosis virus, Arch Virol, 148, 659, 2003. 309. Robinson, H.L., Astrin, S.M., Senior, A.M. & Salazar, F.H., Host Susceptibility to endogenous viruses: defective, glycoprotein- expressing proviruses interfere with infections, J Virol, 40, 745, 1981. 310. McDougall, A.S. et al., Defective endogenous proviruses are expressed in feline lymphoid cells: evidence for a role in natural resistance to subgroup B feline leukemia viruses, J Virol, 68, 2151, 1994. 311. Harris, J.R., Placental endogenous retrovirus (ERV): structural, functional, and evolutionary significance, Bioessays, 20, 307, 1998.

REFERENCES 245

312. Larsson, E., Andersson, A.C. & Nilsson, B.O., Expression of an endogenous retrovirus (ERV3 HERV-R) in human reproductive and embryonic tissues-- evidence for a function for envelope gene products, Ups J Med Sci, 99, 113, 1994. 313. Mi, S. et al., Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis, Nature, 403, 785, 2000. 314. Maksakova, I.A. et al., Retroviral Elements and Their Hosts: Insertional Mutagenesis in the Mouse Germ Line, PLoS Genet, 2, e2, 2006. 315. Best, S., Le Tissier, P., Towers, G. & Stoye, J.P., Positional cloning of the mouse retrovirus restriction gene Fv1, Nature, 382, 826, 1996. 316. Fukumitsu, H., Takase-Yoden, S. & Watanabe, R., Neuropathology of experimental autoimmune encephalomyelitis modified by retroviral infection, Neuropathology, 22, 280, 2002. 317. Dupressoir, A. et al., Syncytin-A and syncytin-B, two fusogenic placenta-specific murine envelope genes of retroviral origin conserved in Muridae, Proc Natl Acad Sci U S A, 102, 725, 2005. 318. van de Lagemaat, L.N., Landry, J.R., Mager, D.L. & Medstrand, P., Transposable elements in mammals promote regulatory variation and diversification of genes with specialized functions, Trends Genet, 19, 530, 2003. 319. Sverdlov, E.D., Retroviruses and primate evolution, Bioessays, 22, 161, 2000. 320. O'Reilly, R.L. & Singh, S.M., Retroviruses and schizophrenia revisited, Am J Med Genet, 67, 19, 1996. 321. Conrad, B. et al., A human endogenous retroviral superantigen as candidate autoimmune gene in type I diabetes, Cell, 90, 303, 1997. 322. Herbst, H., Sauter, M. & Mueller-Lantzsch, N., Expression of human endogenous retrovirus K elements in germ cell and trophoblastic tumors, Am J Pathol, 149, 1727, 1996. 323. Coffin, J.M., Superantigens and endogenous retroviruses: a confluence of puzzles, Science, 255, 411, 1992. 324. Boller, K. et al., Evidence that HERV-K is the endogenous retrovirus sequence that codes for the human teratocarcinoma-derived retrovirus HTDV, Virology, 196, 349, 1993. 325. Wang-Johanning, F. et al., Expression of human endogenous retrovirus k envelope transcripts in human breast cancer, Clin Cancer Res, 7, 1553, 2001. 326. Wang-Johanning, F. et al., Detecting the expression of human endogenous retrovirus E envelope transcripts in human prostate adenocarcinoma, Cancer, 98, 187, 2003. 327. Lindeskog, M. & Blomberg, J., Spliced human endogenous retroviral HERV-H env transcripts in T-cell leukaemia cell lines and normal leukocytes: alternative splicing pattern of HERV-H transcripts, J Gen Virol, 78, 2575, 1997. 328. Karlsson, H. et al., Retroviral RNA identified in the cerebrospinal fluids and brains of individuals with schizophrenia, Proc Natl Acad Sci U S A, 98, 4634, 2001. 329. Clausen, J., Endogenous retroviruses and MS: using ERVs as disease markers, Int MS J, 10, 22, 2003.

