mutation in vmat2 causing a pediatric neurotransmitter disease · 2015. 8. 13. · ii mutation in...

187
Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease by Jennifer Rilstone A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Institute of Medical Science University of Toronto © Copyright by Jennifer Rilstone 2015

Upload: others

Post on 08-Mar-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease

by

Jennifer Rilstone

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Institute of Medical Science University of Toronto

© Copyright by Jennifer Rilstone 2015

Page 2: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

ii

Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease

Jennifer Rilstone

Doctor of Philosophy

Institute of Medical Science University of Toronto

2015

Abstract

This thesis describes a new disease encompassing infantile-onset movement disorder (including

severe parkinsonism and nonambulation), mood disturbance, autonomic instability, and

developmental delay that was identified in eight cousins of a consanguineous Bedouin family in

Saudi Arabia. The autosomal recessive disease was hypothesized to be a disorder of monoamine

neurotransmission, and evidence supporting its causation by a mutation in SLC18A2 (which

encodes the vesicular monoamine transporter 2 [VMAT2]) was acquired by single nucleotide

polymorphism (SNP) genotyping, homozygosity analysis, and exome sequencing. VMAT2

translocates dopamine and serotonin into synaptic vesicles and is essential for motor control,

stable mood, and autonomic function. The loss of VMAT2 function was further confirmed

through biochemical assay of serotonin uptake. Consistent with a defect in vesicular monoamine

transport, treatment of the patients with levodopa was associated with worsening, whereas

treatment with direct dopamine agonists was followed by immediate ambulation, near-complete

correction of the movement disorder, and resumption of development. Understanding the

underlying mechanism of this disorder extends the spectrum of known pediatric neurotransmitter

diseases, serves as the first demonstration of mutation in VMAT2 causing a human phenotype,

and thereby provides new insight into the role of VMAT2 in monoamine homeostasis. This

thesis additionally demonstrates the utility of implementing genomic diagnosis in the clinic, with

Page 3: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

iii

respect to providing simple and effective treatments in a timely manner to improve outcomes for

patients with rare inborn errors of metabolism.

Page 4: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

iv

Acknowledgments I would first like to thank my supervisor, Dr. Berge Minassian, for his consistent support

throughout this degree, and more importantly—infinite patience. He taught me to think foremost

about the patients and their families, and that perspective motivated me when the work itself was

a struggle.

I would also like to thank the members of my advisory committee, Dr. Lucy Osborne and Dr.

Stephen Scherer, for timely strategic guidance and their support—particularly with the decision

to submit to NEJM and the decision to write.

I would additionally like to thank the members of the Minassian laboratory. Thank you to Peter

Wang and Xiaochu Zhao for their incredible technical expertise. I’d also like to thank the

summer students who worked with me over the years for giving me the opportunity to teach and

for demonstrating enthusiasm for this project: Tarek Abdelhalim, John Bilbily, Ari Damla, and

Alex Bilbily.

Support and encouragement through completion of this thesis were appreciated, with particular

thanks due to all those who routinely asked me whether I was done yet.

Finally, I’d like to thank my parents, David and Karen Rilstone, for being unendingly supportive

through a nonlinear path to a place in which I’m settled and happy. I’d also like to thank Katie

Mercer, mostly for existing, also for being curious and driven and inspiring me through example

to pursue new opportunities with confidence.

This work was supported by a Vanier Canada Graduate Scholarship from the Natural Sciences

and Engineering Research Council of Canada (NSERC).

Page 5: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

v

Contributions The intellectual content of this thesis, including the planning and direction of experiments, are

attributable to the author.

In Chapter 2, clinical characterization of the disease was performed in collaboration with Dr.

Reem Alkhater. As described throughout the text of the chapter, Dr. Alkhater identified this

family in clinical practice, performed neurological examinations and coordinated clinical

investigations (including the collection of urine and cerebrospinal fluid), collected written

informed consent for participation in the study, and collected blood samples from members of

the extended family for genetic analyses. Dr. Alkhater maintained contact with the family and

their physicians, providing treatment guidance and obtaining follow-up information. Ethical

approval for the study, including blood collection, was sought by the author. Genetic analyses of

candidate genes for pediatric neurotransmitter disorder were performed by the author.

Interpretations of CSF and urine analyses and the underlying pathophysiology of the disorder

were determined through discussions among Dr. Alkhater, Dr. Berge Minassian, and the author.

In Chapter 3, all genetic investigations were conceived and coordinated by the author. As

described in the methods section of the chapter, single nucleotide polymorphism analysis was

contracted through the University of Helsinki, and whole exome sequencing was performed by

The Center for Advanced Genomics at the Hospital for Sick Children. All mutation screening,

linkage and homozygosity analyses, and analyses of exome data were performed by the author,

as well as database searches and bioinformatic analyses of the described variant.

In Chapter 4, all functional analyses were conceived and performed by the author.

Acknowledgement is due to Dr. Robert Edwards and members of his laboratory for providing

Page 6: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

vi

guidance in performing the serotonin uptake assays, some of which were completed by the

author in a visit to his laboratory.

In Chapter 5, all future experiments have been conceived and planned by the author. Cloning

steps and the construction of cell lines described in this chapter were performed by the author,

with gratitude to Xiaochu Zhao for troubleshooting and completing the final step of the mouse

construct, and to the aforementioned summer students for assistance with cell culture

maintenance and PCR.

Page 7: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

vii

Table of Contents

Acknowledgments ................................................................................................................................ iv

Contributions .......................................................................................................................................... v

Table of Contents .................................................................................................................................vii

List of Tables .......................................................................................................................................... xi

List of Figures ...................................................................................................................................... xiii

List of Abbreviations ...................................................................................................................... xviii

Chapter 1 Literature Review ............................................................................................................. 1

1 Movement Disorder and Monoamine Deficiency ................................................................ 1

1.1 Pathophysiology ..................................................................................................................................... 1 1.2 The Biogenic Amines ............................................................................................................................ 4 1.3 Parkinson’s Disease .............................................................................................................................. 9 1.4 Pediatric Neurotransmitter Diseases ........................................................................................... 10

1.4.1 GTP Cyclohydrase I (GTPCH1) Deficiency ......................................................................................... 12 1.4.2 Sepiapterin Reductase (SR) Deficiency .................................................................................................. 15 1.4.3 6-Pyruvoyl Tetrahydropterin Synthase (PTPS) Deficiency ............................................................. 15 1.4.4 Dihydropteridine Reductase (DHPR) Deficiency ............................................................................... 16 1.4.5 Pterin-4α-Carbinolamine (PCD) Deficiency ......................................................................................... 16 1.4.6 Tyrosine Hydroxylase (TH) Deficiency ................................................................................................. 17 1.4.7 Aromatic Amino Acid Decarboxylase (AADC) Deficiency ............................................................ 18 1.4.8 Dopamine Transporter Deficiency Syndrome (DTDS) ..................................................................... 18 1.4.9 Dopamine β-Hydroxylase Deficiency ..................................................................................................... 19 1.4.10 Secondary neurotransmitter disorders and related diseases ........................................................... 19 1.4.11 Phenotypic Spectrum of Pediatric Neurotransmitter Disorders ................................................... 20 1.4.12 Diagnosis of Monoamine Deficiencies ................................................................................................ 21

2 Vesicular Monoamine Transporters ..................................................................................... 23

2.1 VMAT Isolation ..................................................................................................................................... 24 2.2 VMAT Structure .................................................................................................................................... 24 2.3 VMAT Biochemistry ............................................................................................................................ 25 2.4 Role of VMAT2 in Biogenic Amine Physiology .......................................................................... 27

Page 8: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

viii

2.4.1 Vmat2 Knockout Mice ................................................................................................................................ 28 2.4.2 Vmat2 Heterozygous Mice ....................................................................................................................... 29 2.4.3 Constitutive Overexpression of Vmat2 in Mice ............................................................................... 30 2.4.4 Selective Deletion of Vmat2 in Serotonergic Neurons .................................................................. 31 2.4.5 Selective Expression of Vmat2 in Noradrenergic Neurons ........................................................ 32

2.5 Role of VMAT2 in Disease Processes ............................................................................................ 33 2.5.1 VMAT2 and Parkinson’s Disease ........................................................................................................... 33 2.5.2 VMAT2, Neuropsychiatric Phenotypes, and Drugs of Abuse ..................................................... 36 2.5.3 Genetic Variation in VMAT2 .................................................................................................................... 38

3 Thesis Overview ........................................................................................................................... 39

Chapter 2 Clinical Characterization of a New Pediatric Neurotransmitter Disease ... 42

1 Introduction................................................................................................................................... 42

2 Methods ........................................................................................................................................... 43

2.1 Patients .................................................................................................................................................... 43 2.2 General Investigations ....................................................................................................................... 43 2.3 Clinical Measurement of Neurotransmitter Metabolites ...................................................... 43 2.4 AADC Enzyme Test .............................................................................................................................. 44 2.5 Candidate Gene Sequencing ............................................................................................................. 44

3 Results ............................................................................................................................................. 48

3.1 Patient History and Neurological Examination ........................................................................ 48 3.2 Investigations ........................................................................................................................................ 49 3.3 Family Structure .................................................................................................................................. 53 3.4 Genetic Screening of Candidate Genes ......................................................................................... 55 3.5 Drug Response ...................................................................................................................................... 56 3.6 Summary of Clinical Features and Differential Diagnosis .................................................... 59

4 Discussion....................................................................................................................................... 63

Chapter 3 Identification of Disease-Causing Genetic Variant ............................................. 69

1 Introduction................................................................................................................................... 69

2 Methods ........................................................................................................................................... 70

2.1 Patient Samples .................................................................................................................................... 70 2.2 Genotyping of Single Nucleotide Polymorphisms .................................................................... 70

Page 9: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

ix

2.3 Homozygosity Analysis ...................................................................................................................... 70 2.4 Linkage Analysis .................................................................................................................................. 71 2.5 Mutation Screening ............................................................................................................................. 72 2.6 Whole-Exome Sequencing ................................................................................................................ 75 2.7 TaqMan SNP Genotyping Assay ...................................................................................................... 76

3 Results ............................................................................................................................................. 76

3.1 Homozygosity Analysis ...................................................................................................................... 76 3.2 Linkage Analysis .................................................................................................................................. 79 3.3 Characterization of Genes in the Locus ........................................................................................ 82 3.4 Whole-Exome Sequencing of the Proband .................................................................................. 86 3.5 Identification of VMAT2 Variant as a Causative Candidate .................................................. 86 3.6 Controls ................................................................................................................................................... 89

4 Discussion....................................................................................................................................... 89

Chapter 4 Functional Characterization of the Disease-Causing Variant ......................... 95

1 Introduction................................................................................................................................... 95

2 Methods ........................................................................................................................................... 95 2.1 Construct Design and Site-Directed Mutagenesis .................................................................... 95 2.2 Cell Culture and Transfection.......................................................................................................... 96 2.3 Serotonin Uptake Assay..................................................................................................................... 97 2.4 Western Blotting .................................................................................................................................. 97 2.5 Sucrose Gradient Centrifugation .................................................................................................... 98

3 Results ........................................................................................................................................... 101

3.1 Steady-State Protein Levels of Transfected Protein Unaffected by P387L Variant in

Heterologous System .................................................................................................................................. 101 3.2 Highly Reduced Transport Activity of VMAT2 p.P387L in Heterologous System ...... 104 3.3 Measurable Residual Transport Activity of VMAT2 p.P387L Protein ........................... 107 3.4 Subcellular Localization of Protein in Heterologous System ............................................ 109

4 Discussion..................................................................................................................................... 111

Chapter 5 General Discussion and Future Directions .......................................................... 113

1 Introduction................................................................................................................................. 113

Page 10: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

x

2 Identification of the VMAT2 p.P387L Mutation: Impact on Knowledge of

Monoamine Physiology .................................................................................................................. 114

2.1 Future Directions and Experiments ........................................................................................... 116 2.1.1 Subcellular Localization of VMAT2 p.P387L ................................................................................. 116 2.1.2 Additional Biochemical Characterization of VMAT2 p.P387L................................................ 120 2.1.3 Development of a Mouse Model Expression Vmat2 p.P390L ................................................. 121

3 VMAT2 Mutation in PND: Impact on Understanding of PND Pathogenesis ........... 135 3.1 Future Directions and Experiments ........................................................................................... 136

3.1.1 18F-DOPA and 11C-DTBZ PET scans .................................................................................................... 136 3.1.2 Screening of Patient Cohorts to Identify Novel SLC18A2 Variants ...................................... 137

4 VMAT2 Mutation: Impact on Approaches to PND Diagnosis ...................................... 138

4.1 Future Directions and Experiments ........................................................................................... 139 4.1.1 Development of Methodology to Measure Biogenic Amines Directly in CSF ................... 139 4.1.2 Development of VMAT2 Platelet Activity Assay .......................................................................... 140 4.1.3 Use of Next-Generation Sequencing for Genetic Diagnosis of Rare Monogenic Diseases

141 4.1.4 Knowledge Translation Challenges and Strategies for PND Diagnosis .............................. 145

5 Concluding Statement .............................................................................................................. 149

References ........................................................................................................................................... 150

Page 11: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xi

List of Tables Table 1 Causative genes and modes of inheritance for monoamine neurotransmitter diseases 11

Table 2 Metabolites of biogenic amine neurotransmitters and pterin profiles in the

cerebrospinal fluid of patients with pediatric neurotransmitter diseases ...................................... 23

Table 3 Primer sequences for the amplification of exons of candidate genes associated with

known pediatric neurotransmitter diseases ................................................................................... 45

Table 4 Serum metabolic screen revealed no abnormalities ...................................................... 49

Table 5 Cerebrospinal fluid and urine neurotransmitters and their metabolites measured in a

younger affected sibling of the proband reveal decreased monoamines and elevated metabolites

in urine, but not in cerebrospinal fluid. Values outside reference ranges are presented in bold. . 52

Table 6 Pediatric neurotransmitter disease–associated genes screened in the proband revealing

no putative mutations .................................................................................................................... 56

Table 7 Age at initation of dopamine agonist affects disease course ........................................ 58

Table 8 Clinical features common to all affected individuals in the pedigree ........................... 59

Table 9 Clinical features of the disease organized by category ................................................. 60

Table 10 Comparison of clinical features of the present disease with those of closely related

monoamine deficiency syndromes ................................................................................................ 62

Table 11 Comparison of metabolite profiles in cerebrospinal fluid for pediatric

neurotransmitter diseases, including VMAT2 deficiency ............................................................ 65

Table 12 Primer sequences for amplification of exons of candidate genes located at 10q26 ... 73

Table 13 Primer sequences for amplification of SLC18A2 exons ............................................. 75

Table 14 Genes of known neuronal function or localization present in the disease-associated

bomozygous region ....................................................................................................................... 83

Page 12: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xii

Table 15 Sequence variants identified among eight candidate genes in the disease-associated

locus by direct sequencing of exons ............................................................................................. 85

Table 16 Primers to amplify SLC18A2 cDNA sequence for subcloning into pcDNA3.1 ......... 96

Table 17 Oligonucleotides to introduce cytosine to thymine substitution at position 1160 of

SLC18A2 using site-directed mutagenesis .................................................................................... 96

Table 18 Primers for the integration of the HA epitope at Arg94 of VMAT2 using site-direted

mutagenesis ................................................................................................................................. 119

Table 19 Primer sequences to amplify a probe for Slc18a2 exon 13 in mouse ....................... 130

Table 20 Primer sequences to amplify the upstream and downstream arms of the gene-targeting

construct 130

Table 21 Oligonucleotide sequences for site-directed mutagenesis of gene-targeting construct

to introduce the p.P390L variant in the expressed protein .......................................................... 130

Table 22 Sequencing primer sequences for gene-targeting construct ..................................... 130

Table 23 Primer sequences for generating labelled probes to identify the neomycin selection

cassette by Southern blot ............................................................................................................ 131

Page 13: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xiii

List of Figures Figure 1 Schematic diagram of the direct and proposed indirect pathways of the basal ganglia;

(a) direct/striatonigral pathway, (b) indirect/striatopallidal pathways. The overall effect on motor

thalamus of pathways in (b) are equivalent, and opposite in sign to (a). ........................................ 3

Figure 2 Monoaminergic neurotransmission exists as a balance of several critical processes:

biosynthesis, packaging, release, degradation, and reuptake. VMAT2, vesicular monoamine

transporter 2. ................................................................................................................................... 7

Figure 3 Common and parallel pathways for the synthesis and degradation of biogenic amines.

AADC, aromatic amino acid decarboxylase; BH4, tetrahydrobiopterin cofactor; COMT,

catechol-O-methyltransferase;; DAT, plasma membrane dopamine transporter;; DβH, dopamine β-

hydroxylase; 5-HIAA, 5-hydroxyindoleacetic acid; HVA, homovanillic acid; 5-HTP, 5-

hydroxytryptophan; MAO, monoamine oxidase; MHPG, 3-methoxy-4-hydroxyphenylglycol;

3-OMD, 3-O-methyldopa; qBH2, quinonoid dihydrobiopterin; PNMT, phenylethanolamine

N-methyltransferase; TH, tyrosine hydroxylase; TPH, tryptophan hydroxylase; VMA,

vanillylmandelic acid; VMAT2, vesicular monoamine transporter 2. ........................................... 8

Figure 4 Synthesis and regeneration of tetrahydrobiopterin—the conversion of BH4 to PCBD is

coupled to the generation of dopamine or serotonin. AR, aldose reductase; BH4,

tetrahydrobiopterin; DHPR, dihydropteridine reductase; GTPCH, GTP cyclohydrolase 1; H2NP3,

dihydroneopterin triphosphate; PCBD, tetrahydrobiopterin-α-carbinolamine; PCD, pterin-4α-

carbinolamine dehydratase; 6-PTP, 6-pyruvoyltetrahydropterin; qBH2, quinonoid

dihydrobiopterin; PTPS, 6-pyruvoyltetrahydropterin synthase; SP, sepiapterin; SR, sepiapterin

reductase. 14

Figure 5 T2-weighted magnetic resonance images of proband at age 14 revealed no

abnormalities. ................................................................................................................................ 50

Figure 6 Family structure of kindred demonstrates autosomal recessive mode of inheritance

and consanguineous pedigree structure. Proband is labelled V:6. Cerebrospinal fluid and urine

neurotransmitter analyses were performed on individual V:10. Black, affected; white,

unaffected; square, male; circle, female; diamond, spontaneous abortion. Numbers in squares or

Page 14: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xiv

circles indicate number of male or female offspring, respectively. From New England Journal of

Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular

Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical

Society. Reprinted with permission. ............................................................................................ 54

Figure 7 Single nucleotide polymorphism alleles for all genotyped family members in the

homozygous region uniquely shared by affected family members. Black, affected; white,

unaffected; square, male; circle, female. From New England Journal of Medicine, Rilstone JJ,

Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its

Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with

permission. .................................................................................................................................... 78

Figure 8 Multipoint LOD scores estimated across chromosome 10 ............................................. 80

Figure 9 Multipoint LOD scores estimated across chromosomes 3 and 16 ................................. 81

Figure 10 Genes present within the homozygous region shared by all affected individuals in the

family. C10orf82, chromosome 10 open reading frame 82; CACUL 1, CDK2-assocaited cullin

domain 1; CASC2, cancer susceptibility candidate 2 (non-protein coding); CCDC172, coiled-coil

domain containing 172; EIF3A, eukaryotic translation factor 3 subunit A; EMX2, empty

spiracles homeobox 2; ENO4, enolase family member 4; FAM204A, family with sequence

similarity 204 member A; GFRA1, GDNF family receptor alpha 1; GRK5, G protein–coupled

kinase 5; HSPA12A, heat shock 70 kDa protein 12A; KCNK18, potassium channel subfamily K

member 18; KIAA1598, Shootin1; MIR, microRNA; PDZD8, PDZ domain containing 8;

PNLIP, pancreatic lipase; PNLIPRP, pancreatic lipase–related protein; PRDX3, peroxiredoxin 3;

PRLHR, prolactin-releasing hormone receptor; RAB11FIP1, RAB11 family interacting protein 2

(class I); SFXN4, sideroflexin 4; SLC18A2, solute carrier family 18 member 2; SNORA19,

small nucleolar RNA H/ACA box 19; VAX1, ventral anterior homeobox 1. Image from UCSC

Genome Browser [http://genome.ucsc.edu] .................................................................................. 84

Figure 11 Electropherogram depicting sequence variation in unaffected and affected family

members. Top panel, unaffected sibling with wild-type genomic sequence; middle panel,

asymptomatic parent (IV:3) possesses the SLC18A2 c.1160C→T variant in heterozygous form;;

lower panel, proband (V:6) is homozygous for SLC18A2 c.1160C→T. From New England

Page 15: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xv

Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin

Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts

Medical Society. Reprinted with permission. .............................................................................. 88

Figure 12 Predicted structure of VMAT2 comprises 12 transmembrane domains, a large

lumenal loop including four proposed glycosylation sites, and both N-terminal and C-terminal

cytoplasmic regions. Proline residue 387 is located immediately adjacent to the insertion of

transmembrane domain X. From New England Journal of Medicine, Rilstone JJ, Alkhater RA,

Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368,

543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission. .......... 93

Figure 13 Multiple sequence alignment of VMAT2 in the region of the p.P387L variant. TM9

and TM10 represent sequences associated with transmembrane domains 9 and 10, respectively.

Residues that differ from human VMAT2 sequence are indicated in gray. Amino acid position

387 is indicated by an asterisk. Homo, homo sapiens (human); Patient, proband; Pan, Pan

troglodytes (chimpanzee); Macaca, Macaca mulatta (Rhesus macaque); Mus, Mus musculus

(mouse); Rattus, Rattus norvegicus (rat); Canis, Canis familiaris (dog); Bos, Bos taurus (cow);

Monodelphis, Monodelphis domestica (opossum); Gallus, Gallus gallus (chicken); Tetraodon,

Tetraodon nigroviridis (pufferfish); Danio, Danio rario (zebrafish); Drosophila, Drosophila

melanogaster (fruit fly); C. elegans, Caenorhabditis elegans (nematode); hVMAT1, human

VMAT1 isoform. From New England Journal of Medicine, Rilstone JJ, Alkhater RA,

Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368,

543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission. .......... 94

Figure 14 Visualization of ATP by absorbance at 260 nm to illustrate sucrose gradient

linearity 99

Figure 15 Consistent fractionation demonstrated by absorbance at 280 nm of parallel sucrose

gradient fractionations ................................................................................................................ 100

Figure 16 Markers of cellular compartments in sucrose gradient fractions ............................. 101

Figure 17 Parallel transient transfections of wild-type and p.P387L VMAT2 in Cos7 cells, and

vector-transfected control. GAPDH, protein loading control. .................................................... 102

Page 16: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xvi

Figure 18 Comparison of banding pattern observed for wild-type and p.P387L VMAT2

expressed transiently in Cos7 cells. unt, untransfected; WT, wild-type. .................................... 104

Figure 19 Vesicular uptake of tritiated serotonin by the wild-type and p.P387L human VMAT2

transiently expressed in a heterologous Cos7 cell system. Inset: Expression of VMAT2 in lysates

used for uptake assay. 5-HT, serotonin; wt, wild-type. From New England Journal of Medicine,

Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport

Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society.

Reprinted with permission. ......................................................................................................... 106

Figure 20 Vesicular uptake of tritiated serotonin by wild-type and p.P387L human VMAT2

transiently expressed in a heterologous Cos7 cell system after 10 minutes with and without the

addition of the specific VMAT inhibitor reserpine (10 µM). From New England Journal of

Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular

Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical

Society. Reprinted with permission. .......................................................................................... 108

Figure 21 Sucrose gradient centrifugation of transiently transfected Cos7 lysates ................. 110

Figure 22 Expression of wild-type and p.P387L VMAT2 in stable clones of the Cos7 cell line;

band at 37 kDa represents GAPDH loading control. .................................................................. 118

Figure 23 Expression of wild-type and p.P387L VMAT2 in stable clones of the MN9D cell line

120

Figure 24 Schematic of incorporation of mouse gene targeting construct into mouse genome.

Upper panel, wild type mouse genomic sequence of Slc18a2 exons 11–15 and flanking intronic

sequences. Middle panel, mouse genomic sequence after incorporation of the gene-targeting

construct by homologous recombination with the long arms of the construct in embryonic stem

cells. Slc18a2 c.1169C→T mutation, coding for the P390L amino acid substitution, is present in

exon 13. Lower panel, mouse genomic sequence after expression of Cre recombinase and

excision of the selection cassette. PGKneobpA, neomycin selection cassette; loxP, recombination

sites targeted by Cre recombinase. .............................................................................................. 125

Page 17: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xvii

Figure 25 Gene targeting construct for the introduction of the c.1169C→T mutation into

Slc18a2. AmpR, ampicillin resistance gene; F1_origin, origin of replication; loxP, recombination

site for Cre recombinase; NeoR/KanR, neomycin and kanamycin resistance gene (of the

PGKneobpA selection cassette); NheI, HindIII, restriction sites for insertion of downstream arm

of genomic sequence for recombination; NotI, SacII, restriction sites for insertion of upstream

arm of genomic sequence for recombination. Gray boxes, mouse intronic sequence of Slc18a2;

black boxes, mouse exonic sequence of Slc18a2. ....................................................................... 127

Page 18: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xviii

List of Abbreviations

AADC aromatic amino acid decarboxylase

AR aldose reductase

ATP adenosine triphosphate

BH4 tetrahydrobiopterin

CHO Chinese hamster ovary

CIHR Canadian Institutes of Health Research

COMT catechol-O-methyltransferase

CP cerebral palsy

CSF cerebrospinal fluid

DA dopamine

DAT plasma membrane dopamine transporter

DβH dopamine β-hydroxylase

DDC encodes AADC

DHPR dihydropteridine reductase

DMEM Dulbecco’s modified Eagle medium

Page 19: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xix

DOPAC 3,4-dihydroxyphenylacetic acid

DRD dopa-responsive dystonia

DTBZ dihydrotetrabenazine

DTDS dopamine transporter deficiency syndrome

E epinephrine

EEG electroencephalogram

ELISA enzyme-linked immunosorbant assay

ER endoplasmic reticulum

ES embryonic stem

GAPDH glyceraldehyde 3-phosphate dehydrogenase

GPi globus pallidus internus

GCH1 encodes GTPCH1

GTP guanosine-5'-triphosphate

GTPCH1 GTP cyclohydrolase I

5-HIAA 5-hydroxyindoleacetic acid

HIS histamine

Page 20: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xx

H2NP3 dihydroneopterin triphosphate

HPLC-EC high-performance liquid chromatography with electrochemical detection

5-HT serotonin

5-HTP 5-hydroxytryptophan

HVA homovanillic acid

kDa kilodalton

LC locus coerulus

LC-MS/MS liquid chromatography tandem mass spectrometry

LDCV large dense-core vesicles

LOD logarithm of odds

MAO monoamine oxidase

MFS major facilitator superfamily

MHPG 3-methoxy-4-hydroxyphenylglycol

MPP+ 1-methyl-4-phenylpyridinium; active metabolite of MPTP

MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

MRI magnetic resonance imaging

Page 21: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xxi

NBIA neurodegeneration with brain iron accumulation

NE norepinephrine

NHLBI National Heart, Lung, and Blood Institute

3-OMD 3-O-methyldopa

PBS phosphate-buffered saline

PCBD tetrahydrobiopterin-α-carbinolamine

PCBD encodes PCD

PCD pterin-4α-carbinolamine dehydratase

PD Parkinson’s disease

PET positron emission tomography

PKU phenylketonuria

PLP pyridoxal phosphate

PND pediatric neurotransmitter disorder

PNMT phenylethanolamine N-methyltransferase

PNPO pyridoxamine 5’-phosphate oxidase

6-PTP 6-pyruvoyltetrahydropterin

Page 22: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xxii

PTPS 6-pyruvoyl tetrahydropterin synthase

PTS encodes PTPS

PVDF polyvinylidene fluoride

qBH2 quinonoid dihydrobiopterin

QDPR encodes DHPR

SCA spinocerebellar ataxia

SDS-PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis

SERT serotonin plasma membrane transporter

SLC6A3 encodes DAT

SLC18A2 encodes VMAT2

SNP single nucleotide polymorphism

SNpc substantia nigra pars compacta

SNR substantia nigra pars reticulata

SP sepiapterin

SPR encodes SR

SR sepiapterin reductase

Page 23: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

xxiii

SSV small synaptic vesicles

TBS-T 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 0.05% Tween 20

TfR transferrin receptor

TH tyrosine hydroxylase

TMD transmembrane domain

TPH tryptophan hydroxylase

VAChT vesicular acetylcholine transporter

VMA vanillylmandelic acid

VMAT vesicular monoamine transporter

VTA ventral tegmental area

Page 24: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

1

Chapter 1 Literature Review

1 Movement Disorder and Monoamine Deficiency Movement disorders are a large group of diseases in which patients suffer impairment of

the planning, control or execution of movement (Klein, 2005). The clinical spectrum of

movement disorders includes, but is not limited to, ataxia, blepharospasm, dysphonia,

dystonic disorders, gait disorders, Huntington’s disease, myoclonus, Parkinson’s disease,

spasticity, tardive dyskinesia, tics and Tourette syndrome, and tremor (Klein, 2005).

They are clinically, pathologically, and genetically heterogeneous. Genes for many are

already known (Klein, 2005). Despite this, the pathogenic mechanisms of the vast

majority of these movement disorders are still being elucidated.

For example, the prototypical movement disorder, Parkinson’s disease, is characterized

by neurodegeneration of the dopamine-secreting neurons of the substantia nigra pars

compacta (SNpc), and a deficit of dopamine in the brain (see Section 1.3). This results in

bradykinesia (slowing of physical movement), rigidity, postural instability, and rest

tremor. The molecular mechanisms leading to this pathology are still unclear despite the

discovery of 11 distinct genes causing monogenic forms of the disease, as well as a

number of associations with additional genes and loci associated with sporadic forms of

the disease (Singleton et al., 2013).

1.1 Pathophysiology The underlying pathophysiology of Parkinson’s disease and related movement

disorders involves misregulation of a series of structures comprising the basal ganglia.

Page 25: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

2

The basal ganglia function in the fine modulation of motor activity. Input from the cortex

is received by the striatum (input nucleus), which in turn regulates the output nuclei—the

substantia nigra pars reticula (SNR) and globus pallidus internus (GPi). The output

nuclei tonically inhibit the motor thalamus, and therefore motor activity. By current

models, this occurs through the structures of the basal ganglia via two possible

pathways—the direct (striatonigral) or indirect (striatopallidal) pathway (Figure 1)

(Miller, 2008). Inhibitory striatal neurons can be identified as members of either of these

two pathways by the presence of peptide markers (substance P or enkephalin,

respectively).

Page 26: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

3

Figure 1 Schematic diagram of the direct and proposed indirect pathways of the basal ganglia; (a) direct/striatonigral pathway, (b) indirect/striatopallidal pathways. The overall effect on motor thalamus of pathways in (b) are equivalent, and opposite in sign to (a).

Page 27: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

4

The role of dopamine in the basal ganglia is in regulation of the striatum. Dopaminergic

neurons originate in the SNpc, and project their axons to innervate and regulate the

striatum via D1- and D2-class receptors. At physiological concentrations, dopamine is

sequestered in vesicles of dopaminergic neurons and undergoes regulated exocytotic

release at the synapse. However, it has been demonstrated that at higher, pathological

concentrations, unsequestered cytosolic dopamine can cause toxicity with the formation

of reactive oxygen species generated by dopamine auto-oxidation or monoamine oxidase

(MAO) metabolism (Cubells et al., 1994, Graham, 1978, Hastings et al., 1996). This

natural dopamine toxicity is a suggested mechanism underlying the progressive

nigrostriatal neurodegeneration in Parkinson’s disease (see Section 1.3).

1.2 The Biogenic Amines The biogenic amines (also referred to as monoamines) are a class of neurotransmitter that

are synthesized enzymatically from amino acids. Serotonin is derived from tryptophan,

and is the basis of the serotonergic system. The catecholamines (dopamine,

norepinephrine, and epinephrine) are derived from tyrosine. Dopamine is the basis of the

dopaminergic system, and norepinephrine and epinephrine underlie the adrenergic

system. Histamine is derived from histidine, and is present in neurons of the

hypothalamus. The biogenic amines are modulatory in nature (acting on a timescale of

seconds to minutes), have mixed effects, and are subject to significant crosstalk.

The dopaminergic system comprises the nigrostriatal pathway that projects from the

SNpc to the striatum (caudate and putamen), and the mesolimbic and mesocortical

projections from the ventral tegmental area (VTA) to the nucleus accumbens and cerebral

Page 28: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

5

cortex, respectively. There are also dopaminergic projections comprising the

tuberoinfundibular pathway.

The serotonergic system comprises widespread projections from the dorsal raphe nuclei

to many regions of the brain, including the neocortex, amygdala, hippocampus, thalamus,

hypothalamus, striatum, cerebellum, brain stem, and spinal cord.

The noradrenergic system also involves widespread projections from the locus coeruleus

to the neocortex, amygdala, hippocampus, thalamus, hypothalamus, tectum, cerebellar

cortex, visceral cranial nuclei, and spinal cord. Both norepinephrine and epinephrine also

function peripherally in sympathetic ganglion cells.

