Molecular Characterization of Familial Ataxic, Paraplegic and Related Movement Disorders

Thesis Submitted to the University of the Punjab, Lahore for the Award of Degree of Doctor of Philosophy in Biological Sciences by Huma Tariq

Research Supervisor Dr. Sadaf Naz Professor, School of Biological Sciences University of the Punjab, Lahore

School of Biological Sciences University of the Punjab, Lahore, Pakistan and Institute of Neurology University College London, London, UK 2019

IN THE NAME

OF

ALLAH ALMIGHTY

The Most Merciful, Beneficent &

The Most Gracious!

DEDICATED TO The Soil of my motherland, PAKISTAN

DECLARATION CERTIFICATE

The thesis entitled “Molecular Characterization of Familial Ataxic, Paraplegic and Related Movement Disorders”, being submitted to the University of the Punjab, Lahore for the degree of Doctor of Philosophy in Biological Sciences, does not contain any material which has been submitted for the award of Ph.D. in any University and does not contain any material published except when due reference is made to the source in the text of the thesis.

Huma Tariq Summary

Movement disorders are heterogeneous neurological syndromes affecting voluntary movement, coordination, causing ataxia or some involuntary movements. Many of these disorders have a genetic cause and can have any mode of inheritance. The rate of consanguineous marriages is very high in Pakistan and therefore there is a high probability of recessively inherited rare genetic disorders, including movement disorders. The genetics of movement disorders is not very well studied in Pakistan. Next generation sequencing technology can facilitate diagnosis and identification of novel from affected individuals in families practicing consanguineous marriages.

In this study, sixteen families with multiple affected individuals were recruited from Punjab province of Pakistan. The patients exhibited either all, or combination of symptoms including movement disability, abnormality in voluntary movements, abnormal postures, frequent falls, unusual gait with or without abnormal speech. A standard protocol was adopted for videotaping of participants. Diagnosis was made by neurologists either in Pakistan or UK. The required medical investigations and neuroimaging were performed for the probands in the families.

Whole exome sequencing was performed for one or two members of all the sixteen families. Exome data was analyzed in a sequential way using appropriate filters. Variants having high frequency in public databases (>0.01) were excluded and others were prioritized based on their effect on the encoded . Only homozygous and compound heterozygous variants were considered. Segregation of candidate variants was analyzed by Sanger Sequencing in all family participants. Functional assays and computational analysis were performed for selected missense variants to check their effect on function.

The genetic causes of the disorder in eight families out of sixteen were identified. Most of these were in those genes which play vital roles in genome integrity, DNA repair and membrane trafficking. Novel homozygous and compound heterozygous missense variants in SETX were identified in families RDHT01, RDHT02 and RDHT10. These families were reverse phenotyped and found to suffer from ataxia with oculomotor apraxia type 2.

i

Affected individuals in family RDHT04 were homozygous for an identified pathogenic variant in TFG and significantly extended the phenotypic spectrum of TFG related disorders. Sleep disturbances, poor intellect, undeveloped speech, obesity, and severity of the disability to bed ridden status are the important phenotypic extensions in RDHT04 patients. This report also reinforces the differential impact of same variant to cause the disease.

Another family RDHT05 had a novel pathogenic homozygous variant in ALS2 in affected members. They were found to suffer from infantile onset of spastic paraplegia. The pathomechanism due to this variant is hypothesized to be a truncated protein with loss of important functional domains. This was hypothesized as mRNA was found to be expressed in blood samples of both unaffected and affected members of the family.

Patients of family RDHT06 had a known pathogenic nonsense mutation in ATM which was identified for the first time in homozygous condition. The patients suffered from ataxia telangiectasia with onset at five years of age. One of their sibling was still asymptomatic at the age of six years although he also carried the variant in a homozygous form. Perhaps, he will manifest the disorder later in life or may be a modifier will prevent disease progression.

In family RDHT08 a nonsense homozygous variant in C19orf12 was segregating with mitochondrial protein associated neurodegeneration (MPAN). The disease mechanism of this variant is hypothesized to be reduced mRNA as a result of nonsense mediated mRNA decay. The carriers for the variant exhibited no disease, which together with more than 70 unaffected C19orf12 heterozygous individuals from other reports supports the biallelic nature of neurological disorders due to C19orf12 variants. It further negates the monoallelic nature of disorder as suggested recently.

In family RDHT03 a novel homozygous missense variant in DRD4 was segregating with dystonia phenotype. The variant was not conserved in zebrafish, chicken and pig, but none of the organisms had Arg>Cys substitution at this position. The segregation with dystonia in patients, absence in ethnically control and absence of any other potential homozygous variant in affected individuals, supports the potential association of this variant with dystonia. Observation of the crystalline DRD4 at the position of the variant also supports the importance of this residue in DRD4-ligand interactions. A second homozygous novel variant in GENEA was fully conserved and segregated with

ii phenotype of delayed milestones in this family. Absence of this variant in normal population and high pathogenicity scores support its disease association. Previous association of heterozygous variants in this gene with ataxia has been reported but recessive inheritance is presented for the first time, though with different phenotype.

In family RDHT16, three variants were found to segregate with dopa responsive dystonia like phenotype. However, missense variant p.(Lys231Glu) in GENEB was the most likely candidate. Replication of the study is required in additional patients to establish it as bona fide gene implicated pathomechanism of neurological movement disorders if mutated.

Fibroblasts of a South Asian patient who had a novel compound heterozygous variants in FGD4, associated with Charcot-Marie-Tooth disease, type 4H were collected at Institute of Neurology, University College London, UK. The fibroblasts were grown and their proliferation rates were observed to be normal. The stability and localization of the encoded protein was not affected by these pathogenic mutations which suggests that further exploration of pathomechanism of these variant is required.

In a collaborative project at UCL, a novel bilallelic repeat expansion in RFC1was identified after analysis of whole genome sequencing in cerebellar atrophy, neuropathy and vestibular areflexia syndrome (CANVAS) (OMIM 614575) patients. A cohort of 304 normal individuals was screened by flanking PCR and reverse-prime PCR (RPPCR) for this newly identified expansion. The frequency of this mutant allele expansion was 0.7% in normal population with no homozygous individual for the variant. This work served as a verification that the variant is indeed rare and causes the disorder only in the homozygous condition.

For five families, exome sequencing failed to identify a pathogenic variant. The genetic disorder characterization may be completed in future through whole genome sequencing. This will reveal the underlying genetic cause for the disorder and perhaps will identify some new gene involvement in these disorders.

This study has benefited many families by providing the diagnosis and genetic counselling. At the same time it has revealed clinical and genetic information regarding movement disorders in Pakistani families. The results suggest that clinical and genetic diagnosis should be undertaken together to properly diagnose the disorders. Misdiagnosis undermines the possible treatment options and also negatively impacts the family in

iii search of the right diagnosis. In future, once all genes have been identified, candidate panels could be developed for rapid identification of the involved genes and their variants.

iv

Acknowledgement

All praises to Allah Almighty who blessed me, supported me, helped me, and finally enabled me to complete this work in dignified and honest way. Peace be upon the greatest man Hazrat Muhammad (PBUH) who settled an example of honesty, truthfulness, passion and determination for all humanity instead of all miseries, difficulties and sufferings.

I would like to cordially thank my research supervisor Dr. Sadaf Naz, who has supervised me during my PhD research. Her enthusiasm inspired me to work hard and do everything with dedication. She has always impressed me because of her punctuality and honesty towards her duties. The door to her office was always open and her continuous feedback really taught me to polish and improve my writing skills.

I acknowledge director general Dr. Naeem Rahsid for facilitating my research at School of Biological Sciences. I extend my regards to Professor Dr. Muhammad Akhtar, ex.Director General along with Dr.Javed Iqbal, who made gigantic efforts in establishing a research center at School of Biological Sciences, University of the Punjab, Lahore. I feel honored to be a student of Dr. AR Shakoori, and his lectures were the best lecture hours in my life. I am thankful to all the honorable Professors Emeritus and my teachers at SBS for their contributions to my studies at SBS. My regards to all my previous teachers including Dr. Qadri, for their tremendous support.

I acknowledge Prof. Henry Houlden for providing me with an opportunity to work at UCL Institute of Neurology, University College London, London, UK. I really appreciate him for trusting me and boosting my confidence. His appreciation and acknowledging words will always be alive in my heart.

I cordially acknowledge all participating families for their cooperation and sparing their time for research. Thanks to Dr. Jalil ur Rehman, Dr. Shahid Mukhtar, Dr. Rashid Imran, Dr. Iqra Tariq and others for cooperation in identification of some families and valuable clinical suggestions. I would like to thank Mr. Khalid and Mr. Arif for their tremendous help in sampling. I am thankful to all of my lab fellows, especially Azra, Humera, Faiza, Tayabba and Zunera for making a congenial environment in lab to pursue research.

v

I am highly thankful to all my colleagues and friends at UCL for their cheerful company and especially to Thomas, Ambreen, Stephanie, Erica, Nour, Roisin, Emer, Ilyas, Vincenzo and Ben for being there.

I thank the administration and technical staff at SBS who made things convenient for me in particular thankful to Mr. Abid, Mr. Gul, Mr. Irfan, Mr. Qadeer and Mr. Shakeel.

I express my thanks to all my friends especially Dr. Jalil, Saba, Zain, Atif, Bilal and my advisor, Mr. D.Patel, for being there for me whenever I needed them in facing the challenges in life. Thanks to Ashar for providing valuable guidelines in science. I am obliged to the cheerful company of my lovely friends Saba, Farhana, Khadija and Momina to relax in the stressful moments during this phase.

Last but not the least, I would like to thank my parents and siblings for their love, support, and continuous prayers for me. Their patience particularly during the process of research and thesis writing was exceptional. The cute and sweet words of Minaal have always been an encouragement for me which enabled me to justify every moment. At the end, thanks to all I have named, and all those whom I could not mention here. Final thanks to Higher Education Commission (HEC) of Pakistan who sponsored my research with an indigenous scholarship and IRSIP funding for my PhD studies.

Huma Tariq

March 1st 2019

vi

List of Abbreviation bp °C Degree Centigrade DNA Deoxyribonucleic Acid dNTPS Deoxynucleotide Tri-Phosphate

EDTA Ethylene Diamine Triacetic Acid Kb Kilobase Mb Megabase µg Microgram µl Microliter ml Milliliter mg Milligram ng Nanogram OMIM Online Mendelian Inheritance in Man PCR Polymerase Chain Reaction pmoles Pico Moles RNA Ribonucleic Acid rpm Revolution per Minute SDS Sodium Dodecyl Sulfate STR Short Tandem Repeats Taq Thermus aquaticus aa amino acids Tm Melting Temperature DMEM Dulbecco’s modified Eagles medium PBS Phosphate buffer saline

vii

List of Tables

Table 2.1: Lysis Buffer for DNA Extraction………………………..………………….……58

Table 2.2: TEN Buffer for DNA Extraction……………………………………..……….….58

Table 2.3: Low TE buffer………………………………………………………..……….….58

Table 2.4: 50X TAE Buffer………………………………………………………..…….…..59

Table 2.5: 10 mg ml-1Ethidium Bromide………………………………………………..…...59

Table 2.6: 6X Bromophenol Blue dye (loading dye) ………………………………….……59

Table 2.7: 10X PCR buffer (laboratory prepared) …………………………………………..59

Table 2.8: Precipitation Solution for Sequencing Reaction…………………………...……..60

Table 2.9: Primers used to amplify Microsatellite Markers……………...………………….60

Table 2.10: Growth media for fibroblasts…………………………………………………....61

Table 2.11: Fixative solution for Immunofluorescence ……………………………………..61

Table 2.12: Blocking solution for Immunofluorescence ………………………………..…..61

Table 2.13: RIPA lysis buffer……………………………………………………………...... 62

Table 2.14: Standards of different concentrations to make standard curve for BCA-protein assay…………………………………………………………………………………...... 62

Table 2.15: TRIS Acetate running buffer…………………………….……………………...62

Table 2.16: SDS gel loading sample preparation……………………………………………63

Table 2.17: TRIS GLYCIE transfer buffer………………………………………………….63

Table 2.18: 1X PBST ……………………………………………………………………….63

Table 2.19: Blocking solution for western blotting………………………………………....63

Table 2.20: Primers used for routine short range, long range and reverse-primed PCR to check the frequency of repeat expansion in normal population……………………………..64

Table 3.1: Variants considered and prioritized for family RDHT01………………………..69

viii

Table 3.2: Variants considered and prioritized for family RDHT02……………………..73

Table 3.3: Variant considered and prioritized for family RDHT10………………….…..76

Table 3.4: Variants considered and prioritized for family RDHT03………….………….79

Table 3.5: Clinical Investigations for proband of family RDHT04………………………89

Table 3.6: Molecular Investigations for proband of family RDHT04 …………………...91

Table 3.7: Variants considered and prioritized for family RDHT16…………………….105

Table 3.8: Variants considered and prioritized for family RDHT07…………………….108

Table 3.9: Homozygous variants considered and prioritized for family RDHT09 which were shared by two sibling……………………………………………………………….……112

Table 3.10: Homozygous variants considered and prioritized for family RDHT09 with the possibility of genetic heterogeneity……………………………………………………...113

Table 3.11: Variants considered for the possibility of heterozygous and imprinted allele……………………………………………………………………………………..114

Table 3.12: Variant considered and prioritized for family RDHT14…………………...118

Table 3.13: Sporadic cases collected and sent for molecular diagnosis………………...124

Table 3.14: Compound heterozygous variants responsible for disorder in patient of family UCL-HT01……………………………………………………………………………....126

Table 3.15: Absorbance values for different dilutions of known protein concentration…………………………………………………………………………...130

Table 4.1: Recessive variants of TFG with associated phenotypes……………………150 .

ix

List of Figures

Fig.1.1: CANVAS phenotype, a combination of three neurological conditions……………19

Fig. 1.2: Human Brain. Different parts of the brain ……………………………….………..22

Fig. 1.3: Coronal section of the brain showing the basal ganglia……………………………23

Fig.1.4: A neuron showing different genes associated with spastic paraplegia……………..25

Fig. 2.1: Map of Pakistan …………………….……………………………………………..32

Fig. 2.2: PCR profile for standard and touchdown conditions………………………..……..41

Fig. 2.3: Etched grid on the surface of hemocytometer………………………………….…..48

Fig 3.1: Pedigree, MRI scan and sequence chromatograms for family RDHT01………….. 67

Fig 3.2: Pedigree, MRI scan and sequence chromatogram for family RDHT02……………71

Fig.3.3: Pedigree and Sequence chromatograms for family RDHT10…………………...…75

Fig 3.4: Pedigree and Sequence chromatograms for family RDHT03…………………...... 78

Fig 3.5: DRD4 wild type sequence alignment against the structural counterpart 5WIU on PDB ………………………………………………………………………………………….81 Fig.3.6: Protein structure of 5WIU…………………………………………………………..82

Fig.3.7: Illustration of the Arg106 and the primary interacting partner Asp109 in 5WIU…..83

Fig 3.8: The modelled structure of GENEA and superimposition of wild type-mutant forms………………………………………………………………..………………………..85

Fig 3.9: Selected region of Phyre modelled secondary structure of GENEA.……...... ….....86 Fig 3.10: Pedigree, MRI scans and sequence chromatogram of family RDHT04……...…...88

Fig 3.11: Pedigree and sequence chromatogram of family RDHT05…………………...…..94

Fig. 3.12: ALS2 gene expression in family RDHT05……………………………………...... 95

Fig 3.13: Pedigree, MRI scan and sequence chromatogram of family RDHT06…………....97

Fig.3.14: Pedigree, MRI scans and sequence chromatogram of family RDHT08…………...99 Fig. 3.15: C19orf12 gene expression in family RDHT08…………………………………...100 Fig 3.16: Pedigree and sequence chromatograms of family RDHT16……………….…….102

x

Fig 3.17: Regions of homozygosity on chromosomes 1, in family RDHT16…………..104

Fig 3.18: Regions of homozygosity on chromosomes 6, in family RDHT16………….104

Fig.3.19: Pedigree of family RDHT07………………………………………………….107

Fig.3.20: Pedigree of family RDHT09………………………………………………….110

Fig. 3.21: Shared regions of homozygosity in family RDHT09………………………...111

Fig.3.22: Independent regions of autozygosity in family RDHT09……………………..111

Fig.3.23: Pedigree of family RDHT14…………………………………………………..116

Fig.3.24: Shared regions of homozygosity in family RDHT14………………………....117

Fig.3.25: Pedigree of family RDHT11………………………………………………….120

Fig.3.26: Pedigree of family RDHT12………………………………………………….120

Fig.3.27: Pedigree of family RDHT13…………………………………………………..122 Fig.3.28: Pedigree of family RDHT15…………………………………………………..122

Fig.3.29: Immunostained fibroblasts ……………………………………………………128

Fig.3.30: Standard curve of known BSA concentrations employed to quantify total cell protein……………………………………………………………………………………130

Fig.3.31: Frabin expression in control and affected fibroblast cell cultures ……………131

Fig.3.32: Amplification of 348bp fragment spanning repeat region ……………………134

Fig.3.33: DNA quality assurance PCR product…………………………………………134

Fig. 3.34: RPPCR and Sanger confirmation of repeat expansion ……………………….136

Fig. 3.35: Polymorphic configurations of the repeat expansion locus with allelic distribution………………………………………………………………………………..137

Fig.4.1: Diagrammatic representation of SETX …………………………………………140

Fig. 4.2: Alignment of respective orthologues of SETX, p.His1962 is boxed…………...142 Fig.4.3: Alignment of respective orthologues of SETX, residue 502 is boxed…………..142

Fig.4.4: Alignment of respective orthologues of SETX, p.Arg68 is boxed……………...145 Fig.4.5: Alignment of respective orthologues of SETX, residue 1857 is boxed………...145

xi

Fig.4.6: Diagrammatic presentation of TFG……………………………………………..147

Fig.4.7: ALS2 protein structure…………………………………………………………..154

Fig. 4.8: All monoallelic and some reported biallelic mutations affecting C19orf12……157

Fig.4.9: Demonstration of possible impact of Arg106Cys mutation on DRD4-ligand interactions……………………………………………………………………………….159

Fig.4.10: Alignment of respective orthologues of GENEA, Leu357 is boxed…………..161

Fig.4.11: Alignment of respective orthologues of GENEB, Lys231 is boxed…………..163

Fig.4.12: Alignment of respective orthologues of GENEC, Arg564 is boxed…………..163

Fig.4.13: Frabin structure along with domain organization …………..…………………165

.

xii

Table of Contents

Chapter 01: Introduction and Literature Review ...... 1 Introduction ...... 2 Literature review ...... 4 Ataxia ...... 4 Molecular basis of hereditary ataxia ...... 5 Autosomal recessive cerebellar ataxias (ARCA) ...... 5 Relative frequency, features and diagnosis of ataxia ...... 9 Problems in evaluation of hereditary ataxia ...... 10 Hereditary Spastic Paraplegias ...... 11 Infantile onset ascending spastic paralysis (IAHSP) ...... 12 IAHSP ...... 12 SPOAN like hereditary spastic paraplegia ...... 12 Dystonia ...... 13 Neurodegeneration with brain iron accumulation ...... 15 Parkinson’s disease ...... 16 Charcot–Marie–Tooth (CMT) disease ...... 17 Cerebellar atrophy, neuropathy and vestibular areflexia syndrome (CANVAS) ...... 18 Brain and Movement Disorders ...... 20 Mechanisms of movement disorders ...... 24 Importance of molecular testing and treatment options ...... 26 Methods for genetic research on movement disorders ...... 26 In candidate gene approach, ...... 27 SNP (Single Nucleotide Polymorphism) genotyping ...... 27 Movement Disorders in Pakistan ...... 28 Chapter 02: Materials and Methods ...... 30 Institutional Review Board Approval ...... 31 Recruitment of Families ...... 31 Sporadic cases ...... 31 Control Sample Screening ...... 31 Clinical Evaluation...... 33 a. Videotaping ...... 33

xiii

b. Electromyogram and Nerve Conduction Studies ...... 33 c. Magnetic Resonance Imaging ...... 33 d. Base line investigations...... 34 Blood Samples ...... 34 DNA Extraction from Blood ...... 34 Agarose Gel Electrophoresis...... 36 Blood RNA Extraction ...... 36 cDNA Synthesis ...... 37 Molecular Analysis ...... 37 Whole-Exome Sequencing...... 37 Data Annotation ...... 38 Designing the Primers ...... 39 Segregation analysis: Sanger Sequencing ...... 39 a. PCR Amplification ...... 39 b. Treatment of PCR product by EXOSAP ...... 40 c. Sequencing PCR ...... 42 d1. Precipitation ...... 42 d2. Sephadex cleaning ...... 42 e. Sequencing ...... 43 Microsatellite Markers: Genotyping ...... 43 Capillary Electrophoresis (Fragment Analysis) ...... 43 Expression of Candidate genes in Blood ...... 44 Population Screening ...... 44 Protein Modelling ...... 45 Primary Fibroblast Culture ...... 45 Establishing the fibroblast culture ...... 46 Cell counting assays: growth curves ...... 47 Cell counting using hemocytometer ...... 47 Cell seeding ...... 49 Growth Curve plotting ...... 49 Immunofluorescence/ Confocal Microscopy ...... 50 Fixation: ...... 50 Blocking: ...... 50 Immunostaining: ...... 50

xiv

Microscopy: ...... 51 Western blotting ...... 51 Cell lysis and protein extraction ...... 51 Protein quantification: ...... 52 SDS gel electrophoresis ...... 52 Transfer onto membrane ...... 53 Blocking, Primary and secondary antibody incubation ...... 53 Developing the membrane ...... 54 Biallelic AAGGG repeat in gene RFC1 expansion in CANVAS patients ...... 55 Work performed as part of this thesis ...... 55 Standard Short-range PCR ...... 55 Long-range PCR and Sanger sequencing ...... 56 Reverse-primed PCR ...... 56 4x NuPAGE™ LDS loading dye ...... 63 Chapter 03: Results...... 65 Families, RDHT01, RDHT02 and RDHT10 ...... 66 RDHT01 ...... 66 Clinical/phenotypic characterization ...... 66 Genetic Study ...... 68 RDHT02 ...... 70 Clinical/phenotypic characterization ...... 70 Genetic Study ...... 72 RDHT10 ...... 74 Clinical/phenotypic characterization ...... 74 Genetic Study ...... 74 Family RDHT03 ...... 77 Clinical/phenotypic characterization ...... 77 Genetic Studies ...... 77 Computational work...... 80 Mapping of DRD4 variant in DRD4 structure ...... 80 Modelling of GENEA and its variant ...... 84 Family RDHT04 ...... 87 Clinical/phenotypic characterization ...... 87 Genetic Study ...... 90

xv

Family RDHT05 ...... 92 Clinical/phenotypic characterization ...... 92 Genetic Study ...... 93 Functional study ...... 93 ALS2 expression in RDHT05 ...... 93 Family RDHT06 ...... 96 Clinical/phenotypic characterization ...... 96 Genetic study ...... 96 Family RDHT08 ...... 98 Clinical/phenotypic characterization ...... 98 Genetic Study ...... 98 Functional Study ...... 98 C19orf12 expression in RDHT08 ...... 98 Family RDHT16 ...... 101 Clinical/phenotypic characterization ...... 101 Genetic Study ...... 103 Families RDHT07, RDHT09 and RDHT14 ...... 106 RDHT07 ...... 106 Clinical/phenotypic characterization ...... 106 Genetic Study ...... 106 RDHT09 ...... 109 Clinical/phenotypic characterization ...... 109 Genetic study ...... 109 RDHT14 ...... 115 Clinical/phenotypic characterization ...... 115 Genetic Study ...... 115 Families, RDHT11, RDHT12 ...... 119 RDHT11 ...... 119 Clinical/phenotypic characterization and genetic studies ...... 119 RDHT12 ...... 119 Clinical/phenotypic characterization and genetic studies ...... 119 Families, RDHT13 and RDHT15 ...... 121 RDHT13 ...... 121 RDHT15 ...... 121

xvi

Sporadic cases ...... 123 UCL-HT01 ...... 125 Clinical characterization ...... 125 Genetic studies ...... 125 Functional studies ...... 127 Proliferation rate of fibroblasts with mutant frabin ...... 127 Localization of protein in patient verses normal fibroblasts ...... 127 Western Blotting: expression and quantification of protein of interest ...... 129 Allelic distribution of a short tandem repeat of RFC1 gene in the control population ...... 133 Flanking PCR on all control samples...... 133 Pre-Alu PCR to remove false positive results...... 133 Long range flanking PCR and RPPCR for final screening ...... 135 RPPPCR and long range PCR for affected individuals and carriers ...... 135 Frequency of different allelic conformations ...... 135 Chapter 04: Discussion ...... 138 Novel SETX variants identified in three families ...... 139 TFG variant associated severe congenital disorder ...... 146 A novel homozygous ALS2 frameshift variant associated with IAHSP ...... 152 A pathogenic homozygous ATM variant identified in an asymptomatic individual ...... 155 Are some C19orf12 variants monoallelic for neurological disorders? ...... 155 Two variants segregating with two different phenotypes in one family ...... 158 Family with three segregating variants ...... 162 Frabin stability and localization is not affected by compound pathogenic missense variants ...... 164 Families in which no genetic cause was identified ...... 166 Frequency of AAGGG repeat expansion in RFC1 ...... 167 Conclusion ...... 168 References………………………………………………………………………………….170 Appendices …………………………...……………………………………………………187 Appendix A-1………………………………………………………………………………188 Appendix A-2 (Publications)……………...... ………………………………………...191

xvii

Chapter 01: Introduction and Literature Review

Chapter 01: Introduction and Literature Review

1

Chapter 01: Introduction and Literature Review

Introduction

Movement disorders are neurological genetic diseases which can impair movement, or cause ataxia and spasticity. These disorders include Huntington's disease, Wilson's disease, ataxias, spastic paraplegia, Parkinson’s disease, essential tremors, neuroacanthocytosis and dystonia (Dietz 2000; Jarman and Wood 2002). Myoclonus, tics, athetosis, ballism and chorea are also included in this heterogeneous category (Cossu and Colosimo 2017).

A complicated coordination of brain, spinal cord, nerves and muscles drives all types of voluntary and involuntary movements. An interruption or anatomical defect in any of these components may result in movement disorders (Pizzolato and Mandat 2012). The affected individuals are disabled to various degrees due to different movement disorders. Secondary complications such as cognitive deficits, seizures, autoimmune deficiencies, and psychiatric symptoms may also be observed in some patients. The quality of life is significantly reduced and, in some cases, the life expectancy may also be shortened. Pathogenic variants in different genes have been identified for Huntington's disease, Chorea, neurodegeneration with brain iron accumulation, Wilson's disease, spinocerebellar ataxias, recessive ataxias, hereditary spastic paraplegia, and hereditary dystonia (Marras et al. 2017; Schulz 2007).

The hereditary movement disorders can follow different inheritance patterns in different families. These include autosomal recessive, X-linked recessive, autosomal dominant, X- linked dominant or mitochondrial inheritance. Several genetic loci have been identified for ataxia, dystonia, spastic paraplegia, Parkinsonism, essential tremor, and many other movement disorders. The responsible genetic factors has not been identified for many of these disorders as yet, which suggests that other genes and their variants will be identified in future (Krebs and Paisan-Ruiz 2012). Whole exome sequencing or whole genome sequencing help in rapid identification of disease causing variants by analyzing entire exome and genome, respectively. These technologies have increased the rate of discovery of new genes involved in disorders like ataxia, dystonia as well as other movement disorders (Wang et al. 2016). Traditional and new approaches to investigate inherited diseases will increase understanding of the molecular basis for movement disorders.

2

Chapter 01: Introduction and Literature Review

This understanding will reveal common genes and their variants involved in these disorders in local population. It will help in correct diagnosis in a timely fashion and possible treatment of the disorders (Singleton 2011).

Movement disorders affect individuals worldwide. In Pakistan, the high frequency of consanguineous marriages has given rise to increased incidence of recessively inherited genetic disorders, including movement disorders. At the same time, lack of molecular understanding for the genetic causes, for the disorders may result in misdiagnosis of even some treatable cases. Ataxia with vitamin E deficiency, and many dopa responsive dystonias can be treated or managed adequately if diagnosed in time (Bonello and Ray 2016; Gabsi et al. 2001). Some cases of hyperkinetic movement disorders, including primary dystonia, essential tremor and Parkinson’s disease have also been treated effectively by targeted deep brain stimulation (DBS) (Barbey et al. 2015; Guzzi et al. 2016).

The aim of this work is to molecularly and clinically characterize recessively inherited movement disorders in Pakistani families. The research will yield the basic information about the genetics and biology of these disorders. At the same time, it will provide an exact diagnosis to the participants which in some cases, may lead to better management of their conditions. Individuals will also obtain the information about their own carrier status, which could be used to reduce the incidence of the disorder in their families in future generations.

3

Chapter 01: Introduction and Literature Review

Literature Review

Movement disorders are neurological conditions of two types; hypokinetic (associated with the loss of movements) or hyperkinetic (with excess movements) (Fahn 2011). Hyperkinetic movement disorders are actually dyskinesias which are abnormal or impaired voluntary movements. Chorea, tics, dystonia, myoclonus, stereotypies and tremor are some examples of these (Schlaggar and Mink 2003). Parkinson’s disease is a classic example of a hypokinetic movement disorder (Borrell 2000).

Hereditary movement disorders have profound consequences for affected individuals as well as their relatives. The inheritance pattern for these disorders may follow autosomal dominant, autosomal recessive, or X-linked mode of inheritance. Genetic causes of a large number of movement disorders have been mapped to a specific genomic region. Several genetic loci are known for ataxia, spastic paraplegia, dystonia, and other movement disorders. However, many movement disorders, and related genes remain to be discovered which underscores the need to continue research on genetics of movement disorders.

Ataxia

Ataxia are genetically and clinically, heterogeneous group of neurodegenerative disorders. They are accompanied by cerebellar degeneration with, or without, pyramidal and extrapyramidal features. Variable polyneuropathy may also be present. Phenotype of ataxia patients can be either purely cerebellar, or constitute a cerebellar plus syndrome as it may, or may not have systemic manifestations. Uncoordinated gait, truncal instability, tremors of the body or head, uncontrolled co-ordination of hands, dysarthria, and abnormal eye movements are prominent features of cerebellar ataxia. Extrapyramidal signs, such as retinal, cardiac, muscle or neuronal involvement may also be observed, but are less common (Breedveld et al. 2004; Brunberg 2008). Pure cerebellar ataxias constitute about 20% of all ataxias, and are also clinically and genetically heterogeneous neurodegenerative disorders. Autosomal recessive cerebellar ataxia-1 (ARCA1) is an example of pure cerebellar ataxia. CACNA1A, SPTBN2 and SYNE1 mutations are well known causative factors in pure cerebellar ataxias (Bouhlal et al. 2008; Gros-Louis et al. 2007). A high degree of variation in the age of onset and disease progression is observed in different individuals affected with ataxia and its syndromes (Breedveld et al. 2004; Brunberg 2008).

4

Chapter 01: Introduction and Literature Review

Prevalence of ataxia in children is estimated to be the least in Europe with a prevalence of 26 in 100,000 children (Musselman et al. 2014). According to a global study it is estimated that 1 out of 10,000 individuals is affected with hereditary cerebellar ataxia, or hereditary spastic paraplegia (Ruano et al. 2014).

Ataxias may be sporadic or hereditary, congenital, or non-congenital. Sporadic ataxias are caused by various paraneoplastic, toxic, inflammatory, endocrinal, metabolic, malabsorption conditions or de novo variants. Ataxias of unknown genetic reason are called idiopathic ataxias (Breedveld et al. 2004). Congenital ataxias are nonprogressive disorders involving hypotonia, developmental delay leading to ataxia, dysarthria, mental retardation, atrophy of the cerebellum (Delague et al. 2001). Non-congenital adult onset ataxias are usually progressive and the condition of patients worsens with time. One such case is autosomal recessive cerebellar ataxia type 1 or ARCA1 also called SYNE1-related autosomal recessive cerebellar ataxia. It is characterized by onset of cerebellar ataxia, and dysarthria at a mean age of 31 years, with slow progression and moderate disability (range 17-46 years). Syne-1 is a protein, which plays an important role in anchoring of specialized myonuclei at neuromuscular junctions (NMJ). These regions express increased amount of acetylcholine receptor (AChR) subunit genes (Dupre et al. 2007; Gros-Louis et al. 2007).

Molecular basis of hereditary ataxia

Hereditary ataxias are progressive neurodegenerative disorders characterized by symptoms and signs mainly of the central nervous system (CNS). Spinocerebellar ataxia syndromes also affect the spine in addition to the cerebellum. Many autosomal dominant hereditary ataxia are usually caused by point mutations or expanded trinucleotide or pentanucleotide repeats in the respective genes (Gros-Louis et al. 2007). Autosomal recessive hereditary ataxias have also been clinically characterized, and many responsible genes have been identified.

Autosomal recessive cerebellar ataxias (ARCA) are a heterogeneous group of progressive neurodegenerative disorders associated with cerebellar atrophy and spinal tract dysfunction. Friedreich's ataxia (FA) and ataxia telangiectasia (A-T) are more common as compared to all other forms of recessively inherited ataxias.