REFERENCES 246

330. Voisset, C. et al., Chromosomal distribution and coding capacity of the human endogenous retrovirus HERV-W family, AIDS Res Hum Retroviruses, 16, 731, 2000. 331. Cheynet, V. et al., Synthesis, assembly, and processing of the Env ERVWE1/syncytin human endogenous retroviral envelope, J Virol, 79, 5585, 2005. 332. An, D.S., Xie, Y. & Chen, I.S., Envelope gene of the human endogenous retrovirus HERV-W encodes a functional retrovirus envelope, J Virol, 75, 3488, 2001. 333. Lavillette, D. et al., The envelope glycoprotein of human endogenous retrovirus type W uses a divergent family of amino acid transporters/cell surface receptors, J Virol, 76, 6442, 2002. 334. Blond, J.L. et al., An envelope glycoprotein of the human endogenous retrovirus HERV-W is expressed in the human placenta and fuses cells expressing the type D mammalian retrovirus receptor, J Virol, 74, 3321, 2000. 335. Marin, M., Lavillette, D., Kelly, S.M. & Kabat, D., N-linked glycosylation and sequence changes in a critical negative control region of the ASCT1 and ASCT2 neutral amino acid transporters determine their retroviral receptor functions, J Virol, 77, 2936, 2003. 336. Weiss, M.D., Rossignol, C., Sumners, C. & Anderson, K.J., A pH-dependent increase in neuronal glutamate efflux in vitro: possible involvement of ASCT1, Brain Res, 1056, 105, 2005. 337. Dolinska, M., Zablocka, B., Sonnewald, U. & Albrecht, J., Glutamine uptake and expression of mRNA's of glutamine transporting proteins in mouse cerebellar and cerebral cortical astrocytes and neurons, Neurochem Int, 44, 75, 2004. 338. Furuya, S. & Watanabe, M., Novel neuroglial and glioglial relationships mediated by L-serine metabolism, Arch Histol Cytol, 66, 109, 2003. 339. Weiss, M.D., Derazi, S., Kilberg, M.S. & Anderson, K.J., Ontogeny and localization of the neutral amino acid transporter ASCT1 in rat brain, Brain Res Dev Brain Res, 130, 183, 2001. 340. Zhang, K. et al., Human immunodeficiency virus type 1 envelope-mediated neuronal death: Uncoupling of viral replication and neurotoxicity, J Virol, 77, 6899, 2003. 341. Kawaguchi, Y., Miyazawa, T., Tohya, Y., Takahashi, E. & Mikami, T., Quantification of feline immunodeficiency virus in a newly established feline T- lymphoblastoid cell line (MYA-1 cells), Arch Virol, 111, 269, 1990. 342. van Marle, G., Ethier, J., Silva, C., Mac Vicar, B.A. & Power, C., Human immunodeficiency virus type 1 envelope-mediated neuropathogenesis: targeted gene delivery by a Sindbis virus expression vector, Virology, 309, 61, 2003. 343. Bredenbeek, P.J., Frolov, I., Rice, C.M. & Schlesinger, S., Sindbis virus expression vectors: packaging of RNA replicons by using defective helper RNAs, J Virol, 67, 6439, 1993. 344. Schlesinger, S. & Dubensky, T.W., Alphavirus vectors for gene expression and vaccines, Curr Opin Biotechnol, 10, 434, 1999.