Histamine is present in the hypothalamus, where it plays an integral role in

neurometabolic functions (e.g., wakefulness, appetite regulation, response to pain, and

immunological response).

Monoaminergic neurotransmission exists as a balance of several critical processes:

biosynthesis, packaging, release, degradation, and reuptake. First, transmitters are

synthesized from their amino acid precursors in the cytosol. Next, they are accumulated

into synaptic vesicles through the action of transporters that are driven by the proton

gradient. Further biochemical transformation occurs within the synaptic vesicle in the

case of norepinephrine. Neurotransmission occurs by way of regulated exocytotic release

from the synaptic vesicles into the synaptic cleft in response to physiologic stimuli. At

the synapse, the transmitter interacts with target receptors on the postsynaptic cell to

mediate signal transduction. Transmitter at the synapse is then cleared by both

degradation and reuptake processes. Specific monoamine metabolizing enzymes are

Page 29: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

6

present to inactivate the transmitter. Reuptake into the presynaptic cell and glia occurs

through plasma membrane monoamine transporters that are driven by Na+ and Cl–

gradients. This model of monoamine neurotransmission and homeostasis is illustrated in

Figure 2. The biochemical pathways for the synthesis and degradation of the biogenic

amines are presented in Figure 3.

Page 30: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

7

Figure 2 Monoaminergic neurotransmission exists as a balance of several critical processes: biosynthesis, packaging, release, degradation, and reuptake. VMAT2, vesicular monoamine transporter 2.

Page 31: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

8

Figure 3 Common and parallel pathways for the synthesis and degradation of biogenic amines. AADC, aromatic amino acid decarboxylase; BH4, tetrahydrobiopterin cofactor; COMT, catechol-O-methyltransferase; DAT, plasma membrane dopamine transporter;; DβH, dopamine β-hydroxylase; 5-HIAA, 5-hydroxyindoleacetic acid; HVA, homovanillic acid; 5-HTP, 5-hydroxytryptophan; MAO, monoamine oxidase; MHPG, 3-methoxy-4-hydroxyphenylglycol; 3-OMD, 3-O-methyldopa; qBH2, quinonoid dihydrobiopterin; PNMT, phenylethanolamine N-methyltransferase; TH, tyrosine hydroxylase; TPH, tryptophan hydroxylase; VMA, vanillylmandelic acid; VMAT2, vesicular monoamine transporter 2.

Page 32: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

9

1.3 Parkinson’s Disease Parkinson’s disease (PD) is a common, adult-onset neurodegenerative disease

characterized by the selective and progressive loss of neuronal subtypes—in particular,

the nigrostriatal dopaminergic pathway. PD is primarily characterized by the

deterioration of motor function, evidenced as bradykinesia, rigidity, postural instability,

and rest tremor. In addition, there has been increased recognition of the nonmotor

manifestations of PD. These include anosmia, depression, anxiety, sweating, dyspnea,

orthostatic hypotension, constipation, pain, genitourinary problems, sexual dysfunction,

and sleep disorders (Witjas et al., 2002). These nonmotor symptoms may be grouped

into domains: cognitive and psychiatric, autonomic, and sensory/pain. Many of the

nonmotor manifestations of PD precede the motor symptoms of the disease, and certainly

emerge upon disease progression. Furthermore, the nonmotor symptoms of PD are

reported to have a greater impact on patient quality of life (Barone et al., 2009).

Evidence of the degeneration of other monoaminergic neuronal types (serotonergic

neurons in the raphe nuclei and noradrenergic neurons in the locus coeruleus), as well as

cholinergic neurons in the basal forebrain and midbrain tegmentum, establish a broader

monoaminergic deficiency phenotype in PD (Hirsch et al., 2003).

The etiology and pathogenesis of PD have been the subject of considerable study and

reflect significant complexity. PD is primarily sporadic, although multiple monogenic

forms of the disease exist (Singleton et al., 2013). Familial and sporadic forms of the

disease have nearly identical motor phenotypes. Furthermore, toxins such as 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine (MPTP) can selectively destroy dopaminergic neurons

and acutely cause parkinsonism—providing evidence that environmental exposures may

Page 33: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

10

play a role in the pathogenesis of PD. Notably, the strongest risk factor for PD is

advancing age, possibly implicating the aging process in PD pathogenesis.

Current thinking regarding PD pathogenesis favors a mixture of pathogenic mechanisms.

Multiple avenues of investigation have identified common pathways that may underlie

the disease. These include oxidative stress, protein aggregation, defects in the ubiquitin–

proteasome pathway, autophagy, and alterations in mitochondrial function. The

pathologic hallmark of PD is Lewy bodies—inclusion bodies comprising α-synuclein and

other protein deposits—and there has been some evidence of cell-to-cell transfer of these

deposits that may be associated with the spread of the disease to other neuronal types

(Steiner et al., 2011).

1.4 Pediatric Neurotransmitter Diseases The pediatric neurotransmitter diseases, by contrast, are a group of early onset rare

diseases attributable to a disturbance in neurotransmitter metabolism with causative

defects in enzymes that are involved in the biosynthesis, degradation, or membrane

transport of transmitter. Known disorders specifically of the biogenic amine

neuromediators (dopamine, norepinephrine, epinephrine, and serotonin) comprise defects

in nine enzymes and one transporter—each presenting in early childhood with symptoms

and signs referable to the affected neurotransmitter. Deficiency in dopamine results in

movement disorder, including parkinsonism and dystonia; deficient norepinephrine and

epinephrine cause sympathetic autonomic dysfunction; and serotonin deficiency leads to

sleep disturbance and psychiatric disease. The monoamine neurotransmitter disorders of

known genetic etiology are summarized in Table 1 Causative genes and modes of

inheritance for monoamine neurotransmitter diseases.

Page 34: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

11

Table 1 Causative genes and modes of inheritance for monoamine neurotransmitter diseases

Disorder Causative Gene Mode of Inheritance Autosomal dominant GTP cyclohydrolase I deficiency (Dopa-responsive dystonia or Segawa disease)

GCH1 Autosomal dominant

Autosomal recessive GTP cyclohydrolase I deficiency

GCH1 Autosomal recessive

Sepiapterin reductase deficiency

SPR Autosomal recessive

6-Pyruvoyltetrahydropterin synthase deficiency

PTS Autosomal recessive

Dihydropteridine reductase deficiency

QDPR Autosomal recessive

Pterin-4α-carbinolamine dehydratase deficiency

PCD Autosomal recessive

Tyrosine hydroxylase deficiency

TH Autosomal recessive

Aromatic L-amino acid decarboxylase deficiency

DDC Autosomal recessive

Pyridoxal phosphate-dependent epilepsy (PLP-DE)

PNPO Autosomal recessive

Dopamine transporter deficiency syndrome

SLC6A3 Autosomal recessive

The multiple deficits of PNDs overlap substantially with the combination of

dopaminergic motor dysfunction and the domains of nonmotor symptoms in PD.

Similarly, evidence of degeneration of other monoaminergic neuronal types (serotonergic

neurons in the raphe nuclei and noradrenergic neurons in the locus coeruleus), as well as

cholinergic neurons in the basal forebrain and midbrain tegmentum have established the

involvement of multiple monoaminergic systems in PD (Hirsch et al., 2003). However,

whereas PND are disorders of infancy and childhood, PD is a disease of aging. It thus

Page 35: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

12

reflects distinct underlying pathogenic mechanisms. Similarly, the manifestations of

PNDs are expected to differ from those of PD because the monoaminergic dysfunction

occurs during critical periods of neurological development.

1.4.1 GTP Cyclohydrase I (GTPCH1) Deficiency

The longest recognized monoamine neurotransmitter disorder is autosomal dominant

GTP cyclohydrolase I (GTPCH1) deficiency that was first described in the 1970s

(Segawa et al., 1976), and is more commonly referred to as dopa-responsive dystonia

(DRD) or Segawa disease. DRD results from a defect in tetrahydrobiopterin (BH4)

synthesis—which is a necessary cofactor for the rate-limiting step in dopamine synthesis

(tyrosine hydroxylase conversion of tyrosine to L-DOPA). These patients exhibit

dystonic spasms with diurnal fluctuation that begin in the first decade of life. The disease

is associated with short stature. Adult-onset cases have also been described, in which the

primary features are writer’s cramp, torticollis, or generalized rigid hypertonus with

tremor, but not postural dystonia (Segawa, 2011). The disorder is extremely responsive

to treatment with levodopa in combination with a decarboxylase inhibitor (carbidopa).

The disease is caused by heterozygous mutations in GCH1 at 14q22.1–q22.2 (Ichinose et

al., 1994). More than 100 mutations have been discovered. The autosomal dominant

mutations are dominant negative, as a result of an inability to properly form the native

homodecameric form of the protein. Less than 10% of identified mutations are found in

homozygous or compound heterozygous form, causing an autosomal recessive form of

the disease (Thony and Blau, 2006).

Several other disorders of tetrahydrobiopterin synthesis or regeneration present as

pediatric movement disorder, including defects in 6-pyruvoyl tetrahydropterin synthase

Page 36: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

13

(PTPS; PTS), sepiapterin reductase (SR; SPR), dihydropteridine reductase (DHPR;

QDPR), and pterin-4α-carbinolamine dehydratase (PCD; PCBD) (Longo, 2009). The

pathway for tetrahydrobiopterin synthesis and regeneration is presented in Figure 4.

Tetrahydrobiopterin is an essential cofactor for tyrosine hydroxylase and tryptophan

hydroxylase, as well as phenylalanine hydroxylase, nitric oxide synthases, and glyceryl-

ether monooxygenase. Despite its necessity for phenylalanine hydroxylase activity,

fewer than 2% of patients with phenylketonuria have mutations in PTS, QDPR, GCH1, or

PCBD (Longo, 2009). Laboratory diagnosis of tetrahydrobiopterin deficiency is based

on newborn screening for phenylketonuria, urine or dried blood profiling of pterins, and

the measurement of DHPR enzyme activity in blood.

Page 37: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

14

Figure 4 Synthesis and regeneration of tetrahydrobiopterin—the conversion of BH4 to PCBD is coupled to the generation of dopamine or serotonin. AR, aldose reductase; BH4, tetrahydrobiopterin; DHPR, dihydropteridine reductase; GTPCH, GTP cyclohydrolase 1; H2NP3, dihydroneopterin triphosphate; PCBD, tetrahydrobiopterin-α-carbinolamine; PCD, pterin-4α-carbinolamine dehydratase; 6-PTP, 6-pyruvoyltetrahydropterin; qBH2, quinonoid dihydrobiopterin; PTPS, 6-pyruvoyltetrahydropterin synthase; SP, sepiapterin; SR, sepiapterin reductase.

Page 38: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

15

1.4.2 Sepiapterin Reductase (SR) Deficiency

SR deficiency, like GTPCH1 deficiency, is not associated with hyperphenylalaninemia

but phenylalanine loading tests are often positive. The disease is caused by mutations in

the SPR gene at 2p13, and at least 19 mutations have been identified (Thony and Blau,

2006). Symptoms arise in the first decade of life, including psychomotor retardation,

dystonia, oculogyric crises, choreoathetosis, hypotonia, spasticity, tremor, ataxia,

parkinsonism, seizures, temperature instability, hypersalivation, microcephaly, and

irritability, as well as psychiatric symptoms. These symptoms exhibit diurnal variation,

as with GTPCH1 deficiency. Alternatively, a mild form of the disease with mild motor

symptoms has been reported, associated with a splicing defect leading to a missense

mutation with residual protein function (Arrabal et al., 2011). Metabolite profiles in CSF

reveal a disturbance of dopamine, norepinephrine, and serotonin synthesis. SR activity

can be measured clinically in skin fibroblasts.

1.4.3 6-Pyruvoyl Tetrahydropterin Synthase (PTPS) Deficiency

PTPS deficiency is the most common disorder of BH4 metabolism (Leuzzi et al., 2010),

caused by mutations in the PTS gene at 11q22.3–23.3. More than 50 mutations have

been described (Thony and Blau, 2006). It exists as both severe and peripheral forms.

The severe form manifests in the first few months of life with delayed developmental

milestones and movement disorder, as well as hyperphenylalaninemia and risk of

premature birth and low birth weight. The peripheral form involves

hyperphenylalaninemia but minor or no disturbance in biogenic amine neurotransmitters,

as measured by their metabolites in CSF and a normal neurological course. Genotype–

phenotype correlation is evident in that mutations with high residual enzyme activity

Page 39: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

16

cause the peripheral form of the disease, whereas mutations causing gross disturbances to

protein function (such as frameshift mutations leading to protein truncation or altered

protein zinc binding or oligomerization) lead to the severe form of the disease (Brasil et

al., 2011).

1.4.4 Dihydropteridine Reductase (DHPR) Deficiency

DHPR deficiency is the most severe of the disorders of pterin metabolism, caused by

mutations in the QDPR gene at 4p15.31. DHPR deficiency reflects a defect in the

regeneration of tetrahydrobiopterin. It has neonatal onset, or onset in early infancy.

Patients exhibit feeding difficulties, bulbar dysfunction, hypersalivation, and

microcephaly. They also exhibit delayed motor and cognitive milestones, truncal and

limb hypertonia, dyskinesia, tremor, dystonia, choreoathetosis, and seizures. These

patients are at risk of sudden death. There are often white matter abnormalities and basal

ganglia calcifications visible by MRI (Longhi et al., 1985, Woody et al., 1989). The

severity of this particular defect in biopterin metabolism is thought to be related to the

concomitant accumulation of q-dihydrobiopterin and its inhibitory effects on AADC,

tyrosine hydroxylase, tryptophan hydroxylase, and phenylalanine hydroxylase, as well as

depletion of folate in the brain related to the absence of DHPR and the accumulation of

q-dihydrobiopterin. A mild form of the disease affects only serotonin metabolism, and is

caused by two specific protein mutations (G151S and F212C) (Blau et al., 1992).

1.4.5 Pterin-4α-Carbinolamine (PCD) Deficiency

PCD deficiency is also a defect in the regeneration of tetrahydrobiopterin resulting from

mutations in PCBD at 10q22. It is associated with a mild hyperphenylalaninemia that

Page 40: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

17

normalizes after a few months of life. There is usually no neurotransmitter phenotype,

but transient neonatal hypotonia has been reported in some patients (Thony et al., 1998).

1.4.6 Tyrosine Hydroxylase (TH) Deficiency

A deficiency of tyrosine hydroxylase is a progressive and lethal encephalopathy with

poor prognosis, involving dystonia and other extrapyramidal movement disorder

symptoms. It is a rare autosomal recessive disorder caused by mutations in TH at

11p15.5, and reported in less than 40 patients worldwide thus far. Importantly, mutations

reflecting a complete loss of function (such as premature truncations) have not been

observed in homozygous form, suggesting that a complete lack of tyrosine hydroxylase is

incompatible with life (Willemsen et al., 2010). There are two overlapping subtypes:

type A, a progressive extrapyramidal movement disorder (hypokinetic–rigid syndrome

with dystonia) with onset in infancy or childhood; and type B, a complex encephalopathy

with onset in the neonatal period or early infancy (Willemsen et al., 2010). Type A

comprises approximately 70% of patients who present in infancy with a parkinsonian

phenotype of hypokinesia, bradykinesia, dystonia, and rigidity. There can be diurnal

variation in the dystonia. An onset in the first year is associated with some mild

cognitive impairment. Some patients develop onset of symptoms later (within the first 5

years), with gait instability and difficulty walking. Type B disease has an onset within

the first 3 months of life with hypotonia and severe parkinsonism, in combination with

mental retardation, dystonia, oculogyric crises, myoclonus, tremor, dyskinesia, and

ptosis. Autonomic features (sweating, temperature instability, hyperpyrexia, and

drooling) are also observed. Seizures have also been reported in these patients. Notably,

Page 41: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

18

MRI investigations are usually normal, though some white matter abnormalities and

increased extracerebral CSF spaces have been observed in more severe cases.

1.4.7 Aromatic Amino Acid Decarboxylase (AADC) Deficiency

AADC deficiency disorder, for which there have been less than 100 patients diagnosed

worldwide (Brun et al., 2010), results from mutations in the aromatic amino acid

decarboxylase gene (DDC; also known as dopa decarboxylase) at 7p12.3–12.1. These

patients present with hypotonia in the first six months of life, and further develop

extrapyramidal movement disorder symptoms including dystonia, chorea, blepharospasm,

bulbar dysfunction, and myoclonus. They further exhibit developmental delay,

irritability, sleep disturbances, autonomic manifestations, temperature instability,

irritability, and nasal congestion. Most patients exhibit cognitive impairment. AADC

activity can be assayed clinically in plasma. MRI is generally normal, although some

nonspecific changes have been reported (Brun et al., 2010). Patients generally do not

respond to treatment.

Briefly, pyridoxal phosphate is an essential cofactor for AADC. Deficiency in the

synthesis of this cofactor resulting from mutations in the gene encoding pyridoxamine 5’-

phosphate oxidase (PNPO at 17q21.32) leads to prenatal seizures an a severe, potentially

fatal, anticonvulsant-resistant neonatal encephalopathy (Mills et al., 2005). CSF

metabolite profiles mimic those of AADC deficiency.

1.4.8 Dopamine Transporter Deficiency Syndrome (DTDS)

Whereas the pediatric disorders described thus far reflect deficiencies in the synthesis of

one or more biogenic amine neurotransmitters, dopamine transporter (DAT) deficiency

Page 42: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

19

syndrome (DTDS) is a defect in monoamine transport. Children with homozygous or

compound heterozygous mutations in SLC6A3 (coding for DAT) at 5p15.3 present with a

complex motor disorder in early infancy that comprises hyperkinetic symptoms,

hypokinesia, or a mixed phenotype that involves progressive dystonia, axial hypotonia,

and parkinsonism (Kurian et al., 2011b, Kurian et al., 2009). Eye movement disorder

(ocular flutter, saccade initiation failure, eyelid myoclonus, and oculogyric crises) is also

observed. Patients also exhibit sleeping difficulties, orthopedic complications, and

cardiorespiratory insufficiency. The mean life expectancy for patients with DTDS is 13.6

years.

Defects in presynaptic dopamine reuptake reduce presynaptic dopamine stores and an

accumulation of synaptic dopamine that is susceptible to catabolism. This is reflected by

increased HVA concentrations in the CSF. Increased synaptic dopamine may also lead to

downregulation of postsynaptic receptor expression (Blackstone, 2009).

1.4.9 Dopamine β-Hydroxylase Deficiency

Dopamine β-hydroxylase deficiency is characterized as a primary autonomic failure

stemming from the inability to convert dopamine to norepinephrine or epinephrine

(Senard and Rouet, 2006).

1.4.10 Secondary neurotransmitter disorders and related diseases

Dopamine and serotonin depletion can occur secondary to certain other neurological

disorders, including perinatal asphyxia, disorders of folate metabolism, phenylketonuria,

Lesch-Nyhan disease, mitochondrial disorders, epilepsy and infantile spasms,

Page 43: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

20

opsoclonus-myoclonus, pontocerebellar hypoplasia, leukodystrophies, Rett’s syndrome,

and certain neuropsychiatric disorders (Kurian et al., 2011a).

Additionally, there are several other rare disorders that present in the first decade of life

and involve a combination of dystonia and parkinsonism complicated by pyramidal tract

involvement (Pearl, 2013, Schneider and Bhatia, 2010). These include Wilson’s disease;;

parkinsonism associated with mutations in parkin (PARK2), PINK1 (PARK6), and DJ1

(PARK7); x-linked dystonia-parkinsonism/Lubag (DYT3); rapid-onset dystonia-

parkinsonism (DYT12); DYT16 dystonia; neurodegeneration with brain iron

accumulation (NBIA, including PANK2 and PLA2G6/PARK14); neuroferritinopathy;

Kufor-Rakeb disease; SENDA syndrome; and autosomal recessive spastic paraplegia

with thin corpus collosum (SPG11).

1.4.11 Phenotypic Spectrum of Pediatric Neurotransmitter Disorders

A phenotypic continuum has been proposed for GTPCH1 deficiency and extended to all

pediatric monoamine biosynthetic deficiencies (see Table 3 in Pons, 2009). This

spectrum illustrates the variability in the presentation of pediatric neurotransmitters

disorders that reflects a range of severities stemming from a common underlying

pathophysiology. The severity of presentation depends upon the effect of the mutation on

enzyme function and the importance of the affected enzyme to biogenic amine

availability, on the extent of monoamine deficiency, on the relative involvement of each

of the biogenic amines, and on the age of onset and subsequent effect on neurological

development. Additional variability in presentation is observed between patients with

common defects.

Page 44: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

21

1.4.12 Diagnosis of Monoamine Deficiencies

In addition to clinical signs, several tests may be useful to the diagnosis of pediatric

neurotransmitter disorder. Because tetrahydrobiopterin is also a cofactor for

phenylalanine hydroxylase, some monoamine deficiencies include peripheral

hyperphenylalanemia that may be identified in serum or in neonatal screening. These

include autosomal recessive GTP cyclohydrolase deficiency, pterin-carbinolamine

dehydratase deficiency, dihydropteridine reductase deficiency, and pyruvoyl-

tetrahydropterin synthase deficiency. In the absence of peripheral hyperphenylalanemia,

an oral phenylalanine loading test, in which the ratio of phenylalanine to tyrosine is

measured in serum after loading, may be useful to identify defects in tetrahydrobiopterin

metabolism (Bandmann et al., 2003, Opladen et al., 2010).

Genetic testing of associated genes is reasonably simple, but may not reveal noncoding

mutations with functional relevance, and the functional relevance of novel mutations may

be unclear. Enzyme activity tests are available for GTPCH activity in fibroblasts, SR

activity in fibroblasts, DHPR activity in blood spots, and AADC activity in plasma.

These tests are not considered routine, but may be useful in cases of negative genetic

screening.

Urine metabolite (HVA, 3-OMD, 5-HIAA, VMA; see Figure 3) and pterin profiles

(biopterin and neopterin; see Figure 4) may also aid diagnosis. However, as a

measurement of metabolites and pterins in the periphery, urine results are not conclusive.

An abnormal pterin profile in the urine may help distinguish diseases with peripheral

Page 45: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

22

hyperphenylalanemia from phenylketonuria (PKU; phenylalanine hydroxylase

deficiency).

The most conclusive available diagnostic is CSF neurotransmitter metabolite and pterin

profiles (see Table 2). Metabolite levels are indirect measures of each neurotransmitter,

as well as an indication of turnover. There are, however, several difficulties with this

analysis that prohibit routine testing. CSF collection requires lumbar puncture, with

inherent risks. CSF neurotransmitter analysis must be performed in specialized

laboratories, of which there are few. CSF is labile, and special collection procedures are

necessary, including immediate centrifugation, snap freezing, and preservatives.

Metabolites exhibit diurnal variation, and there is a rostrocaudal gradient of metabolite

and pterin concentrations that requires precise labeling of collected fractions and careful

interpretation of results. In addition, there is variation in concentrations with age, so

reference ranges must be defined in age-matched controls.

Page 46: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

23

Table 2 Metabolites of biogenic amine neurotransmitters and pterin profiles in the cerebrospinal fluid of patients with pediatric neurotransmitter diseases Affected Enzyme

HVA 5-HIAA HVA/5-HIAA MHPG 3-OMD Pterin profile

Disorders of BH4 synthesis (recessive)

decrease decrease normal decrease normal abnormal

GTP cyclohydrolase (dominant)

decrease normal normal normal normal abnormal

Tyrosine hydroxylase

decrease normal normal decrease normal normal

AADC decrease decrease normal decrease increase normal PNPO decrease decrease normal decrease increase normal Dopamine β-hydroxylase

increase normal normal decrease normal normal

Dopamine transporter (Kurian et al., 2009, Kurian et al., 2011b)

increase normal increase NR NR normal

AADC = L-aromatic amino acid decarboxylase; BH4 = tetrahydrobiopterin; GTP = guanosine-5'-triphosphate;; NR = not reported;; PNPO = pyridoxamine 5’-phosphate oxidase; Note: Data in table derived from Hyland 2008, except where otherwise noted.

2 Vesicular Monoamine Transporters The vesicular monoamine transporters are responsible for the efficient uptake of cytosolic

monoamines into storage vesicles. A 10,000-fold concentration of monoamines (up to

0.5 M) can be achieved by active transport. This transport against the concentration

gradient is driven by the transmembrane pH and electrochemical gradient generated by

the vesicular H+-ATPase in the synaptic membrane.

Two closely related vesicular monoamine transporters, VMAT1 and VMAT2, have been

characterized. The VMAT1 protein is primarily expressed in neuroendocrine cells,

including chromaffin and enterochromaffin cells, and localized to large dense core

Page 47: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

24

vesicles. The VMAT2 protein is primarily expressed in monoaminergic neurons of the

central nervous system, as well as the sympathetic nervous system, mast cells, platelets,

β cells of the pancreas, and histaminergic cells of the gut. Both VMAT1 and VMAT2 are

expressed in chromaffin cells of the adrenal medulla (Erickson et al., 1996).

2.1 VMAT Isolation The VMATs were initially isolated and characterized from chromaffin granules.

The SLC18A2 gene encoding VMAT2 was initially identified from a PC12 cDNA library

for its ability to confer resistance to the neurotoxin MPP+ in MPP+-sensitive Chinese

hamster ovary (CHO) cells (Liu et al., 1992). MPTP (of which MPP+ is the active

metabolite) is a neurotoxin that, in humans and primates, as well as other mammals,

produces a very specific phenotype—parkinsonism characterized by oxidative damage

and progressive neurodegeneration in the substantia nigra region of the brain. It is

thought that MPTP represents a greatly accelerated model of natural dopamine toxicity.

2.2 VMAT Structure Both VMAT1 and VMAT2 are acidic glycoproteins with an apparent endogenous

molecular weight of 70 kDa, and they share 63% overall amino acid identity (the related

vesicular acetylcholine transporter, VAChT, has 34% amino acid identity). The predicted

secondary structure of the VMATs comprises 12 transmembrane domains (TMDs)

(Erickson and Eiden, 1993). This includes a large lumenal loop near the N-terminus with

four putative N-linked glycosylation sites. Both the C- and N-termini of the protein are

predicted to be cytoplasmic. These regions are the most variable with respect to species

conservation. The lumenal loop and C- and N-termini are both subject to the greatest

Page 48: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

25

sequence divergence between VMAT1 and VMAT2. The VMATs are part of the broader

DHS12 superfamily of multidrug transporters of the major facilitator superfamily (MFS)

of secondary transporters that include drug-resistance proteins (toxin-extruding

antiporters), sugar uniporter proteins, H+ symporters, antiporters of organic phosphate

esters, and bacterial permeases (Marger and Saier, 1993).

2.3 VMAT Biochemistry The biochemistry of the VMATs has been extensively reviewed (Schuldiner et al., 1995,

Wimalasena, 2011, Eiden and Weihe, 2011, Henry et al., 1994). The inward transport of

each cytosolic monoamine is coupled to the efflux of two protons. The first proton is

proposed to alter transporter conformation to generate a high-affinity amine-binding site

on the cytosolic side. The second proton is proposed to generate a second conformational

change to move the amine from the cytosol to the vesicle lumen (Parsons, 2000).

The VMATs have several natural substrates: serotonin (5-HT), dopamine (DA),

norepinephrine (NE), and epinephrine (E). VMAT2 additionally transports histamine

(HIS). VMAT2 has a consistently higher affinity for all monoamine substrates than

VMAT1, though the rank order of affinity is the same for both isoforms: serotonin,

dopamine, epinephrine (Peter et al., 1994). Other substrates, including MPP+ and

methamphetamine show a similar pattern. The greatest difference in affinity, however, is

for histamine: Km of 3 µM for VMAT2 and 436 µM for VMAT1. Common structural

features among native and non-native substrates and inhibitors of VMAT include a

positive charge and aromatic ring. Hydroxyl, methoxy, and amino substituents in the ring

improve affinity, whereas negative charge in the molecule reduces affinity

Page 49: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

26

(Schuldiner et al., 1995). Specificity for a wide variety of substrates demonstrates

significant plasticity in the binding site.

In synaptic vesicles, uptake is dependent upon the magnitude of the pH and

electrochemical gradients maintained by the vesicular H+-ATPase, as well as the

cytoplasmic concentration of transmitters, the transporter density on the vesicle

membrane, and the composition of the extravesicular media (Pothos et al., 2000, Sulzer

and Pothos, 2000). The stoichiometry of transport (net transport out of the vesicle of two

H+ but only one positive charge) leads to a greater dependency on pH than on membrane

potential.

There are two commonly utilized VMAT inhibitors, reserpine and tetrabenazine.

Biochemical study has revealed that these inhibitors function by two different

mechanisms. The binding of reserpine is modulated by the proton gradient, as is

substrate binding (Darchen et al., 1989), as well as substrate concentration. It is thus

suggested that reserpine interacts with the substrate-binding site of VMAT. However,

tetrabenazine binding is not affected by the proton gradient or substrate concentration

(except at concentrations greater than 100 × Km) and therefore likely binds a unique site

of the VMAT protein. Because tetrabenazine inhibits the binding of reserpine, it is

further suggested that it binds a distinct conformation of the VMAT protein than that

bound by reserpine and monoamine substrates. Notably, VMAT1 is less sensitive to

tetrabenazine than VMAT2 (Peter et al., 1994).

Page 50: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

27

2.4 Role of VMAT2 in Biogenic Amine Physiology Although classically attributed to postsynaptic mechanisms such as receptor number and

sensitivity, evidence that receptors are not saturated under physiological conditions

provides a role for the dynamic regulation of quantal size to affect synaptic transmission.

A “quantum” is defined as a single vesicle filled with transmitter (Katz, 1971). As

reviewed by Edwards (Edwards, 2007), VMAT2 is the key determinant of vesicle filling

for serotonin and catecholamines, and therefore a mediator of quantal size for

catecholaminergic and serotonergic neurotransmission. The activity of VMAT2 may

therefore be a rate-limiting step in monoamine production and release, and its regulation

is thus relevant to the pathophysiology of perturbations in monoamine neurotransmission.

Indeed, the overexpression of VMAT2 increases quantal size (Pothos et al., 2000).

VMAT2 function depends on the cytosolic concentrations of monoamine, the transport

mechanism itself, and nonspecific leakage of monoamine across the vesicle membrane.

Low cytosolic concentrations of monoamine (possibly reflective of cytosolic toxicity) are

compensated by the relatively high affinity of VMAT2 (low Km). Furthermore, the rapid

depletion of vesicular monoamine stores in chromaffin granules with the use of reserpine

suggests substantial nonspecific leakage (Kozminski et al., 1998), which further

emphasizes and explains the importance of VMAT2 to maintaining quantal size.

In addition to its role in synaptic release, VMAT2 has also been found to localize to a

subpopulation of vesicles that undergo regulated exocytotic release in dendrites,

conferring somatodendritic release of retrograde signals (Li et al., 2005).

Page 51: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

28

The physiological consequences of VMAT2 deletion and hypomorphism have been

explored through a number of mouse models.

2.4.1 Vmat2 Knockout Mice

Complete deletion of Slc18a2 (Vmat2-/-) in mice resulted in elimination of exocytotic

release and dramatic reduction of monoamine stores in the brain. Vmat2-/- mice survive

only a few days after birth (Fon et al., 1997, Wang et al., 1997). They have extremely

low monoamine levels relative to wild-type animals (DA, 1.5%; NE, 6%; 5-HT, 1%), and

consequently a severe impairment in monoaminergic signaling that is most likely the

underlying cause of postnatal lethality. The pups feed poorly and move little prior to

dying within the first postnatal week. However, they are born in normal Mendelian ratios

(~1/4) and there are no gross differences in brain morphology. There was also no

difference in TH immunohistochemistry between wild-type and Vmat2-/- mice, indicating

that vesicular monoamine transport (and subsequent exocytotic release) are not required

for normal development of the mesostriatal projections.

In vitro cell cultures derived from mesostriatal cells of Vmat2-/- mice exhibited effectively

no release of dopamine after K+-induced depolarization, demonstrating the requirement

of vesicular transport for exocytotic release. The biogenesis and cycling of empty

synaptic vesicles were unaffected by Vmat2 deletion (Croft et al., 2005).

The motor and feeding phenotype can be partially rescued with amphetamine (Fon et al.,

1997), demonstrating that reverse efflux of monoamines through plasma membrane

transporters can mimic regulated exocytotic release with temporary success. This effect

is most likely related to dopamine. The profound deficit in monoamine levels indicates

Page 52: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

29

an increased ratio of degradation to synthesis, underscoring the importance of vesicular

storage to this balance. Inhibition of monoamine degradation by MAO also rescues

behavior and improves survival (Fon et al., 1997).

Detailed histology of the early postnatal cerebral cortex shows an increase in

developmental programmed cell death in these mice, which is partially rescued by MaoA

inhibition (Vmat2-MaoA double knockout mice) with increased serotonin levels

(Stankovski et al., 2007).

2.4.2 Vmat2 Heterozygous Mice

Mice that are heterozygous for Vmat2 (Vmat2+/-) exhibit a 50% reduction in Vmat2

expression, and normal viability and physiology compared with wild-type littermates.