5

Chapter 01: Introduction and Literature Review

The other frequent forms are autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS), ataxia with oculomotor apraxia type 1 (AOA1), ataxia with oculomotor apraxia type (AOA2), ataxia include ataxia with vitamin E deficiency (AVED) and POLG related syndromes (Storey 2014). Early-onset ARCA mostly have an underlying genetic cause. Adult-onset ARCA can have a genetic cause but may be undiagnosed due to early death of affected individuals.

Friedreich ataxia (FA) with early onset, is the most common inherited ataxia worldwide but a few cases of late onset Friedreich’s ataxia (LOFA) have also been reported. Typical features of this disease are ataxia, areflexia, Babinski sign, sensory loss, weakness, scoliosis and pes cavus (Martinez et al. 2016; Shinnick et al. 2016). Friedreich’s ataxia and ataxia with vitamin E deficiency have a nearly common phenotype. Friedreich's ataxia (FA) is inherited in an autosomal recessive fashion. Homozygous trinucleotide expansions, or heterozygous loss of function variants in FRDA result in disease phenotype (Ben Hamida et al. 1993).

FRDA encodes frataxin, which is translocated to mitochondria where it acts as iron chaperone. It has important roles in iron-sulfur biogenesis and heme biosynthesis. Frataxin also has antioxidant, and tumor suppressor effects (Navarro et al. 2010). In drosophila, localized reduction of this protein from glial cells results in sensitivity to oxidative damage, neurodegeneration with compromised locomotor activity (Schmucker et al. 2008).

Ataxia with vitamin E deficiency (AVED) is an autosomal recessive neurodegenerative disorder caused by mutations in the alpha-tocopherol transfer protein, TTPA. Patients have progressive spinocerebellar symptoms with significantly reduced plasma levels of vitamin E. Visual impairment is also exhibited by few AVED patients due to macular degeneration Truncating and missense mutations in TTPA have been associated with AVED (Mariotti et al. 2004). Treatment of these patients is possible by Vitamin E supplementation therapy which alleviates neurological conditions in most of the patients. Visual impairment is also exhibited by few AVED patients due to macular degeneration (Iwasa et al. 2014; Mariotti et al. 2004).

6

Chapter 01: Introduction and Literature Review

Ataxia telangiectasia (A-T) is a neuro-immunologic disease caused by pathogenic variants in ATM. Ataxia telangiectasia mutated (ATM) encodes a serine-threonine kinase which is crucial in DNA repair and is a master regulator of DNA damage response (Mansfield et al. 2017). Ataxia telangiectasia is a pleiotropic disorder but typical features include cerebellar ataxia, telangiectasia, immunodeficiency, hypersensitivity to radiations and increased risk of malignancies (Wright et al. 1996). Life span of A-T patients is usually shortened due to higher chances of malignancies (Davis et al. 2013). Absence of full length ATM is the most common molecular basis of the disorder but biallelic missense mutations have also been reported (Sandoval et al. 1999; Sasaki et al. 1998). All reported pathogenic variants of ATM are associated with severe neurological symptoms except for the variant c.5585T>A p.(Leu1862His). An individual carrying this variant manifested only mild neurological symptoms (Roohi et al. 2017). In addition, genetic heterozygous carriers of loss of function mutations in ATM are at an increased risk of developing breast cancer (Bernstein and Concannon 2017).

A-T was assumed to be a non-curable disease. Currently, supportive therapy by administration of glucocorticoid drug betamethasone improves cerebellar functions in A- T patients. The efficacy of this therapy demonstrates the importance of antioxidant and anti-inflammatory mechanisms in normal physiological functions (Broccoletti et al. 2011; Buoni et al. 2006; Giardino et al. 2013; Russo et al. 2009).

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is an ataxia with pyramidal features, with onset around two years, but the age of onset can be variable. A very prominent sign of the disease is spastic paraparesis which worsens with time. Polyneuropathy and retinal myelinated fibers on eye fundus are also exhibited by few patients and brain magnetic resonance imaging reveals vermian cerebellar atrophy among these patients (Anheim et al. 2008). Progressive spasticity and amyotrophy of distal muscles are also important features. Biallelic mutations in SACS cause ARSACS (Anheim et al. 2008; Bouslam et al. 2007; Bradshaw et al. ; Criscuolo et al. 2005). SACS encodes sacsin protein, which has particular role in ubiquitin proteasome pathway (Engert et al. 2000; Li et al. 2015).

7

Chapter 01: Introduction and Literature Review

Ataxia with oculomotor apraxia (AOA) Ataxia with oculomotor apraxia (AOA) are recessive ataxias which are caused by pathological defects in DNA repair mechanisms. Characteristic abnormalities and defective control of eye movements and impairment of saccade initiation are prevalent features in this group of ataxia. AOA1 is characterized by early-onset and slowly progressive cerebellar ataxia with peripheral neuropathy and areflexia. Ocular apraxia is observed in early stages of disease whereas hypercholesterolemia, hypoalbuminemia, and cognitive impairment are observed at the adult stages. Biallelic mutations in APTX are responsible for this disorder (D'Arrigo et al. 2008). APTX encodes a protein called aprataxin which is a single strand break DNA repair protein. Absence of functional APTX compromise the repair mechanism and also adversely affect mitochondrial DNA (Rana et al. 2013).

AOA2 is a less common form of ARCA and is clinically characterized by progressive cerebellar atrophy, peripheral neuropathy, oculomotor apraxia and elevated α-fetoprotein levels in serum. The age of onset varies so that a person can be affected anywhere between 10–20 years of age (Lavin et al. 2013). Recessive mutations in SETX cause the disorder. SETX encodes the protein senataxin. Senataxin is a helicase and has been suggested to play important roles in regulation of transcription, mRNA splicing, DNA damage response and gene silencing (Becherel et al. 2013; Choudhury et al. 2017; Lavin et al. 2013; Yeo et al. 2015).

Ataxia with oculomotor apraxia type 3 (AOA3) is caused by mutations in PIK3R5 (Al Tassan et al. 2012). PIK3R5 encodes a protein of 101 KD named as phosphoinositide 3- kinase regulatory subunit 5, which is a subunit of class 1 phosphoinositide 3-kinases (PI3Ks). The important cellular functions attributed by PI3Ks are proliferation, differentiation, growth, and chemotaxis (Brock et al. 2003).

Ataxia with oculomotor apraxia type 4 (AOA4) disorder is caused by mutations in PNKP. AOA4 is a complex movement disorder with progressive hyperkinetic features, eye movement abnormalities, polyneuropathy, cognitive impairment and chorea (Paucar et al. 2016). PNKP encodes a protein polynucleotide kinase 3-prime phosphatase, which has 5' phosphorylation and 3' phosphatase activities, and thus plays an important role in DNA repair (Bernstein et al. 2005).

8

Chapter 01: Introduction and Literature Review

Cayman cerebellar ataxia is a type of congenital ataxia characterized by hypotonia, truncal limb ataxia, dysarthria, psychomotor delay, nystagmus, cerebellar hypoplasia, intention tremors, and non-progressive cerebellar dysfunction (Nystuen et al. 1996). This disorder is caused by mutations in ATCAY. The protein encoded by this gene is called caytaxin which has a CRAL-TRIO domain. ATCAY plays crucial roles in synaptogenesis and glutamate synthesis (Bomar et al. 2003).

Marinesco-Sjoegren syndrome (MSS) is a recessively inherited rare neurodegenerative multisystem disorder. The disease is characterized by cerebellar atrophy, ataxia, cataracts, skeletal muscle myopathy, motor neuropathy, bradykinesia and developmental delay (Byrne et al. 2015). The pathogenic variants are located in a gene called SIL1 which encodes an endoplasmic reticulum (ER) resident co-chaperone protein. This protein regulates cell stress and plays a role in protein translocation into the ER. It also assists in proper folding of the newly translated proteins and regulates degradation of improperly folded proteins (Anttonen et al. 2008a).

Sensory ataxic neuropathy, dysarthria, and ophthalmoparesis (SANDO) is a recessive form of ataxia which is caused by biallelic mutaions in POLG. POLG is a nuclear-encoded DNA polymerase-gamma enzyme. Mutant POLG function results in mitochondrial dysfunction and mtDNA depletion in skeletal muscle and peripheral nerve tissue. SANDO is characterized by severe sensory ataxic neuropathy, dysarthria and chronic progressive external ophthalmoplegia. Lower limb areflexia, positive Romberg sign, peripheral axonal neuropathy, migraine and depression are also reported in patients (Milone and Massie 2010).

Relative frequency, features and diagnosis of ataxia

Friedreich ataxia (FA) is the most common recessive ataxia. Ataxia telangiectasia (A-T) is approximately eight times less frequent than FA. Marinesco-Sjoegren syndrome (MSS), Ataxia with oculomotor apraxia type 2 (AOA2) and autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) are even less frequent than AT. Due to its high prevalence, FRDA is usually categorized as an independent form of ataxia rather than under ARCA. Peripheral neuropathy and lower limb areflexia are more frequent in the Friedreich ataxia as compared to non-Friedreich ataxia.

9

Chapter 01: Introduction and Literature Review

AOA2 and A-T are both autosomal recessive disorders, associated with DNA repair dysfunction. They can be accompanied by oculomotor apraxia, sensorimotor neuropathy or cerebellar atrophy. AOA2 patients may also have pyramidal signs whereas A-T patients are at an increased risk of developing cancer. Alpha feto-protein, AFP levels are elevated in both disorders but are significantly higher in A-T patients (150µg/L in AT as compared to 90µg/L in AOA2 patients and less than 10µg/L in controls). The distinguishing features among the two disorders are telangiectasia, low serum immunoglobin and younger age of onset in A-T patients (8.2±2.5) whereas the age of onset in AOA2 patients is 14.8±2.4, with slow disease progression (Mariani et al. 2017).

Symptomatic ataxia is diagnosed on the basis of typical history and simple laboratory tests. A negative family history does not rule out autosomal recessive or X-linked ataxia. Few autosomal dominant ataxias with reduced penetrance or those due to de novo mutations can also have negative family history.

Investigations of vitamin E, albumin, cholesterol, phytanic acid, lactic acid, creatine kinase, coenzyme Q10, alpha-fetoprotein, copper, ceruloplasmin and chitotriosidase are helpful in reaching a diagnosis of ataxia. Clinical investigations include nerve conduction studies and magnetic resonance imaging to identify the presence and the type of neuropathy and cerebellar atrophy, respectively. These investigations help to establish a suspected diagnosis which is further confirmed by molecular testing and detection of the causative genetic mutation (Bouhlal et al. 2008).

Problems in evaluation of hereditary ataxia

Many factors make evaluation of hereditary ataxia difficult. These include imprinting effects, variable expression, overlapping phenotypes of different types of ataxia, and different phenotypes caused by the same variant. Adoption and non-paternity can also confer problem in evaluation of disease (Breedveld et al. 2004). Some types of ataxias can be associated with myoclonus and epilepsy and are classified as epilepsy disorders. Unverricht-Lundborg disease is an example of such a disease where ataxia is present but the main problem is progressive myoclonus epilepsy (El-Shanti et al. 2006).

10

Chapter 01: Introduction and Literature Review

Hereditary Spastic Paraplegias

Hereditary spastic paraplegias (HSP) are a group of neurodegenerative gait disorders which affect the corticospinal tracts, upper motor neurons and the lower motor neurons. The primary clinical manifestation in HSP is spasticity, immobility and weakness of the lower limbs (Blackstone 2012; Harlalka et al. 2016). Spasticity which is the main and classical clinical manifestation of spastic paraplegia is an increase in muscle tone due to increased excitability of the muscle stretch reflex. It is manifested as an increased resistance conferred by muscles to passive stretching. Spasticity is related to increased tendon reflexes, clonus and flexor/extensor spasms (Mukherjee and Chakravarty 2010). This disease has a prevalence of 2–7.4/ 100000 in most populations (Kara et al. 2016). Involvement of other neurons creates a spectrum of phenotypes ranging from pure HSP to complicated HSPs (Beetz et al. 2013b). These categories are described on the basis of absence (pure) or presence (complicated) of other clinical features. The additional phenotypes can include retinopathy, distal amyotrophy, cognitive dysfunction, ataxia, thin corpus callosum and peripheral neuropathy (Blackstone 2012). HSPs are associated with mutations of more than 70 genes and the genotype-phenotype correlations are difficult to establish (Harlalka et al. 2016; Racis et al. 2014).

Hereditary spastic paraplegias affect several hundred thousand individuals worldwide (Blackstone 2012). Frequencies of dominant and recessive hereditary spastic paraplegias are comparable but the X-linked forms are much rarer (Blackstone 2012). More than 70 genes have been implicated in the genetics of hereditary spastic paraplegias (Harlalka et al. 2016). A large number of HSP cases are caused by mutations in genes involved in membrane trafficking and organelle architecture (Blackstone 2012; Soderblom and Blackstone 2006; Yagi et al. 2016). Some of these include SPAST (OMIM 604277), ATL1 (OMIM 606439), RTN2 (OMIM 603183) and REEP1 (OMIM 609139) among others. Mutations in SPG11 (OMIM 604360), PGN (OMIM 607259), FA2H (OMIM 612319), ZFYVE26 (OMIN 270700), AP5Z1 (OMIM 613647), CYP7B1 (OMIM 270800) and ATP13A2 (OMIM 617225) have been implicated in pathogenesis of complex recessively inherited forms of spastic paraplegias (Kara et al. 2016). Most of these genes are involved in neuronal axonal growth, cargo trafficking, degradation of misfolded proteins, cholesterol biosynthesis and myelin synthesis pathways.

11

Chapter 01: Introduction and Literature Review

Infantile onset ascending spastic paralysis (IAHSP)

IAHSP is a type of HSP which is characterized by infantile onset of spastic paraplegia at the age of ambulation with progression of disease to the upper limbs. The disorder involves upper motor neurons, and is accompanied by dysarthria and sometimes dysphagia. Lower motor neurons (LMNs) are spared but can also be affected which may result in hypotonia and fasciculations (Eymard-Pierre et al. 2002; Wakil et al. 2014). IAHSP (OMIM 607225) is caused by recessive mutations in ALS2 (Amyotrophic Lateral Sclerosis 2) which encodes alsin. Alsin is a putative GTPase regulator and has three important functional domains (Gros-Louis et al. 2003; Wakil et al. 2014). Mutations in ALS2 have been implicated in two other clinically overlapping upper motor neurodegenerative disorders, juvenile amyotrophic lateral sclerosis (ALS2, OMIM 205100) and juvenile primary lateral sclerosis (JPLS, OMIM 606353) (Eymard-Pierre et al. 2002). Progression of motor disability in IAHSP is rapid which may result in confinement to a wheelchair at the mean age of 3.2- 8.4 years (Flor-de-Lima et al. 2014; Racis et al. 2014; Wakil et al. 2014).

SPOAN like hereditary spastic paraplegia

Spastic paraplegia with optic atrophy and neuropathy known as SPOAN is a severe form of complicated HSP in which the patients exhibit spastic paraplegia with optic atrophy and neuropathy (OMIM 609541). This disorder is associated with biallelic mutations in KLC2 (Macedo-Souza et al. 2005; Melo et al. 2015). Other spastic paraplegia disorders without involvement of KLC2 but with similar phenotypes as that of SPOAN are called SPOAN like disorders (Amorim et al. 2014).

12

Chapter 01: Introduction and Literature Review

Dystonia

The Dystonia constitute a heterogenous group of hyperkinetic movement disorders accompanied by involuntary muscular contractions resulting in abnormal body postures. These disorders may have onset in infancy or late adulthood. The prevalence for late onset dystonia is higher as compared to early onset dystonia (Defazio et al. 2004). Prevalence of 16.43 per 100,000 person is estimated for this dystonia which is probably an underestimate (Steeves et al. 2012). On the basis of clinical characteristics, dystonia can be categorized by age of the onset, physical distribution, temporal pattern and presence or absence of additional clinical complications. On the basis of the age of onset it can be infancy dystonia (birth to 2 years), childhood dystonia (3-12 years), adolescence dystonia (13-20 years), early adulthood dystonia (21-40 years) and late adulthood dystonia (>40 years). Dystonia can be focal dystonia (onlyone body part is attected ,.e.g s’rriter cramp ,(.etcsegmental dystonia (adlacent body parts are involved e.g. cranial dystonia), multitocal dystonia (tro or more noncontiguous body parts are affected), hemi-dystonia (one full or partial body side is affected), and generalized dystonia (the trunk and other sites are involved) on the basis of body distribution. Temporal pattern is important in explaining some characteristic features of dystonia like manner of onset (acute or chronic), progression over time (non-progressive or progressive) and short-term variations in symptoms (intermittent, action-specific, diurnal fluctuations, episodic). The disorder may be present in pure form as a sole motor feature without other neurological features referred to as isolated dystonia, or may present with co-occurrence of other neurological problems and movement disorders termed as combined dystonia (Zech et al. 2017).

Dystonia can be inherited or acquired. Acquired dystonia is attributed to environmental factors including brain injury, drugs, and toxins, vascular and neoplastic factors. Familial and sporadic cases of dystonia where genetic factor is unknown are called idiopathic (Jinnah and Factor 2015). Dystonia may follow any pattern of inheritance but the frequency of recessive dystonia is less as compared to dominant dystonia (Albanese et al. 2013).

13

Chapter 01: Introduction and Literature Review

Dystonia is a disorder with heterogeneous genetic basis. Although many genes have been identified, many still remain to be discovered (Domingo et al. 2016). In some cases, dystonia can accompany neurodevelopmental disorders. These complex forms of disease are accompanied by neurodevelopmental delay and intellectual disability (Domingo et al. 2016).

Some forms of dystonia result from dopamine deficiency termed as dopa responsive dystonia (DRD). These are treatable with dopamine therapy. DRD is a syndrome of nigrostriatal dopamine deficiency caused by genetic defects in the dopamine synthetic pathway but without degeneration of nigral cells. DRD phenotype results from deficiencies of enzymes which are important in dopamine synthetic pathway. These deficiencies are caused by pathogenic mutations in GTP cyclohydrolase I (GCH-1 OMIM 128230), tyrosine hydroxylase (TH OMIM 605407), and sepiapterin reductase (SPR OMIM 612716). Disorders due to variants in GCH1, TH and SPR may be recessively or dominantly inherited. Some other disorders which are also responsive to dopaminergic drugs are transportopathies, SOX6 related decreased number ot nigral cells, Parkinson’s disease related degenerative loss of nigral cells and some disorders which do not affect the nigrostriatal dopaminergic system e.g DYT1; GLUT1 deficiency; myoclonus-dystonia; ataxia telangiectasia (Lee et al. 2018).

DRD is usually accompanied by Parkinsonism, which is a complex of symptoms, and sometimes confused with young onset Parkinson's disease (YOPD). DRD and YODP both are caused by dopamine deficiency. Loss of nigral cells causes YOPD and defects in dopamine synthesis cause DRD. Assessment of nigrostriatal dopaminergic system integrity and degeneration is important for making the exact diagnosis. Segawa syndrome; dopa responsive recessive form of dystonia, is caused by deficiency of the tyrosine hydroxylase (TH) of the nigrostriatal dopamine neurons. Tyrosine hydroxylase is a crucial enzyme for the conversion of amino acid tyrosine to dopamine (Segawa et al. 2013). The patients carrying mutation in SLC30A10 suffer from recessive dystonia (OMIM 613280) which is unresponsive to dopamine therapy and patients have elevated manganese levels in their blood. It is a basal ganglia malfunction, caused by the loss of function of manganese transporter (Tuschl et al. 1993).

14

Chapter 01: Introduction and Literature Review

Mutations in genes for neurodevelopmental pathways, calcium homeostasis, dopamine production pathway and other transcriptional pathways have been identified in dystonia patients (Iwabuchi et al. 2013; Kumar 2015). Pathogenic dominant variants in KCTD17 (OMIM 616398), CACNA1B (OMIM 614860), TUBB4A (OMIM 128101), ANO3 (OMIM 615034), THAP1 (OMIM 602629), ATP1A3 (OMIM 128235), SGCE (OMIM 159900) and GNAL (OMIM 615073) whereas recessive variants in HPCA (OMIM 224500) and COL6A3 (OMIM 616411) have been implicated in either pure or combined persistent dystonic phenotypes (Domingo et al. 2016; Lee et al. 2018).

Neurodegeneration with brain iron accumulation

Neurodegeneration with brain iron accumulation are a group of neurodegenerative disorders which usually involve progressive motor and cognitive dysfunction. They can be of early childhood or adulthood onset. They are characterized by spasticity, dystonia, parkinsonism, and usually iron accumulation in the basal ganglia (Dusi et al. 2014). Recessively inherited forms of NBIA are associated with pathogenic mutations in different genes such as PANK2 (OMIM 234200), PLA2G6 (OMIM 610217 ), CP (OMIM 604290 ), FA2H (OMIM 612319 ), COASY (OMIM 615643), REPS1 (OMIM 617916), CRAT (OMIM 617917), and C19orf12 (OMIM 614298) (Hartig et al. 2011; Monfrini et al. 2018).

Oxidative stress plays a significant role in pathogenesis of neurodegenerative diseases. Iron is an essential metal for proper functioning of large number of proteins and enzymes. Since an excess of iron is toxic, free iron generates oxidative stress leading to the toxicity. The brain cells have less number of oxidative stress defense mechanisms which explains pathogenesis of some of the movement disorders; Parkinson's disease, Alzheimer's disease, Huntington's chorea, Amyotrophic Lateral Sclerosis, and neurodegeneration with Brain Iron Accumulation (NBIA) for example. Metal chelating drugs therapies can be considered for management of these disorders (Carocci et al. 2018) .

COASY and PANK2 are genes mutated in two recessive forms of NBIA. COASY and PANK2 encode important enzymes, coenzyme A synthase and pantothenate kinase, respectively which are crucial for coenzyme A synthesis. The compromised synthesis of coenzyme A results in the disorder (Daugherty et al. 2002; Zhou et al. 2001).

15

Chapter 01: Introduction and Literature Review

Heterozygous mutations in FTL (OMIM 606159) cause autosomal dominant form of NBIA whereas dominant mutations in WDR45 (OMIM 300894) cause X-linked dominant form of disorder (Saitsu et al. 2013) (Cremonesi et al. 2004). A subtype of autosomal recessively inherited NBIA which is caused by bi-allelic pathogenic mutations in C19orf12, is called mitochondrial protein associated neurodegeneration (MPAN) (Monfrini et al. 2018).

Parkinson’s disease

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by asymmetric bradykinesia, rigidity, resting tremor and postural instability. PD can also involve many rheumatological, sleep and mood disorders, but sometimes it is misdiagnosed as other Parkinsonian disorders and drug induced Parkinsonism. Careful clinical assessment, follow-up with monitoring of the treatment response along with the history of premotor, non-motor symptoms (sleep disorders and constipation), help in proper diagnosis of PD. Non-motor features in early disease course usually support the diagnosis of PD (Ali and Morris 2015).

PD is classified into two categories, based upon the age of onset. Late-onset PD (LOPD) is common among people of age over 55 years. PD diagnosed in younger individuals between the ages of 21–45 years is called young onset PD (YOPD) (Rana et al. 2016).The tirst genetic mutation knorn to cause Parkinson’s disease (PD) ras described in SNCA (OMIM 605543 ) and since then, 27 genes have been reported to be associated with either autosomal dominant, autosomal recessive, or X-linked PD. Mutations in PRKN (OMIM 600116), ATP13A2 (OMIM 606693), PINK1 (OMIM 605909), DJ1 (OMIM 606324), PLA2G6 (OMIM 612953), FBXO7 (OMIM 260300), DNAJC6 (OMIM 615528), SYNJ1(OMIM 615530) and VPS13C (OMIM 616840) have been associated with recessively inherited forms of PD. PRKN, PINK1 and DJ1 variants are associated with slowly progressing typical early onset PD, whereas other autosomal recessive genes are associated with severe atypical Parkinsonism (Lunati et al. 2018) .

Mitochondrial dysfunction has been implicated in pathogenesis of PD. PINK1, encodes a ubiquitously expressed mitochondrial serine/threonine protein kinase. The kinase function is compromised in early-onset Parkinson's disease (PD) attributed to recessive mutations in PINK1.

16

Chapter 01: Introduction and Literature Review

PINK1 activation can offer advantageous neuroprotective effects for potential treatments for this type of PD. Many small molecules are being tested as possible activators for PINK1 (Lambourne and Mehellou 2018).

Charcot–Marie–Tooth (CMT) disease

Charcot–Marie–Tooth (CMT) disease is a genetically heterogeneous hereditary polyneuropathy characterized by onset of distal predominant motor and sensory loss, muscle wasting, and pes cavus of feet. Diagnostic scheme for these disorders is not complete. Next-generation sequencing (NGS) along with multigene testing panels has led to effective genetic testing for accurate prognosis and genetic counseling. Correct diagnosis has enabled therapies in some cases (Hoyle et al. 2015).

CMT disorders usually follow autosomal dominant inheritance, but X-linked and autosomal recessive forms of disorder also exist. Autosomal recessive inheritance is less frequent in this disorder (Hoyle et al. 2015).

Heterozygous mutations in MPZ (OMIM 118200), DNM2 (OMIM 606482), LITAF (OMIM 601098), MFN2 (OMIM 609260), GJB1 (OMIM 302800), PMP22 (OMIM 118220), INF2 (OMIM 614455), DNM2 (OMIM 606482), YARS (OMIM 608323), KIF1B (OMIM 118210), and GNB4 (OMIM 615185) are associated with dominant forms of CMT whereas pathogenic mutations in JPH1 (OMIM 605266), LRSAM1 (OMIM 610933), MME (OMIM 120520), MFN2 (OMIM 608507), NEFL (OMIM 162280), and GDAP1 (OMIM 606598) are associated with both dominant and recessive forms of CMT. GDAP1 encodes an integral membrane protein of the outer mitochondrial membrane expressed in the nervous system, especially in Schwann cells. Dominant and recessive mutations in this gene cause dominant (OMIM 608340) or recessive (OMIM 214400 ) forms of CMT disease (Niemann et al. 2005)

Variations in SH3TC2 (OMIM 601596), MED25 (OMIM 605589), LMNA (OMIM 605588), KARS (OMIM 613641), COX6A1 (OMIM 616039) and FGD4 (OMIM 609311) are associated with recessive forms of disorder. Biallelic mutations in Frabin encoding gene FGD4 are responsible for autosomal recessive CMT4H demyelinating neuropathy (Houlden et al. 2009).

17

Chapter 01: Introduction and Literature Review

FGD4 is a member of a family of Cdc42-specific guanine nucleotide exchange factors (GEFs) and has crucial role in actin reorganization which is important for cell shape changes and formation of membrane projections like filopodia and lamellipodia (Nakanishi and Takai 2008).

Cerebellar atrophy, neuropathy and vestibular areflexia syndrome (CANVAS)

Late-onset ataxia is not an uncommon neurological condition and postural imbalance and frequent falls are the reasons for neurological consultation. Both acquired and genetic causes of ataxia are known but up to 50% of cases diagnosed with late onset ataxia remain idiopathic e.g idiopathic late-onset cerebellar ataxia (ILOCA) (Muzaimi et al. 2004) (Harding 1981). Notably, these idiopathic cases often show a multisystemic involvement: peripheral neuropathy can frequently be observed in cases with ILOCA (Abele et al. 2002), and vice versa. Cerebellar impairment and peripheral neuropathy can be associated in cases of idiopathic bilateral vestibulopathy (Kirchner et al. 2011), suggesting that existence of a common underlying aetiology.

A spectrum of clinical signs ranging from pure ILOCA and including combined degeneration of the cerebellum and its vestibular and sensory afferences, is named cerebellar atrophy, neuropathy and vestibular areflexia syndrome (CANVAS) (Fig.1.1). (Migliaccio et al. 2004). CANVAS is an adult-onset slowly progressive neurologic disorder characterized by imbalance, sensory neuropathy (neuronopathy), and bilateral vestibulopathy (Szmulewicz et al. 2016). CANVAS frequently presents as a sporadic disease. However, it has occasionally been reported in multiple siblings, raising the possibility of recessive transmission. Initial attempts to identify the underlying genetic defect by whole-exome sequencing were unsuccessful (Migliaccio et al. 2004).

18

Chapter 01: Introduction and Literature Review

Fig.1.1: CANVAS phenotype, a combination of three neurological conditions

19

Chapter 01: Introduction and Literature Review

Brain and Movement Disorders

The brain has three major parts, the cerebrum, the cerebellum, and the brain stem. Neurons and glial cells are important components of nervous system where neurons play signaling role and glial cells play a supporting role. All neurons have a cell body, and branching structures called, dendrites. Dendrites receive the information from other neurons. Axon is a thread like structure which conveys the information signal to next neuron. Brain tissue consists of gray matter and white matter. Gray matter is highly comprised of cell bodies and dendrites whereas white matter is comprised of axons (Nolte 2009).

The cerebrum is comprised of a pair of cerebral hemispheres and diencephalon (Fig. 1.2). Cerebral hemisphere is divided in different lobes; the frontal lobe, the parietal lobe, the limbic lobe, the temporal lobe and the occipital lobe. These lobes contain regions for motor, somatosensory, auditory, visual and emotional responses. The diencephalon is further divided into thalamus and hypothalamus. The thalamus conveys sensory information to the cortex and the hypothalamus is involved in controlling the autonomic nervous system (Nolte 2009).

The cerebellum consists of vermis and cerebellar hemispheres but anatomically is divided into three different lobes. The anterior lobe has important role in coordination of trunk and limbs. The flocculonodular lobe, which is comprised of vermis region, is involved in control of balance and ocular movements. The posterior lobe coordinates voluntary movements (Marsden and Harris 2011).

The part of the brain which joins the brain to the spinal cord is called brainstem. Brainstem is comprised of midbrain, the pons and the medulla (Nolte 2009). The basal ganglia is a bundle of nuclei which are clusters of neuronal cell bodies, embedded in each cerebral hemisphere. The nuclei present in diencephalon is subthalamic nucleus and that in midbrain is the substantia nigra. The primary role of basal ganglia is in motor control (Lanciego et al. 2012).

20

Chapter 01: Introduction and Literature Review

Cerebellum and its connections are mostly affected in ataxic patients. Patients may fall down frequently due to an unsteady gait. Speech and ocular movement are affected in many cases. In patients with vermian atrophy lower limbs are most affected (Marsden and Harris 2011).The oculomotor apraxia in ataxic patients is associated with marked loss of Purkinje cells and peripheral nerve fibers (Taroni and DiDonato 2004). Congenital cerebellar hypoplasia has been associated with Cayman ataxia which signifies the anatomical importance of this region (Akbar and Ashizawa 2015).

Abnormal functioning of the basal ganglia, which helps control coordination of movements results in dystonia. Basal ganglia controls the speed of movement and restricts unwanted movements. Dystonia patients may experience uncontrolled twisting, unwanted movements or abnormal postures. Parkinson's disease involves neurodegeneration in the part of the brain called the substantia nigra. NBIA phenotypes usually result from iron depositions in substantia nigra and globus pallidus of basal ganglia (Fig.1.3). These nerve cells die or become functionally compromised leading to less production of dopamine which results in respective disorders (Neychev et al. 2011).

21

Chapter 01: Introduction and Literature Review

Fig. 1.2: Human Brain. Different parts of the brain. Lobes of cerebral hemisphere are shown (Source: http://www.nature-education.org/brain.html)

22

Chapter 01: Introduction and Literature Review

Fig. 1:3. Coronal section of the brain showing the basal ganglia: Parts are shown in specific colours. Depositions of iron in these regions cause dystonia, PD and NBIA) (Source: https://openi.nlm.nih.gov/detailedresult.php?img=PMC2883978_1744-8069-6-27- 1&query=&req=4&simResults=PMC2883978_1744-8069-6-27-1&npos=1)

23

Chapter 01: Introduction and Literature Review

Mechanisms of movement disorders

Organelle dynamics is a very important aspect for normal physiology of cell. Perturbations in this aspect lead to different disorders. Mechanisms leading to ataxia are diverse and some of these can be because of defective mitochondrial or nuclear proteins. Disruption of the quality control mechanisms of cell can also contribute to movement disorder specific phenotype.

ARSACS is caused by loss of function mutations in SACS. It encodes a modular protein sacsin, essential for normal mitochondrial network organization. SACS knockdown cells show alteration in oxidative phosphorylation and expression of oxidative stress genes (Bradshaw et al.). Comparative mitochondrial bioenergetics studies have demonstrated elevated levels of superoxide in patient cells with reduced levels of sacsin. Dynamin- related protein 1 (Drp1) mediated mitophagy is decreased in sacsin knockdown cells and ARSACS fibroblasts thus compromising mitochondrial health. Neurons with reduced sacsin have impaired mitochondrial quality control which results in ataxia phenotype (Bradshaw et al. ; Grieco et al. 2004).

MARS2 is mutated in autosomal recessive spastic ataxia with leukoencephalopathy (ARSAL) patients. There is reduced mitochondrial Complex I activity, reduced cell proliferation rate and increased reactive oxygen species (ROS) which contribute towards this ataxia (Bayat et al. 2012) Mutations in KIAA0226 which encodes the protein rundataxin, involved in vesicular trafficking and signaling pathways, cause childhood onset gait and limb ataxia with dysarthria (Assoum et al. 2010). Many pathogenic spastic movement disorders are due to variants in genes which control membrane trafficking and organelle architecture. Abnormalities in these pathways may cause axonopathies of corticospinal neurons. Many genetic defects in genes associated with heterogeneous functions in cells have been associated with spastic paraplegia. This aspect expands the pathomechanism of these disorders (Fig.1.4) (Blackstone et al. 2011) (Blackstone 2012; Soderblom and Blackstone 2006; Yagi et al. 2016).