REFERENCES 247

345. Vergote, D. et al., Characterisation of proteins differentially present in the plasma of Biomphalaria glabrata susceptible or resistant to Echinostoma caproni, Int J Parasitol, 35, 215, 2005. 346. Wallace, J.L. et al., Gastric tolerability and prolonged prostaglandin inhibition in the brain with a nitric oxide-releasing flurbiprofen derivative, NCX-2216 [3-[4- (2-fluoro-alpha-methyl-[1,1'-biphenyl]-4-acetyloxy)-3-methoxyphenyl] -2- propenoic acid 4-nitrooxy butyl ester], J Pharmacol Exp Ther, 309, 626, 2004. 347. Guenther, K., Deacon, R.M., Perry, V.H. & Rawlins, J.N., Early behavioural changes in scrapie-affected mice and the influence of dapsone, Eur J Neurosci, 14, 401, 2001. 348. Livak, K.J. & Schmittgen, T.D., Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods, 25, 402, 2001. 349. Butterfield, D.A. & Lauderback, C.M., Lipid peroxidation and protein oxidation in Alzheimer's disease brain: potential causes and consequences involving amyloid beta-peptide-associated free radical oxidative stress, Free Radic Biol Med, 32, 1050, 2002. 350. Bar-Or, A. et al., Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis, Brain, 126, 2738, 2003. 351. Imai, Y., Ibata, I., Ito, D., Ohsawa, K. & Kohsaka, S., A novel gene iba1 in the major histocompatibility complex class III region encoding an EF hand protein expressed in a monocytic lineage, Biochem Biophys Res Commun, 224, 855, 1996. 352. Bansal, R., Warrington, A.E., Gard, A.L., Ranscht, B. & Pfeiffer, S.E., Multiple and novel specificities of monoclonal antibodies O1, O4, and R- mAb used in the analysis of oligodendrocyte development, J Neurosci Res, 24, 548, 1989. 353. Power, C. et al., Cerebral white matter changes in acquired immunodeficiency syndrome dementia: alterations of the blood-brain barrier, Ann Neurol, 34, 339, 1993. 354. West, M.J., Ostergaard, K., Andreassen, O.A. & Finsen, B., Estimation of the number of somatostatin neurons in the striatum: an in situ hybridization study using the optical fractionator method, J Comp Neurol, 370, 11, 1996. 355. Lander, E.S. et al., Initial sequencing and analysis of the human genome, Nature, 409, 860, 2001. 356. Bock, M. & Stoye, J.P., Endogenous retroviruses and the human germline, Curr Opin Genet Dev, 10, 651, 2000. 357. Calabrese, V. et al., Nitric oxide synthase is present in the cerebrospinal fluid of patients with active multiple sclerosis and is associated with increases in cerebrospinal fluid protein nitrotyrosine and S-nitrosothiols and with changes in glutathione levels, J Neurosci Res, 70, 580, 2002. 358. Lu, F. et al., Oxidative damage to mitochondrial DNA and activity of mitochondrial enzymes in chronic active lesions of multiple sclerosis, J Neurol Sci, 177, 95, 2000. 359. John, G.R. et al., Multiple sclerosis: re-expression of a developmental pathway that restricts oligodendrocyte maturation, Nat Med, 8, 1115, 2002.