They also exhibit significant reductions in monoamines (DA, 42%; NE, 23%; 5-HT,

34%) (Fon et al., 1997), demonstrating the importance of vesicular storage capacity to

monoamine content. In vitro cell cultures derived from mesostriatal cells of Vmat2+/-

mice exhibited approximately half (46.8%) the dopamine released from cells of wild-type

mice after K+-induced depolarization, demonstrating the role of vesicular transport in

determining quantal size (Fon et al., 1997). Notably, Vmat2+/- mice are sensitized to

direct and indirect DA agonists such as cocaine and amphetamine (Wang et al., 1997),

and are significantly more vulnerable to MPTP and methamphetamine toxicity

(Takahashi et al., 1997, Gainetdinov et al., 1998, Fumagalli et al., 1999).

Phenotypically, the heterozygotes exhibit moderately elevated heart rate and blood

pressure relative to wild-type littermates (Takahashi et al., 1997). Behaviorally, they

exhibit no difference in locomotor activity, passive avoidance habit, or stress response

Page 53: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

30

(Takahashi et al., 1997). However, they do express a depressive phenotype comprising

locomotor retardation, anhedonia, immobility in forced swim and tail suspension test, and

increased sensitivity to learned helplessness (Fukui et al., 2007). The forced swim and

tail suspension phenotypes were ameliorated by antidepressants with serotonergic,

dopaminergic, and noradrenergic mechanisms (fluoxetine, bupropion, and imipramine,

respectively), suggesting involvement of multiple monoamine systems in the depressive

phenotype (Fukui et al., 2007).

Additionally, the heterozygous mice were reported to prolonged QT syndrome and an

increased rate of sudden death at 2–4 months of age (Fukui et al., 2007, Itokawa et al.,

1999).

2.4.3 Constitutive Overexpression of Vmat2 in Mice

Evidence of the dose dependence of the Vmat2 phenotype was recently complemented by

a study investigating the effects of constitutive Vmat2 overexpression. BAC transgenic

C57BL/6 mice were generated that possess three additional copies of the complete

Slc18a2 gene and flanking regions (Lohr et al., 2014). These mice exhibit 3.5-fold higher

Slc18a2 mRNA levels, and 3-fold higher Vmat2 protein levels, compared with wild type

littermates. The mice exhibited increases both in vesicular uptake and in vesicular

capacity, corresponding to increased striatal vesicle volume and increased vesicle size.

Resultingly, the mice exhibited increased striatal dopamine content, synaptic dopamine

release, and extracellular dopamine.

The locomotor and behavioral phenotypes of the mice also corresponded to these

increases in dopamine neurotransmission—the mice showed increased locomotor activity

Page 54: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

31

and reduced anxiety-like and depressive-like behaviors (measured by marble-burying

assay and forced-swim test, respectively)—although these measures were likely also

confounded by serotonergic and noradrenergic effects. Finally, these mice with increased

Vmat2 expression had demonstrably reduced susceptibility to MPTP neurotoxicity, as

measured by loss of striatal dopamine terminal markers (DAT and TH).

Evidence of the dose dependence of the Vmat2 phenotype demonstrates its role as an

important modulator of monoaminergic function, in contrast to vesicular glutamate

transport in which compensatory decreases in synaptic vesicle release result in no net

increase in neurotransmission (Daniels et al., 2004). Enhancement of Vmat2 function

may therefore be an attractive therapeutic target for disorders resulting from reduced

monoaminergic function, including depression and Parkinson’s disease.

2.4.4 Selective Deletion of Vmat2 in Serotonergic Neurons

Two models of the selective deletion of Vmat2 in serotonergic neurons have been

developed to address the developmental role of serotonin. The Slc18a2 transcript is

expressed transiently in several regions of the brain during early postnatal development in

mice, in spatial and temporal co-expression with the serotonin plasma membrane

transporter (SERT) (Lebrand et al., 1998). In Vmat2sert-cr e mice, the Slc18a2 gene is

selectively deleted in all SERT-expressing cells, causing a profound depletion of central

serotonin stores (~95%), a partial depletion of serotonin in the blood, and normal

serotonin levels in the gut (Narboux-Neme et al., 2011). In Vmat2pet1-cre mice, the

deletion of Slc18a2 is specifically targeted to raphe neurons, producing a partial (~75%)

reduction in brain serotonin levels (Narboux-Neme et al., 2013). In both models, an

overall somatic growth defect was observed, consistent with the phenotype of serotonin

Page 55: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

32

depletion observed in Tph2-/- mice. Additionally, severe serotonin depletion was shown

to cause a mild delay in the development of the upper layers of the cerebral cortex, but no

impairment in barrel cortex development. Interestingly, the barrel cortex development

defects observed in Vmat2-/- mice are not reproduced in these mouse models (Stankovski

et al., 2007), and therefore likely do not derive from serotonin depletion. Generally, the

delay in cortical growth was much more mild than the overall growth defect in these

mice.

Behavioral testing of Vmat2sert-cre mice revealed decreased locomotor activity, decreased

anxiety, and decreased immobility in the tail suspension test; interestingly, the latter

result differs from observations in Vmat2+/- mice in which all monoamines are affected.

These changes were reversed by treatment with a monoamine oxidase inhibitor.

Behavioral correlates of Vmat2 depletion specifically in raphe neurons have not yet been

reported.

2.4.5 Selective Expression of Vmat2 in Noradrenergic Neurons

A Vmat2 transgene was selectively expressed in noradrenergic neurons of Vmat2-/- mice

(Vmat2-/-NE+) to investigate the role of noradrenergic defects in early postnatal lethality of

Vmat2 deletion (Ohara et al., 2013). These mice had a slightly extended lifespan

compared with Vmat2-/- mice (2–3 weeks), but a body weight of approximately half that

of wild-type littermates. Behavioral testing revealed pronounced akinesia and almost

complete elimination of the normal open field exploratory pattern. The data suggest that

noradrenergic function is critical to early neonatal survival, but survival beyond this point

is dependent on other monoaminergic systems.

Page 56: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

33

2.5 Role of VMAT2 in Disease Processes The relationship between VMAT2, affective disorders, and Parkinson’s disease stems

from the early discovery that chronic administration of reserpine—initially introduced for

the treatment of hypertension—causes central nervous system effects including aspects of

depression and Parkinsonism. This observation lead to the monoamine hypothesis of

affective disorders (Freis, 1954), and the recognition of the role of dopamine depletion in

Parkinson’s disease.

2.5.1 VMAT2 and Parkinson’s Disease

Dopamine toxicity is a controversial topic suffering a lack of direct evidence in vivo.

Importantly, toxic dopamine is that displaced from vesicles and subject to oxidation;

greater than 90% of intracellular dopamine is normally sequestered in the reducing

environment of vesicles (Eisenhofer et al., 2004). Dopamine is highly reactive, and can

yield reactive species including hydroxyl radicals, superoxide, hydrogen peroxide, and

dopamine quinones (Graham, 1978, Hastings et al., 1996). Reaction of quinones with

proteins can create cysteinyl adducts; L-DOPA and DOPAC cysteinyl adducts are also

formed. Superoxide further reacts with nitric oxide to produce reactive nitrogen species.

All of these species may contribute to the oxidative stress underlying the multifactorial

pathogenesis of Parkinson’s disease, and the unique vulnerability of nigrostriatal

dopamine neurons (Jenner, 2007). Therefore, VMAT2 function and regulation may be

critical to neuroprotection through its role in maintaining dopamine homeostasis.

Several lines of indirect evidence suggest a role for VMAT2 in Parkinson’s disease

pathogenesis. These include the parkinsonian side effects of the specific VMAT

Page 57: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

34

inhibitor, reserpine; its role in the sequestration of MPP+ conferring protection against

MPTP-induced parkinsonism; evidence of a direct interaction between VMAT2 and

α-synuclein and an increase in cytosolic dopamine and reactive species with α-synuclein

overexpression (Lotharius and Brundin, 2002, Mosharov et al., 2006, Guo et al., 2008);

and the mechanism of methamphetamine toxicity disrupting vesicular dopamine

sequestration (Cubells et al., 1994, Sulzer and Rayport, 1990). Furthermore, a

hypomorphic Vmat2 mouse model appears to recapitulate relevant molecular and

pathological hallmarks of PD.

2.5.1.1 Vmat2 Hypomorphic Mice

In addition to the previously described models of Vmat2 deletion, hypomorphic mice

were studied to assess the effects of significant Vmat2 deficiency in animals that are able

to overcome the barrier of early postnatal lethality exhibited by Vmat2-/- mice. These

mice were created accidentally during the process of generating complete knockout mice,

in which the knockout cassette was stably inserted into intron 3 (Mooslehner et al., 2001).

Termed the KA1 strain, these mice also unintentionally possessed a spontaneous deletion

encompassing the D-synuclein locus (not recognized in the initial reports).

KA1 mice exhibit 95% reduction in Vmat2 protein levels, and corresponding reductions

in tissue monoamines of 92% (dopamine), 87% (norepinephrine), and 82% (serotonin).

Interestingly, in vitro assay of electrically stimulated dopamine release measured only a

70% reduction compared with age-matched wild-type animals (Patel et al., 2003).

Compared with a 95% reduction in protein levels, this result demonstrates the presence of

compensatory mechanisms for the reduction in Vmat2—likely a redistribution of Vmat2

from reserve pools to actively cycling vesicles.

Page 58: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

35

In KA1 mice, no differences were observed in D1/D2 receptor sensitization, but there

was supersensitization of D2/D3 autoreceptors (Colebrooke et al., 2007). In addition,

downregulation of TH phosphorylation was observed at residues involved in feedback

inhibition. No differences in DAT expression or activity were observed (Colebrooke et

al., 2006). Importantly, these mice were found to downregulate substance P expression,

and upregulate enkephalin in the striatum, demonstrating differences in underlying

architecture of the striatum (Mooslehner et al., 2001).

Behaviorally, the KA1 mice exhibit decreased locomotor activity and impairment in

motor coordination (measured by beam walking and rotarod) that worsens with age, but

normal reactivity in the novelty place preference task (Mooslehner et al., 2001). The

motor deficits were responsive to L-DOPA administration. KA1 animals were also

predictably more sensitive to MPTP toxicity and amphetamine.

No evidence of neurodegeneration was evident in Vmat2-deficient KA1 mice at any age,

but this was later attributed to the lack of D-synuclein in these mice. A strain of Vmat2-

deficient mice was therefore created in which wild-type D-synuclein was reintroduced

through selective breeding. These Vmat2-deficient mice exhibited an 85% reduction in

striatal dopamine with associated reductions in DOPAC and HVA (Caudle et al., 2007).

Importantly, age-related declines in striatal dopamine were observed in these mice, and

were associated with several hallmarks of PD (Caudle et al., 2007). These mice exhibited

molecular markers of oxidative stress and damage, including cysteinyl-DOPA and

DOPAC adducts at 2 and 12 months of age, and protein carbonyls and 3-nitrotyrosine at

12 months of age. Progressive cell death and loss of TH-positive neurons was observed

Page 59: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

36

within the SNpc in older animals. More dramatic progressive cell loss was observed in

the locus coerulus (LC), preceding that in the SNpc, consistent with observations in

human patients with PD (Braak et al., 2003). Additional progressive motor deficits were

also observed at 28 months of age, including a shorted forepaw stride length that may

mimic shuffling gait. These motor deficits were responsive to L-DOPA.

Although age-dependent alterations in the nigrostriatal system were observed in both

Vmat2-deficient mouse strains, progressive cell loss was only observed in the Vmat2-

deficient mice with intact D-synuclein, implicating the latter in the severity of the

phenotype and the mechanism of cell death.

Nonmotor deficits analogous to the nonmotor symptoms of PD were also assessed in

these Vmat2-deficient animals. These mice were observed to have progressive deficits in

olfactory discrimination, altered sleep latency, delayed gastric emptying, and anxiety-like

and depressive phenotypes (Taylor et al., 2009). As in PD, these nonmotor deficits

preceded the severe motor deficits, and the pattern of L-DOPA responsiveness was

similar (with the exception of sleep latency, which was L-DOPA–responsive in Vmat2-

deficient mice and is not responsive in human patients with PD) (Taylor et al., 2009,

Taylor et al., 2011).

2.5.2 VMAT2, Neuropsychiatric Phenotypes, and Drugs of Abuse

That the monoaminergic systems play a role in neuropsychiatric phenotypes is well

established, although the precise mechanisms are poorly understood. The particular role

of VMAT2 in this respect, however, has not been clearly investigated. As discussed

above, mouse models of VMAT2 deficiency (Vmat2+/- and hypomorphic Vmat2 mice)

Page 60: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

37

exhibit phenotypes related to depression and anxiety. In addition, SLC18A2 was

identified in multiple genome-wide association studies of schizophrenia, as well as

alcohol dependency and post-traumatic stress disorder (see Section 2.5.3). Nonetheless,

the critical role of VMAT2 in determining quantal size makes it an attractive target for

further study into the perturbations of monoaminergic function that underlie these

phenotypes.

More evidence exists for a role of VMAT2 in the mechanism of action of drugs of abuse.

Amphetamines have multiple effects within the synapse that combine to produce

non-exocytic release of dopamine. Efflux occurs through reverse transport of dopamine

through DAT. The amphetamine- and methamphetamine-induced efflux of dopamine out

of cells, however, first requires the desequestration of dopamine from vesicles.

Amphetamine prompts this by one or both of two proposed mechanisms: VMAT2

inhibition or eliciting DA reverse transport as a VMAT2 substrate, and by collapsing the

synaptic vesicle pH gradient through competition for intravesicular protons (Fleckenstein

et al., 2007, Sulzer et al., 2005). In the case of methamphetamine, desequestration was

shown to be tetrabenazine-sensitive (Volz et al., 2006).

Some evidence has been provided that drug treatments may cause differential

redistribution of VMAT2 within striatal synaptic terminals. Administration of

methamphetamine was shown to redistribute VMAT2 in striatal synaptosome

preparations out of an enriched vesicular fraction in conjunction with a decrease in

vesicular DA content (Riddle et al., 2002, Sandoval et al., 2003, Eyerman and

Yamamoto, 2005). Cocaine and methylphenidate primarily act as inhibitors of dopamine

reuptake. However, both have been shown to redistribute VMAT2 from a membrane-

Page 61: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

38

associated vesicle fraction to a vesicle-enriched, non–membrane-associated fraction

(Sandoval et al., 2002, Riddle et al., 2002).

Importantly, differential sensitivity to the locomotor effects of amphetamine, cocaine,

and ethanol (Takahashi et al., 1997, Wang et al., 1997), and methamphetamine toxicity

(Fumagalli et al., 1999), as well as altered ethanol-associated behaviors (Hall et al., 2003,

Savelieva et al., 2006), have been demonstrated in Vmat2+/- mice. Much work remains

be done to truly understand the role of VMAT2 in the mechanism of action of drugs of

abuse.

2.5.3 Genetic Variation in VMAT2

Although no direct disease-associated VMAT2 mutations have been identified prior to

that reported in this thesis, some associations have been discovered between single

nucleotide polymorphism (SNP) alleles in SLC18A2 introns or promoter and Parkinson’s

disease (Brighina et al., 2013), schizophrenia (Talkowski et al., 2008, Chu and Liu, 2010,

Gutierrez et al., 2007, Simons and van Winkel, 2013), post-traumatic stress disorder

(Solovieff et al., 2014), and liability to or protection against alcohol dependency (Lin et

al., 2005, Fehr et al., 2013).

Gain-of-function promoter haplotypes may also be associated with a protective effect

against Parkinson’s disease in women (Glatt et al., 2006). As expected for traits that are

highly polygenic, the magnitudes of effect in each case are not conclusive, and functional

validation of mechanistic involvement of VMAT2 would be required to draw further

conclusions.

Page 62: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

39

3 Thesis Overview A new pediatric neurotransmitter disorder was discovered in eight cousins of a

consanguineous Bedouin family in Saudi Arabia, and a causative mutation in VMAT2

(SLC18A2) was discovered in the present work. Understanding the underlying

mechanism of this disorder extends the spectrum of known pediatric neurotransmitter

diseases, serves as the first demonstration of mutation in VMAT2 causing a human

phenotype, and thereby provides new insight into the role of VMAT2 in monoamine

homeostasis.

This thesis additionally demonstrates the utility of implementing genomic diagnosis in

the clinic, with respect to providing simple and effective treatments in a timely manner to

improve outcomes for patients with rare inborn errors of metabolism.

My thesis objectives were the following:

(1) To characterize this new pediatric neurotransmitter disorder.

The disease was hypothesized to involve a defect in monoamine neurotransmitter

pathophysiology. To provide evidence of the underlying pathophysiology of the

disease, careful clinical investigations, including genetic analyses of candidate

genes, were undertaken to exclude known diagnoses and produce a

comprehensive clinical description of the disorder and its progression. In

particular, neurotransmitter metabolite profiles were assessed in both CSF and

urine. This data informs the differential diagnosis of this particular disease and

related disorders; however, genetic and treatment evidence were further required

to confirm this hypothesis.

Page 63: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

40

(2) To identify the genetic mutation causing this disease and characterize the effect of

the mutation

I hypothesized that a homozygous genetic mutation was causing this autosomal

recessive disorder in affected individuals of a consanguineous pedigree. To

investigate this hypothesis, whole-genome SNP analysis was performed on 5

patients and 3 unaffected siblings, in combination with linkage and homozygosity

analyses, to identify the region containing the disease-causing mutation. Sanger

sequencing of candidate genes within the locus identified a nonsynonymous

variant in the SLC18A2 gene encoding the vesicular monoamine transporter 2

(VMAT2) protein. The possibility of other variants in the region and in other

candidate genes in the genome was excluded by whole-exome sequencing of the

proband.

(3) To infer treatment targets on the basis of the biochemical and physiological role

of the identified disease-causing mutation

Patients exhibited an initial positive response to treatment with

levodopa/carbidopa followed by a rapid decline and worsening of the phenotype.

This poor response to dopamine replacement is consistent with the presumed

normal dopamine biosynthetic capacity in these patients combined with aberrant

dopamine storage and release. It was then hypothesized on the basis of the known

defect in VMAT2 function that treatment with a dopamine agonist would

stimulate dopamine receptors while bypassing the requirement for vesicular

storage and release. Subsequent treatment with a dopamine agonist, pramipexole,

Page 64: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

41

resulted in a remarkable response in all patients, with the most substantial

recovery occurring in younger patients.

Much of the work contained in this thesis and the major conclusions were published in:

RILSTONE, J. J., ALKHATER, R. A. & MINASSIAN, B. A. 2013. Brain

dopamine-serotonin vesicular transport disease and its treatment. N Engl J Med,

368, 543-50.

Page 65: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

42

Chapter 2 Clinical Characterization of a New Pediatric Neurotransmitter

Disease

1 Introduction The pediatric neurotransmitter diseases (PNDs) mimic the phenotype of other

neurological disorders, and are therefore often misdiagnosed or remain without diagnosis

for years. Common misdiagnoses include cerebral palsy, hypoxic ischemic

encephalopathy, paroxysmal disorders, inherited metabolic diseases, and genetic dystonic

or parkinsonian syndromes. Further hindering accurate and timely diagnosis, several of

the pediatric neurotransmitter diseases are exceedingly rare. The most similar syndromes

to that presented in this thesis (TH deficiency and AADC deficiency) reflect worldwide

diagnoses of just 50 and 100 patients, respectively. Increasing awareness in the clinical

community of this group of disorders and the clinical spectrum of monoamine deficiency

will aid the recognition of these disorders. In this chapter, the clinical investigation and

diagnosis of a new autosomal recessive pediatric neurotransmitter disease is described.

The diagnosis of monoamine neurotransmitter disorders is based on clinical history,

neurological examination, biochemical investigations (including specific cerebrospinal

fluid investigations), enzyme analysis, and genetic analysis. Conclusive diagnosis of

biogenic amine deficiency in the central nervous system is often demonstrated by clinical

measurements of their primary metabolites in cerebrospinal fluid. In the present case,

several lines of evidence coincided—most importantly, the genetic results described in

Chapter 3—to conclusively diagnose this new disease. The relevant investigations are

presented in this chapter to characterize this new syndrome, demonstrate the involvement

Page 66: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

43

of biogenic amines in its etiology, and present a successful treatment strategy.

Additionally, difficulties in diagnostic strategy are discussed, including the pitfalls of

cerebrospinal fluid metabolite measurements as a conclusive diagnostic method.

2 Methods

2.1 Patients The patients in this study were identified in the Khobar province of Saudi Arabia by Dr.

Reem Alkhater, who undertook neurological examinations and relevant investigations.

The study was approved by the Hospital for Sick Children’s research ethics board.

Written informed consent was provided by the parents, with the patients providing their

assent for participation in the study.

2.2 General Investigations Investigations presented here were performed as part of regular clinical care for these

patients, and included neurological examination, bloodwork and metabolic screens,

video-EEG, magnetic resonance imaging (MRI), and MR spectroscopy.

2.3 Clinical Measurement of Neurotransmitter Metabolites Lumbar puncture was performed in a younger affected sibling of the proband at the age

of two to allow measurement of monoamine neurotransmitter metabolites, intermediates,

and precursors. Samples were drawn sequentially into numbered tubes to ensure

collection of the appropriate fraction. The tube for analysis contained 1.0 mL of CSF

collected directly from the tap needle after previously dispensing 1.0 mL of CSF. This

tube contained appropriate antioxidants for preservation of the analytes. The tube was

Page 67: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

44

immediately frozen at –80°C and shipped on dry ice for analysis. The sample contained

no visible blood contamination.

Metabolites were measured at the Service de Génétique Médicale of the Centre

Hospitalier Universitaire de Sherbrooke Fleurimont by liquid chromatography tandem

mass spectrometry (LC-MS/MS) according to established methods.

A urine sample was also collected from the same patient. This sample was analyzed for

relevant neurotransmitter metabolites at the Mayo Clinic in Rochester, MN.

2.4 AADC Enzyme Test The activity of aromatic L-amino acid decarboxylase (AADC) was assayed commercially

by plasma enzymology at Medical Neurogenetics. Plasma was separated immediately

upon collection of a blood sample in an EDTA tube and stored at –80°C before shipping

on dry ice for analysis.

2.5 Candidate Gene Sequencing A total of 5 mL of whole blood was collected in EDTA tubes. Genomic DNA was

isolated at the Hospital for Sick Children’s DNA lab. Commercial sequencing of a panel

of genes associated with spinocerebellar ataxia (SCA) and those associated with

mitochondrial DNA disorders was performed.

Primers were designed to amplify the exons of each gene and approximately 50

bp of flanking intronic sequence (Table 3). The reaction was performed using Picomaxx

DNA polymerase (Stratagene) according to the manufacturer’s specifications. Cycling

conditions incorporated an initial denaturation at 95°C for 5 minutes, followed by 30

cycles of 95°C for 30 s, 55°C for 30 s, and 72°C for 30 s, and a final extension step at

Page 68: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

45

72°C for 10 minutes. Amplification products were visualized by gel electrophoresis, and

purified from bands of the appropriate size using a Qiagen Gel Purification Kit according

to manufacturer’s specifications. Purified PCR products were sequenced using both

forward and reverse primers in separate reactions.

Sequences were compared to reference genome sequence using the BLAT tool in

the UCSC Genome Browser (Meyer et al., 2013). Putative variants were verified by

manual inspection of electropherograms.

Table 3 Primer sequences for the amplification of exons of candidate genes associated with known pediatric neurotransmitter diseases Gene Exon Sequence Size PTS 1 AGC GGA GAC GCA CTT CCT A 389 bp

GAC ACT CCA GCC CCC ATC 2 TTG GTG AGC TAA AGT AAT AAA TTG

G 300 bp

TCC GTA AGT TTT CCC ATT CTT 3 AGC TTT TGG GGA CAG ATC TAA 250 bp

AAG CAA TAC TGA CTG GAA CAG TTT 4 GGA TGA AGG CAA ATG TGC AA 298 bp

CCA GTT CTA TTC ACA AAG TCA TGG 5 GAC AGC TGG GCC TGA CTT TA 380 bp

GAA ATT CTA GTT TCG AAA GAT TTC AT

6 TTT GAT TGT TGT GTG ATT TCT GA 392 bp AAT AGG CAC TCC AGA GCA CAA

SPR 1 CAG ATC CCA AGG GAA CCA G 600 bp GCT GGA ACA ACT AGG GCT TTA

2 TAG GTT CCC CAG CTG TGT CT 575 bp ATC CTG TGG GTT GTT TCC TG

3 AAG ATG ACG TGT TTC CTC TGG 400 bp CCC TAT GGC AGG GTG TGG

QDPR 1 GCC TGG CCG AAG TTA CAG T 428 bp GTG CAA GCA ACA CGA GTC AG

2 TCC CAA AGT GCT GGC ATT AC 298 bp GCC AAA GGA AGA ACA TAC AGC

3 CAA AGC ATT AAT TGC CAG GT 300 bp AGT GCA AAC CCA ATC CTT GT

4 GCC CTG TGC TGT TTG TGT TA 371 bp

Page 69: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

46

TCC TCA TCC CAT GAA AGT GC 5 CGT CTG ACC TGA AGG AGG AG 363 bp

CAG AAC CAG AGG TGA GAG CA 6 TAG CAC CCT CAG TGC CAG A 300 bp

GGG AAC ACA GAC TTG TCC TCT 7 TTA AAC AGT CGC TGC TGT GC 377 bp

GGA GAG CAA ATG CAT ATT ATG TGA GTCPH1 1 GCG TAC CTT CCT CAG GTG AC 561 bp

AGT GAG GCA ACT CCG GAA AC 2 CAG AAA AGG ACT TTG CTA CTT TGA 476 bp

GCC TTC TGC TAC TTT GGT TTT G 3 AAC AGT TCC AGA TGT TTT CAA GG 379 bp

GTA GGG GAC GAG AAG GAA GG 4 GTC CTT TTT GTT TTA TGA GGA AGG C 288 bp

GGT GAT GCA CTC TTA TAA TCT CAG C 5 TGG TGT GTC TTG GCT CTT AAA 300 bp

CCT GGT GCT ACA AAA TAT GAG AA 6 CCG CAG TTA CTT TTG CAT GA 396 bp

CAC ATC TGT AAC AAT TGA AAA TGG A

SLC6A3 1 TGG CTG AAG ACC AAG AGG G 425 bp CTC GTT TCC GTA CGT GCC

2 ATG GAT GGT TGA CTG GGG TA 335 bp TAG CAA AGC AGG GCT GGA T

3 TGG GCT CAG GGT AAT GTC TC 490 bp CCA GAG CAC TAA AGG GAT GG

4 AGT TCC AGG TGG GTT GAC AG 304 bp AGC ACA AAA CCC AAC TGA GG

5 CTC CCA ATC AGA GGA CAA GC 354 bp CCT GGT GTC TGC AAC TCT GA

6 CAC TTC CTG AGG CTG CAT CT 361 bp TCT CAT CCA GGG ACA CCC TA

7 AAA AGT AGC CCC TCC GAA GA 383 bp TGA GGC AGC AAC TCT CAC TG

8 CGC ACC AGC CCT AGT CTC TA 424 bp TCC AGT CAC CAC TCA CTC CA

9 AAA CCC CCT ACC GTG GAT AC 561 bp TGA CTC TGG GAC CAA GCT CT

10 TAC ACG TGG CCC TAA GAA GC 442 bp AGC ACC TCA CTG ATC CCA TC

11 CTT GGG AGT CAG CGA GGA 314 bp AGT CTT GAG GCC CCT GAC TC

12 GAG ACG CTC TGC CAT GAA GT 311 bp CTT TCT GGT GGC CTC ACA CT

13 CTG GCA GTG GGT ACT GGT CT 200 bp

Page 70: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

47

CTG GGG GCT AAG AAC ACT GA 14 CAG GAG GCT GCA AAC TGT CT 2.2 kb

AGG CCG GAA GGA GAG ATG TH 1 GCC TCC CTC CTT CCT CAC 379 bp

GGT TTG CAT GGA CCC TGA 2 AAA TGG GTT TTT ATT TAT GGA CCT T 400 bp

GGG ACT TGG CAG ACA CCT G 3 CTC AAA AAC GTG CTC TCA TCC 500 bp

AGC TGA GGC CTG AGA CTC C 4 AAG AAC GGG ATC TGT GTG CT 479 bp

CAC GGA TGT GTA GCA AAA CG 5, 6 CCC CCG GAA GTC TTG TAG G 500 bp

GGT CCT CCC CTT TGT CCT T 7 CAC CCT CCT GTC CAT CCT C 293 bp

CTC TCC TGT TGT GCC AAG GT 8 ACT GGG GTG GGG CAT TAG 396 bp

CAC TGG AAC ACG CGG AAG 9 GGG GAT GGT CAG CCA AGC 297 bp

CGC GTA GGA GGG AGA AGG 10 ACT CCC CTG AGC CGT GAG 268 bp

AGC AGG CAG CAC ACT TCA C 11 AGG GAA GTG TCC CAG AGA CC 235 bp

AGA GGG TGA GGC CTG GAT T 12 TGT CTC TGG GCT GAT GCT G 250 bp

AGA GCC TGA GTC CTG GAG GT 13 TGG AGT CAG TGA TGC CAT TG 242 bp

CTC AAG GCC AGA AGG AAG G 14 TCT GAG CCA CTG TGA AGG TG 296 bp

GTT GGG AAG GGC CCT CAG DDC 1 GGG GAG GCA GAC ACT CTG T 228 bp

GGA GGA GAA TTC AGC ACA GC 2 TCC TAC AGA CAT GGA GGG AAA 494 bp

TGC CAT AGG GAT TCC TTG AA 3 ACA TTT GGG GAA CTG CAC TC 340 bp

CAG GTC CCT TGT GCA TAG GT 4 TCT GGG CTT TAG TGG AAG GTT 300 bp

GTG CCT CTT TCC CCA CCT 5 GGA CAC AAA ACA ATA TGT CTT CCA 300 bp

TGG TTT GGT TTG AAT TTG ACA 6 CCT TGT GTT TGC AAT GTT GG 300 bp

CCC GAG TAG CTG GGA TTA CA 7 CAT TGG GAT CTC GGC TCA T 300 bp

GGC AAA CCA TCA CAA TAT GAA 8 CAA GAA GGT GCT CAG ACA GG 299 bp

TTG GCT GAA ACA AAC CTC AA

Page 71: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

48

9 CAC TGT GGA TTA GTT GTG CCA TA 296 bp GCA GCA AGC AGT GAG CTA AG

10 CCC AGG TAC TTG GAG CAG AG 390 bp TAC AAG GGC AAA TCC AGG AA

11 GCC TTT GGG CAG TTT TAT TTC 249 bp CGT GCT GAT CAT GAG AGT GG

12 GTT GGC CAC CAG GGA ATC 300 bp GCA GTG AGC TGA GAT TGT GC

13 TGC CAA GAG CGT CTA AAT GA 392 bp CGT GGA AAC AAG GCT GTG TA

14 GTA GGG TTG CCA AGC ACT GT 395 bp CCT GTA GCT GGG TCT GGA CT

15-1 ACC GTG GAA AGA GAG GGA GA 475 bp GTT GCG TGA ACA TTG ATT GC

15-2 GAG GGT TGT GAT TTT GTC TGC 400 bp TGC CGT TTA AAA ACA TCC AA

3 Results

3.1 Patient History and Neurological Examination The index case was a 16-year-old female with global developmental delay and abnormal

movements. She had initially been brought to medical attention at 4 months of age with

hypotonia, loss of acquired head control, and paroxysmal stereotyped episodes of

persistent eye deviation, and crying lasting hours. Video-EEG monitoring had excluded

seizures, and a symptom diagnosis of oculogyric crisis had been made. Development had

been normal initially, but had slowed after presentation. She had developed the ability to

sit at 30 months, crawled at 4 years, and walked at age 13.

Upon presentation at the age of 16, she was experiencing fatigue, excessive diaphoresis,

profuse nasal and oropharyngeal secretions, noisy breathing, hypernasal speech, poor

distal perfusion, cold extremities, disrupted sleep, hypotonia, ataxia, dysarthria, and

incoordination. There was no diurnal variation and no improvement with vitamin B6 or

folinic acid.

Page 72: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

49

Neurological examination of the proband at age 16 revealed ptosis, hypomimia, facial

dyskinesia, and limited upward gaze. She had axial hypotonia and appendicular

hypertonia specifically involving the extensor muscles of the upper and lower

extremities, and her power was 4+/5 in all muscle groups. Deep tendon reflexes were

2+/4 and symmetric. Plantar reflexes were flexor, and there was no clonus. Coordination

testing revealed a fine tremor, and dysdiadochokinesia in the upper and lower extremities.

Her gait was parkinsonian with typical shuffling, her posture was stooped, and her

postural reflexes were diminished. She walked with bilateral alternating dystonia of

hands and feet with intermittent toe walking and foot inversion, and was unable to

tandem walk.

3.2 Investigations The patient’s basic bloodwork and metabolic screens were normal (Table 4). Repeat

overnight video-EEG revealed neither seizures nor interictal abnormalities, and MRI and

MR spectroscopy were normal (Figure 5).

Table 4 Serum metabolic screen revealed no abnormalities

Amino acids Vitamin B12 Biotinidase Copper Ceruloplasmin Lead Very long chain fatty acids Carnitine Ammonia Lactate Pyruvate

Page 73: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

50

Figure 5 T2-weighted magnetic resonance images of proband at age 14 revealed no abnormalities.