24

Chapter 01: Introduction and Literature Review

Fig.1:4. A neuron and different genes associated with spastic paraplegia (Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5584382/),The genes encode important products for neuronal function and axonal protection. These are involved in membrane trafficking and organelle architecture. Pathogenic variants in these genes lead to spastic paraplegia.

25

Chapter 01: Introduction and Literature Review

Importance of molecular testing and treatment options

The exact diagnosis by appropriate genetic testing for ataxia patients is very important as the identification can help to treat some forms of the disease which include abetalipoproteinaemia, ataxia with vitamin E deficiency, cerebrotendinous xanthomatosis, Refsum disease, Niemann-Pick disease and Wilson disease. A small group of metabolic, inflammatory, hereditary, and immune mediated cerebellar ataxias are responsive to targeted therapies (Ramirez-Zamora et al. 2015).

Treatment options for dystonia and PD are usually common. Anticholinergic medications, levodopa and deep brain stimulation (DBS) are treatment options for both of these disorders, yet the stimulation target for DBS may be different for both. Muscle relaxants or antispastic agents and botulinum toxin injections are also used to treat dystonia. Some cases of NBIA are being managed adequately by administration of botulinum toxin so it is important to diagnose the disorder properly (Do et al. 2018). Molecular testing improves the prognosis of PD patient and helps in proper management of depression, associated secondary cardiovascular complications and osteoporosis (Fayyaz et al. 2018). Many patients report the benefits of complimentary therapies such as yoga and meditation if advised in time.

Genetic counseling can play an important role in reducing the frequency of recessively inherited genetic diseases in highly inbred populations. Currently, genetic heterogeneity of a specific disorder and overlapping symptoms in different movement disorders makes this task difficult due to the cost involved in testing the involved genes. Massively parallel sequencing technology will make genetic diagnosis more affordable and available in the near future. This will probably enable carrier testing and counseling possible in a timely fashion.

Methods for genetic research on movement disorders

Different methods have been employed to investigate the genetic causes of movement disorders. These include sib-pair analyses, linkage analyses, whole genome scanning, genome wide scan association studies, candidate gene approaches, and whole exome sequencing (Ezquerra et al. 2011b).

26

Chapter 01: Introduction and Literature Review

In linkage analysis, genetic region responsible for the disorder is identified by employing different short tandem repeat (STR) markers. The segregation of these multiallelic markers, also called microsatellites, is analyzed in the family without knowing the candidate gene, or molecular pathway of disorder. After the region is identified, targeted sequencing of the specific genes located in that region is completed by Sanger sequencing, and causative gene mutation is identified (Kwon and Goate 2000).

Uniformly spaced microsatellites are genotyped in several unaffected and affected family members of the family to find the segregation of any marker. Microsatellites marker close to disease locus co-segregate with disease status and the genes in that region can be sequenced to identify the pathogenic variant. This approach can be used to identify the locus of the gene, even if not the variant. Linkage analysis was employed to establish the linkage of Charcot-Marie-Tooth neuropathy to the Duffy locus on 1 (Bird et al. 1982) .

In candidate gene approach, genetic disorders are identified by selecting candidate genes due to their potential role in disease association. The candidate genes are tested in the affected family and all the coding exons of these candidate genes are checked in the family to find out the causative mutation. The chances are higher to find the genetic cause if it is used in combination with linkage analysis. Pathogenic recessive mutations c.1367T>A p.(Leu456Ter) and c.1370T>C p.(Leu457Pro) in SIL1 responsible for Marinesco– Sjögren syndrome were identified by sequencing all the exons of potential candidate genes (Anttonen et al. 2008b)

SNP (Single Nucleotide Polymorphism) genotyping is a genome wide linkage analysis approach (performed using a DNA chip array) which is used to simultaneously assay hundred thousands of SNPs for each individual in affected families to map the disease loci (Ezquerra et al. 2011a). Genome-wide human SNP array followed by candidate gene sequencing of individuals with dominant ataxia mapped the disease to spinocerebellar ataxia 12 locus. The region contains PPP2R2B, but no variant was identified in exonic or promoter region of this gene, suggesting the involvement of another gene variant responsible for disorder (Sato et al. 2010).

27

Chapter 01: Introduction and Literature Review

The traditional methods of linkage analysis and candidate gene approach are increasingly replaced by next generation sequencing (NGS) or massively parallel sequencing (MPS). This is an efficient way of high throughput sequencing to unravel the underlying genetic cause of different genetic diseases by whole-exome sequencing (WES) or whole genome sequencing (WGS) (Yang et al. 2013). Massively parallel sequencing makes it possible to sequence the whole genome or all exome of an individual at a reasonable cost and speed. For dystonia, pathogenic variants in known disease associated genes as well as novel disease associated genes have been identified by this approach. This has contributed towards the better understanding of pathophysiological mechanisms leading to dystonia. HPCA and COL6A3 are the genes which first associated with recessive forms of isolated dystonia with the help of NGS (Domingo et al. 2016).

Whole-exome sequencing has some drawbacks that it may skip, or inadequately cover the high GC rich exons and it also does not reveal the mutations in regulatory regions which results in failure to identify the genetic cause of disorder (Schneeberger 2014).

Movement Disorders in Pakistan

Preliminary research work has been conducted on movement disorders in Pakistan. The rate of consanguineous marriages is as high as 60% and therefore there is a great probability of families to be affected with recessively inherited disorders (Hussain and Bittles 1998).

Different movement disorders have been described in Pakistan. Spastic paraplegia caused by bi-allelic mutations of ALS2 have been reported in Pakistani families (Daud et al. 2016; Gros-Louis et al. 2003). A unique movement disorder with crawling gait, dystonia, and limited speech was described in a Pakistani family with yet unidentified genetic cause (Arif et al. 2011). Bi-allelic mutations in MRE11A are associated with ataxia talengiectasia like disorder 1 (OMIM 604391) and homozygous c.1879C>T;p.(Arg633Ter) mutation in this gene was identified in consanguineous Pakistani family with ataxia and cerebellar vermis hypoplasia (Chaki et al. 2012). An OPA3 mutation has been identified in a Pakistani family suffering with complex syndrome with chorea, cerebellar ataxia, dystonia and pyramidal tract signs (Arif et al. 2013). Spastic paraplegia and psychomotor retardation with or without seizures (OMIM 616756) is the disorder which has been associated with recessive mutations in HACE1.

28

Chapter 01: Introduction and Literature Review

Bi-allelic loss-of-function truncating and insertion mutations in HACE1 were identified in two families associated with severe recessive neurodevelopmental disorders. HACE1 is an E3 ubiquitin ligase which regulates the activity of cellular GTPases (Hollstein et al. 2015). Bi-allelic novel mutations were identified in SACS associated with atypical ARSACS characterized by cognitive symptoms, peripheral neuropathy and epilepsy in two Pakistani inbred families (Ali et al. 2016). An unrelated disorder, is caused due to bi-allelic mutations in TBC1D23 resulting in pontocerebellar hypoplasia, type 11 (OMIM 617695). Homozygous truncating variants in TBC1D23 were reported in Pakistani patients who had microcephaly, severely delayed psychomotor development, poor speech, poor ocular contact, delayed walking, spastic hypertonia, cerebellar syndrome, and motor weakness (Ivanova et al. 2017).

However, these findings in no way explain the complexity, or the large number of observed cases of movement disorders. Therefore, this clinical and genetic heterogeneity of movement disorders demands further research to identify other genes and their variants associated with different movement disorders. This basic information will ultimately improve the understanding of the pathogenesis of the disease and may suggest possible treatment options in future. This molecular characterization of movement disorders will provide a platform for better prognosis, genetic counseling and therapeutic option for these disorders (Lunati et al. 2018).

29

Chapter 02: Materials and Methods

Chapter 02: Materials and Methods

30

Chapter 02: Materials and Methods

Institutional Review Board Approval

This study was conducted after approval by Institutional review Board at the School of Biological Sciences, University of the Punjab, Lahore Pakistan. Written informed consent was obtained from each adult participant, and from parents on behalf of their minor children.

Recruitment of Families

Sixteen families were recruited from different areas of Punjab (Fig 2.1). The families were identified through personal contacts and by visiting neurology departments of different hospitals. The patients in the families suffered from recessively inherited forms of movement disorders. Patients were clinically evaluated by qualified neurologists. All required clinical investigations were performed for the patients. Videotaping was completed for patients and some unaffected members of the families. Blood samples were drawn from the willing participants. Families were visited a few times to ensure the relationships, request clinical investigations, or extend sampling.

Sporadic cases

Some sporadic cases were also collected in order to provide them with molecular diagnosis and possible counseling in future.

Control Sample Screening

Blood samples were collected from more than 100 control individuals who had no family history of any disorders including dystonia, ataxia or other movement disorders. All control samples were ethnically matched to the participating families.

31

Chapter 02: Materials and Methods

Fig. 2.1: Map of Pakistan. All provinces are colored differently. Some cities are also mentioned in different provinces. Participating families in this study were collected from different areas of Punjab

32

Chapter 02: Materials and Methods

Clinical Evaluation

Neurological and physical examination was completed for all patients. All required clinical and radiological investigations were performed for at least one affected individual from the participating families. Patients were directly evaluated by neurologists and expert clinicians from Pakistan. Dr. Iqra Tariq, Dr. Rashid Imran, Dr. Shahid Mukhtar and Dr. Jalil ur Rehman from Allama Iqbal memorial Hospital, Sialkot and Lahore General hospital, Lahore were the consulting doctors. a. Videotaping

Videos of affected individuals and some unaffected family members were obtained for identification of neurological signs, made according to a standard protocol (de Leon et al. 1991) (Appendix A-1). Videos also included sitting, standing and walking patterns. The upper and lower limbs were focused in these videos whereas voice, gait, eye-blinking, and voluntary movements were also recorded. Some of these videos were then shown to movement disorder experts Dr. Henry Houlden and Dr. Thomas Bourinaris, University College London, UK for diagnosis of different movement disorders. b. Electromyogram and Nerve Conduction Studies

Nerve Conduction Studies (NCS) are a way to measure speed and potential of electrical activity is in a nerve which is reduced in case of nerve damage. Electromyography (EMG) measures how well the muscles respond to those signals. EMG and NCS studies help to diagnose neuromuscular diseases, identify problems in the spine, and neural problems elsewhere in the body. Peripheral nerve problems in arms or legs can also be diagnosed with these tests. c. Magnetic Resonance Imaging

Magnetic Resonance Imaging (MRI) detects anatomical abnormalities of the brain. This helps in diagnosing lesions, neurinomas, atrophy, and edemas, in all brain parts. Expert opinions of radiologists were taken for these MRI scans.

33

Chapter 02: Materials and Methods

d. Base line investigations

Baseline investigations, which varied from case to case; complete blood counts, calcium, vitamin D levels were performed as required,. Copper and ceruloplasmin levels were determined in some patients. Serum alpha fetoprotein levels were determined for some cases of ataxia as advised by the clinicians.

Blood Samples

Blood samples were obtained in BD Vaccutainer® tubes containing EDTA (Becton, Dickinson and Company, NJ, USA). 5ml of blood was withdrawn from all participants by an expert phlebotomist. The blood samples were transported at room temperature and stored at 4°C. Samples were processed for DNA extraction. For samples which were to be processed for RNA, 1ml of the blood sample was mixed in 3ml of TRI Reagent® (Molecular Research Center, Inc., Cincinnati, OH, USA) and was transported at room temperature and were processed on the same day, for RNA extraction.

DNA Extraction from Blood

Standard method was used for extraction of genomic DNA. This method involved steps of cell lysis, proteinase K digestion, salting out, and isopropanol precipitation (Grimberg et al., 1989; Miller et al., 1988). The steps are described as follow:

 5ml of blood was decanted into a 50 ml Falcon® tube with an equal volume of chilled sucrose lysis buffer (Table 2.1). 10ml of cold, autoclaved, distilled water was also added. It was mixed by inversion of tube followed by incubation on ice for next 10-15 minutes.

 The tubes were centrifuged at 3500 rpm for 10-15 minutes at 4°C (Eppendorf® 5804R, Eppendorf, Hamburg, Germany). Reddish supernatant was discarded in 10% bleach solution and the cell pellets were re-suspended in a mixture of lysis buffer with water in 1:3 ratio. Centrifugation was done again, supernatant was discarded and washing was repeated one or two times, until creamy white pellets were obtained.

34

Chapter 02: Materials and Methods

 Protein from the lysed cells was digested by adding 5ml of TEN buffer (Table 2.2), 500µl of 10% SDS (Sodium dodecyl sulfate) and 25µl of proteinase K (20mg/ml) solution (Thermo Scientific®, Thermo Fisher Scientific Inc., Waltham, Massachusetts, USA).

 The cell pellets were vigorously re-suspended followed by incubation at 45°C in a water bath overnight.

 Next day, the samples were taken out from the incubator and were normalized to room temperature. Proteins were precipitated by addition and mixing of 4ml of 5.3M NaCl solution in each sample.

 The centrifugation was done at 4500rpm for 15-20 minutes at 4°C and supernatant was collected in a 15ml Falcon® tube. Pellet containing denatured proteins was discarded.

 The supernatant tubes were re-centrifuged to make sure the clarity, and removal of any residual protein which was left over from the last step. The clear supernatants were transferred into a new 50ml Falcon® tube.

 10ml of chilled isopropanol was added in each sample, and tubes were gently inverted to precipitate the DNA. Precipitated DNA became visible in the form of white thread-like strands.

 The visible threads of DNA were precipitated by centrifugation. DNA was washed with 1ml of 70% ethanol. The ethanol was discarded and the pellets were dried at room temperature.

 The dried DNA pellets were re-suspended in 300µl of TE with low EDTA concentration (Table 2.3). nucleases were inactivated by final heating of extracted DNA at 70°C in a water bath for 1 hour .

 DNA was stored at -20°C.

35

Chapter 02: Materials and Methods

Agarose Gel Electrophoresis

Successful extraction and good quality of DNA was confirmed by agarose gel electrophoresis. A 0.8% agarose gel was prepared in 0.5X TAE buffer (Table 2.4) with 0.5µg/ml ethidium bromide in a final concentration (Table 2.5). The genomic DNA samples were loaded on gel with help of 6X bromophenol blue loading dye (Table 2.6) and were electrophoresed. Control genomic DNA, of known concentration, was also run to estimate the concentration of each sample. The DNA was visualized using a UV transilluminator.

DNA was quantified using NanoDrop (Thermo Fisher Scientific Inc.). The DNA concentrations was quantified in ng/µl. The 260/280 ratio was also monitored to ensure purity of DNA and its value around 1.8 is generally acceptable for purity of DNA.

Blood RNA Extraction

RNA was extracted using TRI Reagent® (Molecular Research Center, Inc., Cincinnati, OH, USA) according to the manutacturer’s protocol. 1ml ot blood and 3ml ot TRI Reagent® was mixed in a 15ml Falcon®. It was mixed gently by inverting three to five times followed by vortexing. 600µl of chloroform was added and mixed until it turned milky. The tubes were kept at room temperature for 5 minutes followed by centrifugation (Eppendorf® 5804R) at 3500rpm for 15 minutes at 4°C. Three layers were observed immediately after centrifugation. The upper clear layer was shifted carefully into a new falcon. 5ml isopropanol was added to the each sample and the tubes were incubated at room temperature for 10 minutes. RNA was precipitated by centrifugation of tubes at 3500rpm for 15min. Pellet was formed on one side of the tube and was Resulting pellet was washed with 75% ethanol followed by re-suspension in RNase free water. RNA quality was checked by gel electrophoresis on a 1% agarose gel. NanoDrop (Thermo Fisher Scientific Inc.) was used to quantify the RNA and the required amount was used for downstream applications.

36

Chapter 02: Materials and Methods

cDNA Synthesis cDNA library was constructed using 1µg of RNA using RevertAidTM Premium First Strand cDNA Synthesis Kit (Thermo Fisher Scientific Inc.) according to the manufacturers protocol. Prior to cDNA synthesis, DNase treatment was done in a reaction volume of 10µl containing, 1µg of RNA, 1µl of Reaction Buffer and 1U of DNase I-RNase free (Thermo Scientitic™)Incubation ras pertormed at 3lah tor halt an hour. Dcase treated RcA . ras then reversetranscribed using 100µM dNTPs, 5µM random hexamer primer, 1X Reaction Buffer, 200U of RevertAidTM M-MuLV Reverse Transcriptase (RT), and 20U of RiboLockTM RNase Inhibitor. Reaction was incubated at 25°C for 5 minutes followed by at 42°C for 60 minutes. The cDNA was stored at -20°C or processed for downstream applications.

Molecular Analysis

Karyotyping, Copy number variation and Clinical exome panel sequencing +DEL/DUP for proband of (RDHT04) were carried out in UK clinical center and hospital.

Whole-Exome Sequencing

Whole-exome sequencing was performed for one affected individual each, for families RDHT04 and RDHT05 by using Agilent v5, 51Mbp (hEx-Av5) enrichment kits. Exome libraries were sequenced on an illumina HiSeq 2000 machine at 50X coverage (Otogenetics, USA). The data was uploaded to the DNAnexus server (http://dnanexus.com) and reads were mapped to the Build GRCh37/hg19 of UCSC Genome Browser. The variants were called by conducting Nucleotide level Variation analysis at DNAnexus and the results were exported in .csv format.

Whole-exome sequencing for one individual each of families RDHT02, RDHT06, and RDHT07 was performed using Agilent V4 enrichment kit (Agilent Technologies, Santa Clara, CA, USA). Paired-end reads were obtained at 100X coverage on an Illumina Hi- Seq 2500 sequencer (Macrogen, South Korea). The 90 bpend reads rere aligned to the -pair .Guild hRhh3lehg1S ot UhSh henome Grorser

37

Chapter 02: Materials and Methods

Samples from nine families were subjected to exome sequencing in collaboration with the UhL Institute ot ceurology, University hollege London, London. One individual’s DcA was processed from famlies RDHT01 RDHT11, and RDHT12 whereas DNA from two affected individuals were processed from families RDHT03, RDHT08, RDHT09, RDHT10, RDHT14, and RDHT16.WES was performed on Illumina Hi-Seq 2500 sequencer (Macrogen, South Korea). The reads were mapped to the Build GRCh37/hg19 of UCSC Genome Browser.

Data Annotation

Exome data was annotated by using wANNOVAR (http://wannovar.usc.edu/). Since the mode of inheritance was recessive, and most of the families were consanguineous, only homozygous variants were considered initially. Data was analyzed and filtering was done against all known variants in the 1000 Genomes (http://www.1000genomes.org/), dbSNP database (http://www.ncbi.nlm.nih.gov/SNP/), Exome Sequencing Project (http://evs.gs.washington.edu/EVS/) and Exome Aggregation Consortium (http://exac.broadinstitute.org/). Variants were filtered so that those with an allele frequency equal to or more than 0.01 in any public database were excluded. Variants located in the exons, and those predicted to affect splicing were considered. Synonymous variants were excluded. Variants were prioritized on the basis of being missense, frameshift, in-frame deletions, in-frame insertions, nonsense, creating stop loss and affecting splice sites. The pathogenicity scores predicted by CADD, FATHMM and PROVEAN were accessed from the ANNOVAR analysis file. In addition, REVEL scores (https://sites.google.com/site/revelgenomics/) and ClinPred scores (Alirezaie et al. 2018) were determined for each missense variant. Regions of autozygosity in the whole exome data were determined by using the software Agile VCF Mapper (http://dna.leeds.ac.uk/agile/AgileVCFMapper/) (Carr et al., 2013). Synonymous variants in these autozygous regions were also considered.

The conservation of variants in respective orthologues was checked through homologene (https://www.ncbi.nlm.nih.gov/homologene), UCSC multizalign (http://genome.ucsc. edu/) and by aligning the protein sequences from different species using Clustal Omega (https://www.ebi.ac.uk/Tools/msa/clustalo/). Compound heterozygous variants were also considered if pathogenic homozygous variants were not identified in disease causing genes in exome data

38

Chapter 02: Materials and Methods

Designing the Primers

An online program Primer3 (v. 0.4.0) (http://bioinfo.ut.ee/primer3-0.4.0/) was used to design primers and respective DNA sequences were retrieved from UCSC genome browser (http://www.genome.ucsc.edu). It was ensured that primers were not designed within polymorphic and repeat region. The forward and reverse primers did not have a melting temperature difference of more than 1°C. In silico PCR on UCSC website was used to check primers specificity to single regions and Blat analysis was done to ensure the appropriate single amplicons.

Segregation analysis: Sanger Sequencing a. PCR Amplification

Targeted regions of genome spanning the variants of interest were amplified via polymerase chain reactions. A total volume of 20µl was used for PCR reaction containing,

1X PCR Buffer (Laboratory prepared Table 2.7) , 200µM dNTPs, 2-4mM MgCl2, 0.1U of Taq polymerase (Thermo Scientific®, Thermo Fisher Scientific Inc.), 6pico mole of each primer and 50ng of DNA. Standard PCR or touchdown PCR program on MyCyclerTM

Thermal Cycler (Bio-Rad Laboratories, Inc., California, USA.) were performed according to the required program. Standard PCR program was, 94°C for 2 minutes, 36 rounds of three subsequent temperatures, denaturation at 94°C, annealing at a temperature regarding the Tm of primers, and extension at 72°C. A single final extension step at 72°C for 10 minutes was also included in program (Fig. 2.2A). In touch down PCR, the annealing temperature was set 10 degrees higher than the final annealing temperature which decreased by 1°C at every subsequent cycle. After that, 28 cycles were provided for amplification and the annealing temperature was set according to Tm of primers (Fig. 2.2B). 5µl of PCR product mixed with 1µl of 6X loading dye was loaded on a 1.5% agarose gel to check the amplification of the product. PCR reactions were optimized to remove nonspecific amplification, and primer dimers.

39

Chapter 02: Materials and Methods

b. Treatment of PCR product by EXOSAP

15µl of PCR amplified products were mixed with 1µl Fast Alkaline phosphatase (FastAP) (ThermoSamples rere .(™and t.eil ot exonuclease I ( xo I) (Thermo Scientitic (™Scientitic incubated tor inactivation ot unused dcTPs and primers. The incubation was carried out at 37oC for 15 minutes followed by inactivation of enzymes for 15 minutes at 85oC. The samples were further used as such, or in some cases were diluted so that one volume of water was added to double volume of EXOSAP PCR mixture before performing sequencing PCR.

40

Chapter 02: Materials and Methods

Fig. 2.2: PCR profile for standard and touchdown conditions (A) Standard PCR profile: X indicates the specific temperature for each primer set based upon the Tm of primers which varied from 55 to 68 degrees Celsius. (B) Touchdown PCR profile usedot yfilpma AND msynfmgor. ↓smismrmgor 1 degree decrease in temperature in subsequent cycle.

41

Chapter 02: Materials and Methods

c. Sequencing PCR

Sequencing of the amplicons was done using BigDye® Terminator v3.1 cycle Sequencing kit (ABI). 10µl of the sequencing reaction was comprised of 2µl of EXOSAP treated PCR product, 0.5X sequencing buffer (ABI), 0.5µM of either forward or reverse primer and 0.5µl of BigDye® Terminator v3.1. Sequencing PCR was performed on MyCyclerTM Thermal Cycler (Bio-Rad) using the conditions: 96oC for 1 minute, 36 repeats of three steps of 96oC for 30 seconds; 55oC for 30 seconds and, 60oC for 4 minutes. The final incubation was set at 60oC for 5 minutes. d1. Precipitation

10µl of the sequencing reaction was precipitated by mixing 32µl of precipitation solution (Table 2.9) to it, followed by incubation at room temperature for 15 minutes in dark. After incubation mixture was centrifuged at 3700rpm (Eppendorf® 5804R) for 45 minutes. The supernatant was discarded carefully followed by washing of pellet using 30µl of 70% ethanol. Pellet was air dried and these dried pellets were then re-suspended in Hi-Di Formamide for subsequent sequencing. Precipitation was done for some samples, and for others the sequencing reaction was directly cleaned using sephadex before sequencing. d2. Sephadex cleaning

Some sequenced samples were processed using sephadex beads. 50ml of sephadex was prepared by gentle mixing of 2.9 gram of fine DNA grade Sephadex G-50 Bioreagent, in 40ml of autoclaved distilled water and was put at room temperature for hydration for about 30 minutes.

Purification plate was prepared by adding 350µl of the well-mixed Sephadex to each well of a horning® Filtr X™ S6 rell tilter polystyrene plates containing t.66mm glass tiber filter. Corning glass plate was put on an empty collection plate and centrifuged for 3 minutes at 750xg with proper balancing plate on other side.

Sephadex bed was established in all wells of corning plate which purifies the sequencing reaction once it is filtered through the bed. Corning plate was put onto a new plate (which goes to the sequencer) and entire volume of the sequencing reaction (10µl) from the sequencing plate was poured carefully using a multichannel pipette, onto the Sephadex columns.

42

Chapter 02: Materials and Methods

The corning plate with sequencing reaction, loaded on the sephadex columns, was fitted over the sequencer plate. It was centrifuged at 910xg for 5 minutes and filtrate was collected in the sequencer plate. The sequencing plate was heat sealed and put in the frame plate of sequencer. This was finally placed in the sequencer for sequencing. e. Sequencing

10µl of sepahdex cleaned sequencing reactions were directly used, or the precipitated pellets were dissolved in 12µl of Hi-Di formamide for final step of sequencing. Sequencing samples were loaded on an ABI 3730xl genetic analyser (Applied Biosystems, Foster City, CA, USA). Sequencing results were viewed and analyzed by using SeqMan Software (DNASTAR Lasergene) and UCSC genome browser (http://genome.ucsc.edu).

Microsatellite Markers: Genotyping

For Family RDHT03, two microsatellite markers spanning the candidate gene were selected based on their close proximity to the gene, and one was self-designed (Table 2.10). ither the torrard or reverse primer ras attached rith universal M13 tail (e′- CACGACGTTGTAAAACGAC-3′) tor indirect tlorescent labelling (Boutin-Ganache et al. 2001). Microsatellite sequences were amplified by using a standard PCR program (Fig. 2.2A) on MyCyclerTM Thermal Cycler (Bio-Rad). PCR reactions contained 1X Taq Buffer

(Taq Buffer with (NH4)2SO4 and MgCl2 Thermo,(™Scientitic0tt iM dNTPs,0.4 pmoles ot each ,primerUt.3ot Taq polymerase (Thermo Scientific®, Thermo Fisher Scientific Inc.), 4% betaine, 1µM of fluorescent labelled dye 6-FAM (M13 tail specific labeled primer .and etng ot DcA (′3-hAhhAhhTThTAAAAhhAh-FAM-6-′e)

Capillary Electrophoresis (Fragment Analysis)

PCR products labeled with 6-FAM fluorescent dye were diluted in the ratio of 1:5 of PCR product and water. The highly deionized formamide (Hi-Di FormamideTM) was used as denaturant for sample preparation. Reaction mixture for one sample contained 10.7µl of

Hi-Di formamideTM (Applied Biosystems) and 0.3µl of an internal size standard LIZ 600.

This mixture and 1µl of PCR product were mixed, spun down and were denatured at 95°C for 5 minutes, and then cooled immediately.

43

Chapter 02: Materials and Methods

Samples were subjected to capillary electrophoresis on an Applied Biosystems 3130xl Genetic Analyzer (Applied Biosystems). Analysis of the results was done using GeneMapper® software Version 4.0.

Expression of Candidate genes in Blood

For families RDHT05 and RDHT08, the expression of the candidate genes was checked in blood by PCR amplification using cDNA template. Primers were designed from two different exons so that the amplicon would span at least one intron when amplified from gDNA. Two rounds of PCR using external and then internal set of primers were used to amplify the variant containing region of the gene. For family RDHT05, external primers, 6F-GGACAGTGGTTCTGACCCCCACA and 12R-TGGCAGGCTTAGCAAGAAGCTGGA were used to amplify 903bp from cDNA. This product was used as template for next round of PCR. Two µl of the product was used as a template for a nested PCR which utilized primers 8F-TGGGCATTCCGATTTTCCAACA and 11R-TCGGCTAGCCACCTCCTGCAA.

For family RDHT08, external primers, 2FEx-TGGAGGACATCATGAAGCTG and 3REx- GGTGACGTAGTTCACCAGCA were used to amplify a 377bp fragment from cDNA. This product was used as a template for the next round of PCR. Two µl of the product was used as a template for a nested PCR which utilized primers 2FInt- ACATCATGAAGCTGCTGTGC and 3RInt-GAACCGGCTTAAACTGTCCA. Expression of candidate genes ras checked trom a control and patients’ cDcA libraries.

Population Screening

The selected variants which were segregating with disease were checked in 100 DNA samples from ethnically matched controls. Samples were screened by Sanger Sequencing and by analysis of institutional database of exome data of unrelated individuals from the same population.

44

Chapter 02: Materials and Methods

Protein Modelling

The possible structural changes induced by genetic variants in genes were characterized by modelling the proteins structures. The wild type and mutant proteins were compared in their structures for possible explanation of causing disease. The sequence of the proteins was obtained from Uniprot (https://www.uniprot.org/). The sequence was then used in Phyre2 online server (http://www.sbg.bio.ic.ac.uk/ ~phyre2/html/page.cgi?id=index) (Kelley et al. 2015) to search for homologs and determination of protein structure. Default modelling parameters were used. For visual analysis of the location of the mutation, the results were downloaded and visualized using VMD (Humphrey et al. 1996). Additional structural annotation was obtained from RCSB PDB (http://www.rcsb.org/).

Primary Fibroblast Culture

In UCL-HT01 family, compound heterozygous missense variants in FGD4 were segregating with phenotype of Charcot Marie tooth disease. Few assays were performed. to evaluate the effect of these variants at protein level.

Skin biopsy of a CMT4H patient was taken by a clinician at UCL institute of neurology and the fibroblast cells were grown from this skin biopsy. The patient and control fibroblasts were r’pglpbpldylla ndlodsml pg ll Adlvmnnt modifiedmynlm medium DMEM)+Glutamax (Thermo 31966-021) supplemented with 10% heat inactivated fetal bovine serum, FBS (Gibco 10500-064) and 1% penicillin/streptomycin (Thermo 15070- 063). The final media also contained 0.05mg/ml filter sterile uridine which was prepared in DMEM aliquot (Table 2.10). The cells were incubated at 37°C and 5% CO2 in a humidified atmosphere.

45

Chapter 02: Materials and Methods

Establishing the fibroblast culture

Skin biopsies were placed in sterile growth medium contained in a sterile falcon tube, wrapped with Parafilm M. These were kept at room temperature or refrigerated. It was preferred to process the biopsy samples soon to establish the primary fibroblast culture. Culture was established by dissection of biopsy in a 10cm sterile Petri dish in a laminar flow hood. Growth medium was removed to ease the dissection process. Biopsies were dissected in such a way that skin and subcutaneous fat tissue were separated. Small cubes of ~1mm2 were dissected and were placed, skin side up, into 5cm sterile Petri dishes that contained 1-1.5ml of pre-warmed growth medium. It was ensured that the biopsy chunks would not be floating and the surface tension of the medium would hold the skin pieces onto the plate surface in order to allow the adherence to the plate. Plate was placed in an incubator at 37°C in a humidified environment containing 5% CO2. Precautions were taken not to vibrate or shake the plate and it was left undisturbed for two days. Media volume was monitored and was filled up to desired volume if it had evaporated.

After 2 days, 100 – 400µL of pre-warmed growth medium was added without disturbing the explants. The same procedure was repeated after another 2 days. Two days after the last procedure, the plates were examined under a microscope, at which time explants showed growth of epithelial cells (small cells that grow in a single continuous layer). The epithelial cells anchored the explants to the plate surface. After another 3–4 days of growth, spindle-like fibroblasts migrations out of the explants were observed on microscopic examination. Once this stage was reached, 2ml of pre-warmed growth medium was carefully added to the plates.

Fibroblast growth and confluence was carefully monitored. After the plates were quite confluent, the cells were trypsinized using 1ml 1X Trypsin with EDTA 4Na, (Invitrogen, Cat No 25300054), and were transferred to a T75 flask for further growth. Epithelial cells were not transferred as these were more adherent to the plate. 1X Dulbecco's Phosphate Buffered Saline (DPBS), liquid, 500ml; Invitrogen, Cat No 14190-094 was used for washing of the cells when required during trypsinization and media change. For the next few weeks the culture was expanded, stored and processed further for subsequent assays.

46

Chapter 02: Materials and Methods

Cell counting assays: growth curves

Equal number of fibroblast cells were seeded in each well of six-well plates. They were allowed to grow and proliferate. The cells were trypsinized and harvested at equal intervals and were counted using hemocytometer to make the growth curve for control cells and patient fibroblasts.

Cell counting using hemocytometer

100µL of cell suspension was mixed in 100µl of 0.4% trypan blue stain (Invitrogen T10282), a composition of 1:1. This mix was kept at room temperature for about 3-5 minutes. 100 µL of mixture was placed at the edge of the cover-slip and allowed to run under it. Then grid of counting chamber was visualized under 100X magnification of TCM-400 microscope (Labomedinc., USA) and cells were counted. The middle large square was targeted as counting area, and number of cells in it was counted and designated as X1. Cells in four corner large squares were also counted (X2-X5) and finally the mean value of cells was estimated which were in one large square (X). Counting grid of hemocytometer is shown in Fig. 2.3.