REFERENCES 248

360. Kanski, J., Aksenova, M., Stoyanova, A. & Butterfield, D.A., Ferulic acid antioxidant protection against hydroxyl and peroxyl radical oxidation in synaptosomal and neuronal cell culture systems in vitro: structure-activity studies, J Nutr Biochem, 13, 273, 2002. 361. Hua, L.L., Kim, M.O., Brosnan, C.F. & Lee, S.C., Modulation of astrocyte inducible nitric oxide synthase and cytokine expression by interferon beta is associated with induction and inhibition of interferon gamma-activated sequence binding activity, J Neurochem, 83, 1120, 2002. 362. Frendo, J.L. et al., Direct involvement of HERV-W Env glycoprotein in human trophoblast cell fusion and differentiation, Mol Cell Biol, 23, 3566, 2003. 363. Calabrese, V. et al., Disruption of thiol homeostasis and nitrosative stress in the cerebrospinal fluid of patients with active multiple sclerosis: evidence for a protective role of acetylcarnitine, Neurochem Res, 28, 1321, 2003. 364. Smith, K.J. & Lassmann, H., The role of nitric oxide in multiple sclerosis, Lancet Neurol, 1, 232, 2002. 365. Danilov, A.I. et al., Nitric oxide metabolite determinations reveal continuous inflammation in multiple sclerosis, J Neuroimmunol, 136, 112, 2003. 366. Lieberman, A.P., Pitha, P.M., Shin, H.S. & Shin, M.L., Production of tumor necrosis factor and other cytokines by astrocytes stimulated with lipopolysaccharide or a neurotropic virus, Proc Natl Acad Sci U S A, 86, 6348, 1989. 367. Rosen, C.L., Bunge, R.P., Ard, M.D. & Wood, P.M., Type 1 astrocytes inhibit myelination by adult rat oligodendrocytes in vitro, J Neurosci, 9, 3371, 1989. 368. Zafra, F., Castren, E., Thoenen, H. & Lindholm, D., Interplay between glutamate and gamma-aminobutyric acid transmitter systems in the physiological regulation of brain-derived neurotrophic factor and nerve growth factor synthesis in hippocampal neurons, Proc Natl Acad Sci U S A, 88, 10037, 1991. 369. Lynch, W.P. & Sharpe, A.H., Differential glycosylation of the Cas-Br-E env protein is associated with retrovirus-induced spongiform neurodegeneration, J Virol, 74, 1558, 2000. 370. Qiang, W. et al., Astrocytes survive chronic infection and cytopathic effects of the ts1 mutant of the retrovirus Moloney murine leukemia virus by upregulation of antioxidant defenses, J Virol, 80, 3273, 2006. 371. Scott, G.S., Virag, L., Szabo, C. & Hooper, D.C., Peroxynitrite-induced oligodendrocyte toxicity is not dependent on poly(ADP-ribose) polymerase activation, Glia, 41, 105, 2003. 372. Ravikumar, A., Arun, P., Devi, K.V., Augustine, J. & Kurup, P.A., Isoprenoid pathway and free radical generation and damage in neuropsychiatric disorders, Indian J Exp Biol, 38, 438, 2000. 373. Gu, Z. et al., S-nitrosylation of matrix metalloproteinases: signaling pathway to neuronal cell death, Science, 297, 1186, 2002. 374. Donald W Paty, J.H.N.a.G.C.E. Diagnosis of Multiple Sclerosis. in Multiple Sclerosis (ed. Ebers, D.W.P.a.G.C.) 68 (FA Davis Company, Philadelphia, 1998). 375. Anlar, O., Tombul, T. & Kisli, M., Peripheral sensory and motor abnormalities in patients with multiple sclerosis, Electromyogr Clin Neurophysiol, 43, 349, 2003.