Page 74: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

51

Lumbar puncture was performed in a younger affected sibling at the age of 2 to measure

monoamine neurotransmitter metabolites, intermediates, and precursors—all of which

were normal (Table 5). The urine neurotransmitter profile, however, revealed increased

monoamine metabolites [5-HIAA = 17.6 µg/dL (reference range: 0–6 µg/dL); HVA =

14.1 µg/mg Cr (0–13.4 µg/mg Cr)] and a decrease in measurable urine monoamines

[norepinephrine = 1.1 µg/dL (4–29 µg/dL); dopamine = 19 µg/dL (40–260 µg/dL)]

(Table 5). Plasma was tested commercially for AADC enzyme activity; this result was

also normal.

Page 75: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

52

Table 5 Cerebrospinal fluid and urine neurotransmitters and their metabolites measured in a younger affected sibling of the proband reveal decreased monoamines and elevated metabolites in urine, but not in cerebrospinal fluid. Values outside reference ranges are presented in bold.

Cerebrospinal Fluid 5-HIAA HVA HVA/5-HIAA 3-OMD 5-OHTrp 5-HT Normal Range 74–345 nM 233–928 nM 1.5–4.1 <150 nM <25 nM no reference Patient 169 nM 314 nM 1.9 20 nM 12 nM <5 nM Urine 5-HIAA Norepinephrine Epinephrine VMA HVA Dopamine Normal Range 0–6 µg/dL 4–29 µg/dL 0.0–6.0 µg/dL 0–12.9 µg/mg Cr 0–13.4 µg/mg Cr 40–260 µg/dL Patient 17.6 µg/dL 1.1 µg/dL 0.5 µg/dL 5.5 µg/mg Cr 14.1 µg/mg Cr 19 µg/dL HIAA, hydroxyindoleacetic acid; HVA, homovanillic acid; OMD, O-methyldopa; OHTrp, hydroxytryptophan; HT, hydroxytyramine; Cr, creatinine; VMA, vanillylmandelic acid.

Page 76: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

53

3.3 Family Structure The proband is a member of a consanguineous Saudi family that includes 6 children

sharing an identical clinical picture of complex movement disorder (Figure 6). The

proband (V:6) was the eldest of 6 siblings, three of whom also presented with the disease

(V:9, V:10, and V:11). Two siblings were unaffected (V:7 and V:8). The parents had

also experienced two spontaneous abortions. An additional affected individual (V:3) is a

second cousin of the proband. A sixth affected individual (VI:2) was a first cousin once

removed of the affected children.

The parents of the affected individuals exhibited no movement disorder. The ratio of

male to female affected individuals is approximately 1:1, and there is no evidence of

maternal inheritance. The mode of inheritance is therefore autosomal recessive.

Page 77: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

54

Figure 6 Family structure of kindred demonstrates autosomal recessive mode of inheritance and consanguineous pedigree structure. Proband is labelled V:6. Cerebrospinal fluid and urine neurotransmitter analyses were performed on individual V:10. Black, affected; white, unaffected; square, male; circle, female; diamond, spontaneous abortion. Numbers in squares or circles indicate number of male or female offspring, respectively. From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 78: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

55

Two additional affected cousins were identified. These two affected individuals were full

siblings of each other (VI:3 and VI:4) and first cousins of one of the affected individuals

(VI:2). At the time of analysis, the eldest was a 31-month-old boy. In addition to the

phenotype of hypotonia, these siblings exhibited bilateral anopthalmia. There is no clear

theoretical relationship between the neutrotransmitter phenotype and this developmental

phenotype. There is also no documented anopthalmia in other pediatric neurotransmitter

diseases.(Kurian et al., 2011a, Pons, 2009) Given the consanguineous background of

these affected individuals, they may simultaneously express an unrelated autosomal

recessive trait caused by an independent genetic mutation. A karyotype should be

performed for these siblings to identify any chromosomal abnormality, and these patients

may further opt to undergo whole-exome sequencing to identify additional homozygous

nonsynonymous variants.

3.4 Genetic Screening of Candidate Genes The clear autosomal recessive mode of inheritance in this consanguineous family belies a

single causative genetic mutation. Initial screening of some candidate genes was

performed in the clinical laboratory setting. Clinical genetic screening of candidate genes

revealed no mutations associated with SCA or mitochondrial disorders.

After the suggestion was made of biogenic amine involvement, the causative

genes for all known dopamine deficiencies were screened. Genes known to be associated

with pediatric neurotransmitter diseases were screened by sequencing of exons and

flanking intronic sequences (Table 6). The causative gene for pterin-4α-carbinolamine

dehydratase deficiency (PCD) was not sequenced because PCD does not present with

Page 79: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

56

movement disorder features, and the causative gene for PLP was not sequenced because

seizures were not a component of the patients’ histories. No coding mutations or putative

splice variants were identified in any of the sequenced candidate genes.

Whole-genome investigations were undertaken to identify the causative mutation,

and a mutation was discovered in the SLC18A2 gene encoding vesicular monoamine

transporter 2 (VMAT2). These genetic studies are described in Chapter 3.

Table 6 Pediatric neurotransmitter disease–associated genes screened in the proband revealing no putative mutations Gene Symbol

Number of Coding Exons

Protein Name Associated Disorder

DDC 9 Aromatic L-amino acid decarboxylase

AADC deficiency

SLC6A3 14 Dopamine transporter (DAT) Dopamine transporter deficiency syndrome (DTDS)

QDPR 7 Dihydropyridine Receptor (DHPR) DHPR deficiency GCH1 6 GTP cyclohydrolase I GTPCH1 deficiency

Segawa disease Dopamine-responsive dystonia (DRD)

PTS 7 6-Pyruvoyltetrahydropterin synthase (PTPS)

PTPS deficiency

SPR 3 Sepiapterin reductase (SR) SR deficiency TH 13 Tyrosine hydroxylase (TH) TH deficiency

3.5 Drug Response On the basis of the parkinsonism and the diminished urine dopamine, the proband and

three of her younger affected siblings were initially treated with L-DOPA/carbidopa

(approximately 1 mg/kg/day), which resulted within 1 week in major deterioration, with

Page 80: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

57

the appearance of intense chorea and worsening of the dystonia. Discontinuation of the

medication led to rapid return to baseline in all four children.

On the basis of the identification of the underlying VMAT2 mutation (described in

Chapter 3), the decision was made to attempt treatment with a direct dopamine receptor

agonist (pramipexole; 0.02 mg/kg/day). This treatment resulted within 1 week in

dramatic and sustained disappearance of parkinsonism and dystonic attacks, and

improvement of other symptoms.

The younger affected siblings were then treated. They exhibited even more remarkable

recoveries; the younger the particular child at the time of initiation of treatment, the

greater the number of symptoms that were corrected and the greater the extent to which

symptoms improved. The effect of treatment at each age is presented in Table 7.

Page 81: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

58

Table 7 Age at initation of dopamine agonist affects disease course

Feature Age at treatment

18 years 11 years 7 years 3 years Cognition and ability to learn

Mildly improved Mildly improved

Moderately improved

Greatly improved. Able to make stories from pictures.

Occulogyric crises

No further events; on a dose higher than her siblings

No further events

No further events

No further events

Dystonia Gait dystonia persists Gait dystonia persists

Gait dystonia persists

Gait dystonia improved

Parkinsonism Improved Improved Improved Improved

Fine motor skills Improved coordination, able to feed self, drink from cup, and hold a pen. Improved handwriting

Improved coordination, learning to hold a pen. Unable to write or read

Learning to hold a pen and drink from a cup independently. Unable to write or read

Able to write, learning to read

Language and speech

Dysarthric No language development

Mama and Papa Normal language development and mild dysarthria

Gait Improved posture and fatigue (had started walking at age 13)

Started walking within days of treatment

Started walking within days of treatment

Started walking within days of treatment

Page 82: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

59

3.6 Summary of Clinical Features and Differential Diagnosis

The salient features of the disorder are summarized in Table 8. The syndrome is onset in

early infancy (approximately 4 months of age) with the appearance of hypotonia and the

loss of motor milestones. As the affected children age, they develop features of

movement disorder, cerebellar symptoms, and autonomic symptoms. As initially

observed in the proband (the eldest patient), the movement disorder phenotype exhibited

in early childhood (dystonia, oculogyric crises, facial dyskinesia, and chorea) evolves by

the age of 11 into parkinsonism characterized by bradykinesia, rigidity, stooped posture,

and shuffling gait.

Table 8 Clinical features common to all affected individuals in the pedigree Features Onset at age 4 months Hypotonia Hypomimia Paucity of movements Oculogyric crises Attacks of dystonia Dysarthria Ataxia and incoordination Excessive diaphoresis Profuse nasal and oropharyngeal secretions Poor distal profusion and cold extremities Disrupted sleep Mild cognitive impairment No diurnal variation Evolution of the movement disorder by age 11 years into a picture closely resembling Parkinson’s disease, with dyskinesias

The composite clinical picture in this family includes features that can be attributed to

deficiencies of particular biogenic amines. A categorization of the clinical symptoms is

Page 83: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

60

presented in Table 9. The movement disorder symptoms, as well as ptosis and

hypersalivation, are considered signs of dopamine deficiency. The manifestations of

serotonin deficiency include temperature instability, sleep disruption, sweating, and

possibly dystonia. Many of the autonomic disturbances experienced by these patients

may be attributable to the deficiency in norepinephrine. Although this model is useful for

understanding, it should be recognized that significant interaction among the biogenic

amine pathways clouds these distinctions. Additionally, some features are difficult to

directly attribute to the actions of the biogenic amines, such as mild cognitive

impairment.

Table 9 Clinical features of the disease organized by category Onset 4–7 months

hypotonia and developmental regression Movement Disorder oculogyric crises, dystonia, facial

dyskinesia, chorea Parkinsonism bradykinesia, rigidity, shuffling gait,

stooped posture Cerebellar symptoms ataxia, dysarthria, tremor Autonomic symptoms ptosis, fatigue, diaphoresis, excessive nasal

and oropharyngeal secretions, noisy breathing, hypernasal speech, disrupted sleep, poor distal perfusion

Medication responsiveness L-DOPA/carbidopa: temporary improvement, rapid decline and worsening of initial phenotype Dopamine agonist (pramipexole): Responsive

The multisystemic constellation of symptoms of this disorder fall within the spectrum of

pediatric monoamine deficiency. A comparison with the most closely related pediatric

monoamine deficiencies is presented in Table 10; as illustrated, a normal CSF

neurotransmitter metabolite profile is the major distinguishing feature of VMAT2

Page 84: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

61

deficiency among these syndromes. As with other PNDs, the motor manifestations

dominate the clinical picture of VMAT2 deficiency, and the autonomic features of the

syndrome may be overlooked. As such, the underlying monoamine pathophysiology may

not be recognized by many physicians, and the most common misdiagnosis would be

cerebral palsy. Furthermore, physicians who suspect biogenic amine involvement may

opt for confirmatory CSF neurotransmitter analysis, which would produce a negative

result, further impeding proper diagnosis. The sole conclusive diagnostic for VMAT2

deficiency at the present time is genetic analysis.

These caveats underscore the importance of including genomic investigations in the

diagnostic protocol for pediatric movement disorder. In addition to being the sole

diagnostic for VMAT2 deficiency, the rate of misdiagnosis (or lack of diagnosis) is high

for the majority of pediatric neurotransmitter diseases, and more broadly, all inborn errors

of metabolism presenting with motor features.

Page 85: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

62

Table 10 Comparison of clinical features of the present disease with those of closely related monoamine deficiency syndromes

Our Family AADC Deficiency Tyrosine Hydroxylase

Deficiency DTDS Age at onset Infancy Infancy Infancy Infancy Muscular hypotonia + + + + Dystonic spasms + + + + Occulogyric crises + + ++ + Parkinsonism + + infantile onset + Delayed motor milestones + + ++ +

Autonomic symptoms + + + + Depression + + – –

CSF neurotransmitters a Normal b

Abnormal (Decreased HVA,

5-HIAA, and increased 3-OMD)

Abnormal (Decreased HVA)

Abnormal (Increased

HVA/5-HIAA)

Treatment dopamine agonist dopamine agonist L-DOPA

dopamine agonist (mild success in some patients)

AADC = aromatic amino acid decarboxylase; CSF = cerebrospinal fluid; DTDS = dopamine transporter deficiency syndrome; 5-HIAA = 5-hydroxyindoleacetic acid; HVA = homovanillic acid; 3-OMD = 3-O-methyldopa; a Complete CSF neurotransmitter metabolite profiles are presented in Table 11. b Based on a single case.

Page 86: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

63

4 Discussion Until the identification of dopamine transporter deficiency syndrome (DTDS) in 2009

(Kurian et al., 2009), the spectrum of PNDs included multiple defects in the biosynthesis

of one or more of the biogenic amines. The gold standard diagnostic test in patients with

suspected diseases of monoamine metabolism is the measurement of monoamine

metabolites in the CSF. Because each specific defect results in a particular metabolite

profile, this single test specifies the disease (Hyland, 2008, Kurian et al., 2011a, Pons,

2009). The particular metabolite profiles of each disease are presented in Table 11.

DTDS was the first PND discovered to result from a defect in biogenic amine transport

(specifically, dopamine uptake at the plasma membrane). This syndrome depicted a

normal serotonergic profile (normal 5-HIAA), but elevated HVA concentrations. This is

also reflected as an increased HVA/5-HIAA ratio, as recently observed in 11 unrelated

children with DTDS (range, 5.0–13.2; normal range, 1.3–4.0) (Kurian et al., 2011b). The

elevation of the primary metabolite of dopamine indicates the production of dopamine in

relevant quantities, as well as an increased rate of dopamine turnover. The latter is

expected to result from increased exposure of dopamine to degradative processes in the

extraneuronal space.

The measurement of CSF neurotransmitters in 1 patient in the present study revealed

metabolite concentrations within the normal ranges (Table 5), including a normal HVA

concentration and HVA/5-HIAA ratio (314 nM and 1.9, respectively). Acknowledging

the limitation in sample size, this result is similarly distinct to that observed for patients

with biosynthetic monoamine deficiencies.

Page 87: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

64

Defects in monoamine transport processes within the central nervous system would be

expected to produce an abnormal distribution of the affected transmitters with respect to

the relative quantities present within the vesicles, the cytosol, and the extraneuronal

space. Vesicular monoamine transport is driven by the pH gradient across the vesicular

membrane; namely, the acidification of the vesicles. In the acidic, reducing environment

of the vesicles, the monoamines are protected from oxidative processes. Additionally,

they are sequestered from degradative enzymes (MAO and COMT). Aberrant

redistribution of the biogenic amines would therefore be expected to increase the rate of

turnover, and subsequently increase the ratio of metabolite to transmitter. Importantly,

Vmat2-/- mice exhibit normal levels of DOPAC, HVA, and 5-HIAA despite dramatically

reduced levels of brain monoamines (Wang et al., 1997, Fon et al., 1997), consistent with

observations in this patient.

The biogenic amines are synthesized in the cytosol (with the exception of norepinephrine,

which is synthesized intravesicularly from dopamine, which is in turn synthesized in the

cytosol). In the case of VMAT2 deficiency, it is possible that the increased cytosolic

concentrations of dopamine and serotonin may provide some feedback inhibition on their

biosynthesis. This presumed difference in distribution relative to DTDS—in which the

neuronal reuptake of monoamines is defective—may underlie the difference in measured

HVA concentrations between DTDS and this case of VMAT2 deficiency (increase in

HVA versus normal HVA, respectively).

It will be necessary to perform metabolite measurements in the CSF of a larger cohort of

patients to conclusively characterize the particular metabolite profile of VMAT2

deficiency. This opportunity may arise as unrelated patients with suspected

Page 88: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

65

neurotransmitter deficiency phenotypes are screened genetically for VMAT2 mutation.

Many of these patients may have previously undergone CSF metabolite profiling, but

remain undiagnosed given normal results. CSF neurotransmitter metabolite analysis has

been instrumental in the discoveries of the monoamine disorders (Hyland, 2008, Kurian

et al., 2011a, Pons, 2009), but not may have captured VMAT2 deficiency because of its

normal CSF profile.

Table 11 Comparison of metabolite profiles in cerebrospinal fluid for pediatric neurotransmitter diseases, including VMAT2 deficiency Affected Enzyme

HVA 5-HIAA MHPG 3-OMD

Disorders of BH4 synthesis (recessive)

decrease decrease decrease normal

GTP cyclohydrolase (dominant)

decrease normal normal normal

Tyrosine hydroxylase

decrease normal decrease normal

AADC decrease decrease decrease increase PNPO decrease decrease decrease increase Dopamine β-hydroxylase

increase normal decrease normal

Dopamine transporter (Kurian et al., 2009, Kurian et al., 2011b)

increase normal NR NR

VMAT2 (single case)

normal normal NR normal

AADC = L-aromatic amino acid decarboxylase; BH4 = tetrahydrobiopterin; GTP = ; NR = not reported; PNPO = ; VMAT2 = vesicular monoamine transporter 2 Note: Data in table derived from Hyland 2008, except for the present case and where otherwise noted.

Page 89: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

66

The specificity of HVA abnormalities in diagnosing primary neurotransmitter disorders

has also recently been called into question by the demonstration of decreased HVA in

15.4% of cases and increased HVA in 4.6% of cases in a cohort of 1388 pediatric patients

with neurological disorder, whereas genetic dopaminergic deficiency was only

discovered in 21 (1.5%) patients by genetic screening (Molero-Luis et al., 2013). The

magnitude of the HVA decrease was greater in this study for patients with genetic

dopaminergic deficiency compared with patients with other neurological disorders, but

the ranges overlapped significantly.

The analysis of monoamines or their metabolites in urine is not reliable in the diagnosis

of monoamine neurotransmitter diseases (Hyland, 2008, Kurian et al., 2011a, Pons,

2009), except in one—AADC deficiency—in which increased 3-O-methyldopa (3-OMD)

with decreased vanillylmandelic acid (VMA) in the proper clinical context is highly

suggestive and generally confirmed by mutation analysis (Brun et al., 2010, Lee et al.,

2012, Pons et al., 2004, Swoboda et al., 1999). In the present condition, urine shows

abnormalities because VMAT2 also functions outside the central nervous system,

including in the peripheral nervous system, adrenal medulla, and platelets (Eiden and

Weihe, 2011). The reason abnormalities are detected in urine and not CSF may pertain to

differences in monoamine and metabolite stabilities, processing, and ranges of normal

values between brain and periphery. In any case, it appears that the pair of metabolically

and clinically similar diseases, the AADC and VMAT2 deficiencies, could be screened

for by urine testing, and then confirmed by corresponding gene sequencing, obviating

lumbar puncturing.

Page 90: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

67

The initial selection of treatment of the affected children on the basis of clinical

phenotype alone (parkinsonism) led to severe, immediate worsening of the movement

disorder. Subsequent identification of the underlying pathophysiology allowed the

rational selection of an appropriate treatment.

It has been suggested that the immediate worsening of the movement disorder observed

in these patients upon treatment with L-DOPA may reflect a severe dyskinesia resulting

from the overactivation of sensitized receptors. In this hypothesis, the supplementation

with exogenous L-DOPA at a high dose would have been sufficient to overcome the

vesicle-loading deficiency, resulting in excessive release of dopamine at the synapse. L-

DOPA responsiveness has been observed in a pair of AADC-deficient siblings

harbouring a G387A mutation (Chang et al., 2004). The mutation affected the binding

site of the AADC protein, causing an approximately 60-fold decrease in affinity for the

L-DOPA substrate. However, saturation of the enzyme by treatment with exogenous L-

DOPA allowed sufficient enzyme activity to improve motor functioning in these patients.

Given the partial functioning of VMAT2-P387L observed in vitro (see Chapter 4,

Section 3.3), it is plausible that the supplementation of cytosolic dopamine would allow

the transport mechanism to come to equilibrium with a sufficient quantal size to allow

neurotransmission. The plausibility of this suggestion is supported by the L-DOPA

responsiveness of the motor phenotype in VMAT2-LO mice (Caudle et al., 2007, Taylor

et al., 2011). Another possibility that may underlie neurotransmission in these patients

under conditions of an extreme excess of cytosolic dopamine include reverse efflux

through the plasma membrane transporter.

Page 91: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

68

Willemsen et al. suggested that, as in patients with TH deficiency (Willemsen et al.,

2010), careful selection of minimal L-DOPA doses may rescue the dopamine-deficient

motor phenotype and avoid extreme dyskinesia. This would have the additional benefit of

restoring norepinephrine neurotransmission and ameliorating the autonomic features of

the syndrome.

As a treatment strategy, smaller doses of L-DOPA may indeed be effective. However,

treatment with L-DOPA for an extended period of time in a setting of disrupted vesicular

storage confers a potential risk of oxidative damage leading to neurodegeneration, and

hypomorphic Vmat2 mice do exhibit age-related neurodegeneration (Caudle et al., 2007,

Taylor et al., 2011).

An additional clinical feature of note is the very high rate of major depression observed

in the patients’ parents. Heterozygous mice possessing a single Vmat2 allele exhibit no

overt motor phenotype, but do express a depressive behavioral phenotype (Fukui et al.,

2007). Major depression is also observed in parents of patients with AADC deficiency

and thought to be caused by clinically significant reductions in serotonin in these

individuals with hemizygous defects in the serotonin pathway (Brun et al., 2010, Lee et

al., 2012, Pons et al., 2004, Swoboda et al., 1999). To what extent mutations in the AADC

and VMAT2 genes contribute to common depression and its heritability remains to be

investigated.

Page 92: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

69

Chapter 3 Identification of Disease-Causing Genetic Variant

1 Introduction In Chapter 2, the clinical features of a new autosomal recessive pediatric neurotransmitter

disease are described, and the difficulties are discussed of determining a precise diagnosis

by clinical characterization alone, comprising clinical history, neurological examination,

biochemical investigations (including specific urine and cerebrospinal fluid

investigations), enzyme analysis, and genetic sequencing of candidate genes.

In this chapter, the discovery of the disease-causing variant is presented—initially by

genome-wide single nucleotide polymorphism (SNP) genotyping, homozygosity analysis,

linkage analysis, and mutation screening. Additionally, the advent of whole-exome

sequencing allowed the exclusion of the possibility mutations in other genes in the locus

by whole-exome sequencing of the proband, as well as demonstrating the possibilities of

genomic diagnosis of rare diseases in a clinically relevant timeframe. This is particularly

relevant in the present case, in which a group of related diseases has an overlapping

phenotypic spectrum that is difficult to discern by clinical investigations alone, but the

determination of the underlying genetic etiology precisely determines the successful

course of treatment.

Page 93: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

70

2 Methods

2.1 Patient Samples The study was approved by the Hospital for Sick Children’s research ethics board and

parents provided informed consent. A total of 5 mL of whole blood was collected in

EDTA tubes. Blood samples were stored at 4°C for up to 1 week.

Genomic DNA was isolated from each sample using a QIAGEN FlexiGene DNA Kit

according to manufacturer’s specifications. Briefly, blood samples were diluted in buffer

and centrifuged to obtain a cell pellet. The cell pellet was resuspended and incubated

with a protease mixture at 65°C for 10 minutes to release genomic DNA into solution.

This genomic DNA was then precipitated by the addition of isopropanol to a final

concentration of 50% and centrifugation. The resulting genomic DNA pellet was then

washed with 70% isopropanol and re-pelleted. After air drying, the genomic DNA was

resuspended in water or Buffer FG3.

2.2 Genotyping of Single Nucleotide Polymorphisms The genotyping of SNPs was performed by the Finnish Genome Center at the University

of Helsinki. A 300K Illumina SNP microarray was used to genotype >300,000 SNPs in

the genomic DNA of eight family members (V:2,3,6,7,8,9;VI:2; see Figure 6).

2.3 Homozygosity Analysis Regions of homozygosity across the genome were identified using PLINK v0.99s

(Purcell et al., 2007). Raw data from the SNP genotyping array was parsed into the input

file format for PLINK using custom PERL scripts. One set of input files was created for

Page 94: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

71

data from each chromosome (i.e., data was divided among 22 sets of input files), and

analysis was performed by chromosome.

The parameters specified for homozygosity analysis were a minimum size of 100 kb for

each homozygous region. Additionally, a SNP density of at least 1 SNP per 50 kb was

mandated.

Regions of homozygosity were identified separately in all eight genotyped individuals,

and overlap among homozygous regions was then identified between family members.

These shared regions of homozygosity were then further classified by the disease

affection status of the individuals.

2.4 Linkage Analysis The complexity of this consanguineous family structure prohibited successful power

simulation. To perform linkage analysis, it was necessary to alter the analyzed family

structure to remove consanguineous connections. A modified family structure lacking

consanguineous marriages was therefore used in this analysis.

A subset of 2500 SNPs was selected for parametric linkage analysis. To ensure a

maximally informative SNP subset, the selected SNPs had a minimal allele frequency of

at least 0.4 in the available HapMap dataset (International HapMap Consortium 2003).

An average spacing of approximately 1.0 Mb was selected to provide sufficient

resolution across the genome. This subset was determined manually, and PERL scripts

were designed to parse the data appropriately.

Page 95: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

72

Parametric linkage analysis was performed using Merlin software (Abecasis et al., 2002).

A fully penetrant autosomal recessive mode of inheritance was assumed. Marker allele

frequencies were derived from marker allele frequencies in the HapMap dataset, and

disease allele frequency was estimated at 1%. Analysis was performed independently for

each chromosome.

2.5 Mutation Screening Sanger sequencing of candidate gene exons was performed to identify sequence variants

within the homozygous region.

Eight genes in the region were selected as candidates for mutation screening on the basis

of neuronal function or localization: GRK5, EMX2, KCNK18, PRLHR, SLC18A2,

GFRA1, VAX1, and NANOS1. Primers were designed to amplify the exons of each gene

and approximately 50 bp of flanking intronic sequence (Table 12 and Table 13). The

reaction was performed using Taq DNA polymerase (Stratagene) according to

manufacturer’s specifications. Cycling conditions incorporated an initial denaturation at

95°C for 5 minutes, followed by 30 cycles of 95°C for 30 s, 55°C for 30 s, and 72°C for

30 s, and a final extension step at 72°C for 10 minutes. Amplification products were

visualized by gel electrophoresis, and purified from bands of the appropriate size using a

Qiagen Gel Purification Kit according to manufacturer’s specifications. Purified PCR

products were sequenced using both forward and reverse primers in separate reactions.

Sequences were compared to reference genome sequence using the BLAT tool in the

UCSC Genome Browser. Putative variants were verified by manual inspection of

electropherograms.

Page 96: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

73

Table 12 Primer sequences for amplification of exons of candidate genes located at 10q26 Gene Exon Primer sequences Size GRFA1 1 TCA CTG GAT GGA GCT GAA CTT 300 bp

CTT CCT TCC ACA TCC ACC AC 2 CGA AGG CTG GTC AAG CAT C 497 bp

AAG TGA GTG GAG GAT GAG ATC AG 3 GGG GAG AAC AAG GAC AAC AA 398 bp

CCA GAA ACA CTG TGC CAT TC VAX1 1 GCG GGG ACA TTC ATT CTT 534 bp

GCC AAC AAC TTT CTC CCA AG 2 GCC CTC CAC ACA GTG TCT TT 463 bp

CCC AAG GGT AGT TCT GTC CA 3/4 TCT GAA GCA AGC GAA AAA CA 496 bp

TCG AGA GCG AAA CAC TCA AG NANOS1 1 AGT GGG CCC GAT AAA AGG 456 bp

GTC GTC GTC CTC GTC GTA GT 2 CGC ACA CCA TCA AGT ACT GC 393 bp

AGC GCC TCT AAG TTG CCA TA EMX2 1-1 ACC CCA AAC AAA CGA GTC C 490 bp

ATG AGC CAG GGG TAG AAG GT 1-2 GGT AGG GGC GTC TAC TCC A 375 bp

CCG CCT AGT TTC CCA ACA G 2 GTG AGC CCT TGG GAG GAC 474 bp

CAG GCG TGG AAC CAG CTA C 3 GGA GGC TGG ACC TTA GGA CT 422 bp

GTG AAC GTG TAT GCG GTT TG KCNK18 1 CAC ACG CAC CAT CCA CTT AG 486 bp

CTT AAA GTG CCC AGG CAT GA 2 CAC CGA GCT TTG GTG TTG AT 392 bp

GGA AGG GAG GGA AGA AGA GA 3-1 GTA TTT TCA AAA ACA ATG TGT AAA

ATG 500 bp

GTT GCA GTG TGT TCT GTT TCT C 3-2 GCA GAT GAA GCT GTC CCT CA 598 bp

TGT CAC CTG AGA GAT AAT GAA ACC PRLHR 1-1 GTT GTT CTG TGG CCG GTT AT 418 bp

AAG TTC GTC ACG TTG TGC AG 1-2 GGC TGA TCG TGC TGC TCT A 427 bp

AGC TCC ACG TGA TAG GTG TG 1-3 CGC TAC GTC GTG CTG GTG 495 bp

GAG CCA GTG GCA GAG CAG 1-4 GGT GAT CGT GGT GGT GTT C 489 bp

CCC AAG CAA AGA GCA AGA CT

Page 97: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

74

GRK5 1 GGA ATA ATG CGG TAG GCA AG 337 bp CTC TCC GAA GTG TCC TGC TC

2 TGG GAG GAA GAG TGT GTG TG 400 bp TGT GTC CAC TGA TTG GCA TT

3 GAG TTT GCC AGT CAC CTT CC 398 bp CCT CAG ACA ATT TTG CAT TCC

4 CAG GAG AAG GGG GTT GGT C 300 bp CAC TTA ACA GCT CCC CAT GT

5 GCT GCC AGA TGT ACC AGC A 350 bp TGG TAT AAC TTG GCT GAA GCT G

6 GCA TCT GCA TGG GTT GAG A 300 bp CGG GAG GTA ACT GAT TTT TAT ATG

7 CTC GAA GGT CCA GTC TCC AG 391 bp CAA GGC CAC TGA CTC TCT CC

8 AGG CAT CAC TGG GTC CTG 391 bp ATT AGA GGA CCA CGC CCT TC

9 ACT GAA AGG GAG GAG CAG GT 498 bp AGG GCT GAT TCC CAG AGC

10 AGG AGG TGG GAA GGA AAC AC 299 bp CAG GTA CCC AGC ACT GAG C

11 GAG TGG GCA TTT TCC TGT GT 399 bp AGT GCA TGA AGA GGC GAG AG

12 GGG CAC AGA GGA GAG TCA TC 484 bp CTC AGA CCC CTG TCC CTT C

13 AGC CCT GAA GCA AGA CCT TT 369 bp CAC ACT CTG CAC CAG CTC AC

14 CAT AGC AGT TCT GGG GGT GT 353 bp CTG CCG CCA AAC TCA TAT TC

15 CCA GTG GCT TTG CTG CTG 299 bp CAA GGG TCT CTA CTG TGG GTC T

16 CTG AGG GGA GAC TGC AAA AG 377 bp GTT CTA CGT CGA CGG GAT G

Page 98: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

75

Table 13 Primer sequences for amplification of SLC18A2 exons SLC18A2 Primer sequences Size 0 GAC TGA CGG AGC CCA CTG 266 bp

ACT GTG CCA CCT CCC AAA CT 1 GCG CTC GAG GCA GGT GAG 438 bp

CCT GGA GGG GAT GTC TTT TT 2 CTC AGC TTC CCA AAG TCC TG 300 bp

CCA CCA TGG ATT TTC CAG AC 3 TTT CAG AAA AGT CCA CCA AAC A 279 bp

CGC CGT CCT TAG GTT GTA TT 4 AAG CAC AGG GTG GCT AAC AT 298 bp

CAA CCA CCA CCA ATA CCT GA 5 AAG GCA CCC ACT TTC CTC TT 365 bp

GGA CCT CTG TGT CAG TGC AA 6-8 CCC AAA GCC TTA TTG GAA CA 595 bp

GCA GCA AAA CGA AAA TCA GC 9 AGG AGG CAG AAG CCA CTA CA 182 bp

GGG GAC AAA TGC TCT TGA AT 10/11 GGG GCT TCG TTT TAT CTG CT 497 bp

AAA TGT TTC TTG GTG ATT CTT GC 12 CCA CCC TTC TTC CTC CTG TT 324 bp

CCT GCA GCC TCC TTC TAA GAT 13/14 CTG GCA GGG TGG TGA GTT TA 599 bp

TTC TCA CCA TTT ATG TTT GAA GG 14 TTG CAT CTT TCA GTC TAC AAG ACA 269 bp

CCA TGA GGA AGC TAT CAG GAA 15 CGA TTG CTC CAA ATG ACT GG 398 bp

CAA TCG ACC ATA ACC ATG GAA

2.6 Whole-Exome Sequencing Whole-exome sequencing was performed to exclude the possibility of additional

mutations present in genes within the homozygous disease-associated region at 10q26.

Exon sequences were first enriched using an Agilent SureSelect V4 50-Mb capture kit

that uses a biotinylated library of RNA baits and streptavidin-coated magnetic beads to

Page 99: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

76

specifically capture exon sequences. The exon-enriched samples were sequenced using

the Illumina Hiseq 2000 next-generation sequencing system. These protocols were

performed by The Centre for Advanced Genomics (TCAG). The resulting sequence data

was filtered for coding variants.