47

Chapter 02: Materials and Methods

Fig. 2.3: Etched grid on the surface of hemocytometer. The cells counted in middle encircled large square were designated as X1 and in corner larger squares, one of which on upper right side is bounded by bold margins, were designated as X2-X5 respectively

48

Chapter 02: Materials and Methods

Then average number of cells in one large square was X= X1+X2+X3+X4+X5/5

Volume of suspension in one large square would be = length x width x depth of large square

So

1mmx1mmx0.1mm = 0.1cmx0.1cmx0.01cm= 0.0001cm3 (10-4cm3 or 10-4ml)

To convert mL in nl, multiply with 106

10-4x106 = 100nl (volume of sample in 1 large square)

Accordingly, number of cells in one large square is equal to number of cells in 100nl,

Number of cells in 100nl sample = X

Number of cells in 1nl sample = X/100

Number of cells in 106nl (1ml) sample (Y) = X/100 x 106

Refractile, bright and colorless cells were counted as live cells and dead cells were stained as blue, and were not counted. Number of cells in 1ml original sample (Z) was multiplied with 2 to compensate for dilution with trypan blue (Invitrogen T10282).

Z = Y x 2 (whereas Z is number of cells in 1mL of actual undiluted suspension)

Cell seeding

The counted cells were plated according to the following scheme:

A general formula 1000/Z x required number of cells was used to calculate and seed the cells in well for the growth curve or for immunofluorescence.

If 1.5x105 cells had to be seeded per well, then 1000/Z x 1.5x105 μl ot original suspension was put in one well and rest of the volume was adjusted with growth media. The seeded cells were allowed to grow for required time.

Growth Curve plotting

Cell counting of the seeded fibroblast from control sample and patient was continued at consecutive time intervals. Graph was plotted with time on x-axis and cell count on y-axis.

49

Chapter 02: Materials and Methods

Immunofluorescence/ Confocal Microscopy

The major steps for immunofluorescence included the following

Fixation:

The fibroblasts cells carrying the normal and mutant FGD4 were grown on the sterilized coverslips in a 24 well plate. The media was decanted and cells were washed briefly with 1X PBS (Invitrogen Cat No 14190-094). The cells were fixed for immunofluorescence with a fixative solution of 4% formaldehyde in 1X PBS (Table 2.11). Fixative solution was removed after 20-30 minutes, and the cells were washed with PBS three times for 5 min each. The cell culture plates were covered properly with aluminum foil and kept at 4 °C till processing for immunofluorescence.

Blocking:

The fixed cells were blocked for 1 hour with 10% goat serum and 0.01%triton X-100 in 1X PBS (Table 2.12) at room temperature. After blocking, the fixed cells were washed with 1X PBS and processed for subsequent immunostaining.

Immunostaining:

Primary antibody, Anti-FGD4 antibody (ab97785) was diluted to the factor (1:150) in blocking solution and fixed cells were incubated in primary antibody dilution O/N at 4°C on a shaker.

After primary antibody, the cells were washed thrice with 1X PBS and were incubated with secondary antibody anti mouse raised in goat (Invitrogen 488 A21042), used at a dilution of 1:1000, for 1 hour at room temperature. After secondary antibody, cells were washed once with 1X PBS and then were incubated with 0.1µg/ml of DAPI in 1XPBS at room temperature for 15 minutes. Cells were washed once with 1X PBS. Cells were incubated with 1:2000 dilution of Alexa Fluor 647 Phalloidin in 1X PBS for 1 hour at room temperature.

50

Chapter 02: Materials and Methods

Three consecutive washings of cells were performed with 1X PBS for five minutes each.

The mounting slides were cleaned and were labelled. A small drop of mounting media was added to the slide, and then the coverslip of stained cells was carefully handled with a forceps. It was blotted on a blotting paper to remove excess PBS, and was mounted on the slide in a way that stained cells were facing down on the mounting media. Extra mounting media was cleaned from edges and slides were dried for next two hours before microscopy.

Microscopy:

The immunostained cells were visualized under appropriate filters of confocal microscope and images were taken for control and patient cells. Zeiss 710 LSM with an integrated META-detection system was used from the core facility to image the cells.

Western blotting

Western blotting was performed to estimate the qualitative and quantitative expression of protein in control versus patient cells. Major steps are listed below:

Cell lysis and protein extraction

 RIPA lysis buffer was prepared and was supplemented with proteinase inhibitor, 1 tablet of proteinase inhibitor was added into 10ml of RIPA buffer and was used subsequently (Table 2.13).  The fibroblasts were trypsinized, poured into falcon tubes and were harvested by centrifugation at 1000xg for 5 minutes. Trypsin containing media was discarded and pelleted fibroblasts were resuspended in 1ml of ice cold D-PBS (Dulbecco PBS without Ca+2 and Mg+2) and transferred to Eppendorf tube. Tubes were centrifuged at 1000 g for e min at 4ah. Supernatant ras aspirated ott and 1ttμl ice cold RIPA lysis buffer (+ proteinase inhibitors) was added to the pellet. Pellet was vortexed and was incubated on ice for 30 mins (vortexing every 10 mins). Tubes were centrifuged at 14000xg for 15 min at 4°C in a temperature regulated centrifuge. The supernatant was carefully poured into a new tube, and was put on ice whereas debris was discarded as the pellet.

51

Chapter 02: Materials and Methods

Protein quantification:

Bradford assay/BCA was performed to quantify the total protein in each of the cell lysate to ensure equal loading for western blotting. BSA standard curve was prepared in triplicate. Standard curve control samples of required concentration were prepared by mixing water, RIPA buffer and BCA stock (Table 2.14). Later, 10µl of each of the standard curve sample was loaded in wells of an ELISA plate.

The experimental samples were also loaded in triplicates after the standard curve samples.

GhA reagent A and reagent G rere mixed in et:1 ratio. 0ttμl ot this mixture ras added to each well of ELISA plate. Each of these contained either the standard curve, or experimental samples. Plate was incubated for 1 hour at 37°C and then was read in an ELISA reader machine at 595nm absorbance.

Standard curve was plotted by plotting mean absorbance on x-axis and BCA concentration on y-axis. The absorbance of experimental samples was plotted to quantify the protein concentration in all cell lysates and equal amount of protein was loaded in SDS gel for western blotting.

SDS gel electrophoresis

 50ml of 20X TRIS-acetate running buffer was added into 950ml of ddH2O and was poured in SDS gel apparatus (Table 2.15).  4-12 % polyacrylamide precasted gel was taken and comb and bottom white sealing strip were removed before loading into running container (wells facing inwards). The central column of apparatus was filled with more TRIS-acetate running buffer. Gel wells were washed with running buffer using a syringe and apparatus was left to run at 100 V for 10 min. After this time, 10 wells were washed again.  The loading mixture for each sample was made in such a way that 1µl of that should contain 1ig ot total protein. eeμg ot protein (based on Gradtord assay results) ras taken and appropriate volume of RIPA buffer was added to make the volume up to 4t.lμl .Later, t.eeμl ot 1M DTT and 13.leμl ot 4X LDS loading dye rere added (Table 0.16). All samples were votexed and were heated at 70°C for 10 minutes.

52

Chapter 02: Materials and Methods

 0eμl ot cell lystaes ras loaded in each rell and each lysate ras loaded in duplicate. 10.eμl of prestained protein standard Sea Blue plus 2 was loaded in first or last well and gel was run at 70V, for 10 mins after which it was run at 100V for 2.5 hours or until the protein standard reached the bottom.

Transfer onto membrane

 2L of TRIS/GLY transfer buffer was prepared (Table 2.17).  SDS transfer apparatus was taken and everything (whatmann filter paper, sponges and nitrocellulose membrane) were soaked in chilled TRIS/GLY transfer buffer before proceeding.  The transfer sandwich was prepared in order from black to red side. Two soaked sponges and two pieces of soaked whatmann filter paper were put on the black side of transfer sandwich which was on the negative side of the power supply so that protein would transfer from negative to positive; from gel to membrane. SDS gel was carefully removed from the castor plates and was put over these filter papers. Soaked nitrocellulose was taken and was carefully placed over the gel in a way that no bubbles were formed or retained between gel and membrane.  Two soaked filter papers and then two soaked sponges were placed over the membrane and sandwich was locked. It was placed in the transfer container in a way that the black side was towards the black side of container. The transfer chamber was filled with transfer buffer and was run at 35V for 2 hrs, in a cold room.

Blocking, Primary and secondary antibody incubation

Membrane was removed, was labelled on the protein side with a pencil. It was washed with 1X PBST (Table 2.18). Membrane was stained temporarily by adding ponceuos stain to confirm good protein transfer.

 Membranes was washed with 1X PBST three times (3 x 10 mins).  Membrane was blocked with 5% w/v skimmed milk in 1X PBST (Table 2.19) for 1h at room temperature in a falcon tube on a rotor.  Membrane was washed once again with PBS-T.

53

Chapter 02: Materials and Methods

 1% w/v milk PBST was prepared and primary monoclonal Frabin (43): sc-136333 was diluted in it in a ratio of 1:1000. The membrane was incubated in 5 ml of primary antibody and was put on a roller overnight at 4°C. Next morning, membrane was washed three times with PBS-T for 10min each. (3 x 10 min).  Membrane was incubated for 1 hour at room temperature, in 5ml of secondary antibody dilution prepared in 1% milk in 1X PBST. Secondary antibody conjugated with anti- mouse HRP was diluted in a ratio of (1:10000). Membrane was washed again three times with PBST for 10min each (3 x 10 min).  Membrane was kept in PBS to prevent drying, and was developed immediately.

Developing the membrane

 Bio-Rad Chemidoc Touch developer machine was turned on. 1ml of developing solution A was mixed with 1ml of developing solution B and was added all over the membrane. Membrane was incubated for 2 mins at RT  Excess developing solution was tapped off. The membrane was wrapped in cling film  Membrane was placed in the system and signal was recorded.  The membrane was washed three times with 1X PBST for 20 minutes each. Membrane ras then sublected to incubation rith anti β-actin antibody for 1hr, was washed three times with 1X PBST, and was incubated with the secondary antibody. It was washed again with 1X PBST and then signal was recorded. This step was an additional step to ensure equal loading of protein in experimental samples.

54

Chapter 02: Materials and Methods

Biallelic AAGGG repeat in gene RFC1 expansion in CANVAS patients

23 patients from 11 families and 33 sporadic cases of CANVAS patients were recruited in this study to identify the possible recessively inherited genetic cause of the disorder. A combination of non-parametric linkage analysis and genome sequencing identified a recessive intronic AAGGG repeat expansion in the RFC1 as a common cause late-onset ataxia. The recessive repeat expansion showed full segregation in 23 cases from 11 families and 21 out of 33 additional sporadic cases screened (collaborative research).

Work performed as part of this thesis

A large cohort of normal population (n=304) was screened to identify the frequency of this expansion in the normal population for validation of the preliminary results. Short range and long range flanking PCR along with reverse-primed PCR (RPPCR) was employed to screen the individuals for the expansion and also to confirm one expanded allele in carriers and biallellic nature in patients. All the control samples along with the patients and the carrier samples were amplified using the primers spanning that repeat region employing short range PCR. Long range PCR was employed (Table 2.20) to amplify the region in carriers and patients. The products were then analysed by Sanger sequencing

Standard Short-range PCR

All samples were processed through short-range standard PCR using Faststart Master Mix 2X (Roche, Switzerland), repeat specific primers in a concentration of 0.5mM, and 50ng of gDNA to amplify the region of interest.

The quality and integrity of the DNA was also ensured by amplification of a small fragment near the region of interest using specific primers. This was employed to exclude any false positive results. Primers and PCR profile for thermocyling are shown in Table 2.20.

55

Chapter 02: Materials and Methods

Long-range PCR and Sanger sequencing

Samples were processed through long-range PCR using Phusion Flash High-Fidelity 2X PCR Master Mix (enzyme included) (Thermo-Fisher) using 0.5mM of each primer and 50ng of gDNA. This was done to identify heterozygotes in normal population who carry the mutant expansion. This PCR was also employed to amplify the targeted region in selected patients, some carriers and normal individuals. All PCR products were sequence verified by Sanger sequencing. Primer sequences and PCR profile are provided in Table 2.20.

Reverse-primed PCR

All the control samples from a cohort of 304 individuals along with samples of patients and carriers were screened using three independent reverse-prime PCRs (912 reactions for 304 samples). Each independent RPPCR was designed in such a way to accommodate all possible allelic conformation at the loci. Forward primer was FAM labelled and designated as Fw.FAM. This primer was used in all three independent reactions. The anchor was also same in all three independent reactions. The reverse primer designated as Rv, was distinct for each independent reaction and was against any of three alleles; (AAAAG)11,

(AAAGG)exp or (AAGGG)exp. Each Rv was a mixture of three primers mixed in 1:1:1 ratio (Rv1:Rv2:Rv3=1:1:1) for each of independent reaction. The frequency of expansion and other allelic conformations were observed by this approach. Phusion Flash RPPCR was completed using 2X High-Fidelity PCR Master Mix (Thermo-Fisher), Fw FAM Primer in 0.5mM concentration, anchor in 0.5mM, specific Rv primer mix in 0.05mM, DMSO in 3% and gDNA in 50ng concentrations. Primers and PCR profile of are provided in Table 2.20. Fragment length analysis was performed on an ABI 3730xl genetic analyser (Applied Biosystems, Foster City, CA, USA), and data were analysed using the GeneMapper software.

56

Chapter 02: Materials and Methods

Tables of materials

57

Chapter 02: Materials and Methods

Table 2.1: Lysis Buffer (DNA Extraction)

Reagents Concentration

Sucrose 0.32M

Tris.base 10mM

MgCl2 5mM

Triton X-100 1%

pH was adjusted to 7.6 with HCl.

Table 2.2: TEN Buffer (for DNA Extraction)

Reagent Concentration

Tris-base 20mM

Na2EDTA 4mM

NaCl 100mM

pH was adjusted to 7.4 with HCl.

Table 2.3: Low TE buffer

Reagent Concentration

Tris-HCl (pH 8) 10mM

Na2EDTA (pH 8) 0.2mM

58

Chapter 02: Materials and Methods

Table 2.4: TAE Buffer (50X)

Reagents Concentration

Tris base 1M

Na2-EDTA(pH 8) 0.5M

Acetic acid 28.55ml

(water upto final volume 500ml)

Table 2.5: Ethidium Bromide (10 mg ml-1)

Reagent Amount

Ethidium Bromide 10mg

Distilled water 1ml

Table 2.6: 6X Bromophenol Blue dye (loading dye)

Reagent Concentration

Bromophenol blue 0.25%

Glycerine 30%

Table 2.7: 10X PCR buffer (lab prepared)

Reagent Concentration

Tris-HCl(pH 9) 450mM

Na2EDTA (pH 8) 45µM

Ammonium Sulphate 10mM

β- Mercaptoethanol 67mM

Bovine Serum Albumin 1100µg/ml

59

Chapter 02: Materials and Methods

Table 2.8: Precipitation Solution for Sequencing Reaction

Reagent Final Concentration

Ethanol 86%

Sodium EDTA 11.3Mm

Table 2.9: Primers used to amplify Microsatellite Markers

Family Gene Markers Primer 1 Primer 2 Product size (bp) RDHT03 ATP7B D13S1325 CACGACGTTGTAAA TGGTGAATGAGAAG 246 ACGACTCTTCTCAC CAGCAC ACCTGCCTCCT

D13S1305 AGGCCACGGTGAG CACGACGTTGTAAA 324 CTATGA ACGACATTATGGTCC AGAAGCCCAAGT

CACGACGTTGTAAA TCCCACCAATCTTAC 169 C13SATP7B ACGACGGAGTGAG CGTCT GTAAATGGCTCCT

60

Chapter 02: Materials and Methods

Table 2.10: Growth media for fibroblasts

Reagents Concentration

1X Dulbecco’sftlpmpml mynlm fmlpdf DMEM)+Glutamax 1X

Heat inactivated fetal bovine serum, FBS 10%

Penicillin/streptomycin 1%

Uridine 0.05mg/ml

Table 2.11: Fixative solution for Immunofluorescence

Reagent Concentration

Phosphate buffer saline 1X

Formaldehyde 4%

Table 2.12: Blocking solution for Immunofluorescence

Reagent Concentration

Phosphate buffer saline 1X

Goat serum 10%

Triton-X 100 0.01%

61

Chapter 02: Materials and Methods

Table 2.13: RIPA lysis buffer

Reagent Concentration

NaCl 150mM

Triton X-100 0.1%

Sodium deoxycholate 0.5%

Sodium dodecyl sulphate (SDS) 0.1%

Table 2.14: Standards of different concentrations to make standard curve for BCA- protein assay

Samples for Volume of stock RIPA added to Water added to Protein standard curve used (Stock make mixture (µl) make mixture (µl) Concentration 2mg/ml) to make in 10μl of different mixture standards added per mixture (µl) well (µg)

1 0 4 36 0

2 2 4 34 0.5 3 5 4 31 1.25 4 6 4 30 1.5 5 8 4 28 2

6 11 4 25 2.75 7 15 4 21 3.75

8 20 4 16 5

Table 2.15: TRIS Acetate running buffer

Reagent Concentration

20X TRIS acetate stock 1X

ddH2O Up to 1L

62

Chapter 02: Materials and Methods

Table 2.16: SDS gel loading sample preparation

Reagent Concentration

Cell lysate 55µg (required volume)

1X 4x NuPAGE™ LDS loading dye

DTT 0.01M

RIPA buffer Volume up to 55µl

Table 2.17: TRIS GLYCIE transfer buffer

Reagent Concentration

10X TRIS/GLYCINE stock 1X

Methanol 20%

ddH2O Up to 2L

Table 2.18: 1X PBST

Reagent Concentration

PBS 1X

Tween 20 0.1%

Table 2.19: Blocking solution for western blotting

Reagent Concentration

PBS 1X

Tween 0.1%

Skimmed milk 5%

63

Chapter 02: Materials and Methods

Table 2.20: Primers used for routine short range, long range and reverse-primed PCR to check the frequency of repeat expansion in normal population

Primers Cycling Short-range Fw: TCAAGTGATACTCCAGCTACACCGTTGC 95C 4 min flanking PCR Rv: GTGGGAGACAGGCCAATCACTTCAG [95C 3 0s 59C 30 s 72C 60 s] x 35 cycles

72C 5 min Long-Range Fw TCAAGTGATACTCCAGCTACACCGTTGC 98C 3 min flanking PCR Rv GTGGGAGACAGGCCAATCACTTCAG [98C 10 s 65C 15 s - Each cycle decreasing by 0.5C 72C 3 min] X18 cycles

[98C 10s 57C 15s 72C 3 min] X18 cycles

72C 5 min Repeat-primed Fw FAM-TCAAGTGATACTCCAGCTACACCGT 98C 3 min PCR Anchor CAGGAAACAGCTATGACC [98C 10s (AAAAG)11 allele 65C 15s Rv1:CAGGAAACAGCTATGACCAACAGAGCAAGAC 72C 60s] x35 cycles TCTGTTTCAAAAAAGAAAAGAAAAGAAAAGAAAA Rv2:CAGGAAACAGCTATGACCAACAGAGCAAGAC 72C 5 min TCTGTTTCAAAAAGAAAAGAAAAGAAAAGAAAA Rv3:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAGAAAAGAAAAGAAAAGAAAA

(AAAGG)exp allele Rv1:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAAAGGAAAGGAAAGGAAAGGAAA Rv2:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAAGGAAAGGAAAGGAAAGGAAA Rv3:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAGGAAAGGAAAGGAAAGGAAA

(AAGGG)exp allele Rv1:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAAAGGGAAGGGAAGGGAAGGGAA Rv2:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAAGGGAAGGGAAGGGAAGGGAA Rv3:CAGGAAACAGCTATGACCAACAGAGCAAGAC TCTGTTTCAAAAGGGAAGGGAAGGGAAGGGAA

64

Chapter 03: Results

Chapter 03: Results

65

Chapter 03: Results

The affected members in families who were enrolled in this study had all or some of the following conditions; walking difficulties, abnormal gait, spasticity, ataxia, dystonia, speech difficulties, developmental delays or improper coordination.

Families, RDHT01, RDHT02 and RDHT10

RDHT01

Clinical/phenotypic characterization

The family is a non-consanguineous Pakistani family with two affected siblings (Fig.3.1A). Proband II-3 was born after an uneventful pregnancy and attained normal early milestones. Later, in his mid-20s, he developed gait instability and walking difficulty. He also reported headaches, vertigo attacks and occasional generalized tremors. Clinical examination was remarkable for cerebellar ataxia, affecting mainly the trunk with broad- based and unstable gait. He also exhibited upper limb dysmetria and dysdiachokinesia. Speech was affected by cerebellar dysarthria. ye movements’ examination revealed nystagmus and slow and jerky pursuit, without signs of severe oculomotor apraxia. The younger brother II-4 also shared the same phenotype as observed in his sibling, but the age of onset of symptoms was mid-teens. MRI imaging of the brain for the proband revealed cerebellar atrophy (Fig. 3.1B) whereas the MRI of the younger brother was unremarkable and within normal limits. This finding suggests the progressive nature of the anatomical abnormalities in the brain.

Other members in the family were apparently unaffected. None of them had any complaints as described above.

66

Chapter 03: Results

Fig. 3.1: Pedigree, MRI scan and sequence chromatograms for family RDHT01 A: Pedigree of family RDHT01. The genotypes for c.5885A>G;p.(His1962Arg) and c.1504C>T;p.(Arg502Trp) are indicated below the individual symbols of all participants. Patient marked with a star is for whom exome sequencing was performed B: MRI scan of the axial section of brain showing the cerebellar atrophy in II-3 C: The sequence chromatograms indicating the wild type allele in normal members of the family and the corresponding pathogenic novel heterozygous missense mutations c.5885A>G;p.(His1962Arg) (Reverse complement of the sequences are shown). D: The sequence chromatograms indicating the wild type allele in normal members of the family and the corresponding pathogenic novel heterozygous missense mutations c.1504C>T;p.(Arg502Trp) (Reverse complement of the sequences are shown).

67

Chapter 03: Results

Genetic Study

Only homozygous variants and subsequently compound heterozygous variants were considered from the exome data of the proband II-3.since the pattern of inheritance of the phenotype was recessive. All scrutinized variants had minor allele frequency (MAF) < 0.01 and those variants were ignored for which any hemizygous or homozygous individuals in public databases were present. No potential pathogenic homozygous variant was found. Analysis of the heterozygous variants, revealed two SETX variants and three DST variants for further consideration. DST variants had low REVEL scores and did not segregate with the phenotype. The missense variant in exon 14 of SETX c.5885A>G;p.(His1962Arg) (Fig.3.1 C) was found in compound heterozygous condition with a missense variant in exon 10 c.1504C>T;p.(Arg502Trp) (Fig. 3.1D) in two affected siblings of family RDHT01. Obligate carriers of the family were heterozygous for one of either variant. The variant c.5885A>G;p.(His1962Arg) was absent in all public databases and also in 350 control chromosomes from ethnically matched population. The second variant c.1504C>T;p.(Arg502Trp) was present in public databases (GnomAD 0.0004) as well as in ethnically matched control population at a low frequency (Table 3.1), but no homozygous individual for the variant was found. The frequency of this variant in South Asians was higher than in other populations (GnomAD SAS 0.0035). The variant p.(His1962Arg) had a REVEL score of 0.876, GERP score 5. The second variant p.(Arg502Trp) had a REVEL score 0.796, GERP score 3.86 (Table. 3.1).

68

Chapter 03: Results

Table 3.1: Variants considered and prioritized for family RDHT01

Gene Position Variant OMIM Conservation REVEL ClinPred Comparisons of Controls Segregation phenotype phenotypes of patients * with previous

DST 6: NM_001144769:c.1 Neuropathy, NA 0.355 0.7873 Not applicable Absent Does not 4161 hereditary segregate 56357764 G>C;p.(Asp4721His sensory and ) autonomic

DST 6: NM_001144769:c.8 Neuropathy, NA 0.11 0.1295 Not applicable Absent Does not 293 hereditary segregate 56420629 C>G;p.(Gln2765Glu sensory and ) autonomic DST 6: NM_001723:c.3716 Neuropathy, NA 0.036 0.045 Not applicable Absent Does not G>A;p.(Arg1239His hereditary segregate 56485116 ) sensory and autonomic

SETX 9: NM_015046.6:c.588 Ataxia with Conserved 0.876 0.9797 Match Absent Segregates 5A>G;p.(His1962Ar oculomotor 13517233 g) apraxia 2 8

SETX 9: NM_015046.6:c.150 Ataxia with Conserved till 0.796 0.7942 Match 0.005 Segregates 4C>T;p.(Arg502Trp oculomotor reptiles 13520548 ) apraxia type 1 2 GnomAD

0.0004 *175 ethnically matched individuals or 350 control chromosomes

69

Chapter 03: Results

RDHT02

Clinical/phenotypic characterization

RDHT02 is a consanguineous family with two affected individuals. A 29 years old female patient IV-8 (Fig.3.2A) and her 24 years old brother IV-3 suffered from cerebellar ataxia. Individual IV-8 was born after an uneventful pregnancy and she achieved early milestones normally. At the age of 20 years her walk became difficult and ataxic and she complained of progressive weakness in her lower limbs and suffered from frequent falls. She had truncal ataxia, was unable to stand with feet placed together and had a positive Romberg sign. She was unable to perform tandem gait. She exhibited a broad based gait with ataxia. She felt a sensation of burning in her feet and also experienced exacerbated heart rate and increased rate of breathing. She had scanning speech and eye focusing difficulties were evident. Extraocular movements were abnormal and horizontal nystagmus was observed. Slow ocular saccade and diplopia were present. There was no sensory loss although neuropathy was present. Vertigo and mild to severe pulsating headaches were also experienced during difficult ataxic walk. Autonomic testing was not performed but there was no evidence of autonomic dysfunction during examination by the neurologist. All cranial nerves were intact and no facial asymmetry was observed. Power in the limbs was normal with negative Babinski sign. However, the tone of the limbs was decreased. No pyramidal signs were observed (Tariq et al. 2018) (Work as part of this thesis).

Results of clinical investigations revealed elevated alpha feto-protein, 51.8IU/ml (normal range 0.49-9.84IU/ml). Electromyogram and nerve conduction studies showed low amplitude of motor nerves (data not shown). Hematology report of the patient revealed microcytic anemia. Vermian atrophy was observed by magnetic resonance imaging of the brain (Fig.3.2B). No enlarged posterior fossa with torculla-lamboid inversion was observed. Myelination was normal and white and grey matter in the brain was intact. Her 25 years brother IV-3 (Fig.3.2A) was similarly affected and had an identical phenotype except that he had dysarthria and experienced pain in eyes during speech (Tariq et al. 2018) (Work as part of this thesis).

Other members in both of the families were apparently unaffected. None of them had any complaints as described above.

70

Chapter 03: Results

Fig.3.2: Pedigree, MRI scan and sequence chromatogram for family RDHT02 A: Pedigree of family RDHT02. The genotypes for c.202 C>T;p.(Arg68Cys) are indicated below the individuals symbols. Patient marked with a star is whose DNA was exome sequenced. B: Axial brain MRI scan showing vermian atrophy and cystic dilation of the fourth ventricle, indicated with an arrow, which is also observed in Dandy-Walker malformation. C: Sequence chromatograms of the respective parts of SETX indicating the wild type allele in a normal control sample, the heterozygous pathogenic variant c.202 C>T;p.(Arg68Cys) in an obligate carrier, and the homozygous missense mutation in an affected member. The arrow indicates the position of the mutation. (Reverse complement of the sequence is shown)

71

Chapter 03: Results

Genetic Study

For the patient in family RDHT02, four variants were identified (Table 3.2) from the filtered exome data which fulfilled the criteria for further analysis. Segregation analysis revealed that variant c.202 C>T;p.(Arg68Cys) in SETX (NM_015046.6) was homozygous in DNA of both affected individuals and segregated with the phenotype (Fig.3.2C). The variant was present at a very low frequency in ExAC database (0.00001977) with no individual homozygous for the variant. The variant was also absent in 100 control chromosomes from ethnically matched individuals. Four pathogenicity prediction software SIFT, LRT prediction, Mutation taster and Polyphen2 predicted this variant to be pathogenic and the REVEL pathogenicity score for the variant was also high (Table 1). The variant was conserved in respective orthologues.Variants c.1318 T>C;p.(Tyr440His) in EIF2B4 and c.368 A>G;p.(Glu123Gly) in E4F1, though rare (0.00003655 for EIF2B4, 0.002 for E4F1) and predicted to be pathogenic, did not segregate with the phenotype (Tariq et al. 2018) (Table 3.2).

72

Chapter 03: Results

Table 3.2: Variants considered and prioritized for family RDHT02

Gene Position Variant OMIM Conservation REVEL ClinPred Comparisons of 100 control Segregation phenotype phenotypes of chromosome patients with s previous E1F2B4 2:27587636 NM_172195.3:c.1382 AR Conserved 0.786 0.2516 Does not match Absent Does not T>C;p.(Tyr461His) vanishing segregate white matter leukoenceph alopathy

SETX 9:135221834 NM_015046.6:c.202 Ataxia with Conserved 0.694 0.9933 Match significantly Absent Segregates C>T;p.(Arg68Cys) oculomotor with somewhat apraxia type phenotypic extension. 2

E4F1 16:2279629 NM_004424.4:c.368 Not any Conserved 0.217 0.2286 Not applicable Absent Does not A>G;p.(Glu123Gly) segregate

ATP2C2 16:84493155 NM_001286527.2:c.23 NA NA - - - - Does not 77 G>T;p.(Gly793Ter) segregate

73

Chapter 03: Results

RDHT10

Clinical/phenotypic characterization

RDHT10 is a consanguineous family and four siblings were affected similarly (Fig.3.3A). The baseline blood investigations of the proband were normal. He had a broad-based unstable gait, dysarthria and frequent falls due to bad coordination. He uses a metallic walker for an assisted walk. He also exhibits oculomotor apraxia and has subtle head tremors. The MRI of brain revealed prominent cerebellar folia, fourth ventricle and cisterna magna with relatively small vermis which suggested vermian atrophy in two of the affected siblings (data not shown). The sisters were also similarly affected. Other members of the family were apparently unaffected. None of them had any complaints as described above.

Genetic Study

For the patient in family RDHT10, only one potential homozygous variant was identified from the filtered exome data which fulfilled the criteria for further analysis. Segregation analysis confirmed that this variant c.5569T>C;p.(Cys1857Arg) in SETX (NM_015046.6), was homozygous in DNA of four affected individuals and segregated with the phenotype (Fig.3.3B). The variant was absent in all public databases. The variant was also absent in 100 control chromosomes from ethnically matched individuals. Four pathogenicity prediction software SIFT, Mutation taster, FATHM, PROVEAN, and Polyphen2 predicted this variant to be pathogenic. The REVEL pathogenicity score and ClinPred score were also high (Table. 3.3).

74

Chapter 03: Results

Fig.3.3: Pedigree and Sequence chromatograms for family RDHT10 A. Pedigree of family RDHT10. The genotypes for c.5569 T>C;p.(Cys1857Arg) are indicated belor the individuals’ symbols. Patient marked with stars are those for whom DNA samples were exome sequenced. B: Sequence chromatograms of the respective parts of SETX indicating the wild type allele in an unaffected individual, the heterozygous pathogenic variant c.5569 T>C;p.(Cys1857Arg) in an obligate carrier, and the homozygous missense mutation in an affected member. The arrow indicates the position of the mutation (reverse complement of the sequence is shown).

75

Chapter 03: Results

Table 3.3: Variant considered and prioritized for family RDHT10

Gene Position Variant OMIM Conservation REVEL ClinPred Comparisons of 100 control Segregation phenotype phenotypes of chromosomes patients

SETX 9:1351736 NM_015046.6: Ataxia with Conserved in all 0.678 0.9399 Matches significantly Absent Segregates 79 c.5569 T > oculomotor vertebrate classes C;p.(Cys1857 apraxia type except fish Arg) 2

76

Chapter 03: Results

Family RDHT03

Clinical/phenotypic characterization

RDHT03 is a consanguineous family. Four children were affected and had neurological complaints (Fig.3.4A). These complaints were classified into two types, dystonia or delayed milestones. The onset of disease in the proband, IV-1, was reported to be in early infancy. At the age of 24 years she had generalized dystonia, dystonic tremors, seizures and tics. The left hand, the foot and her neck had dystonic features. She also exhibited rriter’s cramp. Her younger sister IV-4 had onset of disease in early childhood. At the age of 12 years she exhibited general dystonic features but the phenotype was less severe as compared to that in her sister. She had weakness and facial tics and head tremors characteristic of dystonia. She additionally had delayed milestones of walking as she started to walk at the age of 7 years. Two brothers of the proband, IV-2 and IV-3, had delayed milestones of walking as they started to walk at the age of 9 years and 5 years respectively. At the current ages of 21 and 22 years they had mild ataxia which is indicative of cerebellar involvement. They had onset of intention tremors in early teens. Other members in the family were apparently unaffected. None of them had any complaints as described above.