REFERENCES 249

376. Ogiwara, T. et al., Inhibition of NO production by activated macrophages by phenolcarboxylic acid monomers and polymers with radical scavenging activity, Anticancer Res, 23, 1317, 2003. 377. Armbruester, V. et al., A novel gene from the human endogenous retrovirus K expressed in transformed cells, Clin Cancer Res, 8, 1800, 2002. 378. Larsson, E. et al., Tissue and differentiation specific expression on the endogenous retrovirus ERV3 (HERV-R) in normal human tissues and during induced monocytic differentiation in the U-937 cell line, Leukemia, 11 Suppl 3, 142, 1997. 379. Katsumata, K. et al., Cytokine regulation of env gene expression of human endogenous retrovirus-R in human vascular endothelial cells, Clin Immunol, 93, 75, 1999. 380. Garson, J. et al., MSRV, Syncytin and the role of endogenous retroviral proteins in demyelination, Mult Scler, 11, 249, 2005. 381. Dolei, A. et al., Multiple sclerosis-associated retrovirus (MSRV) in Sardinian MS patients, Neurology, 58, 471, 2002. 382. Sotgiu, S. et al., Multiple sclerosis-associated retrovirus and MS prognosis: an observational study, Neurology, 59, 1071, 2002. 383. Nowak, J. et al., Multiple sclerosis-associated virus-related pol sequences found both in multiple sclerosis and healthy donors are more frequently expressed in multiple sclerosis patients, J Neurovirol, 9, 112, 2003. 384. Bonnaud, B. et al., Evidence of selection on the domesticated ERVWE1 env retroviral element involved in placentation, Mol Biol Evol, 21, 1895, 2004. 385. Dolei, A., MSRV/HERV-W/syncytin and its linkage to multiple sclerosis: the usability and the hazard of a human endogenous retrovirus, J Neurovirol, 11, 232, 2005. 386. Li, Y. et al., Molecular characterization of human immunodeficiency virus type 1 cloned directly from uncultured human brain tissue: identification of replication- competent and -defective viral genomes, J Virol, 65, 3973, 1991. 387. Lazarini, F. et al., Human immunodeficiency virus type 1 DNA and RNA load in brains of demented and nondemented patients with acquired immunodeficiency syndrome, J Neurovirol, 3, 299, 1997. 388. Bell, P., Montaner, L.J. & Maul, G.G., Accumulation and intranuclear distribution of unintegrated human immunodeficiency virus type 1 DNA, J Virol, 75, 7683, 2001. 389. Hartung, H.P. & Rieckmann, P., Pathogenesis of immune-mediated demyelination in the CNS, J Neural Transm Suppl, 50, 173, 1997. 390. Kort, J.J., Kawamura, K., Fugger, L., Weissert, R. & Forsthuber, T.G., Efficient presentation of myelin oligodendrocyte glycoprotein peptides but not protein by astrocytes from HLA-DR2 and HLA-DR4 transgenic mice, J Neuroimmunol, 173, 23, 2006. 391. Mycko, M.P., Papoian, R., Boschert, U., Raine, C.S. & Selmaj, K.W., Microarray gene expression profiling of chronic active and inactive lesions in multiple sclerosis, Clin Neurol Neurosurg, 106, 223, 2004. 392. Wu, J. & Kaufman, R.J., From acute ER stress to physiological roles of the Unfolded Protein Response, Cell Death Differ, 13, 374, 2006.

REFERENCES 250

393. Bauer, J. et al., Endoplasmic reticulum stress in PLP-overexpressing transgenic rats: gray matter oligodendrocytes are more vulnerable than white matter oligodendrocytes, J Neuropathol Exp Neurol, 61, 12, 2002. 394. De Keyser, J., Zeinstra, E. & Wilczak, N., Astrocytic beta2-adrenergic receptors and multiple sclerosis, Neurobiol Dis, 15, 331, 2004. 395. Daniels, M. & Brown, D.R., Astrocytes regulate N-methyl-D-aspartate receptor subunit composition increasing neuronal sensitivity to excitotoxicity, J Biol Chem, 276, 22446, 2001. 396. Rao, R.V. & Bredesen, D.E., Misfolded proteins, endoplasmic reticulum stress and neurodegeneration, Curr Opin Cell Biol, 16, 653, 2004. 397. Yu, Z., Luo, H., Fu, W. & Mattson, M.P., The endoplasmic reticulum stress- responsive protein GRP78 protects neurons against excitotoxicity and apoptosis: suppression of oxidative stress and stabilization of calcium homeostasis, Exp Neurol, 155, 302, 1999. 398. Kondo, S. et al., OASIS, a CREB/ATF-family member, modulates UPR signalling in astrocytes, Nat Cell Biol, 7, 186, 2005. 399. Thompson, K.A., McArthur, J.C. & Wesselingh, S.L., Correlation between neurological progression and astrocyte apoptosis in HIV-associated dementia, Ann Neurol, 49, 745, 2001. 400. Jiang, Y. et al., Retrovirus-Induced Oxidative Stress with Neuroimmunodegeneration Is Suppressed by Antioxidant Treatment with a Refined Monosodium {alpha}-Luminol (Galavit), J Virol, 80, 4557, 2006. 401. Cullinan, S.B. et al., Nrf2 is a direct PERK substrate and effector of PERK- dependent cell survival, Mol Cell Biol, 23, 7198, 2003. 402. Sato, H., Tamba, M., Kuriyama-Matsumura, K., Okuno, S. & Bannai, S., Molecular cloning and expression of human xCT, the light chain of amino acid transport system xc, Antioxid Redox Signal, 2, 665, 2000. 403. Kaleeba, J.A. & Berger, E.A., Kaposi's sarcoma-associated herpesvirus fusion- entry receptor: cystine transporter xCT, Science, 311, 1921, 2006. 404. Sasaki, H. et al., Electrophile response element-mediated induction of the cystine/glutamate exchange transporter gene expression, J Biol Chem, 277, 44765, 2002. 405. Fernandis, A.Z., Cherla, R.P., Chernock, R.D. & Ganju, R.K., CXCR4/CCR5 down-modulation and chemotaxis are regulated by the proteasome pathway, J Biol Chem, 277, 18111, 2002. 406. Perron, H. et al., Human endogenous retrovirus (HERV)-W ENV and GAG proteins: physiological expression in human brain and pathophysiological modulation in multiple sclerosis lesions, J Neurovirol, 11, 23, 2005. 407. Kanai, Y. & Endou, H., Heterodimeric amino acid transporters: molecular biology and pathological and pharmacological relevance, Curr Drug Metab, 2, 339, 2001. 408. Lock, C. et al., Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis, Nat Med, 8, 500, 2002. 409. Murakami, T. et al., Cleavage of the membrane-bound transcription factor OASIS in response to endoplasmic reticulum stress, J Neurochem, 96, 1090, 2006. 410. Bhat, N.R., Feinstein, D.L., Shen, Q. & Bhat, A.N., p38 MAPK-mediated transcriptional activation of inducible nitric-oxide synthase in glial cells. Roles of