2.7 TaqMan SNP Genotyping Assay Genotyping was performed to confirm the absence of the identified mutation in controls.

Differentially fluorescently labelled TaqMan probes were synthesized to represent each

of the SLC18A2 c.1160C and c.1160T alleles. Within an amplification reaction,

fluorescence is released from probes that hybridize to the template DNA through the

5' nuclease activity of DNA polymerase. The ratio of the detected fluorescent labels

thereby determines the genotype of the individual at that locus. The samples were

processed in 96-well format. This assay was performed by TCAG.

3 Results

3.1 Homozygosity Analysis Homozygosity mapping of the microarray results identified a single homozygous 3.0-Mb

interval in 10q25.3–26.11 that was uniquely shared by the affected family members

(chr10:117,937,133–120,972,346), and not homozygous in the unaffected family

members who were genotyped (Figure 7).

Because of the consanguinity of the family structure, a large number of homozygous

regions were expected in individual genomes. Indeed, an average of 30 homozygous

regions of >500 kb were discovered in each genome. These regions of homozygosity in

individual genomes ranged from 780 kb to 68 Mb. Several of these regions were

Page 100: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

77

inherited in their entirety by multiple siblings, and overlapped with homozygous regions

inherited by cousins.

A limitation of this homozygosity analysis was related to the density of SNPs. Whereas

the region at 10q25.3–26.11 was the only homozygous region shared by the affected

individuals, the parameters for the analysis specified a minimum size of 100 kb, and it is

possible that smaller homozygous regions would not have been identified. The average

size of a gene in the human genome is 27 kb (Lander et al., 2001), smaller than the

minimum identifiable region in this analysis. More dense SNP coverage would allow

higher confidence assessment of smaller homozygous regions.

Page 101: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

78

Figure 7 Single nucleotide polymorphism alleles for all genotyped family members in the homozygous region uniquely shared by affected family members. Black, affected; white, unaffected; square, male; circle, female. From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 102: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

79

3.2 Linkage Analysis Parametric linkage analysis produced a significant logarithm of odds (LOD) score of 4.1

within the region of 10q25–26 (Figure 8). One additional significant LOD score of 3.1 on

chromosome 3 did not correspond to a region of homozygosity shared by all affected

individuals. A small positive LOD score was also obtained on chromosome 16, also not

representing a region of homozygosity shared by all affected individuals (Figure 9).

Methodological limitations, however, were several. These included the difficulty in

calculating a logarithm of odds (LOD) score for each putative locus in a highly

consanguineous family. For the purposes of calculation, the consanguineous connections

in the pedigree had to be removed, thereby not accounting for all of the available

inheritance information and reducing the power of the analysis.

Page 103: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

80

Figure 8 Multipoint LOD scores estimated across chromosome 10

Page 104: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

81

Figure 9 Multipoint LOD scores estimated across chromosomes 3 and 16

Page 105: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

82

3.3 Characterization of Genes in the Locus The homozygous region at 10q25.3–26.11 contained 26 known protein-coding genes

(Figure 10), of which eight genes had known neuronal localization or function: EMX2,

GFRA1, GRK5, KCNK18, NANOS1, PRLHR, SLC18A2, and VAX1. The known function

of these eight genes is summarized in Table 14. The exons and exon–intron boundaries of

each of these genes were sequenced in one affected patient, and all identified sequence

variants are listed in Table 15. Additionally, KIAA1598 was not initially identified as a

candidate (and therefore not initially sequenced), but has since been annotated to have

neuronal localization and is included in Table 14. Sequencing revealed two

nonsynonymous variants among the eight sequenced genes: c.847A→G in PRLHR and

c.1160C→T in SLC18A2. The variant in PRLHR was previously identified in the dbSNP

database with a global minor allele frequency of 0.17; this variant is therefore unlikely to

be causative of this disease. The c.1160C→T variant in SLC18A2, however, appears to

be novel and predicts a substitution of proline with leucine at position 387 (p.P387L)

in the VMAT2 protein.

Page 106: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

83

Table 14 Genes of known neuronal function or localization present in the disease-associated bomozygous region Gene Functional Information (Meyer et al., 2013) EMX2 Homeobox-containing transcription factor homologous to ‘empty spiracles’

gene in Drosophila melanogaster. Expressed in the dorsal telencephalon during development and is proposed to pattern the neocortex into defined functional areas.

GFRA1 A receptor for glial cell line–derived neurotrophic factor (GDNF) and neurturin (NTN). Candidate gene for Hirschsprung disease.

GRK5 G protein–coupled receptor kinase 5. Accumulates in Lewy bodies, a histological hallmark of Parkinson’s disease (Arawaka et al., 2006).

KCNK18 Outward-rectifying potassium channel associated with migraine with aura. KIAA1598 Involved in neuronal polarization NANOS1 Zinc-finger RNA-binding protein expressed in the developing nervous

system and adult brain. PRLHR Prolactin-releasing hormone receptor expressed in the brain and pituitary

gland. Transcription of this gene was shown to be regulated by dopamine D2 receptor agonist bromocriptine (Ozawa et al., 2002).

SLC18A2 Vesicular monoamine transporter 2 responsible for packing monoamines into synaptic vesicles.

VAX1 Homeodomain-containing transcription factor that may play a role in development of the anterior ventral forebrain and visual system.

Page 107: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

84

Figure 10 Genes present within the homozygous region shared by all affected individuals in the family. C10orf82, chromosome 10 open reading frame 82; CACUL 1, CDK2-assocaited cullin domain 1; CASC2, cancer susceptibility candidate 2 (non-protein coding); CCDC172, coiled-coil domain containing 172; EIF3A, eukaryotic translation factor 3 subunit A; EMX2, empty spiracles homeobox 2; ENO4, enolase family member 4; FAM204A, family with sequence similarity 204 member A; GFRA1, GDNF family receptor alpha 1; GRK5, G protein–coupled kinase 5; HSPA12A, heat shock 70 kDa protein 12A; KCNK18, potassium channel subfamily K member 18; KIAA1598, Shootin1; MIR, microRNA; PDZD8, PDZ domain containing 8; PNLIP, pancreatic lipase; PNLIPRP, pancreatic lipase–related protein; PRDX3, peroxiredoxin 3; PRLHR, prolactin-releasing hormone receptor; RAB11FIP1, RAB11 family interacting protein 2 (class I); SFXN4, sideroflexin 4; SLC18A2, solute carrier family 18 member 2; SNORA19, small nucleolar RNA H/ACA box 19; VAX1, ventral anterior homeobox 1. Image from UCSC Genome Browser [http://genome.ucsc.edu]

chr10 (q25.3-q26.11) 10p14 10p13 p12.1 10q21.1 21.2 10q21.3 q22.1 q22.3 10q23.1 10q25.1 q25.3 26.13 q26.3

Scalechr10:

1 Mb hg19118,500,000 119,000,000 119,500,000 120,000,000 120,500,000 121,000,000

UCSC Genes (RefSeq, GenBank, CCDS, Rfam, tRNAs & Comparative Genomics)GFRA1GFRA1GFRA1

CCDC172PNLIPRP3

JA611286PNLIP

PNLIPRP1PNLIPRP1

PNLIPRP1PNLIPRP1

PNLIPRP2PNLIPRP2PNLIPRP2

C10orf82

C10orf82

HSPA12AHSPA12A

DQ596646ENO4

ENO4KIAA1598

KIAA1598KIAA1598KIAA1598KIAA1598AK092331

KIAA1598

KIAA1598

VAX1VAX1

BC039338MIR3663

KCNK18SLC18A2SLC18A2

PDZD8EMX2OSEMX2OS

EMX2

EMX2

RAB11FIP2RAB11FIP2

CASC2CASC2CASC2CASC2CASC2

FAM204AFAM204A

PRLHRPRLHR

Mir_584CACUL1CACUL1

NANOS1EIF3AEIF3AEIF3A

SNORA19SNORA19

FAM45BFAM45BFAM45BFAM45BFAM45B

BC042590SFXN4SFXN4SFXN4

PRDX3

PRDX3

GRK5GRK5

GRK5

Page 108: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

85

Table 15 Sequence variants identified among eight candidate genes in the disease-associated locus by direct sequencing of exons Gene Segment Sequence Variation Present in

dbSNP EMX2 Exon 1 none –

Exon 2 none – Exon 3 none –

GRK5 Exon 1 none – Exon 2 none – Exon 3 c.149-6G→A rs2275036

KCNK18 Exon 1 none – Exon 2 none – Exon 3 none –

PRLHR Exon 1 c.847A→G (I283V) rs1613488 SLC18A2 Exon 1 none –

Exon 2 none – Exon 3 none – Exon 4 none – Exon 5 none – Exon 6 c.700+15C→T rs2072362 Exon 7 c.790+54C→T rs363420 Exon 8 c.791-42C→A rs363343 Exon 9 none – Exon 10 none – Exon 11 none – Exon 12 c.1122+20T→C rs363227 Exon 13 c.1160C→T (P387L) – Exon 14 c.1306+43A→G rs363272 Exon 15 none – Exon 16 c.1441-48T→C rs363279

VAX1 Exon 1 none – Exon 2 none – Exon 3 none – Exon 4 none –

GRFA1 Exon 1 none – Exon 2 none – Exon 3 none –

Page 109: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

86

3.4 Whole-Exome Sequencing of the Proband We also performed whole-exome sequencing in the proband, and the data were filtered to

reveal 9,821 coding variants in the genome. Of these, 3,900 coding variants were present

in the homozygous state. This large number reflects the extent of homozygosity in the

genome of this individual resulting from her consanguineous background. A total of 70

homozygous, non-coding variants were not previously represented in the NCBI dbSNP

database. As a member of an isolated, Bedouin population, there may be poor

representation of this patient’s particular ethnic background among the ethnically diverse

samples genotyped to date. This presents a current limitation of the ability to exclude

variants on the basis of their presence in the general population. In the absence of a clear

functional candidate, it would be warranted to perform whole-exome sequencing on

additional affected and unaffected family members to narrow the list of shared

homozygous sequence variants, and identify those that are present homozygously more

broadly within the pedigree.

However, this analysis independently identified the homozygous variant in SLC18A2

(VMAT2-P387L) and the aforementioned c.847A→G variant in PRLHR, and revealed no

other novel non-synonymous variant in the linked region of shared homozygosity at

10q25.3–26.11

3.5 Identification of VMAT2 Variant as a Causative Candidate

The identified novel variant in SLC18A2 (c.1160C→T) was present in exon 13 (Table 15)

and predicts a substitution of proline with leucine at position 387 (p.P387L) in the

VMAT2 protein. To confirm that the variant segregated with the disease, the region was

Page 110: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

87

sequenced in all available affected and unaffected family members, including siblings

and parents. The mutation was present in homozygous form in all affected family

members and was not present, or was present in heterozygous form, in unaffected

siblings (Figure 11). All genotyped parents carried the variant in heterozygous form.

The SLC18A2 c.1160C→T variant has not previously been identified in the literature, and

is not listed in dbSNP or the 1000 Genomes Database.

Page 111: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

88

Figure 11 Electropherogram depicting sequence variation in unaffected and affected family members. Top panel, unaffected sibling with wild-type genomic sequence; middle panel, asymptomatic parent (IV:3) possesses the SLC18A2 c.1160C→T variant in heterozygous form;; lower panel, proband (V:6) is

homozygous for SLC18A2 c.1160C→T. From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 112: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

89

3.6 Controls To confirm that the SLC18A2 c.1160C→T variant is not present in unaffected members

of the extended kindred, TaqMan genotyping was performed on DNA samples isolated

from direct relatives of the patients. The SLC18A2 c.1160C→T variant is not present in

homozygous form in 78 unaffected members of the extended family, 26 of whom do

carry it in the heterozygous state.

In addition, as one of the most studied candidate genes for involvement in Parkinson’s

disease, SLC18A2 was previously screened in 704 healthy individuals of diverse ethnic

backgrounds and 452 Parkinson’s disease patients (Burman et al., 2004, Glatt et al., 2001,

Iwasa et al., 2001), none of whom had the c.1160C→T change. The 1000 Genomes

Database (Abecasis et al., 2010) identifies 44 missense variants in SLC18A2 that are not

associated with an overt phenotype, but none within the same codon.

4 Discussion Collectively, the results presented in this chapter identify and confirm SLC18A2

c.1160C→T as the disease-causing mutation in this family. Consistent with the observed

autosomal recessive mode of inheritance, the mutation is present in homozygous form in

all affected individuals, and those who possess the mutation in heterozygous form do not

exhibit the movement disorder phenotype. All affected individuals are direct descendants

of individuals I:1 and I:2 (see Figure 6). The expression of the movement disorder

phenotype begins in the fifth generation of this consanguineous kindred, consistent with a

founder mutation likely arising in one of these two individuals.

Page 113: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

90

No disease-associated mutations have been identified in SLC18A2 (VMAT2) to date.

There is no evidence of the SLC18A2 c.1160C→T variant in genomic databases of

healthy individuals. Additionally, the 1000 Genomes Database identifies no variants

predicted to cause a premature stop codon in VMAT2. Of 44 identified VMAT2

missense variants in this database, 12 are predicted to be deleterious (“probably

damaging”) by PolyPhen (a further 10 are “possibly damaging”). By the SIFT algorithm,

20 were considered deleterious. In the Exome Variant Server of the NHLBI GO Exome

Sequencing Project, a single frameshift deletion allele was identified, but this deletion

involved the coding sequence for the final amino acid of the protein and may therefore be

benign. Importantly, none of the 25 missense variants identified in this database (a subset

of the 44 missense variants catalogued in the 1000 Genomes Database) were present in

homozygous form.

In addition, the screening of SLC18A2 in 704 healthy individuals of diverse ethnic

backgrounds and 452 Parkinson’s disease patients identified only 5 missense variants

(only two of which are not missense variants represented in the 1000 Genomes Database)

(Burman et al., 2004, Glatt et al., 2001, Iwasa et al., 2001). Four of these missense

variants were functionally screened, and determined to have only modest effects on

protein function (Burman et al., 2004).

It has been suggested that VMAT2 exhibits a low rate of variation relative to genes of

related function, such as the dopamine and serotonin plasma membrane transporters

(DAT and SERT, respectively) (Glatt et al., 2001). This implies functional significance

of the protein, and therefore suggests that a dramatically deleterious mutation in VMAT2

would have correspondingly dramatic phenotypic effects. Indeed, the 1000 Genomes

Page 114: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

91

Database reports 151 missense mutations in DAT and 317 missense mutations in SERT,

both transmembrane proteins of comparable length to VMAT2 and related function in the

central nervous system. Importantly, 726 missense mutations are reported in the 1000

Genomes Database for the non-neuronal VMAT isoform, VMAT1. In comparison with

the respective 44 missense mutations currently identified in VMAT2 in the same sample

set, these figures therefore illustrate that a functionally deleterious mutation in VMAT2

would be predicted to have significant phenotypic effects—particularly if identified in

homozygous form, as observed here for the VMAT2 p.P387L variant.

Proline residues located adjacent to transmembrane domains have major structural

implications, and are overrepresented among disease-causing substitutions in

transmembrane domains (Partridge et al., 2004). The P387L substitution is immediately

adjacent to a transmembrane segment (Figure 12), and the constraints imposed by the

proline residue on the flexibility of the peptide backbone are likely important for

insertion of the transmembrane domain into the membrane to form the proper tertiary

configuration.

Sequence alignment shows that the P387 residue is highly conserved through evolution,

and is therefore of likely structural or functional significance. It is also conserved in the

VMAT1 paralog, as well as in C. elegans CAT-1—the single vesicular monoamine

transporter in nematode (Figure 13) (Duerr et al., 1999). Interestingly, the residue is not

conserved in the vesicular acetylcholine transporter (VAChT), which maintains 39%

identity to VMAT2, implying that P387 may have a specific implication in monoamine

transport.

Page 115: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

92

Furthermore, the P387L substitution is predicted to be “probably damaging” by the

PolyPhen-2 algorithm (Adzhubei et al., 2010) with a score of 1.000. This algorithm

considers eight sequence-based and three structure-based predictive features, and was

developed using large datasets of known damaging (Mendelian disease-causing or protein

destabilizing) and nondamaging alleles.

The evidence presented in this discussion—the segregation of the variant consistent with

an autosomal recessive inheritance pattern, the absence of the variant in healthy

individuals of ethnically diverse populations, evolutionary conservation of the residue, its

potential structural relevance, and the low rate of genetic variation in SLC18A2—together

predicts the likely functional relevance of the p.P387L substitution in VMAT2 (SLC18A2

c.1160C→T).

Page 116: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

93

Figure 12 Predicted structure of VMAT2 comprises 12 transmembrane domains, a large lumenal loop including four proposed glycosylation sites, and both N-terminal and C-terminal cytoplasmic regions. Proline residue 387 is located immediately adjacent to the insertion of transmembrane domain X. From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 117: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

94

Figure 13 Multiple sequence alignment of VMAT2 in the region of the p.P387L variant. TM9 and TM10 represent sequences associated with transmembrane domains 9 and 10, respectively. Residues that differ from human VMAT2 sequence are indicated in gray. Amino acid position 387 is indicated by an asterisk. Homo, homo sapiens (human); Patient, proband; Pan, Pan troglodytes (chimpanzee); Macaca, Macaca mulatta (Rhesus macaque); Mus, Mus musculus (mouse); Rattus, Rattus norvegicus (rat); Canis, Canis familiaris (dog); Bos, Bos taurus (cow); Monodelphis, Monodelphis domestica (opossum); Gallus, Gallus gallus (chicken); Tetraodon, Tetraodon nigroviridis (pufferfish); Danio, Danio rario (zebrafish); Drosophila, Drosophila melanogaster (fruit fly); C. elegans, Caenorhabditis elegans (nematode); hVMAT1, human VMAT1 isoform. From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 118: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

95

Chapter 4 Functional Characterization of the Disease-Causing Variant

1 Introduction The identification of the putative disease-causing mutation is described in Chapter 3. As

presented in that chapter, the SLC18A2 c.1160C→T variant segregates in a manner

consistent with the observed autosomal recessive inheritance of the disease. The

corresponding VMAT2 p.P387L substitution is predicted to have a structural role within

the protein, and the proline residue at this position is conserved in all vesicular

monoamine transporters isoforms in all species.

The phenotype expressed by the patients harboring SLC18A2 c.1160C→T is similar to

that expressed by patients with biosynthetic deficiencies of the biogenic amine

neurotransmitters, predicting a loss of VMAT2 function that would interrupt vesicular

storage in the brain and expose biogenic amines to degradation. The patient CSF

neurotransmitter metabolite profile presented in Chapter 2 is consistent with this

hypothesis, and the hypothesis is tested in the present chapter.

2 Methods

2.1 Construct Design and Site-Directed Mutagenesis Human VMAT2 cDNA sequence was acquired from the Mammalian Gene Collection

and subcloned into pcDNA3.1A (Invitrogen). The primers used for amplification were

designed to incorporate restriction sites flanking the open reading frame (Table 16). The

P387L (c.1160C→T) mutation was introduced by site-directed mutagenesis using the

QuikChange Site-Directed Mutagenesis kit (Stratagene). The oligonucleotide sequences

Page 119: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

96

for site-directed mutagenesis are presented in Table 17. The final sequence of the vector

was confirmed by sequencing using standard M13 primers that are complementary to

vector sequences.

Table 16 Primers to amplify SLC18A2 cDNA sequence for subcloning into pcDNA3.1 Primer Sequence VMAT2-KpnI-F CAT GGT ACC AGT AGT ATG GCC CTG AGC GAG CTG G VMAT2-PmeI-R CAT GTT TAA ACA CTA CTT CAG TCA CTT TCA GAT TCT

TCA TC

Table 17 Oligonucleotides to introduce cytosine to thymine substitution at position 1160 of SLC18A2 using site-directed mutagenesis Primer Sequence VMAT2-P387L Quikchange-F

AAA CAT TTA TGG ACT CAT AGC TCT GAA CTT TGG AGT TGG TTT TGC

VMAT2-P387L Quikchange-R

GCA AAA CCA ACT CCA AAG TTC AGA GCT ATG AGT CCA TAA ATG TTT

2.2 Cell Culture and Transfection The wild type and mutant constructs were expressed in Cos7 cells, as there is no

endogenous VMAT1 or VMAT2 expressed in these cells. Cos7 cells were maintained in

Dulbecco’s modified Eagle medium (DMEM) with 10% fetal bovine serum (FBS) at

37°C and 5% CO2. Cells were passaged by trypsinization, except immediately prior to

transfection for the serotonin uptake assay; cells prior to the uptake assay were removed

from the surface of the plate by scraping. In both cases, cells were diluted 1:10 for

seeding onto fresh plates.

Page 120: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

97

Transient transfection of Cos7 cells was performed at 80% confluency using Fugene HD

transfection reagent (Roche) according to manufacturer specifications. After 48 hours,

cells were washed once in 1× phosphate-buffered saline (PBS), scraped, and pelleted for

further experiments. Cells from three 10-cm plates that had been transfected in parallel

were combined into a single pellet.

2.3 Serotonin Uptake Assay Vesicular serotonin uptake was assayed in a heterologous cell system. VMAT2 transport

activity was measured by incubating membrane preparations with tritiated serotonin,

followed by rapid washing and filtration to retain vesicles with trapped substrate.

Transiently transfected Cos7 cell pellets were immediately resuspended in 320 mM

sucrose-HEPES buffer (pH 7.4) and sonicated by 20 × 1 s pulses. Lysates were

centrifuged at 4,000g for 5 min, and supernatants were stored at –80°C until use.

Reaction buffer contained 150 mM choline gluconate, 10 mM HEPES-Tris (pH 7.4), 2

mM Mg-ATP, and 90 nM 3H-serotonin (New England Biolabs). Where indicated,

reaction buffer also contained 10 µM reserpine. To each tube was added 10 µL of

microsomal lysate, followed by incubation at 30°C for the specified time. Reactions were

stopped by rapid filtration, and retained serotonin was measured by scintillation counting.

Retained serotonin was also measured from samples incubated on ice and filtered

immediately upon addition of the lysate, as well as from reaction buffer alone.

2.4 Western Blotting Western blots were performed by equal loading of protein lysates onto 10% SDS-PAGE

gels. Proteins were transferred to PVDF membrane in a Bio-Rad semidry transfer cell at

Page 121: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

98

1 mA/cm2 for 45 min. Transfer buffer was 200 mM glycine, 25 mM Tris-HCl, pH 7.5,

and 20% methanol. The membrane was blocked for 1 h in 50 mM Tris-HCl, pH 7.5, 150

mM NaCl, and 0.05% Tween 20 (TBS-T) with 5% skim milk. Blots were incubated in

blocking solution with primary antibody for 1 h at 4°C, washed three times with TBS-T,

and then incubated with secondary antibody in blocking solution for 1 h at 4°C, followed

by three TBS-T washing steps and detection by chemiluminescence according to

manufacturer’s recommendation.

The specificity of a commercial antibody against a 20–amino acid C-terminal peptide of

VMAT2 (Abcam) was verified by western blot, showing no non-specific signal in

untransfected or vector-transfected cultures (see inset to Figure 19). Secondary antibody

was horseradish peroxidase–conjugated anti-rabbit antibody.

2.5 Sucrose Gradient Centrifugation To compare the subcellular localization of the wild-type and p.P387L VMAT2 proteins

in Cos7 cells used for serotonin uptake assays, cell lysates were fractionated by sucrose

gradient centrifugation, and the presence of the protein in each fraction was visualized by

western blot.

Transiently transfected Cos7 cell pellets were immediately resuspended in 320

mM sucrose-HEPES buffer (pH 7.4) and sonicated by 20 × 1 s pulses. Lysates were

centrifuged at 4000 g for 5 min, and protein concentrations were measured by Bradford

assay to ensure equal protein loading on gradients tested in parallel. Supernatants were

immediately applied to a linear sucrose gradient of 20%–55% sucrose (approximately

0.6–1.6 M) in HEPES (pH 7.4) and centrifuged at 150 000 g for 16 h at 4°C.

Page 122: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

99

Fractionation was performed using a Brandel Density Gradient Fractionation

system. Aliquots of equal volume were separated by SDS-PAGE on 4%–12% Bis-Tris

gels (Invitrogen). Blots were prepared as described in Section 2.4.

Linear sucrose gradients were generated by tilted tube rotation on a BioComp

Gradient Master. The linearity of select gradients was confirmed by the addition of ATP

in the heavy phase (50% sucrose) prior to gradient generation, and measurement of

absorbance at 260 nm during fractionation (Figure 14).

Figure 14 Visualization of ATP by absorbance at 260 nm to illustrate sucrose gradient linearity

The consistency of gradient fractionation between samples was demonstrated by

the measurement of absorbance at 280 nm during the fractionation process. An initial

peak in the first fraction, likely representing free cytosolic contents, is followed by a

trimodal peak present approximately between fractions 7 and 12. All fractionations

exhibited the same pattern of absorbance, and parallel fractionations produced

absorbances of equal intensity (Figure 15). This demonstrates the reproducibility of the

Page 123: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

100

gradient centrifugation process. Some minor shift in the absorbance peaks relative to

particular numbered fractions resulted from variation in the fractionation process.

Figure 15 Consistent fractionation demonstrated by absorbance at 280 nm of parallel sucrose gradient fractionations

Protein markers of cellular compartments were visualized by western blot. GAPDH was

used as a cytosolic marker, and was present only fractions 2 and 3, verifying a lack of

Page 124: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

101

cytosolic contamination in later fractions. Transferrin receptor (TfR) was used as a

marker for recycling endosomes, and was reproducibly visualized at highest

concentration in fractions 5–9, with faint signal detectable in later fractions. Calnexin

was used as a marker for endoplasmic reticulum, and was present primarily in fractions

9–13, but with signal detectable in all fractions. Overlapping signal demonstrates

incomplete separation of the microsomal components of the cell, but the relative order of

components is as expected.

Figure 16 Markers of cellular compartments in sucrose gradient fractions

3 Results

3.1 Steady-State Protein Levels of Transfected Protein Unaffected by P387L Variant in Heterologous System

To determine the effect of the p.P387L mutation on VMAT2 transport activity, we

transiently expressed wild-type and mutant human VMAT2 in Cos7 cells.

Immunoblotting of membrane preparations confirmed equivalent protein levels of

VMAT2 in parallel transfections, suggesting no major defect in protein processing

(Figure 17).

Page 125: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

102

Figure 17 Parallel transient transfections of wild-type and p.P387L VMAT2 in Cos7 cells, and vector-transfected control. GAPDH, protein loading control.

Page 126: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

103

The wild-type and p.P387L VMAT2 constructs exhibit the same intensity and banding

pattern on western blot, indicative of multiple glycosylation forms. This banding pattern

is better illustrated in Figure 18. In transiently transfected Cos7 cells, bands of

approximately 57 and 45 kDa are most prominent, with a lower molecular weight band of

lesser intensity at approximately 35 kDa. These bands are consistent with those observed

in previous studies by Tong et al., who examined the expression of VMAT2 in autopsied

human brain tissue, and reported four protein bands of approximately 75, 55, 45, and 35

kDa (Tong et al., 2011). A pair of higher molecular weight bands are also visible, but

have a molecular weight of approximately 90 kDa in this heterologous cell system. The

bands less distinct than is observed in brain tissue, and multiple bands of intermediate

size are also visible and appear as a smear—this was interpreted as an artifact of

overexpression in the heterologous system. Because glycosylation does not substantially

affect VMAT transport activity (Yelin et al., 1998), no further investigation of the

identity of the protein bands was performed.

Page 127: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

104

Figure 18 Comparison of banding pattern observed for wild-type and p.P387L VMAT2 expressed transiently in Cos7 cells. unt, untransfected; WT, wild-type.

3.2 Highly Reduced Transport Activity of VMAT2 p.P387L in Heterologous System

The p.P387L variant of VMAT2 was assayed for its ability to transport tritiated serotonin

in a crude vesicle lysate isolated from Cos7 cells transfected in parallel with either the

wild-type or p.P387L VMAT2 construct, or vector alone. After incubation with tritiated

serotonin, samples were rapidly filtered to retain microsomes, and the trapped substrate

retained on the filter was measured by scintillation counting.

Page 128: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

105

The p.P387L variant of VMAT2 showed dramatically decreased serotonin uptake activity

relative to wild-type VMAT2 (Figure 19). The sensitivity of the assay did not permit

quantification of the uptake kinetics of p.P387L VMAT2, or quantification of the relative

decrease in activity compared with the wild-type protein. In parallel time courses, the

activity of the p.P387L mutant of VMAT2 was indistinguishable from background, as

measured in lysates isolated from vector-transfected Cos7 cultures.

Page 129: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

106

Figure 19 Vesicular uptake of tritiated serotonin by the wild-type and p.P387L human VMAT2 transiently expressed in a heterologous Cos7 cell system. Inset: Expression of VMAT2 in lysates used for uptake assay. 5-HT, serotonin; wt, wild-type. From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 130: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

107

3.3 Measurable Residual Transport Activity of VMAT2 p.P387L Protein

Because a complete lack of VMAT2 function would be expected to be lethal on the basis

of mouse models (Wang et al., 1997), the activity of the p.P387L mutant of VMAT2 was

more closely investigated. The specific VMAT inhibitor reserpine was used to better

distinguish VMAT2 transport activity from the nonspecific uptake or retention of

substrate on the filters observed in lysates from vector-transfected cultures.

Microsomal uptake of tritiated serotonin was assayed at the 10-minute timepoint—the

time at which the reaction begins to saturate (Figure 19)—with and without the addition

of 10µM reserpine. The p.P387L mutant VMAT2 exhibited weakly measurable uptake

that was approximately three times the background level measured with the addition of

reserpine (Figure 20). Therefore, the VMAT2 p.P387L mutation reflects a severe, but

not complete, loss of protein function.

Page 131: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

108

Figure 20 Vesicular uptake of tritiated serotonin by wild-type and p.P387L human VMAT2 transiently expressed in a heterologous Cos7 cell system after 10 minutes with and without the addition of the specific VMAT inhibitor reserpine (10 µM). From New England Journal of Medicine, Rilstone JJ, Alkhater RA, Minassian BA, Brain Dopamine–Serotonin Vesicular Transport Disease and Its Treatment, 368, 543–50. Copyright © 2013 Massachusetts Medical Society. Reprinted with permission.

Page 132: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

109

3.4 Subcellular Localization of Protein in Heterologous System

Sucrose gradient centrifugation was performed to compare the subcellular localization of

wild-type VMAT2 protein with that of VMAT2-P387L in the transiently transfected

Cos7 lysates that were used for the serotonin uptake assay. Preliminary data reveals

signal in the majority of gradient fractions for both proteins, with the highest

concentrations of protein in fractions 7–12 (Figure 21). The broad distribution of

VMAT2 protein across gradient fractions is indicative of either broad localization in

multiple microsomal components of the cell, or it is reflective of ER contamination in

multiple fractions. In either case, the lack of confinement of VMAT2 to its expected

compartment—recycling endosomes—is likely a consequence of overexpression in the

transient transfection setting.

Given the expected elution of fractions containing the highest proportion of ER in

fractions 9–13 relative to the earlier elution of recycling endosomes in fractions 4–9

(Figure 16), extensive misfolding of the mutant protein relative to wild-type VMAT2

would be expected to produce a rightward shift in the distribution of VMAT2 across

fractions. No such pattern is observed. This result is therefore inconclusive, but does

suggest no significant difference in localization between the wild-type and mutant

VMAT2 proteins, and likely a lack of specificity of protein localization as a result of

protein overexpression. This preliminary result therefore suggests that the difference in

serotonin uptake between wild-type VMAT2 and VMAT2-p.P387L is not reflective of a

drastic difference in the subcellular distribution of the proteins, but rather a true

difference in transport function. Verification of this observation should be performed

Page 133: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

110

using an alternative technique such as immunofluorescence. In addition, data regarding

the subcellular localization of VMAT2-P387L in the physiological setting should be

pursued using cell lines with stable expression of the constructs to avoid artifacts of

overexpression.

Figure 21 Sucrose gradient centrifugation of transiently transfected Cos7 lysates

Page 134: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

111

4 Discussion Direct characterization of the mutant VMAT2 protein in a heterologous cell system in

this study revealed a severe detriment of vesicular transport function. This could be the

result of poor incorporation of the transporter into vesicle membranes or lost transport

activity; data regarding the relative subcellular distribution of wild-type and p.P387L

VMAT2 was inconclusive, but suggestively not the major factor underlying loss of

function.

The p.P387L substitution is immediately adjacent to transmembrane domain 10 (see

Figure 12). Prolines can have significant structural and functional significance in

transmembrane domains because the constraints imposed by their ring structure induce

kinks in α-helices. Prolines are also preferentially located at the central and terminal

regions of transmembrane segments (Cordes et al., 2002). Though prolines constrain the

angle of the peptide backbone, they also induce flexibility in the region through the

disruption of stabilizing hydrogen bonds in upstream amino acids, and have therefore

been implicated as hinge points with possible functional relevance (Cordes et al., 2002).

Prolines are prevalent in the transmembrane domains of ion channels and transport

proteins, but not the transmembrane segments of proteins without transport function,

inspiring a hypothesis that cis-trans isomerization may underlie the conformational

change inherent in the transport process. Additionally, the disruption of hydrogen bonds

in the peptide backbone that is imposed by proline exposes a carbonyl group, providing a

site for cationic ligand binding or proton translocation (Brandl and Deber, 1986, Williams

Page 135: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

112

and Deber, 1991). Finally, Partridge et al. reported an overrepresentation of proline

substitutions among disease-causing mutations located in transmembrane domains,

indicating a high phenotypic propensity for amino acid substitutions of this nature

(Partridge et al., 2004).