Genetic Studies

Exome data for the two affected members IV-1 and IV-2 was first analyzed comparatively and then individually as well, under a recessive mode of inheritance. No variant was identified which segregated with the phenotype in all affected individuals. Analyses of individual data yielded four missense variants (Table 3.4) which were checked in the family for their possible segregation with either phenotype. COG8 and ADAMTS variants did not segregate with any of the two phenotypes in the family. DRD4 variant c.316C>T;p.(Arg106Cys) segregated with dystonia (Fig.3.4B) and GENEA variant c.1070T>C;p.(Leu357Pro) (Fig.3.4C) segregated with phenotype of delayed milestones.

77

Chapter 03: Results

Fig. 3.4: Pedigree and Sequence chromatograms for family RDHT03 A: Pedigree of family RDHT03. The genotypes for c.316 C>T;p.(Arg106Cys) in DRD4 and for c.1070 T>C;p.(Leu357Pro) in GENEA are indicated belor the individuals’ symbols. Upper halt tilled symbols shor attected individuals with dystonia, lower half filled symbols depicts those with delayed milestones and fully filled symbol denotes affected individual with both phenotypes. Patients marked with a star are those for whom DNA was subjected to exome sequencing B: Sequence chromatograms of the respective parts of DRD4 indicating the wild type allele in a normal control sample, the heterozygous carrier and homozygous missense variant c.316 C>T;p.(Arg106Cys) in an affected member. (Reverse complement of the sequence is shown) The arrow indicates the position of the mutation. C: Sequence chromatograms of the respective parts of GENEA indicating the wild type allele in a normal control sample, the heterozygous carrier and homozygous missense variant c.1070 T>C;p.(Leu357Pro) in an affected member. The arrow indicates the position of the mutation.

78

Chapter 03: Results

Table 3.4: Variants considered and prioritized for family RDHT03

Gene Position Variant OMIM Conservation REVEL ClinPred Comparisons of 350 control Segregation phenotype phenotypes of chromosomes patients with previous DRD4 11:63946 NM_000797:c.3 Autonomic Not conserved 0.239 0.123 New phenotype Absent Segregates 3 16 nervous system in chicken and with C>T;p.(Arg106 dysfunction pig dystonia Cys)

GENEA - c.1070 Spinocerebeller Conserved 0.709 0.950 Recessive Absent Segregates T>C;p.(Leu357P ataxia dominant inheritance with with ro) inheritance new phenotype delayed milestones

COG8 16:69368 NM_032382:c.1 Congenital Conserved 0.352 0.913 - - Does not 831 006 disorder of segregate C>T;p.(Arg336T glycosylation with either rp) phenotype

ADAMTS18 16:77369 NM_199355:c.1 Microcornea, Conserved 0.249 0.1187 - - Does not 780 732 myopic segregate G>A;p.(Val578I chorioretinal with either le) atrophy, and phenotype telecanthus

79

Chapter 03: Results

Computational work

Mapping of DRD4 variant in DRD4 structure

To predict the structural changes incurred from the p.Arg106Cys mutation in function of the wild type DRD4, first, the structure of the DRD4 protein was sought from RCSB PDB (https://www.rcsb.org/). The protein structure with PDB ID 5WIU was found to be the best match to DRD4. Alignment between the wild type DRD4 and its structural counterpart 5WIU-A is shown (Fig.3.5).

Regions between residue 228 and 336 constitute the ICL3 (intracellular loop 3) which was replaced with BRIL (apocytochrome b562RIL) to achieve better protein crystal stability in 5WIU-A. Despite the replacement of ICL3, the structure was considered an accurate representation of DRD4 (Wang et al. 2017). The 5WIU-A protein structure is illustrated below (Fig 3.6). The mutation location in wild type p.(Arg106Cys) is shown in the licorice representation (Fig 3.6). The p.Arg106 residue and its primary interacting partner Asp109 is shown (Fig 3.7)

80

Chapter 03: Results

Fig. 3.5: DRD4 wild type sequence alignment against the structural counterpart 5WIU on PDB. The sequence of wild type DRD4 was aligned well to the 5WIU-A sequence which is a modified DRD4 bound to its ligand nemonapride). 5WIU-A had crystal structure available on PDB. p.Arg106 is aligned between two sequences.

81

Chapter 03: Results

Fig.3.6: Protein structure of 5WIU illustrated with two colours (red, aligned part and blue the ICL3, substituted by BRIL). The mutation p.(Arg106Cys) location in wild type DRD4 is shown in the licorice representation which is different from the cartoon representation of whole protein

82

Chapter 03: Results

Fig. 3.7: Illustration of the Arg106 in blue color and the primary interacting partner Asp109 in aqua blue. Residues 98-106, illustrated in pink, form a loop which may increase in flexibility from Arg106Cys mutation, impacting the loop lining the cavity opening residues 182-189 (orange). Purple colored structure indicates a ligand of DRD4.

83

Chapter 03: Results

Modelling of GENEA and its variant

The wild type structure of GENEA was modelled with significant confidence (Fig.3.8). The template for the structure was 3pfq which is a protein kinase c beta 2. The variant p.(Leu357Pro) affecting the protein is part of a beta strand (Fig.3.9). Analysis of the homolog 3fpq, shows the kinase bound to phosphoamino phosphonic acid adenylate ester in its allosteric site which is lined by p.Leu357.

84

Chapter 03: Results

Fig. 3.8: The modelled structure of GENEA and superimposition of wild type-mutant forms A: The modelled structure of GENEA using phyre2. The model was constructed with great confidence and the secondary structures are indicated with different colors, Purple is helical, yellow is beta sheet, blue is turn and cyan is disordered part of the structure. The residue indicated in beaded conformation is Leu357 which is a part of beta strand. B: The superimposed structures of wild type (in red) and mutant (in cyan). The red colored areas show the structural differences among wild type and mutant protein as during superimposition the structure which are present in wild type (red) but missing or misaligned in mutant (cyan).

85

Chapter 03: Results

Fig. 3.9: Selected region of Phyre modelled secondary structure of GENEA. The residue 357 is located in the predicted beta sheet

86

Chapter 03: Results

Family RDHT04

Clinical/phenotypic characterization

RDHT04 is a consanguineous family (Fig.3.10A) in which two siblings were affected with a recessive form of movement disorder. The patients had no previous diagnosis in spite of extensive clinical and molecular investigations.

A 5 year old boy and a 4 year girl were affected with a congenital disorder characterized by hypotonic body postures, undeveloped speech, severe intellectual disability, severe spasticity in the lower limbs and frequent ankle clonus. The children were severely disabled and were never able to crawl or walk independently. A mixed pattern of hypotonia in a context of intermittent dynamic tone and spasms was exhibited by the patients. Hyperreflexia was observed in all four limbs and the knee joint was hypermobile. Both affected individuals had intermittent episodes of rigidity and stiffening. The tone in upper limbs of the affected individuals was normal. The affected children suffer from sleep disturbances, waking up several times during the night and have to be lulled back to sleep. No dysmorphic facial features were observed. Eye movements were normal and heat and pain sensation were also intact. Both affected individuals were overweight. The obligate carriers were unaffected (Tariq and Naz 2017) (Work as part of this thesis).

Magnetic resonance imaging (MRI) revealed reduced cerebral white matter volume (Fig. 3.10B), generalized delay in myleination and reduction in supratentorial, and to a lesser extent infratentorial volume. There was limited myelination of peri-rolandic white matter, but there was no evidence of myelination within the cerebral white matter. Attenuation of corpus callosum was observed (Fig. 3.10C). CT scan of the brain revealed mild cerebral atrophy and left maxillary sinusitis (Tariq and Naz 2017) (Work as part of this thesis)..

No abnormality was seen in contrast enhanced, CE MRI dorso lumbo sacral spine but loss of cervical lordosis was seen in CE MRI of the cervical spine. EMG studies revealed no evidence of peripheral neuromuscular abnormality (Table 3.5).

87

Chapter 03: Results

Fig. 3.10: Pedigree, MRI scans and sequence chromatogram of family RDHT04 A: Pedigree of family RDHT04. The genotypes for c.64C>T;p.(Arg22Trp) variant in TFG are indicated below the individual symbols. Patient marked with a star is for whom the DNA was exome sequenced B: MRI scan of coronal section of brain. Marked reduction in white matter and myelination of cerebral cortex are evident here. C: MRI scan of brain indicating thin corpus callosum and cerebral atrophy. There is a reduction in supratentorial and to a lesser extent infratentorial volume. Corpus callosum is significantly reduced in size as indicated by the arrows. D: Sequence chromatograms of the respective parts of TFG indicating the wild type allele, heterozygous allele and homozygous missense variant c.64C>T;p.(Arg22Trp). Arrows indicate the position of the mutation.

88

Chapter 03: Results

Table 3.5: Clinical Investigations for proband of family RDHT04

Type Test Comments

Electromyogram, EMG Normal

Abnormal white matter and Magnetic Resonance Imaging, MRI (Brain) myelination

Attenuation of corpus callosum

Computerized axial tomography, CT scan (Brain) Mild cerebral atrophy

Contrast enhanced MRI (Dorso-Lumbo-Sacral spine) Normal Radio imaging investigations Contrast enhanced MRI (Cervical spine) Loss of cervical lordosis

Abdominal ultrasound Normal

Blood and urinary HPLC based quantification of amino acids and estimation of Normal examinations creatinine, ketone bodies, organic acids,

Glycosaminoglycan, alpha-fetoprotein

89

Chapter 03: Results

Genetic Study

Karyotype was normal and the copy number variation analysis and clinical panel exome sequencing revealed no pathogenic variant accounting for the phenotype (Table 3.6). Analysis of the whole-exome data revealed a single likely pathogenic variant c.64C>T;p.(Arg22Trp) which was found to co-segregate with the phenotype after Sanger sequencing and was responsible for the phenotype (Fig. 3.10D). The variant was present in the LOVD (http://www.lovd.nl/3.0/home) database listed as a pathogenic allele (Elsayed et al. 2016) but was absent from all other public databases as well as in 100 control chromosomes from ethnically matched controls from Pakistan. No pathogenic intronic, synonymous and compound heterozygous mutations were identified which could be hypothesized to increase the severity of the phenotype (Tariq and Naz 2017) (Work as part of this thesis).

90

Chapter 03: Results

Table 3.6: Molecular Investigations for proband of family RDHT04

Type Tests Comment

Karyotype 46XY

Copy number variation No pathogenic CNV

Molecular analysis Clinical exome panel sequencing +DEL/DUP No pathogenic variant

Exome sequencing TFG variant

c.64C>T;p.(Arg22Trp)

91

Chapter 03: Results

Family RDHT05

Clinical/phenotypic characterization

RDHT05 is a consanguineous family (Fig.3.11A) in which two siblings were affected with a recessive form of movement disorder.

Individual IV-2 (Fig.3.11A) was born after an uneventful pregnancy. He had neck holding at the age of 8 weeks, started sitting at the age of 6 months and could stand independently at the age of 9 months. At the age of 1-1.5 year he had onset of disease which consisted of inability to walk, and spasticity of legs. His unaffected parents were first degree cousins. At the time of examination, at age of 7 years, he had difficulty in sitting independently and was not able to walk or stand without support. His movement consisted of crawling and a limited and difficult supported walk with a home constructed wheeled wooden frame.

The patient uttered his first words at the age of one year. His speech development was reported to be appropriate for his age until about 18 months at which time his speech started deteriorating. The patient was examined at the age of seven years. His social interaction and understanding were normal. He maintained good eye contact and answered all questions appropriately, though dysarthria was present. Swallowing difficulties were not evident yet, though it is possible that they could develop later. Drooling was also observed. Extraocular movements were normal and no nystagmus was observed. All the cranial nerves were intact and no facial asymmetry was observed. The tone, power and reflexes were normal in the upper limbs. Examination of the lower limbs revealed increased tone. Power in the lower limbs was 3/5 (Ciesla et al. 2011) with positive Babinski sign and bilateral ankle knee clonus. He had good control over his bladder and bowel movements (Tariq et al. 2017) (Work as part of this thesis).

Clinical investigations including blood creatine phosphokinase levels, magnetic resonance imaging of the brain, nerve conduction studies and electromyography were normal. His younger six years old sister IV-3 (Fig.3.11A) was similarly affected and had an identical phenotype. Other members in the family were apparently unaffected. None of them had any complaints as described above.

92

Chapter 03: Results

Genetic Study

Analysis of the exome data of proband IV-2 revealed a single pathogenic variant, c.1918G>A;p.(Arg640Ter) in ALS2 which was found to co-segregate with the phenotype after Sanger sequencing (Fig.3.11B). The variant was absent in all public databases as well as in 100 control chromosomes from ethnically matched individuals (Tariq et al. 2017) (Work as part of this thesis).

Functional study

ALS2 expression in RDHT05

ALS2 gene expression was checked in three individuals of family RDHT05. ALS2 mRNA was detected in all three individuals of the family; a homozygous wild type, a heterozygous carrier and an affected individual homozygous for the variant (Fig 3.12).

93

Chapter 03: Results

Fig. 3.11: Pedigree and sequence chromatogram of family RDHT05 A: Pedigree of family RDHT05. The genotypes for c.1918G>A;p.(Arg640Ter) in ALS2 variant are indicated below the individual symbols. Patient marked with a star is for whom DNA was exome sequenced B: Sequence chromatograms of the respective parts of ALS2 indicating the wild type allele in a normal control sample and homozygous nonsense variant c.1918G>A;p.(Arg640Ter) in an affected member. The arrow indicates the position of the mutation. The reverse complement of the sequence is shown.

94

Chapter 03: Results

Fig. 3.12: ALS2 gene expression in family RDHT05. No difference in expression was observed among affected and unaffected members. Lanes 1-3 shows the nested RT-PCR amplified band of 451bp (arrow on left side) in wild type, carrier and homozygous individuals for the variant, respectively. Lanes 4 and 5 show size standard, lanes 6-8 show the amplification of GAPDH 496bp from the same cDNA samples as used for the detection of ALS2 expression (indicated with an arrow on right side).

95

Chapter 03: Results

Family RDHT06

Clinical/phenotypic characterization

RDHT06 is a consanguineous family (Fig.3.13A) in which two sisters were affected with a recessive form of movement disorder. These included a 13 years old female patient V-2, who suffered from ataxia. The age of onset of symptoms was 5 years and the disease progressed slowly. The patient V-2 has been bedridden since she was 8 years old. She was unable to sit without support. The patient had chorieform movements, severe dysarthria and speech difficulty. No visual and auditory complaints were reported by the patient. Visual acuity was normal and pupils were equal and reactive. There was oculocutaneous telangiectasia. Sensory system of the patient was intact. Her intellect was normal and she did not have complaints of urinary and fecal incontinence. Body tone and bulk was normal. Power in upper limbs was normal 5/5 but it was 3/5 in lower limbs. Deep tendon reflexes were absent. Electroencephalogram (EEG) studies were normal. MRI of the patient showed vermian and cerebellar degeneration (Fig.3.13B). Complete blood count revealed that she was suffering from anemia. Her older sister had an identical phenotype. The obligate carriers in the family were apparently unaffected. None of them had any complaints as described above (Tariq et al. 2018) (Work as part of this thesis).

Genetic study

The analysis of the filtered exome data revealed homozygous c.7327 C>T;p.(Arg2443Ter), rs121434220, a known pathogenic variant in ATM (NM_000051.3). Segregation analyses revealed that the variant was heterozygous in the obligate carriers of family RDHT06 and homozygous in the other affected sister as well (Fig.3.13C). A 6 years old asymptomatic brother, V-3, of the affected individuals (Fig.3.13A) was also homozygous for the variant (Tariq et al. 2018).

96

Chapter 03: Results

Fig. 3.13: Pedigree, MRI scan and sequence chromatogram of family RDHT06 A: Pedigree of family RDHT06. The genotypes for c.7327 C>T;p.(Arg2443Ter) variant in ATM are indicated below the individual symbols. Patient is marked with a star for whom the DNA was exome sequenced B: Axial brain T1-weighted MRI image showing vermian atrophy and atrophy of the cerebellar hemispheres, which is greater on the left side (indicated by an arrow). C: Sequence chromatograms of the respective parts of ATM indicating the wild type allele, obligate carrier heterozygous allele and homozygous nonsense variant c.7327 C>T;p.(Arg2443Ter). Arrows indicate the position of the variant.

97

Chapter 03: Results

Family RDHT08

Clinical/phenotypic characterization

RDHT08 is a non-consanguineous Pakistani family from Punjab region. The proband II-5 (Fig.3.14A) was born after an uneventful pregnancy and achieved his early milestones adequately. He was reported to have sudden onset of frequent falls while walking at the age of eight years. He was examined at the age of 16 years when he exhibited tip toe walking, with hammer pes cavus toes. Power in the upper and lower limbs was normal. The tone was constantly increased in upper limbs and was increased in lower limbs on distraction. The reflexes in the upper limbs were normal whereas in lower limbs there was hyperreflexia with up going plantar response and positive Babinski sign. The patient exhibited a mixed patterns of spasticity and rigidity. Axial and facial rigidity were also observed which affected his speech. Visual acuity was decreased. His 18 years old sister II-4 also experienced mild ataxia and cognitive impairment in addition to the problems faced by her brother. She had sensory neuropathy. Ankle reflex was absent. The third sister II-6, at the age of 15 years, was affected similarly with more significant dystonic postures of hands and feet. Magnetic resonance imaging (MRI) brain of the proband revealed moderate periventricular white matter abnormalities in axial section of the brain (Fig. 3.14B) and hypointesities in the globus pallidus as evident in the coronal section of brain (Fig.3.14C).

Genetic Study

Exome data revealed a single homozygous variant c.199delG;p.(Ala67LeufsTer5) in C19orf12 (NM_001031726) segregating with the phenotype (Fig.3.14D) being homozygous in affected and heterozygous in obligate carriers and other unaffected siblings.

Functional Study

C19orf12 expression in RDHT08

C19orf12 gene expression was checked in three individuals of family RDHT08. C19orf12 mRNA was detected in two carriers of the variant in family but was not detected in the sample from the affected individual (Fig.3.15). GAPDH was amplified from all three individuals showing that cDNA was of good quality.

98

Chapter 03: Results

Fig.3.14: Pedigree, MRI scans and sequence chromatogram of family RDHT08 A: Pedigree of family RDHT08. The genotypes for C19orf12 variant c.199delG;p.(Ala67LeufsTer5) are indicated below the individual symbols of all participants. Patient marked with a star is for whom DNA was exome sequenced. B: MRI of the axial section of brain showing moderate periventricular white matter abnormalities. C: MRI of the coronal section of brain with hypointensities in the globus pallidus (encircled). D: Partial sequence chromatograms indicating the wildtype allele in a normal control, heterozygous family member and homozygous deletion mutation c.199delG;p.(Ala67LeufsTer5) in an affected member with C19orf12 variant. The position of the mutation is indicated by an arrow.

99

Chapter 03: Results

Fig. 3.15: C19orf12 gene expression in family RDHT08. Lane 1 and 2 shows the nested RT-PCR amplified band of 201bp (arrow on left side) in two carrier of variant in family whereas lane 3 shows very reduced or no amplification of required band from cDNA library of homozygous affected member. Lane 4 has size standard, lanes 5-7 show the amplification of GAPDH specific 496bp products from the same cDNA samples (indicated with an arrow on the right side).

100

Chapter 03: Results

Family RDHT16

Clinical/phenotypic characterization

RDHT16 is a consanguineous family with two affected siblings suffering from a recessive form of movement disorder (Fig. 3.16A). The proband had disease onset in late 30s and had the complaints of general tremors, difficult walk, frequent falls, spasticity, tongue fasciculation, poor eyesight and restricted neck mobility. His symptoms showed diurnal fluctuations in severity and subsided to some extent by taking levodopa. He was severely affected without medication and could not walk without it. His elder sister had onset of disease at the age of 40 years and had the complaints of immobility of hands and feet, biting and swallowing difficulty and spasticity at back. Her complaints also subsided by taking levodopa. The unaffected members of the family did not have any complaints described above.

101

Chapter 03: Results

Fig. 3.16: Pedigree and sequence chromatograms of family RDHT16 A: Pedigree of family RDHT16. The genotypes for homozygous variant in GENEB, CGN and GENEC are indicated below the individuals (top to bottom respectively). The patients marked with star are those whom DNA were exome sequenced. B: Sequence chromatograms of the respective parts of GENE B indicating the wild type allele in a normal control sample, heterozygous carrier and homozygous affected (reverse complement sequence is shown) C: Sequence chromatograms of the respective parts of CGN indicating the wild type allele in a normal control sample, heterozygous carrier and homozygous affected, respectively. D. Sequence chromatograms of the respective parts of GENEC indicating the wild type allele in a normal control sample, heterozygous carrier and homozygous affected. The positions of the variants are shown with respective arrows.

102

Chapter 03: Results

Genetic Study

By comparison of exome data of both individuals IV-5 and IV-2 and filtering the data according to the criteria, three variants were checked and all three segregated with phenotype (Fig 3.16 B,C,D). Two of these variants in GENEB and CGN resided in regions of shared homozygosity on chromosome 1 (Fig.3.17) (Table 3.7). The two regions extended from 2.2-5.3Mb and 147-162Mb on chromosome 1.The variant in GENEB was present in region of homozygosity, was fully conserved and was absent in control chromosomes. The variant in CGN although was present in a large region of homozygosity, and was not conserved even in chicken and rhesus. However, the variant in CGN was absent in ethnically matched control chromosomes.

There were two shared regions of homozygosity on chromosome 6, 25-42Mb and 154- 170Mb for the two samples. The third variant in GENEC which segregated with the phenotype resided in the shared homozygosity region on chromosome 6 (Fig.3.18). This variant was also conserved among all vertebrate classes.

Some shared regions of homozygosity were also observed on chromosome 4, 12, 15, 18, and 19 in two affected individuals but these did not carry any potential filtered variant. The GENEB variant along with symptoms was submitted to Genematcher and some individuals with heterozygous copy number variants were found in DECIPHER (https://decipher.sanger.ac.uk/search?q=drd4#consented-patients/results ) CGN and GENEC variants were also deposited in Genematcher. A few homozygous matches were found with partially matching phenotypes but work on these is ongoing.

103

Chapter 03: Results

Fig 3.17: Regions of homozygosity on chromosomes 1 2.2-5.3Mb and 147-162Mb, in family RDHT16. Two of the segregated variants in GENEB and CGN resided in these regions.

Fig 3.18: Regions of homozygosity on chromosomes 6, from 25-42Mb and 154-170Mb in family RDHT16. One of the segregated variants in GENEC resided in the second region (154-170Mb).

104

Chapter 03: Results

Table 3.7: Variants considered and prioritized for family RDHT16

Gene Variant Conservation REVEL ClinPre Control Segregation Frequency in Public d chromosomes (100) database

GENEB c.691A>G;p.(Lys231Glu) Conserved 0.19 0.9797 Absent Segregates 0

CGN NM_020770:c.2096A>G;p.(Glu699Gly) Not conserved 0.198 0.9397 Absent Segregates 0 in Rhesus and

Chicken

GENEC c.1690C>T;p.(Arg564Trp) Conserved 0.278 0.9265 Absent Segregates 0.0003138

105

Chapter 03: Results

Families RDHT07, RDHT09 and RDHT14

RDHT07

Clinical/phenotypic characterization

RDHT07 is a consanguineous Pakistani family (Fig.3.19). Two siblings are affected with a movement disorder. The onset of disorder in the proband IV-1 was at the age of 24 years. She developed gait instability with positive cerebellar signs and ataxia. She experienced frequent falls and could not walk properly and could not lift things while walking. She experienced vertigo, headache and dizziness as well. Her younger sister had early onset of the disorder at the age of 8 years and had frequent falls, gait instability and tremors and severe pain in legs. She had a protruding thumb in her one foot as well. The other members of the family were apparently unaffected and did not exhibit any symptoms as described above.

Genetic Study

Three homozygous variants were identified in the exome data of proband of RDHT07, but none of these segregated with the phenotype. No potential compound heterozygous variants were identified which segregated with the phenotype. As the age of onset of the disease was in late 20s in the proband, so some variants were checked for disease association in spite of some being present in the public databases in individuals who were homozygous (Table 3.8).

106

Chapter 03: Results

Fig.3.19: Pedigree of family RDHT07. DNA of the patient marked with an asterisk was analyzed by exome sequencing

107

Chapter 03: Results

Table 3.8: Variants considered and prioritized for family RDHT07

Gene Position Variant OMIM Conservation REVEL ClinPred Comparisons of Homozygo 250 Segregation phenotype phenotypes of us in control patients with public chromoso previous databases mes PLEKHG5 1: NM_00126559 Spinal muscle Pro>Gln in 0.144 0.050 Matches 3 Absent Does not 2:c.271C>A;p. atrophy, zebrafish significantly segregate 6537598 (Pro91Thr) Charcoat Marie Tooth disease

NPHP4 1: NM_015102:c. Nephronopht Arg>His in 0.414 0.146 Does not match 1 Absent Does not 3758G>A;p.(A hisis 4 opossum segregate 5925220 rg1253Gln)

SACS 13: NM_014363:c. Spastic ataxia Conserved 0.447 0.118 Matches 1 0.008 Does not 972 segregate 23929779 C>A;p.(Asp32 (2 hets) 4Glu)

108

Chapter 03: Results

RDHT09

Clinical/phenotypic characterization

RDHT09 is a consanguineous family and has five siblings affected with recessively inherited movement disorder (Fig.3.20). The proband was reportedly fine until the age of 16 years when suddenly his lower limbs started shivering. This shivering progressed to the whole body and became associated with headaches and abnormal body postures. The patient had a disturbed sleep pattern. The bulk, tone and power were normal in upper and lower limbs. Sensory system was intact and there were no cerebellar signs. Tongue fasciculations were also exhibited by the patient. He experienced diurnal fluctuations and he felt better in the morning. These symptoms were relieved by taking levodopa. All the five siblings were similarly affected and shared the same clinical phenotype. The other members of the family were apparently unaffected and did not manifest any symptoms as described above.

Genetic study

The exome data for individual IV-8 and IV-9 was compared and no likely variant was identified which segregated with the phenotype in all individuals. Regions of shared homozygosity between two siblings were present on chromosome 2, 6, 8 and 10. Two homozygous variants on chromosome 2 were considered for possible segregation (Table 3.9) (Fig.3.21). Other regions of homozygosity did not carry filtered pathogenic variants.

There were different regions of autozygosity on chromosome 11 observed at different positions individually in two siblings (Fig.3.22). Pathogenic variants in those regions were checked for possible segregation of disease assuming genetic heterogeneity but these were only homozygous in a single affected member and in none of the other affected member (Table 3.10).

A heterozygous variant which was shared by two siblings and was in the UTR region of a dystonia associated gene was also checked for possible segregation. However it did not segregate as well (Table 3.11).

109

Chapter 03: Results

Fig.3.20: Pedigree of family RDHT09. DNA of the patients marked with asterisks were used for exome sequencing.

110

Chapter 03: Results

Fig. 3.21: Shared regions of homozygosity on chromosome 2, 46-70Mb, between two affected siblings in RDHT09. MSH2 and CFAP36 variants reside in this region

Fig.3.22: Independent regions of autozygosity on chromosome 11, in two affected siblings in RDHT09. 73-99 Mb in IV-8 and 11-24 Mb plus 28-33Mb in IV-9. TMEM126 and DEUP1 fall in autozygosity region of IV-8 whereas TEAD1 resides in the autozygosity region of IV-9.

111

Chapter 03: Results

Table 3.9: Homozygous variants considered and prioritized for family RDHT09 which were shared by two siblings

Gene Position Variant OMIM phenotype REVEL Clinpred Segregation

MSH2 2:47703620 NM_001258281: Mismatch repair cancer 0.715 0.294676 Homozygous variant in one normal c.1922G>A;p.(Cys641Tyr) syndrome sibling.

CFAP36 2:55746961 NM_080667: c.24 - 0.107 0.091532 Homozygous variant in one normal G>C;p.(Glu8Asp) sibling.

112

Chapter 03: Results

Table 3.10: Homozygous variants considered and prioritized for family RDHT09 with the possibility of genetic heterogeneity.

Gene Position Variant Conservation REVE Clinpred Status Homozygous in Segregation L public database

TEAD1 11:129034 NM_02 Conserved 0.497 0.9978 Homozygous in one of the 0 Homozygous only in 51 1961:c. exome sequenced patients one affected 521 individual C>G;p.( Pro174 Arg)

TMEM126 11:853666 NM_03 Conserved 0.497 0.0404 Homozygous in one of the 2 Homozygous only in 71 2273:c. exome sequenced patients one affected 314G> A;p.(Ar g105Gl n)

PRDM15 21:432217 NM_02 Conserved 0.418 0.3026 Homozygous in one of the 0 Homozygous only in 46 2115:c. exome sequenced patients one affected, and also 4178T> homozygous in an C;p.(Le unaffected individual u1393Pr o) DEUP1 11:931414 NM_18 Arg>Gln in 0.111 0.8117 Homozygous in one of the 0 Homozygous only in 28 1645:c. mouse two exome sequenced one affected 1358G> patients individual T;p.(Ar g453Me t)

113

Chapter 03: Results

Table 3.11: Variants considered for the possibility of heterozygous and imprinted allele

Gene Position Variant OMIM ClinPred Status Frequency Segregation phenotype

THAP1 8:42698406 NM_018105:- Dystonia No Heterozygous in Heterozygous in one 169C>G two sequenced unaffected individual as well. patients 0.000096 UTR5 GnomAD

114

Chapter 03: Results

RDHT14

Clinical/phenotypic characterization

RDHT14 is a consanguineous Pakistani family and has six affected individuals in three marriages (Fig.3.23). Four of these were siblings to each other and the age of onset was mid 3t’s. The proband IV-5 showed the phenotype of Parkinsonism, tremors, and spasticity. Age of onset was same in all other affected members as well. The two brothers of the proband had similar symptoms and the severity of the symptoms subsided by taking levodopa. The eldest sister IV-1 of the proband had the same levodopa responsive Parkinsonism, tremors and spasticity. The drug responsiveness was reduced with time. She had orofacial dystonia and speech difficulty as well. She was bed ridden at the time of examination and also had urinary incontinence. The other members of the family were apparently unaffected and did not have any symptoms as described above.

Genetic Study

The exome data for individual IV-1 and IV-5 was compared. However, no potential homozygous variants were identified. Regions of shared homozygosity between two siblings were present on chromosome 5 and 6 (Fig.3.24). The filtered variants were examined but none was located in the region of homozygosity.

Some independent autozygous regions were observed on chromosomes 10, 11, 12 and 15, 18, 19 and 22 which were not shared between two siblings. A homozygous variant in POLG was present in one of these regions in one patient and was found in one more affected member by segregation analysis (Table 3.12).

115

Chapter 03: Results

Fig.3.23: Pedigree of family RDHT14. DNA of the patients marked with stars were subjected to exome sequencing.

116

Chapter 03: Results

Fig.3.24: Shared regions of homozygosity among two affected siblings of family RDHT14 on chromosome 5, region 78-90Mb (A) and chromosome 6, region 154-166Mb (B)

117

Chapter 03: Results

Table 3.12: Variant considered and prioritized for family RDHT14

Gene Position Variant OMIM Conservation REVEL ClinPred Homozygous Segregation phenotype in public database

POLG 15:89873445 NM_002693:c.722C>Tp.(Pro241Leu) Mitochondrial Not conserved, 0.362 0.1494 0 Homozygous mutant recessive in two affected ataxia p.(Pro>Leu) in siblings. Other two syndrome frog, zebrafish affected individuals and opossum were heterozygous for the variant.

118

Chapter 03: Results

Families, RDHT11, RDHT12

RDHT11

Clinical/phenotypic characterization and genetic studies

RDHT11 was a consanguineous family with 5 affected individuals but only one was alive at the time of collection (Fig.3.25). All the affected individuals had a very severe and similar phenotype of ataxia. The proband V-2 had severe ataxia, tremors, dysphagia, dysarthria and progressing disability. He was always restless and suffered from continuous tremors and jerks. The age of onset of disorder was in early 30s. No potential variants were identified after analysis of whole exome data.

RDHT12

Clinical/phenotypic characterization and genetic studies

RDHT12 was a consanguineous family with two affected siblings sharing the same phenotype (Fig.3.26). Both individuals had delayed milestones of walking. They were never able to speak normally although they had normal hearing. Even at the age of 14 years, their speech was restricted to only 2-3 words. Exome sequencing did not reveal any potential variant associated with the disorder.

119

Chapter 03: Results

Fig.3.25: Pedigree of family RDHT11. The DNA of the patient marked with a star was analyzed by exome sequencing.

Fig.3.26: Pedigree of family RDHT12. The DNA of the patient marked with a star was analyzed by exome sequencing.

120

Chapter 03: Results

Families, RDHT13 and RDHT15

RDHT13

RDHT13 was a consanguineous family with three affected siblings sharing the same phenotype (Fig.3.27). They had short stature, speech problem, walking difficulty, and psychological problems as well. The eldest was disabled and had a hip crawling gait. The unaffected siblings and normal parents did not have any complaints. Exome sequencing is in progress at Johns Hopkins University.

RDHT15

RDHT15 was a consanguineous Pakistani family with two affected individuals (Fig.3.28). The proband had the disease onset at the age of 35 years. He had weakness and difficulty in walking which progressed with time. He also suffered from dysarthria and ataxia. The second affected individual had the disease onset at the age of 60 years. She had dysarthria, rigidity, ataxia and nystagmus. She was also unable to perform tandem gait and exhibited tremors. Exome sequencing is in progress at Johns Hopkins University.