REFERENCES 251

nuclear factors, nuclear factor kappa B, cAMP response element-binding protein, CCAAT/enhancer-binding protein-beta, and activating transcription factor-2, J Biol Chem, 277, 29584, 2002. 411. Kuhlmann, T. et al., Bcl-2-expressing oligodendrocytes in multiple sclerosis lesions, Glia, 28, 34, 1999. 412. Grewer, C. & Grabsch, E., New inhibitors for the neutral amino acid transporter ASCT2 reveal its Na+-dependent anion leak, J Physiol, 557, 747, 2004. 413. Di Battista, J.A., Martel-Pelletier, J. & Pelletier, J., Suppression of tumor necrosis factor (TNF-alpha) gene expression by prostaglandin E(2). Role Of early growth response protein-1 (Egr-1), Osteoarthritis Cartilage, 7, 395, 1999. 414. Kanai, Y. & Hediger, M.A., The glutamate/neutral amino acid transporter family SLC1: molecular, physiological and pharmacological aspects, Pflugers Arch, 447, 469, 2004. 415. Rupprecht, H.D., Akagi, Y., Keil, A. & Hofer, G., Nitric oxide inhibits growth of glomerular mesangial cells: role of the transcription factor EGR-1, Kidney Int, 57, 70, 2000. 416. Cicala, C. et al., R5 and X4 HIV envelopes induce distinct gene expression profiles in primary peripheral blood mononuclear cells, Proc Natl Acad Sci U S A, 103, 3746, 2006. 417. Asensio, V.C. et al., Interferon-independent, human immunodeficiency virus type 1 gp120- mediated induction of CXCL10/IP-10 gene expression by astrocytes in vivo and in vitro, J Virol, 75, 7067, 2001. 418. Wang, Z. et al., Reduced expression of glutamate transporter EAAT2 and impaired glutamate transport in human primary astrocytes exposed to HIV-1 or gp120, Virology, 312, 60, 2003. 419. McCarthy, M. et al., Cellular tropisms and co-receptor usage of HIV-1 isolates from vertically infected children with neurological abnormalities and rapid disease progression, J Med Virol, 67, 1, 2002. 420. Stalder, A.K. et al., Late-onset chronic inflammatory encephalopathy in immune- competent and severe combined immune-deficient (SCID) mice with astrocyte- targeted expression of tumor necrosis factor, Am J Pathol, 153, 767, 1998. 421. Coombs, B.D. et al., Multiple sclerosis pathology in the normal and abnormal appearing white matter of the corpus callosum by diffusion tensor imaging, Mult Scler, 10, 392, 2004. 422. Saccani, A. et al., Redox regulation of chemokine receptor expression, Proc Natl Acad Sci U S A, 97, 2761, 2000. 423. Kekuda, R. et al., Cloning of the sodium-dependent, broad-scope, neutral amino acid transporter Bo from a human placental choriocarcinoma cell line, J Biol Chem, 271, 18657, 1996. 424. Sakai, K., Shimizu, H., Koike, T., Furuya, S. & Watanabe, M., Neutral amino acid transporter ASCT1 is preferentially expressed in L-Ser-synthetic/storing glial cells in the mouse brain with transient expression in developing capillaries, J Neurosci, 23, 550, 2003. 425. Hackel, D. PhD thesis, Magdeburg (2005). 426. Yamamoto, T. et al., Characterization of rapid and high-affinity uptake of L- serine in neurons and astrocytes in primary culture, FEBS Lett, 548, 69, 2003.