With respect to VMAT2 structure specifically, a hydrogen bond between D400 in

transmembrane domain 10 and Y342 in transmembrane domain 8 was predicted in a

homology model of the rat protein, rVMAT2, and the D400 residue was verified

experimentally to be critical to transport activity (Yaffe et al., 2013). The relative

configuration of transmembrane domain 10 and transmembrane domain 8 is therefore

functionally critical, underscoring the likely importance of proline constraints on the

peptide backbone near the insertion of transmembrane domain 10. This, however, does

not exclude the possibility of a role for the P387 residue in conformational change, ligand

binding of the cationic monoamine substrates, or proton translocation.

Direct biochemical characterization in a heterologous system confirms the functional

significance of the P387L substitution. This functional biochemical evidence—in

combination with genetic linkage data, bioinformatic analysis, and the observed treatment

effect—confirms the causative role of the VMAT2 p.P387L mutation in this autosomal

recessive pediatric neurotransmitter disorder.

Page 136: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

113

Chapter 5 General Discussion and Future Directions

1 Introduction This thesis presents the identification of a new pediatric neurotransmitter disease and its

causative mutation in the gene encoding the brain vesicular monoamine transporter

(VMAT2; SLC18A2), as well as demonstrating the utility of next-generation sequencing

for the clinical diagnosis of extremely rare diseases.

This discovery extends the spectrum of pediatric neurotransmitter disease

pathophysiology to include defects in the vesicular transport and storage of biogenic

amines. This is the first demonstration of a clinical phenotype directly associated with

VMAT2 deficiency, providing new insight on the physiological consequences of

perturbed monoamine homeostasis in humans.

The research directions and experiments proposed in this chapter are designed to address

open questions stemming from the results presented here pertaining to the role of

VMAT2 in monoamine physiology (Section 2), the pathophysiology of VMAT2

deficiency (Section 3), and the diagnosis and treatment of pediatric neurotransmitter

diseases (Section 4).

In Section 2, a series of experiments are proposed to further probe the dose dependence

of VMAT2 function and its role as a modulator of monoamine homeostasis, leveraging

the P387L variant to demonstrate the effect of reduced VMAT2 function without the

confounding effects of reduced protein levels or off-target effects of drug inhibitors.

Page 137: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

114

Subcellular localization experiments in relevant cell culture models are proposed to

explore any mechanisms of regulation of VMAT2 in the context of reduced function.

These results will also inform data derived from a proposed mouse model incorporating

the corresponding P390L mutation.

In Section 3, some investigations are proposed to further explore the pathophysiology of

VMAT2 deficiency by obtaining more data in a clinical context. Positron emission

tomography (PET) is proposed to visualize aberrant VMAT2 and dopamine distribution

in vivo. Genetic screening of patient cohorts with neurological disorders is also proposed

to identify additional patients with VMAT2 mutations and better characterize the range of

phenotypes associated with VMAT2 deficiency.

In Section 4, challenges in the diagnosis of VMAT2 deficiency are described and some

methodologies are described that may address the current limitations of clinical

diagnosis. In addition, the promise and challenges of next-generation sequencing for rare

disease diagnosis are outlined.

In sum, this section outlines three different directions for continued research stemming

from the results of this thesis and discusses the relevance of these results to clinical

practice.

2 Identification of the VMAT2 p.P387L Mutation: Impact on Knowledge of Monoamine Physiology

As the first demonstration of a phenotype associated with VMAT2 deficiency in human

patients, the results presented in this thesis confirm that significant loss of VMAT2

Page 138: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

115

function is associated with a phenotype that is within the spectrum of those caused by

biosynthetic monoamine deficiencies.

The VMAT2 p.P387L variant, which is associated with viability despite corresponding to

a near complete—but not complete—loss of VMAT2 function as measured by functional

assay in a heterologous system, provides an interesting opportunity to further probe the

role of VMAT2 in monoamine physiology and homeostasis. In mice, the complete

knockout of Vmat2 results in absent exocytotic monoamine neurotransmission, and

animals that feed poorly and die within days after birth (Wang et al., 1997, Fon et al.,

1997). By contrast, mice that express just 5% of native Vmat2 levels live to adulthood

and only develop comparatively minor age-related motor deficits over time (Mooslehner

et al., 2001). The phenotypic spectrum of VMAT2 deficiency is therefore broad, and

very large decreases in protein function may be required to result in severe, early onset

motor symptoms. However, there may be differences in the phenotypes caused by lower

protein levels and aberrant protein function, possibly stemming from differences in

VMAT2 trafficking and its relative distribution between readily releasable and recycling

or reserve pools of synaptic vesicles. Future research should clarify the mechanism of

loss of function by confirming whether protein localization to synaptic vesicles is

affected by the mutation in vivo. Appropriate cell culture models can be used to

investigate VMAT2 sorting. These experiments are discussed in Section 2.1.1.

Initial confirmation of reduced uptake activity of a single substrate was considered

sufficient in the present work to confirm the functional effect of the VMAT2 p.P387L

variant. More precise characterization of the p.P387L mutation may be of interest for

Page 139: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

116

drawing broader insights on VMAT2 structure–function relationships. Proposed

experiments are described in Section 2.1.2.

Because VMAT2 density on synaptic vesicles determines quantal size for monoamine

neurotransmission (Edwards, 2007, Pothos et al., 2000), and its function is therefore

subject to dose dependence, unique insights may be gleaned into monoamine physiology

by investigating the effect of the corresponding Vmat2 p.P390L mutation in a mouse

model that were not derived from studying one of the existing mouse models. The

proposed mouse model is described in Section 2.1.3.

2.1 Future Directions and Experiments

2.1.1 Subcellular Localization of VMAT2 p.P387L

Subcellular localization of VMAT2-P387L can be assessed in relevant cell culture

models using immunofluorescence colocalization and sucrose gradient centrifugation

using relevant cell compartment markers (e.g., synaptophysin for synaptic vesicles). To

avoid the artifacts of overexpression introduced by transient transfection (as in Chapter 4,

Section 3.4), cell lines should be constructed to stably express either wild-type or mutant

VMAT2 protein.

The following section describes the construction of these cell lines. Stable expression of

VMAT2 (wild-type and P387L) was confirmed in both Cos7 cells and the MN9D

mesencephalic cell line. MN9D cells were originally derived as a fusion of embryonic

ventral mesencephalic and neuroblastoma cells, and are extensively used as a model of

dopamine neurons because they express tyrosine hydroxylase and synthesize and release

Page 140: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

117

dopamine. These cells can be differentiated with exposure to butyric acid, and develop

some morphological characteristics of neurons such as visible processes. Because these

cells possess synaptic vesicles, they have already been used in studies of VMAT2

function, in which it was shown that overexpression of VMAT2 confers additional

protection against MPP+ (Chen et al., 2005). Interestingly, co-expression of

synaptophysin enhanced this effect possibly through increased vesicular capacity. The

co-expression of synaptophysin may therefore also be helpful in these proposed

localization experiments.

2.1.1.1 Generation of Stable Cell Lines Expressing VMAT2 p.P387L

Cell lines were generated to stably express human VMAT2. Independent cell lines were

generated to express wild-type VMAT2 and VMAT2-P387L. Additionally, cell lines

with genomic incorporation of the vector backbone alone were generated for use as

experimental controls.

Stable expression of human VMAT2 was generated in the Cos7 cell line. Cos7 cells were

maintained as described in Chapter 4 (Section 2.2). The pcDNA3.1 vectors containing

wild-type VMAT2 and VMAT2-P387L cDNA sequence, in addition to empty pcDNA3.1

vector, were used in independent transfections. Prior to transfection, the vector was

linearized to optimize genomic incorporation of the construct. Transfection was

performed with Fugene HD transfection reagent (Roche), according to the manufacturer’s

specifications. After 48 hours to allow expression of the neomycin cassette on the vector,

the cells were trypsinized and a series of dilutions from 1:10 to 1:1000 were plated

Page 141: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

118

independently in DMEM containing 500 µg/mL of G418. The selection medium was

replaced each day with fresh medium until the appearance of visible colonies. Colonies

were transferred to 96-well plates containing trypsin and subsequently diluted into

DMEM + 10% FBS + 500 µg/mL G418. Individual wells were eventually passaged into

larger diameter wells and screened for VMAT2 expression by western blot (Figure 22).

Positive clones were expanded and frozen at –80°C

Figure 22 Expression of wild-type and p.P387L VMAT2 in stable clones of the Cos7 cell line; band at 37 kDa represents GAPDH loading control.

MN9D cells were maintained in custom DMEM media at pH 7.2 containing 10% Fetal

Clone 3 (Hyclone) and 50 U/mL penicillin/streptomycin. They were cultured in flasks at

Page 142: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

119

37°C and 5% CO2, and fed every other day by aspiration of the media and replacement

with fresh media. Cells were passaged by trypsinization and seeding into new flasks at

plating densities of 200,000 per T75 flask and grown to confluence. The number of

passages was minimized to avoid morphological changes.

Because MN9D cells endogenously express mouse Vmat2, the human VMAT2 protein

was tagged internally with an HA epitope to distinguish the isoforms. Initial attempts to

incorporate N-terminal and C-terminal FLAG and myc tags led to no detectable protein

by western blot. The HA tag was therefore incorporated into the large lumenal loop

immediately after Arg94, as described by Thiriot and Ruoho (Thiriot and Ruoho, 2001).

The tag was incorporated by site-directed mutagenesis using complementary

oligonucleotide sequences that incorporated the HA epitope (Table 18). The transfection

and selection of clones was performed as described for generation of the Cos7 stable cell

lines. Confirmation of human VMAT2 expression was performed by western blot using

an antibody against the HA epitope (Figure 23).

Table 18 Primers for the integration of the HA epitope at Arg94 of VMAT2 using site-direted mutagenesis

Primer Sequence

VMAT2-Arg94-HAepitope-upR AGC GTA ATC TGG AAC ATC GTA TGG GTA TCT GGT AGC ATT CCC GGT G

VMAT2-Arg94-HAepitope-downF TAC CCA TAC GAT GTT CCA GAT TAC GCT GAC CTG ACA CTT CAT CAG

Page 143: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

120

Figure 23 Expression of wild-type and p.P387L VMAT2 in stable clones of the MN9D cell line

2.1.2 Additional Biochemical Characterization of VMAT2 p.P387L

The precise functional consequences of proline to leucine substitution at residue 387 in

VMAT2 can be further probed by comparing the relative uptake of additional substrates

(i.e., dopamine, norepinephrine, histamine) either directly or through competitive

inhibition of serotonin uptake, and the relative binding of inhibitors (i.e., reserpine,

tetrabenazine). Furthermore, the use of valinomycin (a K+ ionophore) and bafilomycin

(an inhibitor of the vesicular ATPase) can be used to probe the coupling of VMAT2

p.P387L to the electrochemical and proton gradients.

Page 144: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

121

The recent development of live cell functional assays using fluorescent VMAT2

substrates provide additional options for direct visualization of substrate uptake by live

cell microscopy (Bernstein et al., 2012), or quantitative fluorometric plate assay (Hu et

al., 2013), provide additional options for probing mutant protein function.

2.1.3 Development of a Mouse Model Expression Vmat2 p.P390L

The functional defect of the VMAT2 p.P387L mutation (corresponding to p.P390L in

mouse Vmat2) is expected to dramatically alter dopamine homeostasis in the context of

the brain. Thus, the in vitro characterization presented in Chapter 4 should be

complemented by an in vivo assessment of the physiological consequences of the

mutation. I therefore propose to create a knock-in mouse model to address the role of the

p.P390L mutation in Vmat2 expression and subcellular localization, dopamine

homeostasis, and movement disorder phenotype.

As described in Chapter 1, Sections 2.4 and 2.5, existing mouse models of VMAT2

defect include a Vmat2 knockout mouse and a Vmat2-deficient mouse expressing 5% of

natural protein levels. The knockout mice exhibit normal Mendelian ratios, but neonatal

mice feed poorly, move slowly, and die a few days postnatally (Wang et al., 1997).

Detailed histology of the early postnatal cerebral cortex exhibits an increase in

developmental programmed cell death in these mice, which is partially rescued by MaoA

inhibition (Vmat2-MaoA DKO mice) with increased serotonin levels (Stankovski et al.,

2007). The hypomorpic Vmat2 mouse, by contrast, appears to have sufficient Vmat2

function to overcome this developmental barrier, and these mice exhibit adult onset

nigrostriatal neurodegeneration and deficits in motor function associated with several

Page 145: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

122

molecular hallmarks of oxidative stress and Parkinson’s Disease (increased alpha-

synuclein, increased levels of cysteinyl adducts) (Caudle et al., 2007, Mooslehner et al.,

2001). Additional phenotypes have been ascribed to heterozygotes of the knockout

allele, including prolonged QT syndrome, an increased rate of sudden death at 2–4

months of age, depressive-like behaviours, and reduced locomotor activity (Fukui et al.,

2007, Itokawa et al., 1999).

Because of the early onset and debilitating motor symptoms of the human disease, the

Vmat2 p.P390L mice would be expected to produce a more severe defect than that

observed with the hypomorphic Vmat2 allele. However, with full survival, normal MRI,

and mostly normal cognitive abilities in human patients harbouring the p.P387L

mutation, the effect of p.P390L is expected to be distinct from the severe developmental

defects exhibited by full Vmat2 knockout in mouse.

Given the potential for dose dependence of Vmat2 function and its multiple substrate

specificities, and that the existing mouse models do not clearly recapitulate the clinical

syndrome of pediatric neurotransmitter disorder, there is value in pursuing additional

studies of Vmat2 function in the mouse. The P390L mutation may represent an allele

with novel properties for vesicular monoamine transport functions, or provide more data

on the dose dependence of Vmat2 function, and would therefore provide a valuable

mouse model in addition to those thus far characterized.

The gene-targeting construct for Vmat2-P390L knock-in mouse model was designed,

constructed, and confirmed. The construction of this gene-targeting vector and the

Page 146: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

123

planned strategy for generating the mouse model, as well as the experimental plan for the

mouse model, are described below.

2.1.3.1 Design of Gene-Targeting Construct

The gene-targeting strategy for creation of the Vmat2 P390L knock-in mouse is presented

schematically in Figure 24. The c.1169C→T mutation will be introduced into mouse

sequence adjacent to a neomycin selection cassette. The neomycin selection cassette will

be used for the selection of positive recombinants. The cassette will present in the intron

to avoid disruption to Slc18a2 coding sequence. This cassette is also flanked by loxP

recombination sequences. In cells that express the Cre recombinase protein, the loxP

sequences will recombine in vivo and the intervening selection cassette will be excised.

This step will ensure minimal disruption of Slc18a2 expression. The c.1169C→T

mutation would be present at a distance of 1 kb from the neomycin selection cassette. At

this distance, the mutation is expected to be incorporated into the mouse genome in

combination with the selection cassette at a high frequency, generating a high proportion

of neomycin-selected clones that will be positive for the c.1169C→T mutation.

A schematic of the relevant features of the gene-targeting vector is presented in Figure

25. The pGKneolox2DTA vector designed by Soriano was used as the basis for the

construction of this gene-targeting vector (Soriano, 1997), named pGKneolox2DTA-

Vmat2-P390L. The vector was designed to include two long arms of Slc18a2 sequence

to undergo homologous recombination with the mouse genome in mouse embryonic stem

cells. The neomycin selection cassette is present between the flanking arms of Slc18a2

sequence. The c.1169C→T mutation encoding the P390L amino acid substitution was

Page 147: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

124

introduced into exon 13, present in the downstream arm. The construction of this vector

is discussed in the subsequent section.

Page 148: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

125

Figure 24 Schematic of incorporation of mouse gene targeting construct into mouse genome. Upper panel, wild type mouse genomic sequence of Slc18a2 exons 11–15 and flanking intronic sequences. Middle panel, mouse genomic sequence after incorporation of the gene-targeting construct by homologous recombination with the long arms of the construct in embryonic stem cells. Slc18a2 c.1169C→T mutation, coding for the P390L amino acid substitution, is present in exon 13.

Lower panel, mouse genomic sequence after expression of Cre recombinase and excision of the selection cassette. PGKneobpA, neomycin selection cassette; loxP, recombination sites targeted by Cre recombinase.

Page 149: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

126

Page 150: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

127

Figure 25 Gene targeting construct for the introduction of the c.1169C→T mutation into Slc18a2. AmpR, ampicillin

resistance gene; F1_origin, origin of replication; loxP, recombination site for Cre recombinase; NeoR/KanR, neomycin and kanamycin resistance gene (of the PGKneobpA selection cassette); NheI, HindIII, restriction sites for insertion of downstream arm of genomic sequence for recombination; NotI, SacII, restriction sites for insertion of upstream arm of genomic sequence for recombination. Gray boxes, mouse intronic sequence of Slc18a2; black boxes, mouse exonic sequence of Slc18a2.

Page 151: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

128

Page 152: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

129

2.1.3.2 Construction of Gene-Targeting Construct

Primers were designed to amplify a 1-kb fragment from mouse genomic DNA that encompassed

exon 13 of Slc18a2. This fragment was used to probe the RP23 C57BL/6 BAC library (Table

19). A clone was identified that encompassed the Slc18a2 gene (clone 338J16).

Regions upstream and downstream of the insertion site for the floxed TK-neomycin cassette

were amplified using Roche Long Template (LT) Taq polymerase. The conditions for

amplification were as follows: 95°C for 5 min; 30 cycles of 95°C for 30 s, 57°C for 30s, and

72°C for 2 min; 72°C for 10 min. Primers for amplification incorporated restriction sites for

cloning, and are listed in Table 20. The fragments were initially cloned into pcDNA3.1 for

further subcloning.

After cloning of the downstream fragment into pcDNA3.1, the exon 13 C→T nucleotide

substitution corresponding to P390L was incorporated using the Quikchange Site-Directed

Mutagenesis Kit (Stratagene) according to manufacturer’s specifications. Oligonucleotide

sequences for site-directed mutagenesis are presented in Table 21. The incorporation of the

mutation was confirmed by direct sequencing of the plasmid.

The upstream and downstream fragments were subcloned appropriately into the

pGKneolox2DTA vector to flank the neomycin cassette. The relevant restriction sites used for

subcloning are displayed in the schematic of the gene-targeting vector presented in Figure 25.

The correct orientation and sequence of the fragments were confirmed by direct sequencing

(primer sequences in Table 22).

Page 153: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

130

Table 19 Primer sequences to amplify a probe for Slc18a2 exon 13 in mouse Primer Sequence Size Slc18a2-MsExon13-Probe-F ATT GAA GCT GAG GGT GGA TG 1018 bp Slc18a2-MsExon13-Probe-R TCC TCT GGA GGG TCA GAA AA

Table 20 Primer sequences to amplify the upstream and downstream arms of the gene-targeting construct Primer Sequence Size Ms-mod-upstream-SacII-F GCC AGG CCG CGG TTC CAG GAT CAG

CCA TTA GG 3677 bp

Ms-mod-upstream-NotI-R TCA GCA GCG GCC GCC AGC ACT TGC AAG AAT GAG G

Ms-mod-downstream-NheI-F

ACG GCT GCT AGC AGG CAT GGC TGA TGA AAC ATT ACA G

5306 bp

Ms-mod-downstream-HindIII-R

TAG CAT AAG CTT AGC AGT TCA CTA CCA GCT GTG ACT C

Table 21 Oligonucleotide sequences for site-directed mutagenesis of gene-targeting construct to introduce the p.P390L variant in the expressed protein Primer Sequence Mouse-Vmat2-P390L Quikchange-F AAA TAT CTA CGG ACT CAT CGC TCT

CAA CTT TGG AGT TGG TTT TGC Mouse-Vmat2-P390L Quikchange-R GCA AAA CCA ACT CCA AAG TTG AGA

GCG ATG AGT CCG TAG ATA TTT

Table 22 Sequencing primer sequences for gene-targeting construct Primer Sequence pGKneolox2DTA-upstream-F GTT TTC CCA GTC ACG ACG TT pGKneolox2DTA-upstream-R GGT TCC GGA TCC ACT AGT TCT pGKneolox2DTA-downstream-F GCT GAT CCG GAA CCC TTA AT pGKneolox2DTA-downstream-R CTA AAG CGC ATG CTC CAG AC Ms-SLC18A2-exon11-F ACC GTG GTG TCC GTG ATT AT Ms-SLC18A2-exon11-R TCA AAG ACC CAG ATC ATT CTC A Ms-SLC18A2-exon12-F TGC ATC TTG GGA CAC ACA GT Ms-SLC18A2-exon12-R GAG CTT CAG TCC CCA CAC TC Ms-SLC18A2-exon13-F GGT CCA CTA GGC CCA GAT AA Ms-SLC18A2-exon13-R CCG TCT GTT CAC TCT TCA CG Ms-SLC18A2-exon14-F CAC AGT CGA TGA CAT CTA GCC Ms-SLC18A2-exon14-F TCT GTA CCA CAT TAA AGT AAA GGT TTT Ms-SLC18A2-exon15-F TGG CTT TCA GGA AAA CCT TTA Ms-SLC18A2-exon15-R GGC CAT GGA GAA ATG ATT G

Page 154: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

131

2.1.3.3 Gene Targeting

Gene targeting will be accomplished by electroporation of the gene-targeting construct into

embryonic stem (ES) cells of the G4 cell line (129×C57BL/6 F1 hybrid). Clones can be selected

using G418. Primers were designed to amplify probe sequences for screening by Southern blot

(Table 23). Clones will be screened for the presence of both the neomycin cassette and adjacent

sequence to ensure incorporation of the cassette by homologous recombination. The

incorporation of the c.1169C→T mutation will be confirmed by sequencing.

Confirmed clones will be used to inject C57BL/6 blastocysts. Because the presence of the

selection cassette in intron 12 may affect Vmat2 expression, progeny will be bred to C57BL/6

mice expressing Cre recombinase to ensure removal of the cassette.

Experiments in protein expression and subcellular localization, as well as gross histology/MRI

will be performed primarily on F2 progeny. Because of significant strain differences in

monoaminergic phenotypes (Mhyre et al., 2005), for consistency the mice will be backbred for

five generations to C57BL/6 mice before performing behavioral and motor performance tests

(comparable to similar tests in hypomorphic Vmat2 mice), and catecholamine level and MPTP

sensitivity measurements. On the basis of the autosomal recessive human clinical phenotype for

this mutation, the homozygous mutation is not anticipated to be lethal. If this is the case, tests

will be performed on heterozygotes.

Table 23 Primer sequences for generating labelled probes to identify the neomycin selection cassette by Southern blot Primer Sequence Size Neo-Probe-F AGA CAA TCG GCT GCT CTG AT Neo-Probe-R GCG ATG CAA TTT CCT CAT TT Adj-Probe-F AAT GCA GTC CTG CTT GTG TG 880 bp Adj-Probe-R GAG CTT CAG TCC CCA CAC TC

Page 155: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

132

2.1.3.4 In Vivo and Subcellular Localization

Despite measurement of Vmat2 wild type and P390L levels in heterologous cell culture models,

the regulation of protein expression and localization may depend on factors only present in vivo.

Thus, the quantitation of Vmat2 wild type and P390L levels in mouse brain segments is

necessary to determine the physiologic effect of the mutation.

Relevant areas of the brain should be dissected to measure Vmat2 levels in each (as well as from

whole brain) using western blot of lysates and 3H-TBZ binding assays.

The subcellular localization of VMAT2 in vivo is of paramount importance in determining the

site and regulation of release of neurotransmitter. The cell culture models described in

Section 2.1.1 have significant caveats for the assessment of subcellular localization—Cos7 is not

a neuronal cell model, and in MN9D, the total quantity of available vesicles is diminished. In

dopaminergic axon terminals within the rat striatum, it has been shown that Vmat2

predominantly localizes to small synaptic vesicles (SSVs), but is also present on large dense core

vesicles (LDCVs) (Nirenberg et al., 1997). Using the same methodology, immunogold labeling

and electron microscopy, the subcellular localization of Vmat2 P390L should be studied in

sections from striata of homozygous and wild-type mice. Concurrent labeling of DAT or TH

will distinguish between dopaminergic from serotonergic Vmat2-containing axons.

2.1.3.5 Histology and MRI

MRI represents the best methodology for determining subtle changes in shape and volume of

small brain regions (Chen et al., 2006). MRI will be used on homozygous, heterozygous, and

wild type mice to look for volumetric differences in the cerebral cortex, thalamus, basal ganglia,

striatum, cerebellar cortex, hippocampus, and entorhinal cortex.

Page 156: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

133

Histology of the substantia nigra and early postnatal cortex will reveal any dopaminergic

neurodegeneration or developmental defects, respectively. Proper assessment of degeneration of

the substantia nigra would involve quantitation of TH-immunopositive fibres. Further, in situ

hybridization should be used to quantify the relative amounts of substance P-positive and

enkephalin-positive terminals in the striatum, to distinguish the effect of the mutation on the

development of the direct and indirect striatal output pathways, respectively.

2.1.3.6 Catecholamine Concentrations

Parkinsonism is associated with a depletion of striatal and whole brain dopamine. Hypomorphic

Vmat2 mice similarly indicate a depletion of monoamines in the adult brain (Mooslehner et al.,

2001). The total content of monoamines (DA, 5-HT, NE) and relevant metabolites (DOPAC,

HVA, 5-HIAA) in whole brains and striata of Vmat2 P390L homozygous, heterozygous, and

wild type mice will be measured using high pressure liquid chromatography with

electrochemical detection (HPLC-EC) of perchloric acid lysates, following established protocols

(Guevara-Guzman et al., 1994). Dopamine turnover can be measured as the ratio of

DOPAC/DA and HVA/DA, and serotonin turnover expressed as 5-HIAA/5-HT. Further, to

measure the amount of available dopamine relevant to neurotransmission, the extracellular

concentration of dopamine in the striatum may be assessed using in vivo microdialysis using

implanted dialysis probes, with fractions analyzed immediately by the same HPLC-EC method.

Each method should be performed on 10–15 mice of each genotype and data analyzed by

Student’s t-test.

2.1.3.7 Motor Performance and Behavioral Testing

The motor performance of Vmat2 P390L mutant mice is the most relevant phenotype. Because

locomotor activity shows vast strain differences (Mhyre et al., 2005), it will be necessary to

Page 157: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

134

breed these mice to C57BL/6 for 10 generations to ensure homogeneity of the genetic

background. Initially, mice would be examined for evidence of tremor or dystonia (abnormal

postures or twisting movements). Dystonia also manifests as hindlimb retraction or trunk flexion

in response to tail suspension. Abnormal gait should be assessed by pawprint pattern of mice

walking along a narrow runway (measurement of stride length and width). Locomotor hyper- or

hypoactivity can be assessed in open field with measurement of both horizontal locomotion

(crossing) and vertical locomotion (rearing). A beam walking task and accelerating rotarod test

should be performed as sensitive tests of motor coordination and balance. Grip strength tests

should be performed to measure associated muscle weakness. Results can be compared to those

published for the hypomorphic Vmat2 mice, who showed reduced vertical and horizontal

locomotor activity and slower times and more slips in the beam walking task (Mooslehner et al.,

2001). The Vmat2-KO mice do not survive long enough to be tested for motor performance, but

the heterozygotes do show reduced horizontal and vertical locomotor activity in open field

(Fukui et al., 2007).

It is further important to assess behavioral phenotypes of these mice, given the role of VMAT2

in serotonin vesicular storage, and the depression side effects of reserpine treatment in humans.

The mice should be tested in forced swim, learned helplessness, novelty-suppressed feeding, and

open field exploration, as described for the Vmat2-KO heterozygote mice (Fukui et al., 2007).

2.1.3.8 MPTP Resistance

To assess the contribution of the Vmat2 p.P390L mutation to MPTP-induced nigrostriatal

degeneration, homozygous, heterozygous, and wild type mice should be injected

intraperitoneally with the MPTP toxin. After 8 days, animals can be sacrificed and the relative

Page 158: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

135

sizes of the substantia nigra pars compacta and TH-immunopositive cell counts (vs. control

saline injections) can be assessed in brain sections.

3 VMAT2 Mutation in PND: Impact on Understanding of PND Pathogenesis

The patients treated in this study show a progressive motor phenotype with age, and demonstrate

differential response to treatment on the basis of the age at which treatment was initiated. This

suggests that anatomical differences may underlie the expression of the disease at different ages,

illustrating that monoamine deficiency influences normal neurological development and that the

direct effects of monoamine deficiency are overlaid on abnormal functional architecture in the

basal ganglia. The hypomorphic Vmat2 mouse model exhibited decreased substance P and

increased enkephalin expression, consistent with this hypothesis (Mooslehner et al., 2001). What

is not yet evident, however, is whether there are any downstream neurodegenerative effects of

impaired vesicular monoamine storage over the course of patients’ lifetimes, as were observed in

hypomorphic Vmat2 mice (Caudle et al., 2007). Careful patient follow-up comprising both MRI

and neurological examination should be maintained to monitor this possibility.

The results presented in this thesis have clearly demonstrated a functional effect of the p.P387L

mutation on VMAT2 protein function, but the precise physiological consequences in human

patients remain to be clarified. Studies in a mouse model (as described in Section 2.1.3) may

help clarify the pathophysiological consequences of this mutation relative to complete deletion or

other hypomorphic Vmat2 mice, but there is limited ability to extrapolate these results to human

physiology. Positron emission tomography (PET) is a methodology that would allow direct

visualization of both VMAT2 protein levels and the vesicular retention of monoamines in

patients and control subjects. PET studies are proposed in Section 3.1.1.

Page 159: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

136

Potential refinement or expansion of the clinical phenotypes associated with VMAT2

dysfunction can be further explored through genetic screening of additional patient cohorts to

identify novel SLC18A2 variants, as proposed in Section 3.1.2.

3.1 Future Directions and Experiments

3.1.1 18F-DOPA and 11C-DTBZ PET scans

Specific VMAT2 ligands have been developed as imaging agents for early diagnosis and

monitoring of neurodegenerative disease and neuropsychiatric conditions with monoaminergic

involvement. Derivatives of tetrabenazine are the primary agents developed for positron

emission tomography (PET). In mice, it was demonstrated that after intravenous injection,

[11C]TBZ is rapidly taken up in the brain where it localizes primarily to the striatum and

hypothalamus, as well as less strongly in the hippocampus, cortex, and cerebellum (DaSilva and

Kilbourn, 1992).

3.1.1.1 [11C]Dihydrotetrabenazine ([11C]DTBZ)

There are several potential limitations to the direct imaging of VMAT2 levels using

tetrabenazine analogs. Because there is evidence of overlap between the binding site of

tetrabenazine and the substrate-binding region of VMAT2, the technique is effectively

visualizing the number of available VMAT2 binding sites in the brain. Competition in binding

between the tetrabenazine analog and natural substrates would therefore make the accuracy of

the technique subject to differences of in vivo monoamine levels. Comparison of [11C]DTBZ

signal between patients with a missense mutation in VMAT2 and control subjects would

therefore be confounded by a relative increase in binding site availability (per VMAT2

molecule) as a result of the drastically decreased availability of natural monoamine substrate. A

quantitative study in rats confirmed a statistically significant increase in [11C]DTBZ binding in

Page 160: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

137

vivo with significant reduction in brain dopamine levels without a concomitant increase in

protein levels (Kilbourn et al., 2010). Similarly, a human PET study observed a rapid decrease

in [11C]DTBZ binding with the administration of levodopa (exogenous dopamine) (de la Fuente-

Fernandez et al., 2009). Additionally, the natural plasticity of VMAT2 is the subject of some

controversy, and fluctuations in VMAT2 protein levels may exist among healthy individuals and

mask clinically relevant fluctuations in VMAT2 concentrations. Given these confounding

factors, however, a dramatic decrease in VMAT2 signal would clearly indicate mutant protein

instability or aberrant conformation in vivo.

3.1.1.2 6-[18F]fluoro-L-dopa ([18F]-DOPA)

In conjunction with [11C]DTBZ scans, 6-[18F]fluoro-L-dopa ([18F]-DOPA) scans would provide

more information. The retention of [18F]-DOPA for visualization within the synapse is

dependent upon both presynaptic uptake, dopa decarboxylation, and vesicular uptake—namely,

the proper functioning of DAT, AADC, and VMAT2. This imaging substrate would therefore

provide a more holistic visual indication of dopaminergic functioning in these patients, and avoid

the confounding issues discussed above for [13C]DTBZ binding.

The radiation exposure for a PET scan is modest (7–8 mSv). This noninvasive assessment of

both VMAT2 binding and dopaminergic functioning in patients and siblings would provide a

better understanding of the effect of the P387L variant on VMAT2 availability and dopamine

sequestration in vivo.

3.1.2 Screening of Patient Cohorts to Identify Novel SLC18A2 Variants

An additional research direction of interest is genetic screening in patient cohorts to identify

novel variants in SLC18A2 with the aim of refining—and possibly expanding—the phenotypic

spectrum associated with VMAT2 defect. Patients with possible undiagnosed VMAT2

Page 161: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

138

deficiency may be identified among those previously screened for pediatric neurotransmitter

disease with normal CSF neurotransmitter metabolite profiles. They may also be identified

among patients diagnosed with cerebral palsy (CP) by exclusion. Furthermore, a broader

approach to screening should comprise patients with any phenotype related to biogenic amine

function, including movement disorder, neuropsychiatric phenotypes, addiction phenotypes, and

autonomic dysfunctions. Variants identified through screening should be confirmed through

bioinformatic and functional analyses analogous to those performed in this thesis.