121

Chapter 03: Results

Fig.3.27: Pedigree of family RDHT13

Fig.3.28: Pedigree of family RDHT15

122

Chapter 03: Results

Sporadic cases

Samples from sporadic individuals with different movement disorders (Table 3.13) were collected. All of them were offspring of consanguineous parents. The DNA samples are in process of exome sequencing at John Hopkins

123

Chapter 03: Results

Table 3.13: Sporadic cases collected and sent for molecular diagnosis

Sr. No. Sample ID Age of onset Clinical Phenotype

1 SPO-HT01 55y Cervical dystonia ,tremors

2 SPO-HT02 19y Rigidity, Wilson disease, headache, shivering

3 SPO-HT03 14y Cerebeller ataxia, bilateral hearing loss, difficult speech, bed ridden status

4 SPO-HT04 15y Shivering, jerks, frothing mouth, tonic, clonic, tremors

5 SPO-HT05 17y Shivering, cannot walk, cannot speak, progressive, hand foot deformities

6 SPO-HT06 68y Essential tremors

7 SPO-HT07 45y Weakness in legs, tremors, falls, multiple sclerosis, dystonic postures, pseudobulbar

8 SPO-HT08 12y Wilson disease, fam history, others deceased

9 SPO-HT09 23y General dystonia

10 SPO-HT10 55y Ataxia, walking difficulty, protruding toe of feet

124

Chapter 03: Results

UCL-HT01

Clinical characterization

The patient was diagnosed with Charcot-Marie-Tooth disease, type 4H and had typical signs of high arched feet, slim calves and clawed toes. She felt subjective differences in the power of her right and left leg but objectively on examination she had normal power. Patient was areflexive with flexor plantars. Pinprick sensation was intact but light touch sensation was absent to the base of the toes. Vibration sense was absent at the ankles. Joint position sense was preserved. She had muscles wasting near ankles. She is taking vitamin supplements and exercising to limit the exacerbation of her neuropathy and general health.

Genetic studies

The patient had compound heterozygous variants in FGD4 found through panel sequencing. The variants segregated with phenotype. Both of these variants were at the residues which are conserved among different vertebrate classes and had high REVEL and ClinPred scores (Table 3.14).

125

Chapter 03: Results

Table 3.14: Compound heterozygous variants responsible for disorder in patient of family UCL-HT01

Gene Position Variant OMIM Conservation REVEL ClinPred Comparisons of 100 control Segregation association phenotypes of patients chromosomes for Gene with previous

FGD4 12:327786 c.1730G>A;p.(Ar Charcoat Conserved 0.338 0.9407 Matches significantly Absent Segregates 82 g577Gln) Marie Tooth with

disease phenotype

FGD4 12:327865 c.1859G>T;p.(Gl As above Conserved 0.577 0.9848 Matches significantly Absent Segregates 80 y620Val) with phenotype

126

Chapter 03: Results

Functional studies

The fibroblasts cells from that patient were taken and grown under standard conditions. The impact of mutated frabin was checked upon the cell proliferation rate via cell counting assays. The possible impact of above mentioned variants was also checked on the frabin stability and localization through immunofluorescence and frabin specific western blotting. Patient cells are named “cav” and control cells are named as “Red108” and “Red3S”.

Proliferation rate of fibroblasts with mutant frabin

Six individual growth curve cell counting assays were performed after the optimization of tibroblast culturing conditions. Fibroblasts trom the patient “cav” and trom tro control individuals from different families were included in these assays. The values were normalized and growth curves were constructed (data no shown). There were no differences among the three samples and the fibroblasts from the patient did not show any reduced or compromised rate of proliferation.

Localization of protein in patient verses normal fibroblasts

Immunofluorescence

The cells were stained for FGD4 encoded frabin protein, and also for actin using monoclonal antibodies and actin specific chemical compounds. Frabin and actin both have cytoplasmic expression. The expression and localization of frabin was not changed in patient fibroblasts (Fig.3.29 G-I) as compared to control 1 (Fig.3.29A-C) and control 2 (Fig.3.29 D-F).

127

Chapter 03: Results

Fig.3.29: Immunostained fibroblasts of control Red128, Red39 and patient Nav with anti-frabin and anti- actin antibodies/compounds. A, D and G show the expression of frabin (green color) in fibroblasts of Red128, Red39 and Nav respectively. B, E and H show the expression and localization of actin (red color) in fibroblasts of Red128, Red39 and Nav respectively. C, F and I show the expression and localization of frabin and actin simultaneously in fibroblasts of Red128, Red39 and Nav respectively. The expression and localization of frabin was not changed in patient fibroblasts as compared to control 1 (Red128) and control 2 (Red39).

128

Chapter 03: Results

Western Blotting: expression and quantification of protein of interest

Total protein in the cell lysates of controls Red39, Red128 and patient Nav was quantified. After the standard curve was plotted for standard samples of known concentration (Table 3.15, Fig.3.30), the value of mean absorbance for experimental and control samples was plotted on graph to find the concentration of total protein in those samples.

Red128 had 4.66µg/µl, Red39 had 2.509µg/µl and Nav had 3.98µg/µl of protein in cell lysates. 25µg of total protein was loaded in each respective well and was processed for subsequent steps of western blotting. Frabin protein quantity, in Red128, Red39 and Nav was similar and no difference was found in protein expression among control and patient fibroblasts (Fig.3.31).

129

Chapter 03: Results

Table 3.15: Absorbance values for different dilutions of known protein concentration

Sr. No. Stock* volume Final concentration of Mean absorbance of added protein standards three readings

µl µg/µl

1 0 0 0.0950

2 0.5 0.0047 0.2093

3 1.25 0.01190 0.3506

4 1.5 0.0143 0.3880

5 2 0.0191 0.4783

6 2.75 0.0262 0.5883

7 3.75 0.0357 0.7040

8 5 0.0476 0.9077

Stock concentration* 2µg/µl

0.06

0.05 y = 0.06x - 0.008 R² = 0.9915 0.04

0.03

0.02

[BSA] µg/µl / [BSA] 0.01

0 0 0.2 0.4 0.6 0.8 1 -0.01 Mean Abs. / AU

Fig.3.30: Standard curve of known BSA concentrations employed to quantify total cell protein.

130

Chapter 03: Results

Fig.3.31: Frabin expression in control and affected fibroblast cell cultures is same. Lanes 2, 5, 8 show control Red128, lanes 3, 6, 9 show control Red39 and lanes 4,7,10 show sample Nav respectively in three independent experiments. 25 µg of total protein was loaded in each well. The band shown by an arrow in lane 10 is the band of interest for frabin. Upper part is labelled with anti-frabin antibody and lower part of the figure is the membrane labelled with anti-actin.

131

Chapter 03: Results

Collaborative Project completed in UCL

132

Chapter 03: Results

Allelic distribution of a short tandem repeat of RFC1 gene in the control population

Flanking PCR on all control samples

Routine short range flanking PCR for control population amplified the locus region where the normal allele was present (Fig.3.32). All the control cohort of 304 samples were subjected to standard PCR. The expanded mutant allele was not amplified with the biallelic mutant expansion due to the large size of the product in homozygous individuals. This screening provided initial clue for further investigation. The samples which gave no product on first PCR were double checked for the integrity and quality of the DNA. Patients with homozygous mutant expansion were used as negative controls since they yielded no product for this PCR because of the large size of expansion.

Pre-Alu PCR to remove false positive results

This PCR was used to ensure the quality of the DNA for those samples for which no amplification was observed with short range flanking PCR. It amplified a small fragment in all samples where the DNA quality was good (Fig.3.33). This showed that the quality of the DNA was good for all those samples which yielded a product. However, the samples which did not yield a product after pre-Alu PCR were discarded since their quality was judged to be poor. In some cases these samples were recollected, DNA extracted and used for screening as described.

133

Chapter 03: Results

Fig.3.32: Amplification of a 348bp fragment spanning the repeat region to screen control population. Lane 3 shows no amplification, which may suggest either mutant allele expanded in a homozygous fashion or poor quality of DNA.

Fig.3.33: Pre-Alu PCR product. Amplification of genomic fragments near one side of repeat expansion was used to ensure good quality of DNA. Lane 4 shows one sample, for which the quality of DNA was low. It was therefore excluded from the control sample cohort.

134

Chapter 03: Results

Long range flanking PCR and RPPCR for final screening

Long range flanking PCR was employed to amplify the expanded repeat region from a patient and selected control population. The products were sequenced (Fig.3.34). 304 controls along with three affected and carrier samples were screened by repeat-primed PCR (RPPCR) using three sets of primers, each targeting a unique allelic conformation at the repeat locus. Homozygous AAGGG expansion were not observed in 304 healthy controls screened.

RPPPCR and long range PCR for affected individuals and carriers

Repeat-primed PCR (RPPCR) with primers targeting the mutant AAGGG pentanucleotide unit was performed and it confirmed the presence of a homozygous AAGGG repeat expansion in all affected members from 11 families, as well as heterozygous expansion in unaffected carriers. Flanking PCR using standard conditions failed to amplify the region in all patients suggesting the presence of a large expansion on both alleles, as opposed to their unaffected siblings for whom at least one allele could be amplified by PCR.

Frequency of different allelic conformations

RPPCR analysis targeting the AAGGG repeat showed that 0.7% (4 out of 608 chromosomes tested) carried an AAGGG expansion in heterozygous state. The locus, where the expansion resides, was shown to be highly polymorphic in the normal population and, besides the rare AAGGG expansion allele (AAGGG)exp, three other conformations were observed: (AAAAG)11, (AAAAG)exp, (AAAGG)exp (Fig.3.35).

An allelic carrier frequency of 0.7% for the expanded AAGGG repeat together with Southern blotting for sizing the repeats, suggests that this expansion in RFC1 represents a frequent cause of late-onset ataxia, with clinical similarities and disease frequency equal to that of Friedreich’s ataxia.

135

Chapter 03: Results

Fig. 3.34: RPPCR and by Sanger sequencing confirmation of repeat expansion A: Repeat-primed PCR targeting the mutated AAGGG repeated unit. FAM-labelled PCR products are separated on an ABI3730 DNA Analyzer. Electropherograms are visualized on GENEMAPPER at 2,000 relative fluorescence units.

Representative plots from a patient carrying the AAGGG repeat expansion and one non-carrier (control) carrying (AAAAG)11 are shown. B: Sanger sequencing of long-range PCR reactions confirms in patients (upper trace) the AAAAG to AAGGG nucleotide change of the repeated unit.

136

Chapter 03: Results

Fig.3.35: Polymorphic configurations of the repeat expansion locus and allelic distribution in healthy controls A. Schematic representation of the repeat expansion locus in intron 2 of Replication factor C subunit 1 (RFC1) and its main allelic variants. B. Estimated allelic frequencies in 608 chromosomes from 304 healthy controls.

137

Chapter 04: Discussion

Chapter 04: Discussion

138

Chapter 04: Discussion

Novel SETX variants identified in three families

Four SETX biallelic variants were identified in three families RDHT01, RDHT02 and RDHT10 as cause of the disorder. Three of the variant were previously unreported (Fig.4.1).

SETX as a RNA/DNA helicase is known to play an important role in transcription, neurogenesis and DNA damage responses (Groh et al. 2016; Richard et al. 2013). Variants of SETX have been implicated in two clinically characterized progressive neurodegenerative disorders (Lavin et al. 2013). Dominant gain-of-function mutations in this gene are usually associated with amyotrophic lateral sclerosis (ALS4, OMIM 602433). ALS4 is characterized by atrophy and weakness of distal muscles but without the involvement of the sensory system. Two rare phenotypes also reported to be associated with dominantly inherited mutations in SETX include tremor ataxia syndrome (TAS) (Bassuk et al. 2007) and autosomal dominant proximal spinal muscular atrophy (ADSMA) (Rudnik-Schoneborn et al. 2015). TAS patients do not experience peripheral neuropathy but have cerebellar features and tremors. ADSMA is associated with muscular atrophy, weakness and increased creatine kinase activity (Lavin et al. 2013; Rudnik-Schoneborn et al. 2012). Recessively inherited mutations in SETX are associated with ataxia with oculomotor apraxia type 2 (AOA2 OMIM # 606002). AOA2 is characterized by cerebellar features along with peripheral neuropathy. The characteristic phenotypes of AOA2 include peripheral neuropathy, progressive cerebellar atrophy, oculomotor apraxia and elevated alpha fetoprotein in serum (Becherel et al. 2013).

139

Chapter 04: Discussion

Fig.4.1: Diagrammatic representation of SETX. Novel missense variants in SETX identified in this study are shown above and below the protein structure. Rectangles show homozygous variants and those in ovals indicate compound heterozygous variants. aa, amino acids.

140

Chapter 04: Discussion

The protein interaction domain of SETX is located between N-terminal amino acids 64- 593. Most pathogenic variants located in this region were hypothesized to result in recessively inherited AOA2 (Duquette et al. 2005). However, many variants including c.814C>G;p.(His272Asp) (Kenna et al. 2013) and c.1166T>C;p.(Leu389Ser) (Chen et al. 2004) which affect this region are associated with dominantly inherited ALS4 disorder. Similarly, AOA2 causing variants are scattered throughout the gene making it impossible to assign genotype-phenotype correlation for these disorders. Some of the loss of function variants which lead to AOA2 and gain-of-function variants which result in ALS4 have been reported to induce differential transcriptional changes in cells. This difference in transcriptional sceneries has been suggested to explain phenotypic differences of these patients (Fogel et al. 2014).

Two of the SETX variants were present in compound heterozygous condition in patients of family RDHT01. One of these variants c.5885A>G;p.(His1962Arg) was present in the helicase domain, whereas c.1504C>T;p.(Arg502Trp) affects the N-terminal protein interaction domain (Fig.4.1). The variant c.5885A>G;p.(His1962Arg) was absent in public databases and control chromosomes from ethnically matched population and fully conserved among different vertebrate classes (Fig. 4.2).

The second variant c.1504C>T;p.(Arg502Trp) was present in public databases at a low frequency 0.0004 (GnomAD). It was conserved in vertebrates including reptiles but not in amphibians and fish (Fig.4.3). One explanation which can be considered for non- conservation in these species could be the inherent differences which have arisen during the course of evolution. Therefore, nearly 30, SETX sequences from various organisms belonging to all classes of vertebrates were aligned and observed (data not shown). Arg502 was not fully conserved but Arg>Trp variant was not present in any organism. This variant has been previously reported to be pathogenic in heterozygous form in a patient suffering from hereditary motor neuropathy with pyramidal signs (HMNP) (Drew et al. 2015). This variant was also present in the heterozygous form in the unaffected father in the participating family RDHT01 presented in the current work. Therefore, either this variant, does not cause HMNP in the unaffected father, or the phenotype was very mild and clinically undetectable even in at the age of 60. The report of the same variant in involvement of HMNP in heterozygous form and a biallelic form in AOA2 is interesting and remains to be investigated further.

141

Chapter 04: Discussion

Fig. 4.2: Clustal omega alignment of respective orthologues of SETX from different classes of vertebrates. The p.His1962 is conserved (boxed).

Fig.4.3: Clustal omega alignment of respective orthologues of SETX from different classes of vertebrates. Residue 502 is boxed. Although amphibians and fish have other amino acid residue at the respective position, none of the organisms had the Arg>Trp variant.

142

Chapter 04: Discussion

A SETX homozygous variant c.202C>T;p.(Arg68Cys) was identified in patients of family RDHT02. This novel variant was present in the protein interaction domain of SETX (Fig.4.1). The usual characteristics of cerebellar atrophy, nystagmus, peripheral neuropathy, elevated alpha fetoprotein, weakness in limbs were observed in the patients. Patients of this family exhibited Dandy-Walker resembling radiological findings because of severe vermian atrophy. The unusual features which were observed in the patients of this family included mild to severe headaches, excessive breathing, exacerbated heart rates, and a feeling of fever in both patients.(Tariq et al. 2018) (Work as part of this thesis).

For family, RDHT10, a homozygous variant c.5569T>C;p.(Cys1857Arg) in SETX (NM_015046) was identified. It affects a region near the helicase domain of SETX, It was associated with AOA2 phenotype in the patients. The Cys1857 residue was conserved in all vertebrates except fish (Fig.4.5). Thirty SETX sequences from different classes of vertebrates were aligned but none of them had Arginine at the corresponding position (data not shown). High pathogenic scores, segregation in a large family and absence in normal population makes this variant a likely candidate for disease association in family RDHT10.

Many previously reported AOA2 and ALS4 associated variants are also not conserved in vertebrates which emphasizes that complete conservation is not necessary for SETX pathogenic variants in causing disease. For example, pathogenic variants p.(Arg332Trp) and p.(Ile466Met) have been reported where the corresponding amino acids in the zebrafish orthologue are replaced by Glutamine and Valine respectively (Ghrooda et al. 2012; Moreira et al. 2004). Similarly, the corresponding amino acid for pathogenic variant p.(Gln190Pro) (Ghrooda et al. 2012) is substituted by Lysine or Arginine in respective orthologues of frog and zebrafish, respectively. In case of the reported AOA2 associated variant p.(His435Arg) (Fogel and Perlman 2006), the corresponding amino acid is replaced by Tyrosine in fish orthologues. Another variant of SETX, p.Thr918Ile (Vantaggiato et al. 2014) is also not conserved, being replaced by Valine in chicken and frog, Isoleucine in alligator, and Aspartate in fish orthologues. Another variant p.(Lys992Arg) (Fogel and Perlman 2006) presents an example of a pathogenic variant where the corresponding amino acid is not conserved even in cats where it is replaced by Valine.

143

Chapter 04: Discussion

In addition, many heterozygous SETX variants implicated in ALS4, affect amino acid of SETX which are also not well conserved during evolution. These include p.Thr3, p.Thr14, p.Pro311 and p.Glu623 (HGMD, http://www.hgmd.cf.ac.uk/ac/all.php, accessed February 2019).

It is hypothesized that the AOA2 causing SETX variants disturb the sumoylation profile of the gene whereas ALS4 causing SETX mutations do not. This disruption in sumoylation, in turn disrupts the SUMO-dependent interactions between senataxin and the exosome. The disrupted interaction modulates the normal targeting of the exosome to the sites of DNA damage and disturbs neuroprotection subsequently leading to neurodegeneration (Richard et al. 2013). A pathogenic variant in SETX c.193G>A;p.(Glu65Lys), identified previously in compound heterozygous condition causing in AOA2 phenotype (Duquette et al. 2005), was reported to disrupt the SUMO-dependent interactions of SETX with the exosome (Richard et al. 2013). The novel variant c.202C>T;p.(Arg68Cys) identified in RDHT02 is in close proximity to the c.193G>A; p.(Glu65Lys) variant and resides within the protein interaction domain. Therefore, it can be hypothesized that this variant may adversely affect the SUMO-dependent interactions of SETX. Two of the other identified variants in the current work, c.5885A>G;p.(His1962Arg) and c.5569T>C;p.(Cys1857Arg) were located in or near the helicase domain, and perhaps are not involved in sumoylation dependent pathomechanism.(Tariq et al. 2018) (Work as part of this thesis)

The findings in RDHT01, RDHT02 and RDHT10 extend the allelic spectrum of SETX associated recessive disorder AOA2.

144

Chapter 04: Discussion

Fig.4.4: Clustal omega alignment of respective orthologues of SETX in different classes of vertebrates showing absolute conservation of residue p.Arg68 (boxed).

Fig.4.5: Clustal omega alignment of respective orthologues of SETX from different classes of vertebrates. Residue 1857 is boxed. None of the aligned sequences had arginine at the corresponding position.

145

Chapter 04: Discussion

TFG variant associated severe congenital disorder

A pathogenic homozygous variant c.64C>T;p.(Arg22Trp) was identified in TFG to be responsible for a severe form of disorder in patients of family RDHT04.

TRK-fused gene encodes a protein, which has PB1 and coiled-coil functional domains (Fig.4.6). This protein functions at sites of vesicle biogenesis on the endoplasmic reticulum and plays a crucial role in the sorting of secretory cargoes in cells (Witte et al. 2011). One of the physiological roles of TFG is in long term axonal maintenance which is critical for neuroprotection (Alavi et al. 2015; Beetz et al. 2013b; Yagi et al. 2016). Axonopathy of the corticospinal tract causes neurological disorders characterized by lower limb spasticity and includes hereditary spastic paraplegia (HSP) and amyotrophic lateral sclerosis (Tariq and Naz 2017).

A heterozygous missense variant, c.854C>T;p.(Pro285Leu) of TFG was first identified as a cause of autosomal dominant hereditary motor and sensory neuropathy with proximal dominant involvement (HMSN-P) (OMIM 604484) (Ishiura et al. 2012). Patients exhibited widespread fasciculations, had proximal-predominant muscle weakness, and atrophy followed by distal sensory involvement. The c.854_855 CpG dinucleotide is a mutational hotspot. Therefore, the same variant c.854C>T has been reported in different families from Japan, Korea and Iran (Alavi et al. 2015; Ishiura et al. 2012; Ishiura and Tsuji 2013), but without clinical variability of the associated phenotype. Another novel heterozygous mutation c.806G>T;p.(Gly269Val) was identified as the cause of dominant axonal Charcot-Marie-Tooth disease type 2 (OMIM 609260) in a Taiwanese family (Tsai et al. 2014). However, the same variant was later reported in an Iranian family accounting for autosomal dominant hereditary motor and sensory neuropathy with proximal dominant involvement (HMSN-P) phenotype (Khani et al. 2016).

146

Chapter 04: Discussion

Fig.4.6: Diagrammatic presentation of TFG. Crucial domains are marked. Pathogenic variants identified in this gene are indicated above and below the representation of protein. Rectangles depict heterozygous variants whereas circles indicate biallelic homozygous variants. The variant identified in this work is shown in shaded circle. aa, amino acids.

147

Chapter 04: Discussion

Spastic paraplegia with optic atrophy and neuropathy known as SPOAN is a severe form of complicated HSP in which the patients exhibit spastic paraplegia with optic atrophy and neuropathy (OMIM 609541). This disorder has been associated with biallelic mutations in KLC2 (Macedo-Souza et al. 2005; Melo et al. 2015). Other spastic paraplegia disorders without involvement of KLC2 but with similar phenotypes as that of SPOAN are called SPOAN like disorders (Amorim et al. 2014). TFG associated recessively inherited SPOAN like hereditary spastic paraplegia SPG57 was first reported six years ago (Beetz et al. 2013b). A homozygous missense mutation c.316C>T;p.(Arg106Cys) in TFG was identified in patients of two Indian families suffering from SPOAN like HSP (Beetz et al. 2013b; Harlalka et al. 2016). In contrast, a new variant affecting the same codon, c.317G>A;p.(Arg106His) was reported to be associated with a pure form of Hereditary Spastic Paraplegia without optic atrophy and neuropathy in a third family of Pakistani origin (Harlalka et al. 2016). Haplotype analysis revealed evidence for c.316C>T;p.(Arg106Cys) mutation as a founder allele in India. Secondly, bisulfate sequencing revealed that the c.316_317 CpG dinucleotide constitutes a mutational hotspot (Harlalka et al. 2016).

The c.64C>T;p.(Arg22Trp) variant identified in family RDHT04 has been reported in a Sudanese family to be associated with hereditary spastic paraplegia SPG57 without optic atrophy (Elsayed et al. 2016). This variant has been shown to affect oligomerization of TFG (Elsayed et al. 2016). The DNA of Sudanese family patients was not available for haplotype analysis. It is hypothesized that as there is no known common ancestry between people of Pakistan and Sudan, the mutation arose independently in the two populations. Therefore, the c.64_65 CpG dinucleotide may be a mutational hotspot in TFG (Tariq and Naz 2017) (Work as part of this thesis).

Assembling of TFG complexes in accurate conformations is critical for normal functioning of cell. Mutation p.(Arg106Lys) in coiled-coil domain and the mutation p.(Arg22Trp) in PB1 domain of TFG have been reported to alter compaction of TFG ring complexes changing the secretory protein trafficking. TFG promotes L1CAM on the axonal surface, and these mutations in TFG cause the partial inhibition of TFG function leading to defects in axon bundling (Slosarek et al. 2018).

148

Chapter 04: Discussion

The findings in family RDHT04 extend the phenotypic spectrum of the disease associated with TFG variants (Table 4.1). The affected individuals of this family exhibited no spasticity in the upper limbs and none of them had microcephaly, unlike the affected members of the Sudanese family in which all three had spasticity in the upper limbs and two of the three had microcephaly as well. No extrapyrmidal signs were described in the Sudanese family carrying the same pathogenic variant. In family RDHT04 a frequent clonus was observed in both affected children, which has not been reported before with TFG variants. Additionally, the children had poor intellect, undeveloped speech, sleep disturbances and were also overweight. The disability of the three individuals of the Sudanese family was progressive but was clinically less severe even at the age of 15 years, unlike the two patients in family RDHT04 who were severely affected soon after birth. Most significantly, the affected individuals have never been able to crawl or walk in contrast to the Sudanese patients who could walk without aid (Tariq and Naz 2017).

It remains to be determined how the same mutation can cause different phenotypes in patients belonging to different ethnic groups. This phenomenon of phenotypic heterogeneity has also been reported for another TFG homozygous variant p.(Arg106Cys) in two Indian and one Italian family associated with phenotypes of complicated HSP and infantile neuroaxonal dystrophy, respectively (Beetz et al. 2013a; Catania et al. 2018).

A combination of severe movement disability, undeveloped speech, clonus, poor intellect, obesity and sleep disturbances adds to the phenotypic spectrum associated with TFG variants. This information could be helpful in diagnosis of other individuals with TFG mutation

149

Chapter 04: Discussion

Table 4.1: Recessive variants of TFG with associated phenotypes

A.A Domain Phenotype Ethnic Region Reference ** Pathomechanism change*

Arg22Trp PB1  Complex HSP Sudan PMID:27601211 Reduced domain oligomerization of TFG  Progressive

 Microcephaly

 Walk without assistance

 Spasticity in upper limbs

 No clonus

 Mild white matter hyperintesities

 Speech ?

 Sleep disturbances ?

Arg22Trp PB1  Complex HSP Pakistan This report domain  Static

 No microcephaly

 Bed ridden, cannot crawl or walk

 No spasticity in upper limbs

 Frequent ankle clonus Bulk reduction in white matter

 Undeveloped speech

 Sleep disturbances present

Arg106Cys Coiled Complicated HSP with India PMID:23479643 Collapse of the coil peripheral neuropathy endoplasmic reticulum domain and optic atrophy network and India PMID:27492651 mitochondrial fragmentation

Infantile neuroaxonal Altered mitochondrial dystrophy Italy PMID:29971521 network and inner membrane potential

150

Chapter 04: Discussion

A.A Domain Phenotype Ethnic Region Reference ** Pathomechanism change* Arg106His Coiled Pure HSP without optic Pakistan PMID:27492651 Mitochondrial coil atrophy (British fragmentation but less domain nationals) severe as compared to that due to p.(Arg106Cys)

*Nomenclature according to NP_001007566.1. HSP, hereditary spastic paraplegia, PB1, Phox and Bem 1, **PubMed identifier

151

Chapter 04: Discussion

A novel homozygous ALS2 frameshift variant associated with IAHSP

A novel homozygous truncating c.1918G>A;p.(Arg640Ter) variant in ALS2 involved in a neurological progressive disorder was identified in two siblings of a consanguineous Pakistani family RDHT05. The phenotypic and the clinical findings of the patients correspond to progressive infantile onset ascending hereditary spastic paralysis (IAHSP) (Eymard-Pierre et al. 2002; Xie et al. 2015). The characteristic IAHSP (Orrell 1993) with infantile onset of the disorder with spasticity, brisk reflexes, sustained clonus of the lower limbs and wheelchair dependence were observed in both patients. Severe spastic tetraparesis with pseudobulbar syndrome may also be observed in future years as the disease progression continues (Tariq et al. 2017) (Work as part of this thesis).

ALS2 (Amyotrophic Lateral Sclerosis 2) is composed of 34 exons and encodes a protein termed alsin. Three alternatively spliced variants have been described for ALS2, the longest of which encodes a 1657 amino acid protein. Alsin is a putative GTPase regulator and has three important functional domains RCC1, DH/PH and VPS9 (Gros-Louis et al. 2003; Wakil et al. 2014). Three motor neurodegenerative and progressive recessively inherited disorders are caused by ALS2 mutations (Daud et al. 2016; Flor-de-Lima et al. 2014; Xie et al. 2015). These include juvenile amyotrophic lateral sclerosis (ALS2, OMIM 205100), juvenile primary lateral sclerosis (JPLS, OMIM 606353), and infantile onset ascending spastic paralysis (IAHSP, OMIM 607225) (Eymard-Pierre et al. 2002). IAHSP has infantile onset, usually sparing lower motor neurons (LMNs), and manifests without oculomotor signs. Juvenile primary lateral sclerosis does not affect the lower motor neurons but involves oculomotor signs with earlier onset of bulbar manifestations. Progression of motor disability in IAHSP is rapid as compared to JPLS. ALS2 has a juvenile onset of symptoms due to the involvement of lower motor neurons which results in fasciculations and areflexia (Flor-de-Lima et al. 2014; Racis et al. 2014; Wakil et al. 2014). The age of onset, involvement of the lower motor neurons and clinical history are thus important factors in making the exact diagnoses for patients carrying mutations in ALS2. It is still challenging to correctly diagnose individuals with ALS2 mutation as an overlap of phenotypic manifestations usually occurs (Flor-de-Lima et al. 2014; Orrell 1993).

152

Chapter 04: Discussion

Truncating, missense and frameshift mutations have been associated with all three disorders described for ALS2 (Racis et al. 2014; Verschuuren-Bemelmans et al. 2008).The nature and position of these pathogenic mutations in ALS2 are not related to the resulting disorder so a genotype-phenotype correlation cannot be established (Eymard-Pierre et al. 2006; Panzeri et al. 2006).

Apart from the variant identified in this work, three other pathogenic variants in ALS2 have been reported which segregate with IAHSP in families from Pakistan (Daud et al. 2016; Gros-Louis et al. 2003). These include a frameshift, nonsense and a missense variant (Fig. 4.7). Premature termination codons before penultimate exons can lead to disease phenotype by either the production of truncated proteins, or by complete loss of protein if mRNA is processed through nonsense-mediated mRNA decay (NMD) (Kurosaki and Maquat 2016). The pathomechanism of c.1918G>A;p.(Arg640Ter) mutation could be the former since ALS2 mRNA escapes NMD as suggested by the current study. The variant affects exon 9 of the total 23 exons of ALS2, and translation of the mRNA will result in absence of important functional domains DH/PH and VPS9 from the protein. This may result in loss of function of the protein or complete absence of protein if it is degraded due to its unstability.

Comparison of the phenotypes in all four Pakistan families with ALS2 mutations reveals consistency regarding the age of onset, walking difficulties, dysarthria, absence of oculomotor signs and rapid progression of the disease. These results further emphasize the clinical homogeneity associated with ALS2 mutations in IAHSP. To date, no variants have been identified for other ALS2 related phenotypes in patients from Pakistan. Continued research will reveal additional pathogenic alleles in the Pakistani population associated with IAHSP and other ALS2 related phenotypes. This will enhance our understanding of the role of the gene variants in all three disorders.

153

Chapter 04: Discussion

Fig.4.7: ALS2 protein structure. It has three functional domains; Regulator of Chromosomal Condensation 1 (RCC1), Dbl homology/Plekstrin homology (DH/PH) and Vacuolar sorting protein 9 (VPS9). IAHSP associated with variants in the Pakistani population are indicated above the protein depiction. Variant identified in this study is boxed. For reference, residue numbers of some amino acids are indicated below the protein depiction.

154

Chapter 04: Discussion

A pathogenic homozygous ATM variant identified in an asymptomatic individual

A homozygous pathogenic nonsense variant c.7327C>T;p.(Arg2334Ter) in exon 52 of ATM was identified in family RDHT06 as cause of the disorder. DNA damage responses are vital for genomic stability and cell survival and ATM maintain the integrity of the genome by playing roles in oxidative stress and DNA damage repair responses (Nishida et al. 2017) (Berger et al. 2017; Lavin et al. 2013). This variant was previously reported in a compound heterozygous form with a second deletion variant in African American (Telatar et al. 1998; Wright et al. 1996). The same variant was also identified as one of the mutant alleles in patients of two different German families. Subsequent haplotype analyses revealed that this region has an independent origin and suggested it to be a mutational hotspot because of a CpG dinucleotide (Sandoval et al. 1999). Since individuals in Pakistan have no shared ancestry with either Africans or Germans, therefore the observation of the same variant in ethnically distinct populations further supports an independent mutational event and confirms this position as a mutational hotspot (Tariq et al. 2018).

A younger brother of the affected siblings who was clinically normal at the age of 6 years, was also homozygous for the variant. It is possible that the onset of disease is delayed because of involvement of modifiers of disease onset of expressivity. Future follow up of the individuals will clarify this point.

Are some C19orf12 variants monoallelic for neurological disorders?