REFERENCES 252

427. Sundaram, K.S. & Lev, M., Inhibition of sphingolipid synthesis by cycloserine in vitro and in vivo, J Neurochem, 42, 577, 1984. 428. Kudo, Y. & Boyd, C.A., Changes in expression and function of syncytin and its receptor, amino acid transport system B(0) (ASCT2), in human placental choriocarcinoma BeWo cells during syncytialization, Placenta, 23, 536, 2002. 429. Storck, T., Schulte, S., Hofmann, K. & Stoffel, W., Structure, expression, and functional analysis of a Na(+)-dependent glutamate/aspartate transporter from rat brain, Proc Natl Acad Sci U S A, 89, 10955, 1992. 430. Uchiyama, T., Matsuda, Y., Wada, M., Takahashi, S. & Fujita, T., Functional regulation of Na+-dependent neutral amino acid transporter ASCT2 by S- nitrosothiols and nitric oxide in Caco-2 cells, FEBS Lett, 579, 2499, 2005. 431. Dickhout, J.G. et al., Peroxynitrite causes endoplasmic reticulum stress and apoptosis in human vascular endothelium: implications in atherogenesis, Arterioscler Thromb Vasc Biol, 25, 2623, 2005. 432. Tatarowicz, W.A. et al., Repression of the HSV-1 latency-associated transcript (LAT) promoter by the early growth response (EGR) proteins: involvement of a binding site immediately downstream of the TATA box, J Neurovirol, 3, 212, 1997. 433. Korner, H. et al., Critical points of tumor necrosis factor action in central nervous system autoimmune inflammation defined by gene targeting, J Exp Med, 186, 1585, 1997. 434. FitzGerald, U.F., Gilbey, T., Brodie, S. & Barnett, S.C., Transcription factor expression and cellular redox in immature oligodendrocyte cell death: effect of Bcl-2, Mol Cell Neurosci, 22, 516, 2003. 435. Nishie, M. et al., Multinucleated astrocytes in old demyelinated plaques in a patient with multiple sclerosis, Neuropathology, 24, 248, 2004. 436. Jana, M. & Pahan, K., Redox regulation of cytokine-mediated inhibition of myelin gene expression in human primary oligodendrocytes, Free Radic Biol Med, 39, 823, 2005. 437. Yeh, M.W. et al., Cytokine-stimulated, but not HIV-infected, human monocyte- derived macrophages produce neurotoxic levels of l -cysteine, J Immunol, 164, 4265, 2000. 438. Fujinami, R.S., von Herrath, M.G., Christen, U. & Whitton, J.L., Molecular mimicry, bystander activation, or viral persistence: infections and autoimmune disease, Clin Microbiol Rev, 19, 80, 2006. 439. Brudek, T., Christensen, T., Hansen, H.J., Bobecka, J. & Moller-Larsen, A., Simultaneous presence of endogenous retrovirus and herpes virus antigens has profound effect on cell-mediated immune responses: implications for multiple sclerosis, AIDS Res Hum Retroviruses, 20, 415, 2004. 440. Kudo, Y., Boyd, C.A., Sargent, I.L. & Redman, C.W., Hypoxia alters expression and function of syncytin and its receptor during trophoblast cell fusion of human placental BeWo cells: implications for impaired trophoblast syncytialisation in pre-eclampsia, Biochim Biophys Acta, 1638, 63, 2003. 441. Cheng, Y.H., Richardson, B.D., Hubert, M.A. & Handwerger, S., Isolation and characterization of the human syncytin gene promoter, Biol Reprod, 70, 694, 2004.