4 VMAT2 Mutation: Impact on Approaches to PND Diagnosis

The clinical investigations presented in Chapter 2 demonstrated the challenges in diagnosis of

VMAT2 deficiency related to limitations in CSF neurotransmitter metabolite analysis as a

conclusive diagnostic for monoamine deficiency. Because the sole conclusive diagnostic for

VMAT2 deficiency at present is genetic analysis, mutation screening of SLC18A2 is

recommended in addition to both CSF and urine neurotransmitter metabolite analysis in

suspected cases of monoamine deficiency. For the comprehensive investigation of suspected

monoamine deficiency, and given the relative ease with which a multigene panel could be

assayed, genetic analysis of all known disorders with primary or secondary neurotransmitter

phenotype should be performed in parallel.

To improve functional confirmation of a genetic diagnosis of VMAT2 deficiency, the

development of methodology to directly measure biogenic amines in CSF is proposed in

Section 4.1.1, and the development of a platelet activity assay for VMAT2 is proposed in

Section 4.1.2.

Page 162: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

139

Finally, considerations relevant to the clinical utility of next-generation sequencing for the

diagnosis of pediatric neurotransmitter disorders and other rare monogenic diseases is discussed

in Section 4.1.3, and knowledge translation challenges and strategies for the diagnosis of

pediatric neurotransmitter disorders and related diseases are discussed in Section 4.1.4.

4.1 Future Directions and Experiments

4.1.1 Development of Methodology to Measure Biogenic Amines Directly in CSF

The direct measurement of dopamine, serotonin, and norepinephrine in CSF is unfeasible by LC-

MS/MS because it is below the limit of detection of the technique and monoamine

neurotransmitters are less stable than their respective metabolites. The measurement of

metabolites, however, is accompanied by numerous caveats. In cases of defective biosynthesis,

it is reasonable to assume that the metabolites accurately reflect monoamine deficiencies. The

linearity of this response, however, remains to be demonstrated. Transporter defects, by contrast,

effectively influence the turnover of the biogenic amines. The relationship between endogenous

concentrations of amine and metabolite is therefore more complex in these cases.

The biogenic amines can be measured in physiologic samples using commercially available

ultra-sensitive enzyme-linked immunosorbant assay (ELISA) kits according to manufacturer’s

specifications. For example, serotonin can be measured in physiologic samples to a

concentration of 0.05 ng/mL (Labor Diagnostika Nord BA 10-5900).

For this technique, then, the preservation of endogenous amines during CSF collection becomes

paramount. Samples should be collected directly into 0.1% w/v ascorbic acid to prevent

oxidation, and stored immediately at –80°C. Given the risk associated with lumbar puncture, the

collection of control samples will need to occur in the clinic secondary to lumbar puncture

Page 163: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

140

performed for the diagnosis of unrelated conditions. This will have to be designed prospectively

to ensure proper sample collection in these cases.

Accurate determination of endogenous monoamine concentrations in these patients will provide

better quantitative understanding of the deficiencies in neurotransmission.

4.1.2 Development of VMAT2 Platelet Activity Assay

Because the CSF neurotransmitter metabolite profile appears to be normal in VMAT2

deficiency, alternative strategies are needed to confirm the diagnosis in suspected cases.

Serotonin in the blood is taken up by platelets and released to aid clotting. Both VMAT2 and

SERT are expressed in platelets, and platelets are therefore an accessible peripheral cell type in

which to assay VMAT2 function. As presented in Chapter 2, Section 2.4, AADC enzyme

function is assayed in serum in suspected cases of AADC deficiency.

Some studies have attempted to measure differences in platelet VMAT2 density in various

neuropsychiatric disorders using a tritiated dihydrotetrabenazine ([3H]-TBZOH) binding assay,

with moderate but statistically significant results (Zucker et al., 2002a, Zucker et al., 2002b,

Toren et al., 2005, Schwartz et al., 2005b, Laufer et al., 2005, Schwartz et al., 2005a, Schwartz et

al., 2007, Ben-Dor et al., 2007, Sala et al., 2010, Zalsman et al., 2011a, Zalsman et al., 2011b).

Whereas those studies were intended to probe the regulation of VMAT2 expression as an indirect

indicator of perturbations in monoamine homeostasis, I propose to develop a standardized assay

of tritiated serotonin uptake to assay VMAT2 function. Platelet-rich plasma would be separated

from blood cells by low speed centrifugation, and platelets would be disrupted by sonication in

sucrose buffer. A serotonin uptake assay would be performed on the resulting lysate as

described in Chapter 4, Section 2.3. In this way, a pathogenic loss of VMAT2 function could be

assayed from a blood sample. The caveat of this approach is that previous studies required a

Page 164: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

141

large blood volume (25 mL) to assay [3H]-TBZOH binding, which is a large blood volume for a

pediatric patient. Guidelines on acceptable blood volumes to be collected from pediatric patients

suggest a maximum of 3 mL/kg within 24 hours (approximately 3.8% of total blood volume)—

prohibiting the use of this test for infants under 8.5 kg (Howie, 2011). Although this test would

provide significantly less risk to the patient than a lumbar puncture, it is specific only to this

particular very rare disease, and would therefore likely be pursued only in the event of a normal

CSF metabolite profile.

4.1.3 Use of Next-Generation Sequencing for Genetic Diagnosis of Rare Monogenic Diseases

Beyond targeted genetic screening of individual candidate genes or multigene panels, the results

presented in this thesis demonstrate that there is now the ability to diagnose a new monogenic

disorder in a single family using genomic methods within a timeframe of relevance to the

treatment of individual patients. At the initiation of this project, the identification of disease-

causing mutations was accomplished by investigating the segregation of particular genomic

regions in association with the inheritance of the disease. Microsatellite markers were still

primarily used, although the availability of single nucleotide polymorphism arrays was

increasing. These techniques were used initially in the present case to identify the disease-

associated locus using a combination of linkage and homozygosity analyses. Subsequently,

mutation screening of a manageable set of candidate genes (selected on the basis of known

function or localization) was performed to identify variants.

Experience and improved resources to pursue these methods allowed the initial identification of

the variant in a matter of months rather than years, as was the timeframe for gene hunting during

the previous decade; however, the clinical utility of this information was limited by its

identification in only a single family. Without functional confirmation from the identification of

Page 165: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

142

an unrelated family exhibiting a similar phenotype and harboring a variant in the same gene,

there was an increased burden of proof for confirming the functional relevance of the variant

before translating these insights to the clinic. Namely, we pursued the confirmation of a

functional effect of the variant, the absence of the variant in a significant number of unaffected

family members, and the absence of further variants in the region. The success of these

approaches in finding the causative mutation in a single family was dependent upon the size of

the pedigree, the mode of inheritance, and the number of affected individuals—a significant

limitation for genomic diagnosis by these methods in isolated cases that present to a clinic. In

addition, de novo mutations cannot be identified by positional cloning approaches.

The more recent availability of next-generation sequencing methodology, including whole-

exome sequencing, addresses many limitations of gene hunting using linkage and association

with genomic markers with respect to clinical diagnosis. First, sequence results for the whole

genome can be obtained very quickly and cost is no longer strictly prohibitive. A list of genetic

variants can be generated for a single patient and compared with databases of presumably benign

variants present in an expanding number of healthy individuals. In a small family with more

than one affected individual, shared variants can be identified and subsequently screened in

unaffected family members. The list of variants can then be filtered on the basis of the proposed

mode of inheritance (e.g., the presence of two variants in a single gene for an autosomal

recessive disorder). One recent review estimated that 234 novel rare disease genes have been

discovered by either whole-exome or whole genome sequencing (Boycott et al., 2014).

Whole-exome sequencing of the proband in the present case allowed the exclusion of exonic

mutations in other genes within the shared homozygous region, as well as the exclusion of

candidate genes throughout the genome. The ability to exclude the involvement of other

Page 166: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

143

candidate genes increased our confidence in the involvement of the SLC18A2 variant in the

disease.

Awareness of the pediatric neurotransmitter disorders has increased in the clinical community,

but the majority of neurologists who may encounter such a patient will not have prior experience

with the phenotypic breadth of this group of rare disorders. The identification of putative

causative variants by whole genome or whole-exome sequencing—if not conclusive—can focus

the search for related cases in the literature, and thereby aid differential diagnosis. For example,

one treatable pediatric neurotransmitter disorder, sepiapterin reductase deficiency (see Chapter 1,

Section 1.4.2)—which is very severe when untreated—was recently diagnosed by whole genome

sequencing in a pair of siblings who had long remained undiagnosed and therefore untreated

because of the common difficulties in precise diagnosis of rare diseases. Diagnosis allowed

treatment and recovery of these children (Bainbridge et al., 2011).

Whereas the clinical utility of genomic diagnosis has been illustrated here, many limitations

remain. Sequence data is high resolution, and therefore it is necessary to filter and interpret a

large dataset to derive a clinically meaningful subset. This would be beyond the expertise of

many clinicians. There will therefore need to be an infrastructure in place before whole-exome

sequencing and data interpretation can become routine, including clinical geneticists who receive

specific training and specialization in the interpretation of next-generation sequencing data, and

continuous improvement to bioinformatic methods for filtering variants. Notably, a Canadian

research consortium (Finding of Rare Disease Genes [FORGE] Canada) providing such

centralized infrastructure recently reported the identification of disease-causing variants in 146

of 264 disorders under study (Beaulieu et al., 2014).

Page 167: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

144

Additionally, the success of the technique would vary with the inferred mode of inheritance. In

the present case, an autosomal recessive mode of inheritance in a consanguineous family

narrowed the search to homozygous variants. Without clear consanguinity, compound

heterozygous mutations in a single gene would be expected. A dominant mode of inheritance, in

contrast, would require examining an extensive list of heterozygous variants, which would be

facilitated by parallel sequencing of both parents and available siblings. De novo dominant

mutations, however, may be more straightforward to identify with the provision of parental

genome information. In both cases, genetic diagnosis would necessitate the availability of

family members, their consent to provide their genomes, and would incur additional costs. This

also produces a unique challenge in seeking reimbursement, with respect to defining the minimal

number of patient and family genomes required for a particular diagnostic application.

Whereas whole-exome sequencing is suitable for the discovery of protein-coding variants, other

genetic mechanisms are increasingly recognized to have phenotypic consequences, such as

splicing variants, promoter mutations, and epigenetic modifications. Mutations outside the

exome may be better addressed as the cost of whole-genome sequencing decreases and becomes

competitive with the cost of whole-exome sequencing. However, the availability of predictive

bioinformatics tools with sufficient confidence to identify disease-causing variants with

regulatory significance will need to be developed before this becomes feasible in a clinical,

rather than research, setting.

An additional challenge inherent in using whole-exome or whole-genome methods for clinical

diagnosis derives from the vast quantity of data generated by each individual test. Multiple

testing of this magnitude increases the number of incidental findings, making it difficult to

identify true causative mutations. Attributing confidence to identified variants will involve the

Page 168: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

145

incorporation of functional data, through understanding of the underlying pathophysiology, that

of related diseases, or metabolic data derived through other clinical investigations or

metabolomic screening performed in conjunction with genomics. Additional ethical challenges

may result from the identification of incidental findings with clinical significance that are

unrelated to the reason for testing. For these reasons, initial analyses based on disease-targeted

subsets of data may have a role in clinical utilization of next-generation sequencing.

In summary, although cost and time are no longer prohibitive factors to the clinical use of

genomics for the diagnosis of rare diseases in individual patients and small kindreds, significant

challenges remain in the implementation. However, the use of next-generation sequencing

technologies allow a large volume of information to be brought to bear on diagnostic challenges

that were previously not amenable to investigation by a uniform methodology.

4.1.4 Knowledge Translation Challenges and Strategies for PND Diagnosis

There are nine pediatric neurotransmitter disorders among more than 80 identified inborn errors

of metabolism with neurological correlates (van Karnebeek and Stockler, 2012). As with

VMAT2 deficiency, many of these diseases are readily treatable with already available

medications, diet, or vitamin or co-factor supplementation. The challenge lies in accurate and

timely diagnosis. Individual disorders are exceedingly rare, and the rate of discovery of these

disorders has increased significantly with the advance of genomic technologies to identify

disease-causing mutations in single families. The volume of data, combined with the rate of

increase of the dataset and the potential relevance of extremely recent data, places a significant

information burden on individual clinicians who may encounter such patients infrequently in

their career. Given the rarity of individual disorders, this situation is no less challenging for

specialists. The huge potential impact of a correct diagnosis on patient outcome emphasizes the

Page 169: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

146

importance of improving the ability of clinicians to access the appropriate information as a

standard part of clinical practice.

The publication of research findings in peer-reviewed literature is an inefficient and ineffective

strategy for the communication of relevant information to the primary users of the information.

Although clinicians are familiar with the language and presentation of research findings in the

literature and many have relevant journal subscriptions, the information is not organized in a

consistent and meaningful way. Database searches, such as PubMed, are often incomplete

because they rely on keyword searches, and consistency in the presentation of concepts is not

ensured. Linkages between related studies on the basis of citations are useful, but biased by the

perspectives of individual authors. As a result of these factors, information presented in the

scientific literature is not properly synthesized within a broader body of knowledge. The mere

availability of data to potential knowledge users is not sufficient to ensure its application in

practice. For information with such critical value to patients’ lives, the traditional avenues for

dissemination of research findings are no longer acceptable.

The Canadian Institutes of Health Research (CIHR) define knowledge translation as “a dynamic

and iterative process that includes synthesis, dissemination, exchange, and ethically-sound

application of knowledge to improve the health of Canadians, provide more effective health

services and products, and strengthen the health care system (CIHR, 2013).” Lavis provides a

framework for knowledge translation in the form of five questions: “What should be transferred

to decision makers?”, “To whom should research knowledge be transferred?”, “By whom should

research knowledge be transferred?”, “How should research knowledge be transferred?”, and

“With what effect should research knowledge be transferred?” (Lavis et al., 2003). This section

aims to discuss these elements in the context of improving the timely diagnosis of rare diseases.

Page 170: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

147

The challenge for PNDs and inborn errors of metabolism—and more broadly, all rare diseases—

is the synthesis of disparate and rapidly expanding information in a manner that can be directly

used by clinicians. Van Karnebeek and colleagues produced a synthesis of information for

inborn errors of metabolism in the form of a systematic literature review of inborn errors of

metabolism that present with intellectual disability as a major feature (van Karnebeek and

Stockler, 2012). In this manner, they arranged a vast collection of information (features and

treatment of 81 separate disorders) around a single unifying principle—the clinical presentation

of intellectual disability. In doing so, they presented the information in a context relevant to its

use.

Beyond synthesis, the manner of delivery of information is key. Van Karnebeek et al. further

derived a two-step diagnostic protocol to identify underlying inborn errors of metabolism

presenting with intellectual disability. The protocol was implemented through a web and mobile

app, Treatable-ID (http://www.treatable-id.org/) (van Karnebeek et al., 2012). Information on

the 81 diseases is presented in multiple ways: biochemical categories, neurologic and non-

neurologic signs and symptoms, diagnostic investigations, therapies, effects on outcomes, and

available evidence. A disease page for each condition connects clinicians to online resources and

primary literature. In this way, the information synthesized by the systematic review is made

intuitively navigable in a real-life clinical context. The use of web and mobile technology makes

the information broadly accessible and allows for updates to be made at the pace of discovery.

This is a model of the approach to be applied more broadly for clusters of rare diseases. Tools

should be developed that arrange information about rare conditions around their presentation,

providing consolidated diagnostic strategies and treatment information. In the case of VMAT2

deficiency and other PNDs, the primary presentation is movement disorder rather than

Page 171: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

148

intellectual disability (although there is overlap), and a focus may be placed on motor features,

with respect to the phenotypic spectrum of PNDs and age-specific presentation. Because

movement disorder is a highly specialized field, physician education materials (diagrams and

video) may be integrated into the app to enable them to better classify the relevant disease

features. Finally, it is key that any diagnostic app provide links to primary information sources

and contact information for affiliated clinicians and investigators to promote collaboration. A

long-term vision of an integrated diagnostic tool, rather than a collection of independent apps,

should be pursued for use in regular clinical practice. Furthermore, the adoption of this

technology should be aggressively advertised through medical associations, international

meetings, and physician education programs.

Beyond improving the resources available to physicians, consideration should be given to

developing a framework for presenting analogous information in a publicly accessible manner.

Information accessibility in both format and language fosters patient and caregiver agency in the

diagnostic process; providing a framework for information curation and presentation may better

focus the existing epidemic of internet self-diagnosis. In recognizing the multiple demands on a

clinician’s time, the availability of well-organized rare disease information to patients (and their

families and caregivers) can leverage the motivation of these stakeholders to explore potential

diagnoses. In conjunction with the clinical judgment of the physician through targeted

discussions, this would reflect an evolution toward a collaborative model of rare disease

diagnosis in which information may be more efficiently utilized in the clinical setting through the

involvement of multiple stakeholders in the diagnostic process, made possible through effective

knowledge translation.

Page 172: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

149

5 Concluding Statement This thesis described a new autosomal recessive pediatric neurotransmitter disorder discovered

in eight cousins of a consanguineous Bedouin family in Saudi Arabia, and identified its causative

mutation in the vesicular monoamine transporter 2 (VMAT2; SLC18A2). Biochemical

confirmation of loss of function underscored the importance of vesicular transport in monoamine

homeostasis, extending the spectrum of known pediatric neurotransmitter diseases and providing

the first demonstration of mutation in VMAT2 causing a clinical phenotype. Unremarkable CSF

monoamine metabolite findings demonstrate the inability of this single test to diagnose all

monoamine deficiencies, and prompt reconsideration of the diagnostic strategy.

For pediatric neurotransmitter disorders and other inborn errors of metabolism, precise

and timely diagnosis leads to significantly better patient outcomes with the availability of simple

and effective treatments for many. The rapid identification of causative mutations in single

families with previously undiagnosed rare genetic disease is now possible with the advancement

of next-generation sequencing technologies for whole-genome and whole-exome sequencing.

Diagnostic challenges in genomic data interpretation and dissemination of research findings

remain to be addressed.

Page 173: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

150

References ABECASIS, G. R., ALTSHULER, D., AUTON, A., BROOKS, L. D., DURBIN, R. M., GIBBS,

R. A., HURLES, M. E. & MCVEAN, G. A. 2010. A map of human genome variation from population-scale sequencing. Nature, 467, 1061-73.

ABECASIS, G. R., CHERNY, S. S., COOKSON, W. O. & CARDON, L. R. 2002. Merlin--rapid analysis of dense genetic maps using sparse gene flow trees. Nat Genet, 30, 97-101.

ADZHUBEI, I. A., SCHMIDT, S., PESHKIN, L., RAMENSKY, V. E., GERASIMOVA, A., BORK, P., KONDRASHOV, A. S. & SUNYAEV, S. R. 2010. A method and server for predicting damaging missense mutations. Nat Methods, 7, 248-9.

ARAWAKA, S., WADA, M., GOTO, S., KARUBE, H., SAKAMOTO, M., REN, C. H., KOYAMA, S., NAGASAWA, H., KIMURA, H., KAWANAMI, T., KURITA, K., TAJIMA, K., DAIMON, M., BABA, M., KIDO, T., SAINO, S., GOTO, K., ASAO, H., KITANAKA, C., TAKASHITA, E., HONGO, S., NAKAMURA, T., KAYAMA, T., SUZUKI, Y., KOBAYASHI, K., KATAGIRI, T., KUROKAWA, K., KURIMURA, M., TOYOSHIMA, I., NIIZATO, K., TSUCHIYA, K., IWATSUBO, T., MURAMATSU, M., MATSUMINE, H. & KATO, T. 2006. The role of G-protein-coupled receptor kinase 5 in pathogenesis of sporadic Parkinson's disease. J Neurosci, 26, 9227-38.

ARRABAL, L., TERESA, L., SANCHEZ-ALCUDIA, R., CASTRO, M., MEDRANO, C., GUTIERREZ-SOLANA, L., ROLDAN, S., ORMAZABAL, A., PEREZ-CERDA, C., MERINERO, B., PEREZ, B., ARTUCH, R., UGARTE, M. & DESVIAT, L. R. 2011. Genotype-phenotype correlations in sepiapterin reductase deficiency. A splicing defect accounts for a new phenotypic variant. Neurogenetics, 12, 183-91.

BAINBRIDGE, M. N., WISZNIEWSKI, W., MURDOCK, D. R., FRIEDMAN, J., GONZAGA-JAUREGUI, C., NEWSHAM, I., REID, J. G., FINK, J. K., MORGAN, M. B., GINGRAS, M. C., MUZNY, D. M., HOANG, L. D., YOUSAF, S., LUPSKI, J. R. & GIBBS, R. A. 2011. Whole-genome sequencing for optimized patient management. Sci Transl Med, 3, 87re3.

BANDMANN, O., GOERTZ, M., ZSCHOCKE, J., DEUSCHL, G., JOST, W., HEFTER, H., MULLER, U., ZOFEL, P., HOFFMANN, G. & OERTEL, W. 2003. The phenylalanine loading test in the differential diagnosis of dystonia. Neurology, 60, 700-2.

BARONE, P., ANTONINI, A., COLOSIMO, C., MARCONI, R., MORGANTE, L., AVARELLO, T. P., BOTTACCHI, E., CANNAS, A., CERAVOLO, G., CERAVOLO, R., CICARELLI, G., GAGLIO, R. M., GIGLIA, R. M., IEMOLO, F., MANFREDI, M., MECO, G., NICOLETTI, A., PEDERZOLI, M., PETRONE, A., PISANI, A., PONTIERI, F. E., QUATRALE, R., RAMAT, S., SCALA, R., VOLPE, G., ZAPPULLA, S., BENTIVOGLIO, A. R., STOCCHI, F., TRIANNI, G. & DOTTO, P. D. 2009. The PRIAMO study: A multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson's disease. Mov Disord, 24, 1641-9.

Page 174: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

151

BEAULIEU, C. L., MAJEWSKI, J., SCHWARTZENTRUBER, J., SAMUELS, M. E., FERNANDEZ, B. A., BERNIER, F. P., BRUDNO, M., KNOPPERS, B., MARCADIER, J., DYMENT, D., ADAM, S., BULMAN, D. E., JONES, S. J., AVARD, D., NGUYEN, M. T., ROUSSEAU, F., MARSHALL, C., WINTLE, R. F., SHEN, Y., SCHERER, S. W., FRIEDMAN, J. M., MICHAUD, J. L. & BOYCOTT, K. M. 2014. FORGE Canada Consortium: outcomes of a 2-year national rare-disease gene-discovery project. Am J Hum Genet, 94, 809-17.

BEN-DOR, D. H., ZIMMERMAN, S., SEVER, J., ROZ, N., APTER, A., REHAVI, M. & WEIZMAN, A. 2007. Reduced platelet vesicular monoamine transporter density in Tourette's syndrome pediatric male patients. Eur Neuropsychopharmacol, 17, 523-6.

BERNSTEIN, A. I., STOUT, K. A. & MILLER, G. W. 2012. A fluorescent-based assay for live cell, spatially resolved assessment of vesicular monoamine transporter 2-mediated neurotransmitter transport. J Neurosci Methods, 209, 357-66.

BLACKSTONE, C. 2009. Infantile parkinsonism-dystonia: a dopamine "transportopathy". J Clin Invest, 119, 1455-8.

BLAU, N., HEIZMANN, C. W., SPERL, W., KORENKE, G. C., HOFFMANN, G. F., SMOOKER, P. M. & COTTON, R. G. 1992. Atypical (mild) forms of dihydropteridine reductase deficiency: neurochemical evaluation and mutation detection. Pediatr Res, 32, 726-30.

BOYCOTT, K. M., DYMENT, D. A., SAWYER, S. L., VANSTONE, M. R. & BEAULIEU, C. L. 2014. Identification of genes for childhood heritable diseases. Annu Rev Med, 65, 19-31.

BRAAK, H., DEL TREDICI, K., RUB, U., DE VOS, R. A., JANSEN STEUR, E. N. & BRAAK, E. 2003. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging, 24, 197-211.

BRANDL, C. J. & DEBER, C. M. 1986. Hypothesis about the function of membrane-buried proline residues in transport proteins. Proc Natl Acad Sci U S A, 83, 917-21.

BRASIL, S., VIECELLI, H. M., MEILI, D., RASSI, A., DESVIAT, L. R., PEREZ, B., UGARTE, M. & THONY, B. 2011. Pseudoexon exclusion by antisense therapy in 6-pyruvoyl-tetrahydropterin synthase deficiency. Hum Mutat, 32, 1019-27.

BRIGHINA, L., RIVA, C., BERTOLA, F., SARACCHI, E., FERMI, S., GOLDWURM, S. & FERRARESE, C. 2013. Analysis of vesicular monoamine transporter 2 polymorphisms in Parkinson's disease. Neurobiol Aging, 34, 1712 e9-13.

BRUN, L., NGU, L. H., KENG, W. T., CH'NG, G. S., CHOY, Y. S., HWU, W. L., LEE, W. T., WILLEMSEN, M. A., VERBEEK, M. M., WASSENBERG, T., REGAL, L., ORCESI, S., TONDUTI, D., ACCORSI, P., TESTARD, H., ABDENUR, J. E., TAY, S., ALLEN, G. F., HEALES, S., KERN, I., KATO, M., BURLINA, A., MANEGOLD, C., HOFFMANN, G. F. & BLAU, N. 2010. Clinical and biochemical features of aromatic L-amino acid decarboxylase deficiency. Neurology, 75, 64-71.

Page 175: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

152

BURMAN, J., TRAN, C. H., GLATT, C., FREIMER, N. B. & EDWARDS, R. H. 2004. The effect of rare human sequence variants on the function of vesicular monoamine transporter 2. Pharmacogenetics, 14, 587-94.

CAUDLE, W. M., RICHARDSON, J. R., WANG, M. Z., TAYLOR, T. N., GUILLOT, T. S., MCCORMACK, A. L., COLEBROOKE, R. E., DI MONTE, D. A., EMSON, P. C. & MILLER, G. W. 2007. Reduced vesicular storage of dopamine causes progressive nigrostriatal neurodegeneration. J Neurosci, 27, 8138-48.

CHANG, Y. T., SHARMA, R., MARSH, J. L., MCPHERSON, J. D., BEDELL, J. A., KNUST, A., BRAUTIGAM, C., HOFFMANN, G. F. & HYLAND, K. 2004. Levodopa-responsive aromatic L-amino acid decarboxylase deficiency. Ann Neurol, 55, 435-8.

CHEN, C. X., HUANG, S. Y., ZHANG, L. & LIU, Y. J. 2005. Synaptophysin enhances the neuroprotection of VMAT2 in MPP+-induced toxicity in MN9D cells. Neurobiol Dis, 19, 419-26.

CHEN, X. J., KOVACEVIC, N., LOBAUGH, N. J., SLED, J. G., HENKELMAN, R. M. & HENDERSON, J. T. 2006. Neuroanatomical differences between mouse strains as shown by high-resolution 3D MRI. Neuroimage, 29, 99-105.

CHU, T. T. & LIU, Y. 2010. An integrated genomic analysis of gene-function correlation on schizophrenia susceptibility genes. J Hum Genet, 55, 285-92.

CIHR. 2013. More About Knowledge Translation at CIHR [Online]. Available: http://www.cihr-irsc.gc.ca/e/39033.html [Accessed 17 November 2013].

COLEBROOKE, R. E., CHAN, P. M., LYNCH, P. J., MOOSLEHNER, K. & EMSON, P. C. 2007. Differential gene expression in the striatum of mice with very low expression of the vesicular monoamine transporter type 2 gene. Brain Res, 1152, 10-6.

COLEBROOKE, R. E., HUMBY, T., LYNCH, P. J., MCGOWAN, D. P., XIA, J. & EMSON, P. C. 2006. Age-related decline in striatal dopamine content and motor performance occurs in the absence of nigral cell loss in a genetic mouse model of Parkinson's disease. Eur J Neurosci, 24, 2622-30.

CORDES, F. S., BRIGHT, J. N. & SANSOM, M. S. 2002. Proline-induced distortions of transmembrane helices. J Mol Biol, 323, 951-60.

CROFT, B. G., FORTIN, G. D., CORERA, A. T., EDWARDS, R. H., BEAUDET, A., TRUDEAU, L. E. & FON, E. A. 2005. Normal biogenesis and cycling of empty synaptic vesicles in dopamine neurons of vesicular monoamine transporter 2 knockout mice. Mol Biol Cell, 16, 306-15.

CUBELLS, J. F., RAYPORT, S., RAJENDRAN, G. & SULZER, D. 1994. Methamphetamine neurotoxicity involves vacuolation of endocytic organelles and dopamine-dependent intracellular oxidative stress. J Neurosci, 14, 2260-71.

Page 176: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

153

DANIELS, R. W., COLLINS, C. A., GELFAND, M. V., DANT, J., BROOKS, E. S., KRANTZ, D. E. & DIANTONIO, A. 2004. Increased expression of the Drosophila vesicular glutamate transporter leads to excess glutamate release and a compensatory decrease in quantal content. J Neurosci, 24, 10466-74.

DARCHEN, F., SCHERMAN, D. & HENRY, J. P. 1989. Reserpine binding to chromaffin granules suggests the existence of two conformations of the monoamine transporter. Biochemistry, 28, 1692-7.

DASILVA, J. N. & KILBOURN, M. R. 1992. In vivo binding of [11C]tetrabenazine to vesicular monoamine transporters in mouse brain. Life Sci, 51, 593-600.

DE LA FUENTE-FERNANDEZ, R., SOSSI, V., MCCORMICK, S., SCHULZER, M., RUTH, T. J. & STOESSL, A. J. 2009. Visualizing vesicular dopamine dynamics in Parkinson's disease. Synapse, 63, 713-6.

DUERR, J. S., FRISBY, D. L., GASKIN, J., DUKE, A., ASERMELY, K., HUDDLESTON, D., EIDEN, L. E. & RAND, J. B. 1999. The cat-1 gene of Caenorhabditis elegans encodes a vesicular monoamine transporter required for specific monoamine-dependent behaviors. J Neurosci, 19, 72-84.

EDWARDS, R. H. 2007. The neurotransmitter cycle and quantal size. Neuron, 55, 835-58.

EIDEN, L. E. & WEIHE, E. 2011. VMAT2: a dynamic regulator of brain monoaminergic neuronal function interacting with drugs of abuse. Ann N Y Acad Sci, 1216, 86-98.

EISENHOFER, G., KOPIN, I. J. & GOLDSTEIN, D. S. 2004. Catecholamine metabolism: a contemporary view with implications for physiology and medicine. Pharmacol Rev, 56, 331-49.

ERICKSON, J. D. & EIDEN, L. E. 1993. Functional identification and molecular cloning of a human brain vesicle monoamine transporter. J Neurochem, 61, 2314-7.

ERICKSON, J. D., SCHAFER, M. K., BONNER, T. I., EIDEN, L. E. & WEIHE, E. 1996. Distinct pharmacological properties and distribution in neurons and endocrine cells of two isoforms of the human vesicular monoamine transporter. Proc Natl Acad Sci U S A, 93, 5166-71.

EYERMAN, D. J. & YAMAMOTO, B. K. 2005. Lobeline attenuates methamphetamine-induced changes in vesicular monoamine transporter 2 immunoreactivity and monoamine depletions in the striatum. J Pharmacol Exp Ther, 312, 160-9.

FEHR, C., SOMMERLAD, D., SANDER, T., ANGHELESCU, I., DAHMEN, N., SZEGEDI, A., MUELLER, C., ZILL, P., SOYKA, M. & PREUSS, U. W. 2013. Association of VMAT2 gene polymorphisms with alcohol dependence. J Neural Transm, 120, 1161-9.

FLECKENSTEIN, A. E., VOLZ, T. J., RIDDLE, E. L., GIBB, J. W. & HANSON, G. R. 2007. New insights into the mechanism of action of amphetamines. Annu Rev Pharmacol Toxicol, 47, 681-98.

Page 177: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

154

FON, E. A., POTHOS, E. N., SUN, B. C., KILLEEN, N., SULZER, D. & EDWARDS, R. H. 1997. Vesicular transport regulates monoamine storage and release but is not essential for amphetamine action. Neuron, 19, 1271-83.

FREIS, E. D. 1954. Mental depression in hypertensive patients treated for long periods with large doses of reserpine. N Engl J Med, 251, 1006-8.

FUKUI, M., RODRIGUIZ, R. M., ZHOU, J., JIANG, S. X., PHILLIPS, L. E., CARON, M. G. & WETSEL, W. C. 2007. Vmat2 heterozygous mutant mice display a depressive-like phenotype. J Neurosci, 27, 10520-9.

FUMAGALLI, F., GAINETDINOV, R. R., WANG, Y. M., VALENZANO, K. J., MILLER, G. W. & CARON, M. G. 1999. Increased methamphetamine neurotoxicity in heterozygous vesicular monoamine transporter 2 knock-out mice. J Neurosci, 19, 2424-31.