In family RDHT08, a homozygous pathogenic variant, c.199delG;p.(Ala67LeufsTer5), in C19orf12 was identified to be responsible for disease progression. This variant was previously identified as one of the compound heterozygous alleles in a single French NBIA4/MPAN (OMIM 614298) patient with second allele being a missense mutation c.416 A>G;p.(Tyr139Cys). The same frameshift variant has also been identified in homozygous form in two affected MPAN patients (Gagliardi et al. 2015; Hogarth et al. 2013). This variant is predicted to produce a truncated protein.

155

Chapter 04: Discussion

Biallelic variants in this gene are reported to cause the disorder but recently the possibility of monoallic variants to cause the disorder was also suggested (Monfrini et al. 2018). The unaffected heterozygous carriers of frameshift variant in family RDHT08, together with more than 70 other reported heterozygous unaffected individuals emphasize the fact that presence of half of the normal levels of functional protein is enough to rescue the wild- type phenotype in heterozygous frameshift variant carriers of C19orf12. Moreover, the biallelic pathogenic variants c.191insG;p.(Val64GlyfsTer18), c.404insT; p.(Met135IlefsTer15), c.436-437insG;p.(Ala146GlyfsTer6) and c.199delG; p.(Ala67LeufsTer5) are predicted to add unique amino acids to the protein before truncation (Fig.4.8). However, no clinical symptoms are observed in the heterozygous carriers of these variants as well. This argues against monoallelic nature of the disorder and suggests the lack of a dominant-negative effect for these variants.

Detailed review of the reports where monoallelic variants in heterozygotes were suggested to be solely responsible for disease phenotype, shows the limitations in methodology of the studies. For example, the presence of monoallelic C19orf12 variant c.244A>T;p.(Lys82Ter) in NBIA patient and in her unaffected mother as well, weakens the possibility of monoallelic nature of disorder due to variants in this gene. There is also a high chance that in some studies, the second pathogenic allele in the same gene was missed, perhaps due to it being located in the regulatory region, or due to large deletions within the second allele. Digenic inheritance could be another, although a remote possibility for the disorder in these individuals affected with MPAN and having monoallelic C19orf12 variants. Follow up of patients with monoallelic C19orf12 variants may reveal a second pathogenic variant in the same or a different gene.

156

Chapter 04: Discussion

Fig 4.8: All monoallelic and some reported biallelic mutations affecting C19orf12. The variants mentioned on upper side were reported as monoallelic. Variants shown below the gene structure are those which are known to cause disorder when they are biallelic. Frameshift variants are shown in red colour, whereas missense and nonsense variants are in black. The variant shown in blue has been reported to cause disease both in a monoallelic as well as in biallelic condition. Variant reported in this study is indicated with an asterisk. (PMID 23269600, 22704260, 25962551, 29295770, 22508347, 23166001, 21981780). E1, E2, E3 represent exons 1 to 3 respectively of C19orf12. Slanting lines depict introns, and the horizontal black lines upstream and downstream the coding regions depict the 5' and 3' untranslated regions, respectively.

157

Chapter 04: Discussion

Two variants segregating with two different phenotypes in one family

In RDHT03, two phenotypes of dystonia and delayed milestones were observed. Homozygous DRD4 variant segregated with the dystonia phenotype and homozygous GENEA variant segregated with the phenotype of delayed milestones. Though DRD4 variant was not fully conserved in vertebrates (nearly 35 sequences were aligned-data not shown) but none of the orthologues had Cysteine at this position. Arginine at this position was either replaced with valine (in chicken), thereonine (zebrafish), alanine (chameleon) or glycine (in pig). The neighboring amino acids in the region were also observed and it was noted that in case of Arg>Gly, Arg>Val and Arg>Thr substitutions, the neighboring amino acids were also changed which probably could make these variants tolerable in other species. Another explanation can be that as Cysteine can form disulphide bonds in protein, it may induce unacceptable structural differences in mutant protein.

The DRD4 structure was obtained from PDB in bound form with its ligand. The variant p.(Arg106Cys) in the structure was visualized. Ser104, Pro105, Leu107, Cys108, Asp109, Ala110, Leu171, Cys172 and Gly173 were observed within 5 Å neighborhood of Arg106. The primary interaction of Arg106 is expected to be with Asp109 because of opposite charges. Interaction of Arg106 and Asp109 is very important in maintaining a good flexibility of helices lining the cavity for ligand binding. All these interactions are expected to be disturbed by Arg106Cys mutation due to the loss of a charged residue interacting with Asp109 and relatively poor helical propensity of cysteine (Fujiwara et al. 2012). The binding mode for all interactions of DRD4 is not known. However, from the binding mode of nemonapride, a ligand of this protein, it can be conjectured that cavity opening lined by loop forming residues 182-189 may become blocked due to this variant (Fig 4.9).

158

Chapter 04: Discussion

Figure 4.9: A. Side view of the DRD4 illustrating the position of nemonapride (purple) inside the cavity, lined by multiple helices. Nemonapride is a ligand of this protein. B. Illustration of the arginine106 in blue color and the primary interacting partner Asp109 in aqua blue. Residues 98- 106, illustrated in pink, form a loop which may increase in flexibility from Arg106Cys mutation, impacting the loop lining the cavity opening residues 182-189 (orange).

159

Chapter 04: Discussion

Another homozygous variant which segregated with the phenotype of delayed milestones in family RDHT03 was p.(Leu357Pro) affecting GENEA. The segregation of this variant with the phenotype, its evolutionary conservation (Fig.4.10), its absence in control population and its high pathogenicity score makes it a promising candidate for disease association in the family. Dominantly inherited variants in same gene have been associated with ataxia. This work implicates GENEA variant for the first time in a recessively inherited phenotype, though further research is required.

The structure of GENEA was modelled with great confidence via Phyre2. Leu357 is a part of the beta strand, and substitution of proline at this position may disrupt the hydrogen bonding impacting the dynamics of the allosteric binding site. The propensity value for beta strand is very high for leucine as compared to proline.

160

Chapter 04: Discussion

Fig.4.10: Clustal Omega Alignment of respective orthologues of GENEA from different classes of vertebrates. Leu357 is absolutely conserved (boxed).

161

Chapter 04: Discussion

Family with three segregating variants

Three variant in different genes were found to segregate with the phenotype in family RDHT16. GENEB variant was fully conserved in evolution (Fig.4.11) and was absent in controls and public databases. Some heterozygous copy number variants in this gene were found in DECIPHER (https://decipher.sanger.ac.uk/), where the patients had somewhat overlapping neurological phenotypes. Protein atlas indicates that the protein localizes to the nucleus. This residue p.(Lys231Glu) is located near the C-terminal of the protein and could potentially play an important role in localization of protein which remains to be verified. The mouse knockout model for this gene exhibits metabolic phenotype of decreased circulating insulin level in International Mouse Phenotyping Consortium (IMPC) (http://www.mousephenotype.org/). Some movement disorders are known to be neuro-metabolic in nature and are known to cause dystonia (Eggink et al. 2014; Gouider- Khouja et al. 2010). High pathogenicity scores, absence in normal control and absolute conservation during evolution makes it an attractive candidate for disease association.

The second segregating variant p.(Arg564Trp) in GENEC is also conserved (Fig.4.12 ). The variant was absent in control chromosomes but was present in public database at a low frequency with no homozygous individual. The human protein atlas indicates this protein to be an intracellular plasma membrane protein. The heterozygous exon deleted mouse model for the gene was present in International Mouse Phenotyping Consortium (IMPC) (http://www.mousephenotype.org/) but no phenotypic deviation was detected in comparison to wild type animals. This is consistent with the findings presented in this work that the heterozygous carriers of variant have no phenotype. Further work needs to be carried out in order to see the contribution of homozygous variants in this gene to phenotypes in both mouse and humans.

CGN variant p.(Glu699Gly) was not conserved even in chicken and rhesus though it was absent in normal controls and public databases. This gene was also submitted to Genematcher (https://www.genematcher.org/). One match with an individual homozygous for variant in CGN was found. He had the phenotype of short stature, developmental delay and hirsutism. Human protein atlas indicates this protein to be a transmembrane protein which is also present in cell junctions in low intensities. However, due to non-conservation in rhesus (data not shown), this variant is not likely to cause the disorder in patients.

162

Chapter 04: Discussion

Fig.4.11: Clustal Omega Alignment of respective orthologues of GENEB from different classes of vertebrates. Lys231 is absolutely conserved (boxed).

Fig.4.12: Clustal Omega Alignment of respective orthologues of GENEC from different classes of vertebrates. Arg564 is absolutely conserved (boxed).

163

Chapter 04: Discussion

Frabin stability and localization is not affected by compound pathogenic missense variants

The genetic cause in a patient of family UCL-HT01 were compound heterozygous variants in FGD4. Biallelic mutations in this gene have been associated with recessive form of Charcot-Marie-Tooth disease, type 4H, CMT4H (OMIM 609311).

Frabin encoded by FGD4 is a protein of 766 amino acids (Fig.4.13). It has five important functional domains and has been reported to play important roles in organization of actin cytoskeleton and microspikes formation. FAB/ABD domain is important for binding directly to actin. CRD/FYVE domain has an important role in activation of JNK pathway via activation of cdc42 (Ono et al. 2000).

Frabin is an actin interacting and GDT/GTP exchange protein. Frabin dependent cdc42 activation and frabin-actin interaction synergistically work to organize the actin cytoskeleton in cells. The dynamic reorganization of cytoskeleton is critical for many cellular functions including cell shape changes, motility and adhesion (Nakanishi and Takai 2008). The possible effect of compound heterozygous variants on frabin protein stability and localization was estimated in this study. They were found not to be affected. The fibroblasts of frabin patient had same proliferation rates as those of normal cells as observed in this study. There is a possibility that some membrane trafficking assays would highlight further how these variants lead to disease onset and progression.

164

Chapter 04: Discussion

Fig.4.13: Frabin structure along with domain organization. The variants reported here boxed. ABD indicates actin binding domain, DH indicates Dbl homology domain, PH1 and PH2 indicate Plekstrin homology domain 1 and 2, FYVE indicates a cysteine rich domain which is named after identification of this domain in four proteins, Fab1, YOTB, Vac 1 and EEA1. aa, amino acids.

165

Chapter 04: Discussion

Families in which no genetic cause was identified

In families, RDHT07, RDHT09, RDHT14, RDHT11 and RDHT12, exome data was filtered, analysed and some variants were checked but genetic cause was not identified for the disease phenotype. For family RDHT07, none of the variants segregated with phenotype in both patients. One of these variants which was homozygous in the elder sister was c.972C>A;p.(Asp324Glu) in SACS. This residue was not conserved in pika and alpaca where it was replaced with asparagine or serine respectively. This variant has an allele frequency of 0.008 and one individual homozygous for the variant has been reported in GnomAD database. Therefore, it is not a likely disease causing variant, though there is a significant match of phenotype of this patient with those of patients having SACS variants in homozygous form (OMIM 270550).

There is a possibility that the pathogenic variant in these families is located in regions which were missed during exome capture. Secondly, the variants could be present in regulatory regions which are also not targeted by whole exome sequencing. Whole genome sequencing and subsequent data analysis will probably solve these yet unsolved cases.

166

Chapter 04: Discussion

Frequency of AAGGG repeat expansion in RFC1

Cerebellar atrophy, neuropathy and vestibular areflexia syndrome (CANVAS) (OMIM 614575) is a complex phenotype characterized by cerebellar ataxia, sensory neuropathy and bilateral vestibulopathy. The patients suffering from this disorder were found to be homozygous for a novel repeat expansion identified in RFC1 by whole genome sequencing (Cortese et al. in press). 304 control individuals were assessed for the allelic conformation at this locus using the flanking PCR and RPPCR. None of the individual carried the expansion in homozygous form. Allelic frequency for this pathogenic expansion was found to be 0.7%, although the locus showed hypermutability.

Some samples which gave no product on flanking PCR and tested negative for RPPCR which targeted AAAAG, AAAGG, or AAGGG repeat units, although the DNA integrity was not compromised. This suggests the existence of other possible allelic conformations in 3% (n=18) of tested chromosomes. These results together with Southern blotting and studies on RFC1 expression at RNA and protein level led to the discovery of a novel cause of late onset ataxia as RFC1 repeat expansion (Cortese et al. in press). The results suggest that the recessive AAGGG expansion in RFC1 represents a frequent cause of late-onset ataxia, rith clinical similarities and disease trequency to that ot Friedreich’s ataxia in European population.

167

Conclusion

Conclusion

Movement disorders constitute a large group of neurological disorders with significant overlap of phenotype. Sometimes these disorders are misdiagnosed and make the management and treatment nearly impossible. This study demonstrated the genetic basis of such disorders which are recessively inherited in the participating families.

Out of total 16 families, 8 received a molecular diagnosis. Allelic spectrum of known disease causing genes was expanded. Four novel biallelic variants were identified in SETX and ALS2 as cause of the disorder in four families. A nonsense ATM variant was identified for the first time in homozygous form in a family confirming the pathogenicity of the variant in biallelic form. A known nonsense homozygous C19orf12 variant segregating in a large family with normal heterozygotes negates the monoallelic nature of disorder. A known TFG variant identified in a family extended the phenotypic spectrum significantly and suggested the different impact caused by the same variant.

DRD4 recessive missense variant segregating with dystonia phenotype in a family suggests dystonia association of DRD4 for the first time. The association of homozygous variant in GENEA with delayed milestones and its recessive inheritance is also suggested for the first time. This expands the inheritance pattern from dominant to recessive for GENEA variants. However, proof for association of GENEB and GENEC variants in genetics of dystonia requires support from additional families with similar phenotypes segregating mutations in these genes.

In one family collected at UCL, two novel biallelic variants were identified in FGD4 as cause of Charcot-Marie-Tooth disease, type 4H in a South Asian patient. The normal growth rates of fibroblast cells derived from the patient, the normal localization and stability of the resulting frabin protein underscores the need for additional membrane trafficking assays to understand the disease mechanism due to these variants.

168

Conclusion

Screening for a novel bilallelic repeat expansion in RFC1 in a cohort of 304 unaffected individuals by flanking PCR and RPPCR demonstrated the frequency of this expansion to be 0.7% in normal population with no individual homozygous for the variant. This screening validated the association of the repeat expansion in RFC1 with late onset ataxia in the European population for the first time.

In future, the families in which no genetic cause is identified yet, will be explored further through the technology of whole genome sequencing. This will perhaps yield findings related to genetic, allelic diversity and perhaps phenotypic heterogeneity of such disorders.

169

References

References

Abele M, Burk K, Schols L, Schwartz S, Besenthal I, Dichgans J, Zuhlke C, Riess O, Klockgether T. 2002. The aetiology of sporadic adult-onset ataxia. Brain. 125 (Pt 5): 961-968. Akbar U, Ashizawa T. 2015. Ataxia. Neurol Clin. 33 (1): 225-248. Al Tassan N, Khalil D, Shinwari J, Al Sharif L, Bavi P, Abduljaleel Z, Abu Dhaim N, Magrashi A, Bobis S, Ahmed H, et al. 2012. A missense mutation in PIK3R5 gene in a family with ataxia and oculomotor apraxia. Hum Mutat. 33 (2): 351-354. Alavi A, Shamshiri H, Nafissi S, Khani M, Klotzle B, Fan JB, Steemers F, Elahi E. 2015. HMSN-P caused by p.Pro285Leu mutation in TFG is not confined to patients with Far East ancestry. Neurobiol Aging. 36 (3): 1606 e1601-1607. Albanese A, Bhatia K, Bressman SB, Delong MR, Fahn S, Fung VS, Hallett M, Jankovic J, Jinnah HA, Klein C, et al. 2013. Phenomenology and classification of dystonia: a consensus update. Mov Disord. 28 (7): 863-873. Ali K, Morris HR. 2015. Parkinson's disease: chameleons and mimics. Pract Neurol. 15 (1): 14-25. Ali Z, Klar J, Jameel M, Khan K, Fatima A, Raininko R, Baig S, Dahl N. 2016. Novel SACS mutations associated with intellectual disability, epilepsy and widespread supratentorial abnormalities. J Neurol Sci. 371: 105-111. Alirezaie N, Kernohan KD, Hartley T, Majewski J, Hocking TD. 2018. ClinPred: Prediction Tool to Identify Disease-Relevant Nonsynonymous Single-Nucleotide Variants. Am J Hum Genet. 103 (4): 474-483. Amorim S, Heise CO, Santos S, Macedo-Souza LI, Zatz M, Kok F. 2014. Nerve conduction studies in spastic paraplegia, optic atrophy, and neuropathy (SPOAN) syndrome. Muscle & nerve. 49 (1): 131-133. Anheim M, Chaigne D, Fleury M, Santorelli FM, De Seze J, Durr A, Brice A, Koenig M, Tranchant C. 2008. [Autosomal recessive spastic ataxia of Charlevoix-Saguenay: study of a family and review of the literature]. Rev Neurol (Paris). 164 (4): 363-368. Anttonen AK, Siintola E, Tranebjaerg L, Iwata NK, Bijlsma EK, Meguro H, Ichikawa Y, Goto J, Kopra O, Lehesjoki AE. 2008a. Novel SIL1 mutations and exclusion of functional candidate genes in Marinesco-Sjogren syndrome. Eur J Hum Genet. 16 (8): 961-969.

170

References

Anttonen AK, Siintola E, Tranebjaerg L, Iwata NK, Bijlsma EK, Meguro H, Ichikawa Y, Goto J, Kopra O, Lehesjoki AE. 2008b. Novel SIL1 mutations and exclusion of functional candidate genes in Marinesco-Sjogren syndrome. Eur J Hum Genet. 16 (8): 961-969. Arif B, Grunewald A, Fatima A, Ramirez A, Ali A, Bruggemann N, Wurfel J, Rolfs A, Lohmann K, Malik A, et al. 2011. An unusual neurological syndrome of crawling gait, dystonia, pyramidal signs, and limited speech. Mov Disord. 26 (12): 2279-2283. Arif B, Kumar KR, Seibler P, Vulinovic F, Fatima A, Winkler S, Nurnberg G, Thiele H, Nurnberg P, Jamil AZ, et al. 2013. A novel OPA3 mutation revealed by exome sequencing: an example of reverse phenotyping. JAMA Neurol. 70 (6): 783-787. Assoum M, Salih MA, Drouot N, H'Mida-Ben Brahim D, Lagier-Tourenne C, AlDrees A, Elmalik SA, Ahmed TS, Seidahmed MZ, Kabiraj MM, et al. 2010. Rundataxin, a novel protein with RUN and diacylglycerol binding domains, is mutant in a new recessive ataxia. Brain. 133 (Pt 8): 2439-2447. Barbey A, Bloch J, Vingerhoets FJ. 2015. DBS in Dystonia and Other Hyperkinetic Movement Disorders. Curr Treat Options Neurol. 17 (9): 373. Bassuk AG, Chen YZ, Batish SD, Nagan N, Opal P, Chance PF, Bennett CL. 2007. In cis autosomal dominant mutation of Senataxin associated with tremor/ataxia syndrome. Neurogenetics. 8 (1): 45-49. Bayat V, Thiffault I, Jaiswal M, Tetreault M, Donti T, Sasarman F, Bernard G, Demers- Lamarche J, Dicaire MJ, Mathieu J, et al. 2012. Mutations in the mitochondrial methionyl-tRNA synthetase cause a neurodegenerative phenotype in flies and a recessive ataxia (ARSAL) in humans. PLoS Biol. 10 (3): e1001288. Becherel OJ, Yeo AJ, Stellati A, Heng EY, Luff J, Suraweera AM, Woods R, Fleming J, Carrie D, McKinney K, et al. 2013. Senataxin plays an essential role with DNA damage response proteins in meiotic recombination and gene silencing. PLoS Genet. 9 (4): e1003435. Beetz C, Johnson A, Schuh AL, Thakur S, Varga RE, Fothergill T, Hertel N, Bomba-Warczak E, Thiele H, Nurnberg G, et al. 2013a. Inhibition of TFG function causes hereditary axon degeneration by impairing endoplasmic reticulum structure. Proc Natl Acad Sci U S A. 110 (13): 5091-5096. Beetz C, Johnson A, Schuh AL, Thakur S, Varga RE, Fothergill T, Hertel N, Bomba-Warczak E, Thiele H, Nurnberg G, et al. 2013b. Inhibition of TFG function causes hereditary axon degeneration by impairing endoplasmic reticulum structure. Proceedings of the National Academy of Sciences of the United States of America. 110 (13): 5091-5096. 171

References

Ben Hamida M, Belal S, Sirugo G, Ben Hamida C, Panayides K, Ionannou P, Beckmann J, Mandel JL, Hentati F, Koenig M, et al. 1993. Friedreich's ataxia phenotype not linked to chromosome 9 and associated with selective autosomal recessive vitamin E deficiency in two inbred Tunisian families. Neurology. 43 (11): 2179-2183. Berger ND, Stanley FKT, Moore S, Goodarzi AA. 2017. ATM-dependent pathways of chromatin remodelling and oxidative DNA damage responses. Philos Trans R Soc Lond B Biol Sci. 372 (1731). Bernstein JL, Concannon P. 2017. ATM, radiation, and the risk of second primary breast cancer. Int J Radiat Biol. 1-7. Bernstein NK, Williams RS, Rakovszky ML, Cui D, Green R, Karimi-Busheri F, Mani RS, Galicia S, Koch CA, Cass CE, et al. 2005. The molecular architecture of the mammalian DNA repair enzyme, polynucleotide kinase. Mol Cell. 17 (5): 657-670. Bird TD, Ott J, Giblett ER. 1982. Evidence for linkage of Charcot-Marie-Tooth neuropathy to the Duffy locus on chromosome 1. Am J Hum Genet. 34 (3): 388-394. Blackstone C. 2012. Cellular pathways of hereditary spastic paraplegia. Annual review of neuroscience. 35: 25-47. Blackstone C, O'Kane CJ, Reid E. 2011. Hereditary spastic paraplegias: membrane traffic and the motor pathway. Nat Rev Neurosci. 12 (1): 31-42. Bomar JM, Benke PJ, Slattery EL, Puttagunta R, Taylor LP, Seong E, Nystuen A, Chen W, Albin RL, Patel PD, et al. 2003. Mutations in a novel gene encoding a CRAL-TRIO domain cause human Cayman ataxia and ataxia/dystonia in the jittery mouse. Nat Genet. 35 (3): 264-269. Bonello M, Ray P. 2016. A Case of Ataxia with Isolated Vitamin E Deficiency Initially Diagnosed as Friedreich's Ataxia. Case Rep Neurol Med. 2016: 8342653. Borrell E. 2000. Hypokinetic movement disorders. J Neurosci Nurs. 32 (5): 254-255. Bouhlal Y, Zouari M, Kefi M, Ben Hamida C, Hentati F, Amouri R. 2008. Autosomal recessive ataxia caused by three distinct gene defects in a single consanguineous family. J Neurogenet. 22 (2): 139-148. Bouslam N, Bouhouche A, Benomar A, Hanein S, Klebe S, Azzedine H, Di Giandomenico S, Boland-Auge A, Santorelli FM, Durr A, et al. 2007. A novel locus for autosomal recessive spastic ataxia on chromosome 17p. Hum Genet. 121 (3-4): 413-420. Boutin-Ganache I, Raposo M, Raymond M, Deschepper CF. 2001. M13-tailed primers improve the readability and usability of microsatellite analyses performed with two different allele-sizing methods. Biotechniques. 31 (1): 24-26, 28. 172

References

Bradshaw TY, Romano LE, Duncan EJ, Nethisinghe S, Abeti R, Michael GJ, Giunti P, Vermeer S, Chapple JP. A reduction in Drp1-mediated fission compromises mitochondrial health in autosomal recessive spastic ataxia of Charlevoix Saguenay. Hum Mol Genet. Breedveld GJ, van Wetten B, te Raa GD, Brusse E, van Swieten JC, Oostra BA, Maat-Kievit JA. 2004. A new locus for a childhood onset, slowly progressive autosomal recessive spinocerebellar ataxia maps to chromosome 11p15. J Med Genet. 41 (11): 858-866. Broccoletti T, Del Giudice E, Cirillo E, Vigliano I, Giardino G, Ginocchio VM, Bruscoli S, Riccardi C, Pignata C. 2011. Efficacy of very-low-dose betamethasone on neurological symptoms in ataxia-telangiectasia. Eur J Neurol. 18 (4): 564-570. Brock C, Schaefer M, Reusch HP, Czupalla C, Michalke M, Spicher K, Schultz G, Nurnberg B. 2003. Roles of G beta gamma in membrane recruitment and activation of p110 gamma/p101 phosphoinositide 3-kinase gamma. J Cell Biol. 160 (1): 89-99. Brunberg JA. 2008. Ataxia. Am J Neuroradiol. 29 (7): 1420-1422.

Buoni S, Zannolli R, Sorrentino L, Fois A. 2006. Betamethasone and improvement of neurological symptoms in ataxia-telangiectasia. Arch Neurol. 63 (10): 1479-1482. Byrne S, Dlamini N, Lumsden D, Pitt M, Zaharieva I, Muntoni F, King A, Robert L, Jungbluth H. 2015. SIL1-related Marinesco-Sjoegren syndrome (MSS) with associated motor neuronopathy and bradykinetic movement disorder. Neuromuscul Disord. 25 (7): 585- 588. Carocci A, Catalano A, Sinicropi MS, Genchi G. 2018. Oxidative stress and neurodegeneration: the involvement of iron. Biometals. Catania A, Battini R, Pippucci T, Pasquariello R, Chiapparini ML, Seri M, Garavaglia B, Zorzi G, Nardocci N, Ghezzi D, et al. 2018. R106C TFG variant causes infantile neuroaxonal dystrophy "plus" syndrome. Neurogenetics. 19 (3): 179-187. Chaki M, Airik R, Ghosh AK, Giles RH, Chen R, Slaats GG, Wang H, Hurd TW, Zhou W, Cluckey A, et al. 2012. Exome capture reveals ZNF423 and CEP164 mutations, linking renal ciliopathies to DNA damage response signaling. Cell. 150 (3): 533-548. Chen YZ, Bennett CL, Huynh HM, Blair IP, Puls I, Irobi J, Dierick I, Abel A, Kennerson ML, Rabin BA, et al. 2004. DNA/RNA helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis (ALS4). Am J Hum Genet. 74 (6): 1128-1135. Choudhury SD, Vs A, Mushtaq Z, Kumar V. 2017. Altered translational repression of an RNA- binding protein, Elav by AOA2-causative Senataxin mutation. Synapse. 71 (5).

173

References

Ciesla N, Dinglas V, Fan E, Kho M, Kuramoto J, Needham D. 2011. Manual muscle testing: a method of measuring extremity muscle strength applied to critically ill patients. J Vis Exp. (50). Cortese A, Simone R, Sullivan R, Vandrovcova J, Tariq H, Yau W, Humphrey J, Jaunmuktane J, Sivakumar P, Ilyas M, et al. in press. Expansion of a Recessive Intronic AAGGG Repeat in the RFC1 Gene is a common cause of Late-Onset Ataxia. . Nature Genetics. Cossu G, Colosimo C. 2017. Hyperkinetic Movement Disorder Emergencies. Curr Neurol Neurosci Rep. 17 (1): 6. Cremonesi L, Cozzi A, Girelli D, Ferrari F, Fermo I, Foglieni B, Levi S, Bozzini C, Camparini M, Ferrari M, et al. 2004. Case report: a subject with a mutation in the ATG start codon of L-ferritin has no haematological or neurological symptoms. J Med Genet. 41 (6): e81. Criscuolo C, Sacca F, De Michele G, Mancini P, Combarros O, Infante J, Garcia A, Banfi S, Filla A, Berciano J. 2005. Novel mutation of SACS gene in a Spanish family with autosomal recessive spastic ataxia. Mov Disord. 20 (10): 1358-1361. D'Arrigo S, Riva D, Bulgheroni S, Chiapparini L, Castellotti B, Gellera C, Pantaleoni C. 2008. Ataxia with oculomotor apraxia type 1 (AOA1): clinical and neuropsychological features in 2 new patients and differential diagnosis. J Child Neurol. 23 (8): 895-900. Daud S, Kakar N, Goebel I, Hashmi AS, Yaqub T, Nurnberg G, Nurnberg P, Morris-Rosendahl DJ, Wasim M, Volk AE, et al. 2016. Identification of two novel ALS2 mutations in infantile-onset ascending hereditary spastic paraplegia. Amyotroph Lateral Scler Frontotemporal Degener. 17 (3-4): 260-265. Daugherty M, Polanuyer B, Farrell M, Scholle M, Lykidis A, de Crecy-Lagard V, Osterman A. 2002. Complete reconstitution of the human coenzyme A biosynthetic pathway via comparative genomics. J Biol Chem. 277 (24): 21431-21439. Davis MY, Keene CD, Swanson PD, Sheehy C, Bird TD. 2013. Novel mutations in ataxia telangiectasia and AOA2 associated with prolonged survival. J Neurol Sci. 335 (1-2): 134-138. de Leon D, Moskowitz CB, Stewart C. 1991. Proposed guidelines for videotaping individuals with movement disorders. J Neurosci Nurs. 23 (3): 191-193. Defazio G, Abbruzzese G, Livrea P, Berardelli A. 2004. Epidemiology of primary dystonia. Lancet Neurol. 3 (11): 673-678.

174

References

Delague V, Bareil C, Bouvagnet P, Salem N, Chouery E, Loiselet J, Megarbane A, Claustres M. 2001. Nonprogressive autosomal recessive ataxia maps to chromosome 9q34-9qter in a large consanguineous Lebanese family. Ann Neurol. 50 (2): 250-253. Dietz V. 2000. Spastic movement disorder. Spinal Cord. 38 (7): 389-393. Do HK, Jo GY, Kwon JK, Kim WJ. 2018. Botulinum Toxin-A Injection in the Treatment of Spasticity in a Infantile-Onset Neurodegeneration With Brain Iron Accumulation: A Case Report. Ann Rehabil Med. 42 (2): 363-367. Domingo A, Erro R, Lohmann K. 2016. Novel Dystonia Genes: Clues on Disease Mechanisms and the Complexities of High-Throughput Sequencing. Mov Disord. 31 (4): 471-477. Drew AP, Zhu D, Kidambi A, Ly C, Tey S, Brewer MH, Ahmad-Annuar A, Nicholson GA, Kennerson ML. 2015. Improved inherited peripheral neuropathy genetic diagnosis by whole-exome sequencing. Mol Genet Genomic Med. 3 (2): 143-154. Dupre N, Bouchard JP, Gros-Louis F, Rouleau GA. 2007. [Mutations in SYNE-1 lead to a newly discovered form of autosomal recessive cerebellar ataxia]. Med Sci. 23 (3): 261- 262. Duquette A, Roddier K, McNabb-Baltar J, Gosselin I, St-Denis A, Dicaire MJ, Loisel L, Labuda D, Marchand L, Mathieu J, et al. 2005. Mutations in senataxin responsible for Quebec cluster of ataxia with neuropathy. Ann Neurol. 57 (3): 408-414. Dusi S, Valletta L, Haack TB, Tsuchiya Y, Venco P, Pasqualato S, Goffrini P, Tigano M, Demchenko N, Wieland T, et al. 2014. Exome sequence reveals mutations in CoA synthase as a cause of neurodegeneration with brain iron accumulation. Am J Hum Genet. 94 (1): 11-22. Eggink H, Kuiper A, Peall KJ, Contarino MF, Bosch AM, Post B, Sival DA, Tijssen MA, de Koning TJ. 2014. Rare inborn errors of metabolism with movement disorders: a case study to evaluate the impact upon quality of life and adaptive functioning. Orphanet J Rare Dis. 9: 177. El-Shanti H, Daoud A, Sadoon AA, Leal SM, Chen S, Lee K, Spiegel R. 2006. A distinct autosomal recessive ataxia maps to chromosome 12 in an inbred family from Jordan. Brain Dev. 28 (6): 353-357. Elsayed LE, Mohammed IN, Hamed AA, Elseed MA, Johnson A, Mairey M, Mohamed HE, Idris MN, Salih MA, El-Sadig SM, et al. 2016. Hereditary spastic paraplegias: identification of a novel SPG57 variant affecting TFG oligomerization and description of HSP subtypes in Sudan. Eur J Hum Genet.