REFERENCES 253

442. Cockerill, P.N., Osborne, C.S., Bert, A.G. & Grotto, R.J., Regulation of GM-CSF gene transcription by core-binding factor, Cell Growth Differ, 7, 917, 1996. 443. McQualter, J.L. et al., Granulocyte macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis, J Exp Med, 194, 873, 2001. 444. Wang, D., Yamamoto, S., Hijiya, N., Benveniste, E.N. & Gladson, C.L., Transcriptional regulation of the human osteopontin promoter: functional analysis and DNA-protein interactions, Oncogene, 19, 5801, 2000. 445. Chabas, D. et al., The influence of the proinflammatory cytokine, osteopontin, on autoimmune demyelinating disease, Science, 294, 1731, 2001. 446. Galey, D. et al., Differential transcriptional regulation by human immunodeficiency virus type 1 and gp120 in human astrocytes, J Neurovirol, 9, 358, 2003. 447. Koppal, T. et al., Peroxynitrite-induced alterations in synaptosomal membrane proteins: insight into oxidative stress in Alzheimer's disease, J Neurochem, 72, 310, 1999. 448. Gaudin, P. et al., Infrequency of detection of particle-associated MSRV/HERV-W RNA in the synovial fluid of patients with rheumatoid arthritis, Rheumatology (Oxford), 39, 950, 2000. 449. Mallet, F. et al., The endogenous retroviral locus ERVWE1 is a bona fide gene involved in hominoid placental physiology, Proc Natl Acad Sci U S A, 101, 1731, 2004. 450. Zawada, M. et al., MSRV pol sequence copy number as a potential marker of multiple sclerosis, Pol J Pharmacol, 55, 869, 2003. 451. Hirt, B., Selective extraction of polyoma DNA from infected mouse cell cultures, J Mol Biol, 26, 365, 1967. 452. Lucke-Huhle, C., Hinrichs, S. & Speit, G., DHFR gene amplification in cultured skin fibroblasts of ataxia telangiectasia patients after methotrexate selection, Carcinogenesis, 8, 1801, 1987. 453. Xu, C., Bailly-Maitre, B. & Reed, J.C., Endoplasmic reticulum stress: cell life and death decisions, J Clin Invest, 115, 2656, 2005. 454. Pierig, R., Belliveau, J., Amouri, R., Menard, A. & Rieger, F., Association of a gliotoxic activity with active multiple sclerosis in US patients, Cell Mol Biol (Noisy-le-grand), 48, 199, 2002. 455. Barca, O. et al., Interferon beta promotes survival in primary astrocytes through phosphatidylinositol 3-kinase, J Neuroimmunol, 139, 155, 2003. 456. Benjelloun, N. et al., Case report: DNA fragmentation in glial cells in a cerebral biopsy from a multiple sclerosis patient, Cell Mol Biol (Noisy-le-grand), 44, 579, 1998. 457. Gveric, D., Cuzner, M.L. & Newcombe, J., Insulin-like growth factors and binding proteins in multiple sclerosis plaques, Neuropathol Appl Neurobiol, 25, 215, 1999. 458. Lucchinetti, C. et al., Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination, Ann Neurol, 47, 707, 2000. 459. Franklin, R.J., Crang, A.J. & Blakemore, W.F., Transplanted type-1 astrocytes facilitate repair of demyelinating lesions by host oligodendrocytes in adult rat spinal cord, J Neurocytol, 20, 420, 1991.