175

References

Engert JC, Berube P, Mercier J, Dore C, Lepage P, Ge B, Bouchard JP, Mathieu J, Melancon SB, Schalling M, et al. 2000. ARSACS, a spastic ataxia common in northeastern Quebec, is caused by mutations in a new gene encoding an 11.5-kb ORF. Nat Genet. 24 (2): 120-125. Eymard-Pierre E, Lesca G, Dollet S, Santorelli FM, di Capua M, Bertini E, Boespflug-Tanguy O. 2002. Infantile-onset ascending hereditary spastic paralysis is associated with mutations in the alsin gene. American journal of human genetics. 71 (3): 518-527. Eymard-Pierre E, Yamanaka K, Haeussler M, Kress W, Gauthier-Barichard F, Combes P, Cleveland DW, Boespflug-Tanguy O. 2006. Novel missense mutation in ALS2 gene results in infantile ascending hereditary spastic paralysis. Ann Neurol. 59 (6): 976-980. Ezquerra M, Compta Y, Marti MJ. 2011a. Identifying the genetic components underlying the pathophysiology of movement disorders. Appl Clin Genet. 4: 81-92. Ezquerra M, Compta Y, Marti MJ. 2011b. Identifying the genetic components underlying the pathophysiology of movement disorders. Appl Clin Genet. 4: 81-92. Fahn S. 2011. Classification of movement disorders. Mov Disord. 26 (6): 947-957. Fayyaz M, Jaffery SS, Anwer F, Zil EAA, Anjum I. 2018. The Effect of Physical Activity in Parkinson's Disease: A Mini-Review. Cureus. 10 (7): e2995. Flor-de-Lima F, Sampaio M, Nahavandi N, Fernandes S, Leao M. 2014. Alsin related disorders: literature review and case study with novel mutations. Case Rep Genet. 2014: 691515. Fogel BL, Cho E, Wahnich A, Gao F, Becherel OJ, Wang X, Fike F, Chen L, Criscuolo C, De Michele G, et al. 2014. Mutation of senataxin alters disease-specific transcriptional networks in patients with ataxia with oculomotor apraxia type 2. Hum Mol Genet. 23 (18): 4758-4769. Fogel BL, Perlman S. 2006. Novel mutations in the senataxin DNA/RNA helicase domain in ataxia with oculomotor apraxia 2. Neurology. 67 (11): 2083-2084. Fujiwara K, Toda H, Ikeguchi M. 2012. Dependence of alpha-helical and beta-sheet amino acid propensities on the overall protein fold type. BMC Struct Biol. 12: 18. Gabsi S, Gouider-Khouja N, Belal S, Fki M, Kefi M, Turki I, Ben Hamida M, Kayden H, Mebazaa R, Hentati F. 2001. Effect of vitamin E supplementation in patients with ataxia with vitamin E deficiency. Eur J Neurol. 8 (5): 477-481. Gagliardi M, Annesi G, Lesca G, Broussolle E, Iannello G, Vaiti V, Gambardella A, Quattrone A. 2015. C19orf12 gene mutations in patients with neurodegeneration with brain iron accumulation. Parkinsonism Relat Disord. 21 (7): 813-816. 176

References

Ghrooda S, Borys A, Spriggs E, Hegde M, Mhanni A. 2012. SETX gene novel mutations in a non-French Canadian with ataxia-oculomotor apraxia type 2. Parkinsonism Relat Disord. 18 (5): 700-701. Giardino G, Fusco A, Romano R, Gallo V, Maio F, Esposito T, Palamaro L, Parenti G, Salerno MC, Vajro P, et al. 2013. Betamethasone therapy in ataxia telangiectasia: unraveling the rationale of this serendipitous observation on the basis of the pathogenesis. Eur J Neurol. 20 (5): 740-747. Gouider-Khouja N, Kraoua I, Benrhouma H, Fraj N, Rouissi A. 2010. Movement disorders in neuro-metabolic diseases. Eur J Paediatr Neurol. 14 (4): 304-307. Grieco GS, Malandrini A, Comanducci G, Leuzzi V, Valoppi M, Tessa A, Palmeri S, Benedetti L, Pierallini A, Gambelli S, et al. 2004. Novel SACS mutations in autosomal recessive spastic ataxia of Charlevoix-Saguenay type. Neurology. 62 (1): 103-106. Groh M, Albulescu LO, Cristini A, Gromak N. 2016. Senataxin: Genome Guardian at the Interface of Transcription and Neurodegeneration. J Mol Biol. Gros-Louis F, Dupre N, Dion P, Fox MA, Laurent S, Verreault S, Sanes JR, Bouchard JP, Rouleau GA. 2007. Mutations in SYNE1 lead to a newly discovered form of autosomal recessive cerebellar ataxia. Nat Genet. 39 (1): 80-85. Gros-Louis F, Meijer IA, Hand CK, Dube MP, MacGregor DL, Seni MH, Devon RS, Hayden MR, Andermann F, Andermann E, et al. 2003. An ALS2 gene mutation causes hereditary spastic paraplegia in a Pakistani kindred. Ann Neurol. 53 (1): 144-145. Guzzi G, Della Torre A, Chirchiglia D, Volpentesta G, Lavano A. 2016. Critical reappraisal of DBS targeting for movement disorders. J Neurosurg Sci. 60 (2): 181-188. Harding AE. 1981. "Idiopathic" late onset cerebellar ataxia. A clinical and genetic study of 36 cases. J Neurol Sci. 51 (2): 259-271. Harlalka GV, McEntagart ME, Gupta N, Skrzypiec AE, Mucha MW, Chioza BA, Simpson MA, Sreekantan-Nair A, Pereira A, Gunther S, et al. 2016. Novel Genetic, Clinical, and Pathomechanistic Insights into TFG-Associated Hereditary Spastic Paraplegia. Hum mutat.37(11):1157-1161. Hartig MB, Iuso A, Haack T, Kmiec T, Jurkiewicz E, Heim K, Roeber S, Tarabin V, Dusi S, Krajewska-Walasek M, et al. 2011. Absence of an orphan mitochondrial protein, c19orf12, causes a distinct clinical subtype of neurodegeneration with brain iron accumulation. Am J Hum Genet. 89 (4): 543-550.

177

References

Hogarth P, Gregory A, Kruer MC, Sanford L, Wagoner W, Natowicz MR, Egel RT, Subramony SH, Goldman JG, Berry-Kravis E, et al. 2013. New NBIA subtype: genetic, clinical, pathologic, and radiographic features of MPAN. Neurology. 80 (3): 268-275. Hollstein R, Parry DA, Nalbach L, Logan CV, Strom TM, Hartill VL, Carr IM, Korenke GC, Uppal S, Ahmed M, et al. 2015. HACE1 deficiency causes an autosomal recessive neurodevelopmental syndrome. J Med Genet. 52 (12): 797-803. Houlden H, Hammans S, Katifi H, Reilly MM. 2009. A novel Frabin (FGD4) nonsense mutation p.R275X associated with phenotypic variability in CMT4H. Neurology. 72 (7): 617-620. Hoyle JC, Isfort MC, Roggenbuck J, Arnold WD. 2015. The genetics of Charcot-Marie-Tooth disease: current trends and future implications for diagnosis and management. Appl Clin Genet. 8: 235-243. Humphrey W, Dalke A, Schulten K. 1996. VMD: visual molecular dynamics. J Mol Graph. 14 (1): 33-38, 27-38. Hussain R, Bittles AH. 1998. The prevalence and demographic characteristics of consanguineous marriages in Pakistan. J Biosoc Sci. 30 (2): 261-275. Ishiura H, Sako W, Yoshida M, Kawarai T, Tanabe O, Goto J, Takahashi Y, Date H, Mitsui J, Ahsan B, et al. 2012. The TRK-fused gene is mutated in hereditary motor and sensory neuropathy with proximal dominant involvement. Am J Hum Genet. 91 (2): 320-329. Ishiura H, Tsuji S. 2013. [Hereditary motor and sensory neuropathy with proximal dominant involvement (HMSN-P) is caused by a mutation in TFG]. Rinsho Shinkeigaku. 23 (11): 1203-1205. Ivanova EL, Mau-Them FT, Riazuddin S, Kahrizi K, Laugel V, Schaefer E, de Saint Martin A, Runge K, Iqbal Z, Spitz MA, et al. 2017. Homozygous Truncating Variants in TBC1D23 Cause Pontocerebellar Hypoplasia and Alter Cortical Development. Am J Hum Genet. 101 (3): 428-440. Iwabuchi S, Koh JY, Wang K, Ho KW, Harata NC. 2013. Minimal Change in the cytoplasmic calcium dynamics in striatal GABAergic neurons of a DYT1 dystonia knock-in mouse model. PLoS One. 8 (11): e80793. Iwasa K, Shima K, Komai K, Nishida Y, Yokota T, Yamada M. 2014. Retinitis pigmentosa and macular degeneration in a patient with ataxia with isolated vitamin E deficiency with a novel c.717 del C mutation in the TTPA gene. J Neurol Sci. 345 (1-2): 228-230. Jarman PR, Wood NW. 2002. Genetics of movement disorders and ataxia. J Neurol Neurosurg Psychiatry. 73 Suppl 2: II22-26. 178

References

Jinnah HA, Factor SA. 2015. Diagnosis and treatment of dystonia. Neurol Clin. 33 (1): 77-100. Kara E, Tucci A, Manzoni C, Lynch DS, Elpidorou M, Bettencourt C, Chelban V, Manole A, Hamed SA, Haridy NA, et al. 2016. Genetic and phenotypic characterization of complex hereditary spastic paraplegia. Brain. 139 (Pt 7): 1904-1918. Kelley LA, Mezulis S, Yates CM, Wass MN, Sternberg MJ. 2015. The Phyre2 web portal for protein modeling, prediction and analysis. Nat Protoc. 10 (6): 845-858. Kenna KP, McLaughlin RL, Byrne S, Elamin M, Heverin M, Kenny EM, Cormican P, Morris DW, Donaghy CG, Bradley DG, et al. 2013. Delineating the genetic heterogeneity of ALS using targeted high-throughput sequencing. J Med Genet. 50 (11): 776-783. Khani M, Shamshiri H, Alavi A, Nafissi S, Elahi E. 2016. Identification of novel TFG mutation in HMSN-P pedigree: Emphasis on variable clinical presentations. J Neurol Sci. 369: 318-323. Kirchner H, Kremmyda O, Hufner K, Stephan T, Zingler V, Brandt T, Jahn K, Strupp M. 2011. Clinical, electrophysiological, and MRI findings in patients with cerebellar ataxia and a bilaterally pathological head-impulse test. Ann N Y Acad Sci. 1233: 127-138. Krebs CE, Paisan-Ruiz C. 2012. The use of next-generation sequencing in movement disorders. Front Genet. 3: 75. Kumar KR. 2015. Mutations in hippocalcin and autosomal recessive dystonia: a role for perturbed calcium signaling? Mov Disord. 30 (7): 911. Kurosaki T, Maquat LE. 2016. Nonsense-mediated mRNA decay in humans at a glance. J Cell Sci. 129 (3): 461-467. Kwon JM, Goate AM. 2000. The candidate gene approach. Alcohol Res Health. 24 (3): 164- 168. Lambourne OA, Mehellou Y. 2018. Chemical Strategies for Activating PINK1, a Protein Kinase Mutated in Parkinson's Disease. Chembiochem. Lanciego JL, Luquin N, Obeso JA. 2012. Functional neuroanatomy of the basal ganglia. Cold Spring Harb Perspect Med. 2 (12): a009621. Lavin MF, Yeo AJ, Becherel OJ. 2013. Senataxin protects the genome: Implications for neurodegeneration and other abnormalities. Rare Diseases. 1: e25230. Lee WW, Jeon B, Kim R. 2018. Expanding the Spectrum of Dopa-Responsive Dystonia (DRD) and Proposal for New Definition: DRD, DRD-plus, and DRD Look-alike. J Korean Med Sci. 33 (28): e184.

179

References

Li X, Menade M, Kozlov G, Hu Z, Dai Z, McPherson PS, Brais B, Gehring K. 2015. High- Throughput Screening for Ligands of the HEPN Domain of Sacsin. PLoS One. 10 (9): e0137298. Lunati A, Lesage S, Brice A. 2018. The genetic landscape of Parkinson's disease. Rev Neurol (Paris). Macedo-Souza LI, Kok F, Santos S, Amorim SC, Starling A, Nishimura A, Lezirovitz K, Lino AM, Zatz M. 2005. Spastic paraplegia, optic atrophy, and neuropathy is linked to chromosome 11q13. Annals of neurology. 57 (5): 730-737. Mansfield SA, Pilarski R, Agnese DM. 2017. ATM mutations for surgeons. Fam Cancer. 16 (3): 407-410. Mariani LL, Rivaud-Pechoux S, Charles P, Ewenczyk C, Meneret A, Monga BB, Fleury MC, Hainque E, Maisonobe T, Degos B, et al. 2017. Comparing ataxias with oculomotor apraxia: a multimodal study of AOA1, AOA2 and AT focusing on video-oculography and alpha-fetoprotein. Sci Rep. 7 (1): 15284. Mariotti C, Gellera C, Rimoldi M, Mineri R, Uziel G, Zorzi G, Pareyson D, Piccolo G, Gambi D, Piacentini S, et al. 2004. Ataxia with isolated vitamin E deficiency: neurological phenotype, clinical follow-up and novel mutations in TTPA gene in Italian families. Neurol Sci. 25 (3): 130-137. Marras C, Lang A, van de Warrenburg BP, Sue CM, Tabrizi SJ, Bertram L, Mercimek- Mahmutoglu S, Ebrahimi-Fakhari D, Warner TT, Durr A, et al. 2017. Nomenclature of genetic movement disorders: Recommendations of the International Parkinson and Movement Disorder Society task force. Mov Disord. 32 (5): 724-725. Marsden J, Harris C. 2011. Cerebellar ataxia: pathophysiology and rehabilitation. Clin Rehabil. 25 (3): 195-216. Martinez AR, Moro A, Abrahao A, Faber I, Borges CR, Rezende TJ, Martins CR, Jr., Moscovich M, Munhoz RP, Segal SL, et al. 2016. Nonneurological Involvement in Late-Onset Friedreich Ataxia (LOFA): Exploring the Phenotypes. Cerebellum. Melo US, Macedo-Souza LI, Figueiredo T, Muotri AR, Gleeson JG, Coux G, Armas P, Calcaterra NB, Kitajima JP, Amorim S, et al. 2015. Overexpression of KLC2 due to a homozygous deletion in the non-coding region causes SPOAN syndrome. Hum Mol Genet. 24 (24): 6877-6885. Migliaccio AA, Halmagyi GM, McGarvie LA, Cremer PD. 2004. Cerebellar ataxia with bilateral vestibulopathy: description of a syndrome and its characteristic clinical sign. Brain. 127 (Pt 2): 280-293. 180

References

Milone M, Massie R. 2010. Polymerase gamma 1 mutations: clinical correlations. Neurologist. 16 (2): 84-91. Monfrini E, Melzi V, Buongarzone G, Franco G, Ronchi D, Dilena R, Scola E, Vizziello P, Bordoni A, Bresolin N, et al. 2018. A de novo C19orf12 heterozygous mutation in a patient with MPAN. Parkinsonism Relat Disord. 48: 109-111. Moreira MC, Klur S, Watanabe M, Nemeth AH, Le Ber I, Moniz JC, Tranchant C, Aubourg P, Tazir M, Schols L, et al. 2004. Senataxin, the ortholog of a yeast RNA helicase, is mutant in ataxia-ocular apraxia 2. Nat Genet. 36 (3): 225-227. Mukherjee A, Chakravarty A. 2010. Spasticity mechanisms - for the clinician. Front Neurol. 1: 149. Musselman KE, Stoyanov CT, Marasigan R, Jenkins ME, Konczak J, Morton SM, Bastian AJ. 2014. Prevalence of ataxia in children: a systematic review. Neurology. 82 (1): 80-89. Muzaimi MB, Thomas J, Palmer-Smith S, Rosser L, Harper PS, Wiles CM, Ravine D, Robertson NP. 2004. Population based study of late onset cerebellar ataxia in south east Wales. J Neurol Neurosurg Psychiatry. 75 (8): 1129-1134. Nakanishi H, Takai Y. 2008. Frabin and other related Cdc42-specific guanine nucleotide exchange factors couple the actin cytoskeleton with the plasma membrane. J Cell Mol Med. 12 (4): 1169-1176. Navarro JA, Ohmann E, Sanchez D, Botella JA, Liebisch G, Molto MD, Ganfornina MD, Schmitz G, Schneuwly S. 2010. Altered lipid metabolism in a Drosophila model of Friedreich's ataxia. Hum Mol Genet. 19 (14): 2828-2840. Neychev VK, Gross RE, Lehericy S, Hess EJ, Jinnah HA. 2011. The functional neuroanatomy of dystonia. Neurobiol Dis. 42 (2): 185-201. Niemann A, Ruegg M, La Padula V, Schenone A, Suter U. 2005. Ganglioside-induced differentiation associated protein 1 is a regulator of the mitochondrial network: new implications for Charcot-Marie-Tooth disease. J Cell Biol. 170 (7): 1067-1078. Nishida K, Kuwano Y, Nishikawa T, Masuda K, Rokutan K. 2017. RNA Binding Proteins and Genome Integrity. Int J Mol Sci. 18 (7). Nolte J. 2009. The Human Brain: An Introduction to Its Functional Anatomy (6th ed.). Mosby/Elsevier. Nystuen A, Benke PJ, Merren J, Stone EM, Sheffield VC. 1996. A cerebellar ataxia locus identified by DNA pooling to search for linkage disequilibrium in an isolated population from the Cayman Islands. Hum Mol Genet. 5 (4): 525-531.

181

References

Ono Y, Nakanishi H, Nishimura M, Kakizaki M, Takahashi K, Miyahara M, Satoh-Horikawa K, Mandai K, Takai Y. 2000. Two actions of frabin: direct activation of Cdc42 and indirect activation of Rac. Oncogene. 19 (27): 3050-3058. Orrell RW. 1993. ALS2-Related Disorders. GeneReviews(R), Seattle (WA). Panzeri C, De Palma C, Martinuzzi A, Daga A, De Polo G, Bresolin N, Miller CC, Tudor EL, Clementi E, Bassi MT. 2006. The first ALS2 missense mutation associated with JPLS reveals new aspects of alsin biological function. Brain. 129 (Pt 7): 1710-1719. Paucar M, Malmgren H, Taylor M, Reynolds JJ, Svenningsson P, Press R, Nordgren A. 2016. Expanding the ataxia with oculomotor apraxia type 4 phenotype. Neurol Genet. 2 (1): e49. Pizzolato G, Mandat T. 2012. Deep brain stimulation for movement disorders. Front Integr Neurosci. 6: 2. Racis L, Tessa A, Pugliatti M, Storti E, Agnetti V, Santorelli FM. 2014. Infantile-onset ascending hereditary spastic paralysis: a case report and brief literature review. European journal of paediatric neurology : EJPN : official journal of the European Paediatric Neurology Society. 18 (2): 235-239. Ramirez-Zamora A, Zeigler W, Desai N, Biller J. 2015. Treatable causes of cerebellar ataxia. Mov Disord. 30 (5): 614-623. Rana AQ, Khan OA, Akthar R. 2013. Progressive ataxia associated with ocular apraxia type 1 (AOA1) with a presence of a novel mutation on the aprataxin gene. Ann Indian Acad Neurol. 16 (2): 269-271. Rana AQ, Saleh M, Yousuf MS, Mansoor W, Hussaini S, Rahman M, Iqbal Z. 2016. DOPA- sparing strategy in the treatment of young onset Parkinson's disease. J Neurosci Rural Pract. 7 (1): 67-69. Richard P, Feng S, Manley JL. 2013. A SUMO-dependent interaction between Senataxin and the exosome, disrupted in the neurodegenerative disease AOA2, targets the exosome to sites of transcription-induced DNA damage. Genes Dev. 27 (20): 2227-2232. Roohi J, Crowe J, Loredan D, Anyane-Yeboa K, Mansukhani MM, Omesi L, Levine J, Revah Politi A, Zha S. 2017. New diagnosis of atypical ataxia-telangiectasia in a 17-year-old boy with T-cell acute lymphoblastic leukemia and a novel ATM mutation. J Hum Genet. 62 (5): 581-584. Ruano L, Melo C, Silva MC, Coutinho P. 2014. The global epidemiology of hereditary ataxia and spastic paraplegia: a systematic review of prevalence studies. Neuroepidemiology. 42 (3): 174-183. 182

References

Rudnik-Schoneborn S, Arning L, Epplen JT, Zerres K. 2012. SETX gene mutation in a family diagnosed autosomal dominant proximal spinal muscular atrophy. Neuromuscul Disord. 22 (3): 258-262. Rudnik-Schoneborn S, Eggermann T, Kress W, Lemmink HH, Cobben JM, Zerres K. 2015. Clinical utility gene card for: Proximal spinal muscular atrophy (SMA) - update 2015. Eur J Hum Genet. 23 (11). Russo I, Cosentino C, Del Giudice E, Broccoletti T, Amorosi S, Cirillo E, Aloj G, Fusco A, Costanzo V, Pignata C. 2009. In ataxia-teleangiectasia betamethasone response is inversely correlated to cerebellar atrophy and directly to antioxidative capacity. Eur J Neurol. 16 (6): 755-759. Saitsu H, Nishimura T, Muramatsu K, Kodera H, Kumada S, Sugai K, Kasai-Yoshida E, Sawaura N, Nishida H, Hoshino A, et al. 2013. De novo mutations in the autophagy gene WDR45 cause static encephalopathy of childhood with neurodegeneration in adulthood. Nat Genet. 45 (4): 445-449, 449e441. Sandoval N, Platzer M, Rosenthal A, Dork T, Bendix R, Skawran B, Stuhrmann M, Wegner RD, Sperling K, Banin S, et al. 1999. Characterization of ATM gene mutations in 66 ataxia telangiectasia families. Hum Mol Genet. 8 (1): 69-79. Sasaki T, Tian H, Kukita Y, Inazuka M, Tahira T, Imai T, Yamauchi M, Saito T, Hori T, Hashimoto-Tamaoki T, et al. 1998. ATM mutations in patients with ataxia telangiectasia screened by a hierarchical strategy. Hum Mutat. 12 (3): 186-195. Sato K, Yabe I, Fukuda Y, Soma H, Nakahara Y, Tsuji S, Sasaki H. 2010. Mapping of autosomal dominant cerebellar ataxia without the pathogenic PPP2R2B mutation to the locus for spinocerebellar ataxia 12. Arch Neurol. 67 (10): 1257-1262. Schlaggar BL, Mink JW. 2003. Movement disorders in children. Pediatr Rev. 24 (2): 39-51. Schmucker S, Argentini M, Carelle-Calmels N, Martelli A, Puccio H. 2008. The in vivo mitochondrial two-step maturation of human frataxin. Hum Mol Genet. 17 (22): 3521- 3531. Schneeberger K. 2014. Using next-generation sequencing to isolate mutant genes from forward genetic screens. Nat Rev Genet. 15 (10): 662-676. Schulz JB. 2007. [Hereditary movement disorders]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz. 50 (12): 1524-1530.

183

References

Segawa M, Nomura Y, Hayashi M. 2013. Dopa-responsive dystonia is caused by particular impairment of nigrostriatal dopamine neurons different from those involved in Parkinson disease: evidence observed in studies on Segawa disease. Neuropediatrics. 44 (2): 61-66. Shinnick JE, Isaacs CJ, Vivaldi S, Schadt K, Lynch DR. 2016. Friedreich Ataxia and nephrotic syndrome: a series of two patients. BMC Neurol. 16: 3. Singleton AB. 2011. Exome sequencing: a transformative technology. Lancet Neurol. 10 (10): 942-946. Slosarek EL, Schuh AL, Pustova I, Johnson A, Bird J, Johnson M, Frankel EB, Bhattacharya N, Hanna MG, Burke JE, et al. 2018. Pathogenic TFG Mutations Underlying Hereditary Spastic Paraplegia Impair Secretory Protein Trafficking and Axon Fasciculation. Cell Rep. 24 (9): 2248-2260. Soderblom C, Blackstone C. 2006. Traffic accidents: molecular genetic insights into the pathogenesis of the hereditary spastic paraplegias. Pharmacol Ther. 109 (1-2): 42-56. Steeves TD, Day L, Dykeman J, Jette N, Pringsheim T. 2012. The prevalence of primary dystonia: a systematic review and meta-analysis. Mov Disord. 27 (14): 1789-1796. Storey E. 2014. Genetic cerebellar ataxias. Semin Neurol. 34 (3): 280-292. Szmulewicz DJ, Roberts L, McLean CA, MacDougall HG, Halmagyi GM, Storey E. 2016. Proposed diagnostic criteria for cerebellar ataxia with neuropathy and vestibular areflexia syndrome (CANVAS). Neurol Clin Pract. 6 (1): 61-68. Tariq H, Imran R, Naz S. 2018. A Novel Homozygous Variant of SETX Causes Ataxia with Oculomotor Apraxia Type 2. J Clin Neurol. 14 (4): 498-504. Tariq H, Mukhtar S, Naz S. 2017. A novel mutation in ALS2 associated with severe and progressive infantile onset of spastic paralysis. J Neurogenet. 31 (1-2): 26-29. Tariq H, Naz S. 2017. TFG associated hereditary spastic paraplegia: an addition to the phenotypic spectrum. Neurogenetics. 18 (2): 105-109. Taroni F, DiDonato S. 2004. Pathways to motor incoordination: the inherited ataxias. Nat Rev Neurosci. 5 (8): 641-655. Telatar M, Teraoka S, Wang Z, Chun HH, Liang T, Castellvi-Bel S, Udar N, Borresen-Dale AL, Chessa L, Bernatowska-Matuszkiewicz E, et al. 1998. Ataxia-telangiectasia: identification and detection of founder-effect mutations in the ATM gene in ethnic populations. Am J Hum Genet. 62 (1): 86-97.

184

References

Tsai PC, Huang YH, Guo YC, Wu HT, Lin KP, Tsai YS, Liao YC, Liu YT, Liu TT, Kao LS, et al. 2014. A novel TFG mutation causes Charcot-Marie-Tooth disease type 2 and impairs TFG function. Neurology. 83 (10): 903-912. Tuschl K, Clayton PT, Gospe SM, Jr., Mills PB. 1993. Dystonia/Parkinsonism, Hypermanganesemia, Polycythemia, and Chronic Liver Disease. GeneReviews(R), Seattle (WA). Vantaggiato C, Cantoni O, Guidarelli A, Romaniello R, Citterio A, Arrigoni F, Doneda C, Castelli M, Airoldi G, Bresolin N, et al. 2014. Novel SETX variants in a patient with ataxia, neuropathy, and oculomotor apraxia are associated with normal sensitivity to oxidative DNA damaging agents. Brain Dev. 36 (8): 682-689. Verschuuren-Bemelmans CC, Winter P, Sival DA, Elting JW, Brouwer OF, Muller U. 2008. Novel homozygous ALS2 nonsense mutation (p.Gln715X) in sibs with infantile-onset ascending spastic paralysis: the first cases from northwestern Europe. European journal of human genetics 16 (11): 1407-1411. Wakil SM, Ramzan K, Abuthuraya R, Hagos S, Al-Dossari H, Al-Omar R, Murad H, Chedrawi A, Al-Hassnan ZN, Finsterer J, et al. 2014. Infantile-onset ascending hereditary spastic paraplegia with bulbar involvement due to the novel ALS2 mutation c.2761C>T. Gene. 536 (1): 217-220. Wang S, Wacker D, Levit A, Che T, Betz RM, McCorvy JD, Venkatakrishnan AJ, Huang XP, Dror RO, Shoichet BK, et al. 2017. D4 dopamine receptor high-resolution structures enable the discovery of selective agonists. Science. 358 (6361): 381-386. Wang Y, Pan X, Xue D, Li Y, Zhang X, Kuang B, Zheng J, Deng H, Li X, Xiong W, et al. 2016. Application of next generation sequencing technology in Mendelian movement disorders. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 41 (2): 197-205. Witte K, Schuh AL, Hegermann J, Sarkeshik A, Mayers JR, Schwarze K, Yates JR, 3rd, Eimer S, Audhya A. 2011. TFG-1 function in protein secretion and oncogenesis. Nat Cell Biol. 13 (5): 550-558. Wright J, Teraoka S, Onengut S, Tolun A, Gatti RA, Ochs HD, Concannon P. 1996. A high frequency of distinct ATM gene mutations in ataxia-telangiectasia. Am J Hum Genet. 59 (4): 839-846. Xie F, Cen ZD, Xiao JF, Luo W. 2015. Novel compound heterozygous ALS2 mutations in two Chinese siblings with infantile ascending hereditary spastic paralysis. Neurological sciences : official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology. 36 (7): 1279-1280. 185

References

Yagi T, Ito D, Suzuki N. 2016. TFG-Related Neurologic Disorders: New Insights Into Relationships Between Endoplasmic Reticulum and Neurodegeneration. J Neuropathol Exp Neurol. 75 (4): 299-305.

Yang Y, Muzny DM, Reid JG, Bainbridge MN, Willis A, Ward PA, Braxton A, Beuten J, Xia F, Niu Z, et al. 2013. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med. 369 (16): 1502-1511. Yeo AJ, Becherel OJ, Luff JE, Graham ME, Richard D, Lavin MF. 2015. Senataxin controls meiotic silencing through ATR activation and chromatin remodeling. Cell Discov. 1: 15025. Zech M, Boesch S, Jochim A, Weber S, Meindl T, Schormair B, Wieland T, Lunetta C, Sansone V, Messner M, et al. 2017. Clinical exome sequencing in early-onset generalized dystonia and large-scale resequencing follow-up. Move Disord. 32(4):549-559

Zhou B, Westaway SK, Levinson B, Johnson MA, Gitschier J, Hayflick SJ. 2001. A novel pantothenate kinase gene (PANK2) is defective in Hallervorden-Spatz syndrome. Nat Genet. 28 (4): 345-349.

186

Appendices

Appendices

187

Appendices

Appendix A-1: Standardized Videotape Protocol-Dystonia

Beth Israel Medical Center: Movement Disorder Research Center, Department of Neurology

Roll-up sleeves to elbow, remove socks and shoes, roll pant legs up half-way to the knee, make intro card: On a piece of paper write:

 Videotape ID Code & your initials (A, G, h…. in the order ot appearance on the tape)  Date of Videotaping Please film entire segment with study participant sitting directly opposite you, i.e. Head on shot.  VIDEOTAPE THE INTRO CARDS for 10 seconds.

WHOLE BODY: Film participants sitting in a chair (preferably a chair without arms) at rest, feet flat on the floor, arms relaxed and sitting on each thigh. (Film whole body. Zoom into  different body parts, including the face).  HEAD: (Zoom in on face)

a. Slowly turn head to the left, to the right, up, straight ahead and downward. b. Looking straight ahead, slowly tilt head to the right, touching ear to shoulder. c. Repeat to the left.

188

Appendices

 ARMS AND HANDS: (zoom in on upper part of body)

a. Arms outstretched straight in front of you, with fingers spread out and palms down. b. Slowly turn hands over so that your palms face the ceiling. c. Slowly turn palms down again. d. Make a “Wing” position: (both elbors straight out to the side, each elbor bent with palms of hands icing the floor and both hands facing toward each other; close but not touching)  FINGER-TO-NOSE: (Videotape head-on, getting a close-up of the arm, elbow and finger)

a. Have someone hold their tinger out in tront ot you about an arm’s length aray you’re your tace. b. Using your index finger, touch your nose, then SLOWLY extend your arm to touch the other person’s tinger. (Make sure that you are extending your arm FULLY rhen reaching to touch the other person’s tinger) c. Do this 5 times with each hand.  RAPID SUCCESSIVE MOVEMENTS: (10 times each)

a. (zoom in on hands) Make big, quick taps with thumbs and index fingers. b. Fully open and close hands in tight fists. c. (zoom in on feet) Stomp foot, lifting the entire foot off the floor. Tap toe on the floor keeping the heel on the floor. Alternate tapping the heel and toe. Repeat each movement with other foot. STANDING: (zoom the posture)

a. Cross your arms over your chest and stand up unassisted, uncross arms. b. Face the camera, then make 4 quarter turns, stopping after each turn for video (Videotape whole body posture)

189

Appendices

PULL TEST: (Videotape the entire body).

In standing position, relaxed, with feet spread slightly apart. Have another individual stand about 0 teet behind person being videotaped: Place hiseher hands on the person’s shoulders and gently pull back, allowing the individual to catch his/her balance. (Be prepared to catch that  person before he/she falls on the floor). Repeat 2-3 times.

WALKING: Walk up and down halfway, going back and forth 4-5 times. (Video whole body,  zoom into different body parts, especially the feet. Try to videotape head-on).  VOICE:

a. Without mentioning names and relatedness, speak about any subject for about 1 minute. b. Repeat (Translated in Urdu)  Lovely yellow lilies  He saw half a seashell at the seashore  Ambling along rainy island avenue c. Take deep breath before each:  Hold a long “eeeeeee” sound tor 1t seconds  Hold a long “aaahhh” tor 1t seconds  WRITING:

Show entire upper body including arm, shoulder and neck for most of this segment.

Zoom in to show hand posture. Also videotape feet during the writing process.

. “Today is a sunny day in halitornia” (three times) (Translated in Urdu) . n entire line of connected cursive lower-case L’s (loops) (three times) 

190

Appendices A-2: Publications

Appendix A-2: Publications

191

Appendices A-2: Publications

192

Appendices A-2: Publications

193

Appendices A-2: Publications

194

Appendices A-2: Publications

195

Appendices A-2: Publications

196

Appendices A-2: Publications

197

Appendices A-2: Publications

198

Appendices A-2: Publications

199

Appendices A-2: Publications

200

Appendices A-2: Publications

201

Appendices A-2: Publications

202

Appendices A-2: Publications

203

Appendices A-2: Publications

204

Appendices A-2: Publications

205

Appendices A-2: Publications

206

Appendices A-2: Publications

207

Appendices A-2: Publications

208

Appendices A-2: Publications

209

Appendices A-2: Publications

210

Appendices A-2: Publications

211

Appendices A-2: Publications

212

Appendices A-2: Publications

213

Appendices A-2: Publications

214

Appendices A-2: Publications

215

Appendices A-2: Publications

216

Appendices A-2: Publications

217

Appendices A-2: Publications

218

Appendices A-2: Publications

219

Appendices A-2: Publications

220

Appendices A-2: Publications

221

Appendices A-2: Publications

222

Appendices A-2: Publications

223

Appendices A-2: Publications

224

Appendices A-2: Publications

225

Appendices A-2: Publications

226