GAINETDINOV, R. R., FUMAGALLI, F., WANG, Y. M., JONES, S. R., LEVEY, A. I., MILLER, G. W. & CARON, M. G. 1998. Increased MPTP neurotoxicity in vesicular monoamine transporter 2 heterozygote knockout mice. J Neurochem, 70, 1973-8.

GLATT, C. E., DEYOUNG, J. A., DELGADO, S., SERVICE, S. K., GIACOMINI, K. M., EDWARDS, R. H., RISCH, N. & FREIMER, N. B. 2001. Screening a large reference sample to identify very low frequency sequence variants: comparisons between two genes. Nat Genet, 27, 435-8.

GLATT, C. E., WAHNER, A. D., WHITE, D. J., RUIZ-LINARES, A. & RITZ, B. 2006. Gain-of-function haplotypes in the vesicular monoamine transporter promoter are protective for Parkinson disease in women. Hum Mol Genet, 15, 299-305.

GRAHAM, D. G. 1978. Oxidative pathways for catecholamines in the genesis of neuromelanin and cytotoxic quinones. Mol Pharmacol, 14, 633-43.

GUEVARA-GUZMAN, R., EMSON, P. C. & KENDRICK, K. M. 1994. Modulation of in vivo striatal transmitter release by nitric oxide and cyclic GMP. J Neurochem, 62, 807-10.

GUO, J. T., CHEN, A. Q., KONG, Q., ZHU, H., MA, C. M. & QIN, C. 2008. Inhibition of vesicular monoamine transporter-2 activity in alpha-synuclein stably transfected SH-SY5Y cells. Cell Mol Neurobiol, 28, 35-47.

GUTIERREZ, B., ROSA, A., PAPIOL, S., ARRUFAT, F. J., CATALAN, R., SALGADO, P., PERALTA, V., CUESTA, M. J. & FANANAS, L. 2007. Identification of two risk haplotypes for schizophrenia and bipolar disorder in the synaptic vesicle monoamine transporter gene (SVMT). Am J Med Genet B Neuropsychiatr Genet, 144B, 502-7.

HALL, F. S., SORA, I. & UHL, G. R. 2003. Sex-dependent modulation of ethanol consumption in vesicular monoamine transporter 2 (VMAT2) and dopamine transporter (DAT) knockout mice. Neuropsychopharmacology, 28, 620-8.

Page 178: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

155

HASTINGS, T. G., LEWIS, D. A. & ZIGMOND, M. J. 1996. Role of oxidation in the neurotoxic effects of intrastriatal dopamine injections. Proc Natl Acad Sci U S A, 93, 1956-61.

HENRY, J. P., BOTTON, D., SAGNE, C., ISAMBERT, M. F., DESNOS, C., BLANCHARD, V., RAISMAN-VOZARI, R., KREJCI, E., MASSOULIE, J. & GASNIER, B. 1994. Biochemistry and molecular biology of the vesicular monoamine transporter from chromaffin granules. J Exp Biol, 196, 251-62.

HIRSCH, E. C., ORIEUX, G., MURIEL, M. P., FRANCOIS, C. & FEGER, J. 2003. Nondopaminergic neurons in Parkinson's disease. Adv Neurol, 91, 29-37.

HOWIE, S. R. 2011. Blood sample volumes in child health research: review of safe limits. Bull World Health Organ, 89, 46-53.

HU, G., HENKE, A., KARPOWICZ, R. J., JR., SONDERS, M. S., FARRIMOND, F., EDWARDS, R., SULZER, D. & SAMES, D. 2013. New Fluorescent Substrate Enables Quantitative and High-Throughput Examination of Vesicular Monoamine Transporter 2 (VMAT2). ACS Chem Biol.

HYLAND, K. 2008. Clinical utility of monoamine neurotransmitter metabolite analysis in cerebrospinal fluid. Clin Chem, 54, 633-41.

ICHINOSE, H., OHYE, T., TAKAHASHI, E., SEKI, N., HORI, T., SEGAWA, M., NOMURA, Y., ENDO, K., TANAKA, H., TSUJI, S. & ET AL. 1994. Hereditary progressive dystonia with marked diurnal fluctuation caused by mutations in the GTP cyclohydrolase I gene. Nat Genet, 8, 236-42.

INTERNATIONAL HAPMAP CONSORTIUM 2003. The International HapMap Project. Nature, 426, 789-96.

ITOKAWA, K., SORA, I., SCHINDLER, C. W., ITOKAWA, M., TAKAHASHI, N. & UHL, G. R. 1999. Heterozygous VMAT2 knockout mice display prolonged QT intervals: possible contributions to sudden death. Brain Res Mol Brain Res, 71, 354-7.

IWASA, H., KURABAYASHI, M., NAGAI, R., NAKAMURA, Y. & TANAKA, T. 2001. Multiple single-nucleotide polymorphisms (SNPs) in the Japanese population in six candidate genes for long QT syndrome. J Hum Genet, 46, 158-62.

JENNER, P. 2007. Oxidative stress and Parkinson's disease. Handb Clin Neurol, 83, 507-20.

KATZ, B. 1971. Quantal mechanism of neural transmitter release. Science, 173, 123-6.

KILBOURN, M. R., BUTCH, E. R., DESMOND, T., SHERMAN, P., HARRIS, P. E. & FREY, K. A. 2010. In vivo [11C]dihydrotetrabenazine binding in rat striatum: sensitivity to dopamine concentrations. Nucl Med Biol, 37, 3-8.

KLEIN, C. 2005. Movement disorders: classifications. J Inherit Metab Dis, 28, 425-39.

Page 179: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

156

KOZMINSKI, K. D., GUTMAN, D. A., DAVILA, V., SULZER, D. & EWING, A. G. 1998. Voltammetric and pharmacological characterization of dopamine release from single exocytotic events at rat pheochromocytoma (PC12) cells. Anal Chem, 70, 3123-30.

KURIAN, M. A., GISSEN, P., SMITH, M., HEALES, S., JR. & CLAYTON, P. T. 2011a. The monoamine neurotransmitter disorders: an expanding range of neurological syndromes. Lancet Neurol, 10, 721-33.

KURIAN, M. A., LI, Y., ZHEN, J., MEYER, E., HAI, N., CHRISTEN, H. J., HOFFMANN, G. F., JARDINE, P., VON MOERS, A., MORDEKAR, S. R., O'CALLAGHAN, F., WASSMER, E., WRAIGE, E., DIETRICH, C., LEWIS, T., HYLAND, K., HEALES, S., JR., SANGER, T., GISSEN, P., ASSMANN, B. E., REITH, M. E. & MAHER, E. R. 2011b. Clinical and molecular characterisation of hereditary dopamine transporter deficiency syndrome: an observational cohort and experimental study. Lancet Neurol, 10, 54-62.

KURIAN, M. A., ZHEN, J., CHENG, S. Y., LI, Y., MORDEKAR, S. R., JARDINE, P., MORGAN, N. V., MEYER, E., TEE, L., PASHA, S., WASSMER, E., HEALES, S. J., GISSEN, P., REITH, M. E. & MAHER, E. R. 2009. Homozygous loss-of-function mutations in the gene encoding the dopamine transporter are associated with infantile parkinsonism-dystonia. J Clin Invest, 119, 1595-603.

LANDER, E. S., LINTON, L. M., BIRREN, B., NUSBAUM, C., ZODY, M. C., BALDWIN, J., DEVON, K., DEWAR, K., DOYLE, M., FITZHUGH, W., FUNKE, R., GAGE, D., HARRIS, K., HEAFORD, A., HOWLAND, J., KANN, L., LEHOCZKY, J., LEVINE, R., MCEWAN, P., MCKERNAN, K., MELDRIM, J., MESIROV, J. P., MIRANDA, C., MORRIS, W., NAYLOR, J., RAYMOND, C., ROSETTI, M., SANTOS, R., SHERIDAN, A., SOUGNEZ, C., STANGE-THOMANN, N., STOJANOVIC, N., SUBRAMANIAN, A., WYMAN, D., ROGERS, J., SULSTON, J., AINSCOUGH, R., BECK, S., BENTLEY, D., BURTON, J., CLEE, C., CARTER, N., COULSON, A., DEADMAN, R., DELOUKAS, P., DUNHAM, A., DUNHAM, I., DURBIN, R., FRENCH, L., GRAFHAM, D., GREGORY, S., HUBBARD, T., HUMPHRAY, S., HUNT, A., JONES, M., LLOYD, C., MCMURRAY, A., MATTHEWS, L., MERCER, S., MILNE, S., MULLIKIN, J. C., MUNGALL, A., PLUMB, R., ROSS, M., SHOWNKEEN, R., SIMS, S., WATERSTON, R. H., WILSON, R. K., HILLIER, L. W., MCPHERSON, J. D., MARRA, M. A., MARDIS, E. R., FULTON, L. A., CHINWALLA, A. T., PEPIN, K. H., GISH, W. R., CHISSOE, S. L., WENDL, M. C., DELEHAUNTY, K. D., MINER, T. L., DELEHAUNTY, A., KRAMER, J. B., COOK, L. L., FULTON, R. S., JOHNSON, D. L., MINX, P. J., CLIFTON, S. W., HAWKINS, T., BRANSCOMB, E., PREDKI, P., RICHARDSON, P., WENNING, S., SLEZAK, T., DOGGETT, N., CHENG, J. F., OLSEN, A., LUCAS, S., ELKIN, C., UBERBACHER, E., FRAZIER, M., et al. 2001. Initial sequencing and analysis of the human genome. Nature, 409, 860-921.

LAUFER, N., ZUCKER, M., HERMESH, H., MAROM, S., GILAD, R., NIR, V., WEIZMAN, A. & REHAVI, M. 2005. Platelet vesicular monoamine transporter density in untreated patients diagnosed with social phobia. Psychiatry Res, 136, 247-50.

Page 180: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

157

LAVIS, J. N., ROBERTSON, D., WOODSIDE, J. M., MCLEOD, C. B. & ABELSON, J. 2003. How can research organizations more effectively transfer research knowledge to decision makers? Milbank Q, 81, 221-48, 171-2.

LEBRAND, C., CASES, O., WEHRLE, R., BLAKELY, R. D., EDWARDS, R. H. & GASPAR, P. 1998. Transient developmental expression of monoamine transporters in the rodent forebrain. J Comp Neurol, 401, 506-24.

LEE, H. C., LAI, C. K., YAU, K. C., SIU, T. S., MAK, C. M., YUEN, Y. P., CHAN, K. Y., TAM, S., LAM, C. W. & CHAN, A. Y. 2012. Non-invasive urinary screening for aromatic L-amino acid decarboxylase deficiency in high-prevalence areas: a pilot study. Clin Chim Acta, 413, 126-30.

LEUZZI, V., CARDUCCI, C. A., CARDUCCI, C. L., POZZESSERE, S., BURLINA, A., CERONE, R., CONCOLINO, D., DONATI, M. A., FIORI, L., MELI, C., PONZONE, A., PORTA, F., STRISCIUGLIO, P., ANTONOZZI, I. & BLAU, N. 2010. Phenotypic variability, neurological outcome and genetics background of 6-pyruvoyl-tetrahydropterin synthase deficiency. Clin Genet, 77, 249-57.

LI, H., WAITES, C. L., STAAL, R. G., DOBRYY, Y., PARK, J., SULZER, D. L. & EDWARDS, R. H. 2005. Sorting of vesicular monoamine transporter 2 to the regulated secretory pathway confers the somatodendritic exocytosis of monoamines. Neuron, 48, 619-33.

LIN, Z., WALTHER, D., YU, X. Y., LI, S., DRGON, T. & UHL, G. R. 2005. SLC18A2 promoter haplotypes and identification of a novel protective factor against alcoholism. Hum Mol Genet, 14, 1393-404.

LIU, Y., PETER, D., ROGHANI, A., SCHULDINER, S., PRIVE, G. G., EISENBERG, D., BRECHA, N. & EDWARDS, R. H. 1992. A cDNA that suppresses MPP+ toxicity encodes a vesicular amine transporter. Cell, 70, 539-51.

LOHR, K. M., BERNSTEIN, A. I., STOUT, K. A., DUNN, A. R., LAZO, C. R., ALTER, S. P., WANG, M., LI, Y., FAN, X., HESS, E. J., YI, H., VECCHIO, L. M., GOLDSTEIN, D. S., GUILLOT, T. S., SALAHPOUR, A. & MILLER, G. W. 2014. Increased vesicular monoamine transporter enhances dopamine release and opposes Parkinson disease-related neurodegeneration in vivo. Proc Natl Acad Sci U S A.

LONGHI, R., RIVA, E., VALSASINA, R., PACCANELLI, S. & GIOVANNINI, M. 1985. Phenylketonuria due to dihydropteridine reductase deficiency: presentation of two cases. J Inherit Metab Dis, 8 Suppl 2, 97-8.

LONGO, N. 2009. Disorders of biopterin metabolism. J Inherit Metab Dis, 32, 333-42.

LOTHARIUS, J. & BRUNDIN, P. 2002. Impaired dopamine storage resulting from alpha-synuclein mutations may contribute to the pathogenesis of Parkinson's disease. Hum Mol Genet, 11, 2395-407.

Page 181: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

158

MARGER, M. D. & SAIER, M. H., JR. 1993. A major superfamily of transmembrane facilitators that catalyse uniport, symport and antiport. Trends Biochem Sci, 18, 13-20.

MEYER, L. R., ZWEIG, A. S., HINRICHS, A. S., KAROLCHIK, D., KUHN, R. M., WONG, M., SLOAN, C. A., ROSENBLOOM, K. R., ROE, G., RHEAD, B., RANEY, B. J., POHL, A., MALLADI, V. S., LI, C. H., LEE, B. T., LEARNED, K., KIRKUP, V., HSU, F., HEITNER, S., HARTE, R. A., HAEUSSLER, M., GURUVADOO, L., GOLDMAN, M., GIARDINE, B. M., FUJITA, P. A., DRESZER, T. R., DIEKHANS, M., CLINE, M. S., CLAWSON, H., BARBER, G. P., HAUSSLER, D. & KENT, W. J. 2013. The UCSC Genome Browser database: extensions and updates 2013. Nucleic Acids Res, 41, D64-9.

MHYRE, T. R., CHESLER, E. J., THIRUCHELVAM, M., LUNGU, C., CORY-SLECHTA, D. A., FRY, J. D. & RICHFIELD, E. K. 2005. Heritability, correlations and in silico mapping of locomotor behavior and neurochemistry in inbred strains of mice. Genes Brain Behav, 4, 209-28.

MILLER, R. 2008. A theory of the basal ganglia and their disorders, Boca Raton, CRC Press.

MILLS, P. B., SURTEES, R. A., CHAMPION, M. P., BEESLEY, C. E., DALTON, N., SCAMBLER, P. J., HEALES, S. J., BRIDDON, A., SCHEIMBERG, I., HOFFMANN, G. F., ZSCHOCKE, J. & CLAYTON, P. T. 2005. Neonatal epileptic encephalopathy caused by mutations in the PNPO gene encoding pyridox(am)ine 5'-phosphate oxidase. Hum Mol Genet, 14, 1077-86.

MOLERO-LUIS, M., SERRANO, M., ORMAZABAL, A., PEREZ-DUENAS, B., GARCIA-CAZORLA, A., PONS, R. & ARTUCH, R. 2013. Homovanillic acid in cerebrospinal fluid of 1388 children with neurological disorders. Dev Med Child Neurol.

MOOSLEHNER, K. A., CHAN, P. M., XU, W., LIU, L., SMADJA, C., HUMBY, T., ALLEN, N. D., WILKINSON, L. S. & EMSON, P. C. 2001. Mice with very low expression of the vesicular monoamine transporter 2 gene survive into adulthood: potential mouse model for parkinsonism. Mol Cell Biol, 21, 5321-31.

MOSHAROV, E. V., STAAL, R. G., BOVE, J., PROU, D., HANANIYA, A., MARKOV, D., POULSEN, N., LARSEN, K. E., MOORE, C. M., TROYER, M. D., EDWARDS, R. H., PRZEDBORSKI, S. & SULZER, D. 2006. Alpha-synuclein overexpression increases cytosolic catecholamine concentration. J Neurosci, 26, 9304-11.

NARBOUX-NEME, N., ANGENARD, G., MOSIENKO, V., KLEMPIN, F., PITYCHOUTIS, P. M., DENERIS, E., BADER, M., GIROS, B., ALENINA, N. & GASPAR, P. 2013. Postnatal growth defects in mice with constitutive depletion of central serotonin. ACS Chem Neurosci, 4, 171-81.

NARBOUX-NEME, N., SAGNE, C., DOLY, S., DIAZ, S. L., MARTIN, C. B., ANGENARD, G., MARTRES, M. P., GIROS, B., HAMON, M., LANFUMEY, L., GASPAR, P. & MONGEAU, R. 2011. Severe serotonin depletion after conditional deletion of the vesicular monoamine transporter 2 gene in serotonin neurons: neural and behavioral consequences. Neuropsychopharmacology, 36, 2538-50.

Page 182: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

159

NIRENBERG, M. J., CHAN, J., LIU, Y., EDWARDS, R. H. & PICKEL, V. M. 1997. Vesicular monoamine transporter-2: immunogold localization in striatal axons and terminals. Synapse, 26, 194-8.

OHARA, A., KASAHARA, Y., YAMAMOTO, H., HATA, H., KOBAYASHI, H., NUMACHI, Y., MIYOSHI, I., SCOTT HALL, F., UHL, G. R., IKEDA, K. & SORA, I. 2013. Exclusive expression of VMAT2 in noradrenergic neurons increases viability of homozygous VMAT2 knockout mice. Biochem Biophys Res Commun.

OPLADEN, T., OKUN, J. G., BURGARD, P., BLAU, N. & HOFFMANN, G. F. 2010. Phenylalanine loading in pediatric patients with dopa-responsive dystonia: revised test protocol and pediatric cutoff values. J Inherit Metab Dis, 33, 697-703.

OZAWA, A., YAMADA, M., SATOH, T., MONDEN, T., HASHIMOTO, K., KOHGA, H., HASHIBA, Y., SASAKI, T. & MORI, M. 2002. Transcriptional regulation of the human PRL-releasing peptide (PrRP) receptor gene by a dopamine 2 Receptor agonist: cloning and characterization of the human PrRP receptor gene and its promoter region. Mol Endocrinol, 16, 785-98.

PARSONS, S. M. 2000. Transport mechanisms in acetylcholine and monoamine storage. Faseb J, 14, 2423-34.

PARTRIDGE, A. W., THERIEN, A. G. & DEBER, C. M. 2004. Missense mutations in transmembrane domains of proteins: phenotypic propensity of polar residues for human disease. Proteins, 54, 648-56.

PATEL, J., MOOSLEHNER, K. A., CHAN, P. M., EMSON, P. C. & STAMFORD, J. A. 2003. Presynaptic control of striatal dopamine neurotransmission in adult vesicular monoamine transporter 2 (VMAT2) mutant mice. J Neurochem, 85, 898-910.

PEARL, P. L. 2013. Monoamine neurotransmitter deficiencies. Handb Clin Neurol, 113, 1819-25.

PETER, D., JIMENEZ, J., LIU, Y., KIM, J. & EDWARDS, R. H. 1994. The chromaffin granule and synaptic vesicle amine transporters differ in substrate recognition and sensitivity to inhibitors. J Biol Chem, 269, 7231-7.

PONS, R. 2009. The phenotypic spectrum of paediatric neurotransmitter diseases and infantile parkinsonism. J Inherit Metab Dis, 32, 321-32.

PONS, R., FORD, B., CHIRIBOGA, C. A., CLAYTON, P. T., HINTON, V., HYLAND, K., SHARMA, R. & DE VIVO, D. C. 2004. Aromatic L-amino acid decarboxylase deficiency: clinical features, treatment, and prognosis. Neurology, 62, 1058-65.

POTHOS, E. N., LARSEN, K. E., KRANTZ, D. E., LIU, Y., HAYCOCK, J. W., SETLIK, W., GERSHON, M. D., EDWARDS, R. H. & SULZER, D. 2000. Synaptic vesicle transporter expression regulates vesicle phenotype and quantal size. J Neurosci, 20, 7297-306.

Page 183: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

160

PURCELL, S., NEALE, B., TODD-BROWN, K., THOMAS, L., FERREIRA, M. A., BENDER, D., MALLER, J., SKLAR, P., DE BAKKER, P. I., DALY, M. J. & SHAM, P. C. 2007. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet, 81, 559-75.

RIDDLE, E. L., TOPHAM, M. K., HAYCOCK, J. W., HANSON, G. R. & FLECKENSTEIN, A. E. 2002. Differential trafficking of the vesicular monoamine transporter-2 by methamphetamine and cocaine. Eur J Pharmacol, 449, 71-4.

SALA, G., BRIGHINA, L., SARACCHI, E., FERMI, S., RIVA, C., CARROZZA, V., PIROVANO, M. & FERRARESE, C. 2010. Vesicular monoamine transporter 2 mRNA levels are reduced in platelets from patients with Parkinson's disease. J Neural Transm, 117, 1093-8.

SANDOVAL, V., RIDDLE, E. L., HANSON, G. R. & FLECKENSTEIN, A. E. 2002. Methylphenidate redistributes vesicular monoamine transporter-2: role of dopamine receptors. J Neurosci, 22, 8705-10.

SANDOVAL, V., RIDDLE, E. L., HANSON, G. R. & FLECKENSTEIN, A. E. 2003. Methylphenidate alters vesicular monoamine transport and prevents methamphetamine-induced dopaminergic deficits. J Pharmacol Exp Ther, 304, 1181-7.

SAVELIEVA, K. V., CAUDLE, W. M. & MILLER, G. W. 2006. Altered ethanol-associated behaviors in vesicular monoamine transporter heterozygote knockout mice. Alcohol, 40, 87-94.

SCHNEIDER, S. A. & BHATIA, K. P. 2010. Rare causes of dystonia parkinsonism. Curr Neurol Neurosci Rep, 10, 431-9.

SCHULDINER, S., SHIRVAN, A. & LINIAL, M. 1995. Vesicular neurotransmitter transporters: from bacteria to humans. Physiol Rev, 75, 369-92.

SCHWARTZ, K., HERMAN, I., PEER, G., WEIZMAN, A. & REHAVI, M. 2007. Elevated platelet vesicular monoamine transporter 2 in former heroin addicts maintained on methadone. J Neural Transm, 114, 281-4.

SCHWARTZ, K., IANCU, I., STRYJER, R., CHELBEN, J., KOTLER, M., WEIZMAN, A. & REHAVI, M. 2005a. Reduced platelet vesicular monoamine transporter density in smoking schizophrenia patients. Eur Neuropsychopharmacol, 15, 557-61.

SCHWARTZ, K., WEIZMAN, A. & REHAVI, M. 2005b. Decreased platelet vesicular monoamine transporter density in habitual smokers. Eur Neuropsychopharmacol, 15, 235-8.

SEGAWA, M. 2011. Hereditary progressive dystonia with marked diurnal fluctuation. Brain Dev, 33, 195-201.

SEGAWA, M., HOSAKA, A., MIYAGAWA, F., NOMURA, Y. & IMAI, H. 1976. Hereditary progressive dystonia with marked diurnal fluctuation. Adv Neurol, 14, 215-33.

Page 184: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

161

SENARD, J. M. & ROUET, P. 2006. Dopamine beta-hydroxylase deficiency. Orphanet J Rare Dis, 1, 7.

SIMONS, C. J. & VAN WINKEL, R. 2013. Intermediate phenotype analysis of patients, unaffected siblings, and healthy controls identifies VMAT2 as a candidate gene for psychotic disorder and neurocognition. Schizophr Bull, 39, 848-56.

SINGLETON, A. B., FARRER, M. J. & BONIFATI, V. 2013. The genetics of Parkinson's disease: progress and therapeutic implications. Mov Disord, 28, 14-23.

SOLOVIEFF, N., ROBERTS, A. L., RATANATHARATHORN, A., HALOOSIM, M., DE VIVO, I., KING, A. P., LIBERZON, I., AIELLO, A., UDDIN, M., WILDMAN, D. E., GALEA, S., SMOLLER, J. W., PURCELL, S. M. & KOENEN, K. C. 2014. Genetic Association Analysis of 300 Genes Identifies a Risk Haplotype in SLC18A2 for Post-traumatic Stress Disorder in Two Independent Samples. Neuropsychopharmacology, 39, 1872-9.

SORIANO, P. 1997. The PDGF alpha receptor is required for neural crest cell development and for normal patterning of the somites. Development, 124, 2691-700.

STANKOVSKI, L., ALVAREZ, C., OUIMET, T., VITALIS, T., EL-HACHIMI, K. H., PRICE, D., DENERIS, E., GASPAR, P. & CASES, O. 2007. Developmental cell death is enhanced in the cerebral cortex of mice lacking the brain vesicular monoamine transporter. J Neurosci, 27, 1315-24.

STEINER, J. A., ANGOT, E. & BRUNDIN, P. 2011. A deadly spread: cellular mechanisms of alpha-synuclein transfer. Cell Death Differ, 18, 1425-33.

SULZER, D. & POTHOS, E. N. 2000. Regulation of quantal size by presynaptic mechanisms. Rev Neurosci, 11, 159-212.

SULZER, D. & RAYPORT, S. 1990. Amphetamine and other psychostimulants reduce pH gradients in midbrain dopaminergic neurons and chromaffin granules: a mechanism of action. Neuron, 5, 797-808.

SULZER, D., SONDERS, M. S., POULSEN, N. W. & GALLI, A. 2005. Mechanisms of neurotransmitter release by amphetamines: a review. Prog Neurobiol, 75, 406-33.

SWOBODA, K. J., HYLAND, K., GOLDSTEIN, D. S., KUBAN, K. C., ARNOLD, L. A., HOLMES, C. S. & LEVY, H. L. 1999. Clinical and therapeutic observations in aromatic L-amino acid decarboxylase deficiency. Neurology, 53, 1205-11.

TAKAHASHI, N., MINER, L. L., SORA, I., UJIKE, H., REVAY, R. S., KOSTIC, V., JACKSON-LEWIS, V., PRZEDBORSKI, S. & UHL, G. R. 1997. VMAT2 knockout mice: heterozygotes display reduced amphetamine-conditioned reward, enhanced amphetamine locomotion, and enhanced MPTP toxicity. Proc Natl Acad Sci U S A, 94, 9938-43.

Page 185: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

162

TALKOWSKI, M. E., KIROV, G., BAMNE, M., GEORGIEVA, L., TORRES, G., MANSOUR, H., CHOWDARI, K. V., MILANOVA, V., WOOD, J., MCCLAIN, L., PRASAD, K., SHIRTS, B., ZHANG, J., O'DONOVAN, M. C., OWEN, M. J., DEVLIN, B. & NIMGAONKAR, V. L. 2008. A network of dopaminergic gene variations implicated as risk factors for schizophrenia. Hum Mol Genet, 17, 747-58.

TAYLOR, T. N., CAUDLE, W. M. & MILLER, G. W. 2011. VMAT2-Deficient Mice Display Nigral and Extranigral Pathology and Motor and Nonmotor Symptoms of Parkinson's Disease. Parkinsons Dis, 2011, 124165.

TAYLOR, T. N., CAUDLE, W. M., SHEPHERD, K. R., NOORIAN, A., JACKSON, C. R., IUVONE, P. M., WEINSHENKER, D., GREENE, J. G. & MILLER, G. W. 2009. Nonmotor symptoms of Parkinson's disease revealed in an animal model with reduced monoamine storage capacity. J Neurosci, 29, 8103-13.

THIRIOT, D. S. & RUOHO, A. E. 2001. Mutagenesis and derivatization of human vesicle monoamine transporter 2 (VMAT2) cysteines identifies transporter domains involved in tetrabenazine binding and substrate transport. J Biol Chem, 276, 27304-15.

THONY, B. & BLAU, N. 2006. Mutations in the BH4-metabolizing genes GTP cyclohydrolase I, 6-pyruvoyl-tetrahydropterin synthase, sepiapterin reductase, carbinolamine-4a-dehydratase, and dihydropteridine reductase. Hum Mutat, 27, 870-8.

THONY, B., NEUHEISER, F., KIERAT, L., BLASKOVICS, M., ARN, P. H., FERREIRA, P., REBRIN, I., AYLING, J. & BLAU, N. 1998. Hyperphenylalaninemia with high levels of 7-biopterin is associated with mutations in the PCBD gene encoding the bifunctional protein pterin-4a-carbinolamine dehydratase and transcriptional coactivator (DCoH). Am J Hum Genet, 62, 1302-11.

TONG, J., BOILEAU, I., FURUKAWA, Y., CHANG, L. J., WILSON, A. A., HOULE, S. & KISH, S. J. 2011. Distribution of vesicular monoamine transporter 2 protein in human brain: implications for brain imaging studies. J Cereb Blood Flow Metab, 31, 2065-75.

TOREN, P., REHAVI, M., LUSKI, A., ROZ, N., LAOR, N., LASK, M. & WEIZMAN, A. 2005. Decreased platelet vesicular monoamine transporter density in children and adolescents with attention deficit/hyperactivity disorder. Eur Neuropsychopharmacol, 15, 159-62.

VAN KARNEBEEK, C. D., HOUBEN, R. F., LAFEK, M., GIANNASI, W. & STOCKLER, S. 2012. The treatable intellectual disability APP http://www.treatable-id.org: a digital tool to enhance diagnosis & care for rare diseases. Orphanet J Rare Dis, 7, 47.

VAN KARNEBEEK, C. D. & STOCKLER, S. 2012. Treatable inborn errors of metabolism causing intellectual disability: a systematic literature review. Mol Genet Metab, 105, 368-81.

VOLZ, T. J., HANSON, G. R. & FLECKENSTEIN, A. E. 2006. Measurement of kinetically resolved vesicular dopamine uptake and efflux using rotating disk electrode voltammetry. J Neurosci Methods, 155, 109-15.

Page 186: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

163

WANG, Y. M., GAINETDINOV, R. R., FUMAGALLI, F., XU, F., JONES, S. R., BOCK, C. B., MILLER, G. W., WIGHTMAN, R. M. & CARON, M. G. 1997. Knockout of the vesicular monoamine transporter 2 gene results in neonatal death and supersensitivity to cocaine and amphetamine. Neuron, 19, 1285-96.

WILLEMSEN, M. A., VERBEEK, M. M., KAMSTEEG, E. J., DE RIJK-VAN ANDEL, J. F., AEBY, A., BLAU, N., BURLINA, A., DONATI, M. A., GEURTZ, B., GRATTAN-SMITH, P. J., HAEUSSLER, M., HOFFMANN, G. F., JUNG, H., DE KLERK, J. B., VAN DER KNAAP, M. S., KOK, F., LEUZZI, V., DE LONLAY, P., MEGARBANE, A., MONAGHAN, H., RENIER, W. O., RONDOT, P., RYAN, M. M., SEEGER, J., SMEITINK, J. A., STEENBERGEN-SPANJERS, G. C., WASSMER, E., WESCHKE, B., WIJBURG, F. A., WILCKEN, B., ZAFEIRIOU, D. I. & WEVERS, R. A. 2010. Tyrosine hydroxylase deficiency: a treatable disorder of brain catecholamine biosynthesis. Brain, 133, 1810-22.

WILLIAMS, K. A. & DEBER, C. M. 1991. Proline residues in transmembrane helices: structural or dynamic role? Biochemistry, 30, 8919-23.

WIMALASENA, K. 2011. Vesicular monoamine transporters: structure-function, pharmacology, and medicinal chemistry. Med Res Rev, 31, 483-519.

WITJAS, T., KAPHAN, E., AZULAY, J. P., BLIN, O., CECCALDI, M., POUGET, J., PONCET, M. & CHERIF, A. A. 2002. Nonmotor fluctuations in Parkinson's disease: frequent and disabling. Neurology, 59, 408-13.

WOODY, R. C., BREWSTER, M. A. & GLASIER, C. 1989. Progressive intracranial calcification in dihydropteridine reductase deficiency prior to folinic acid therapy. Neurology, 39, 673-5.

YAFFE, D., RADESTOCK, S., SHUSTER, Y., FORREST, L. R. & SCHULDINER, S. 2013. Identification of molecular hinge points mediating alternating access in the vesicular monoamine transporter VMAT2. Proc Natl Acad Sci U S A, 110, E1332-41.

YELIN, R., STEINER-MORDOCH, S., AROETI, B. & SCHULDINER, S. 1998. Glycosylation of a vesicular monoamine transporter: a mutation in a conserved proline residue affects the activity, glycosylation, and localization of the transporter. J Neurochem, 71, 2518-27.

ZALSMAN, G., ASLANOV-FARBSTEIN, D., REHAVI, M., ROZ, N., VERMEIREN, R., LAOR, N., WEIZMAN, A. & TOREN, P. 2011a. Platelet vesicular monoamine transporter 2 density in the disruptive behavior disorders. J Child Adolesc Psychopharmacol, 21, 341-4.

ZALSMAN, G., REHAVI, M., ROZ, N., LAOR, N., WEIZMAN, A. & TOREN, P. 2011b. Altered affinity of the platelet vesicular monoamine transporter 2 to dihydrotetrabenazine in children with major depression. J Neural Transm, 118, 1383-7.

ZUCKER, M., AVIV, A., SHELEF, A., WEIZMAN, A. & REHAVI, M. 2002a. Elevated platelet vesicular monoamine transporter density in untreated patients diagnosed with major depression. Psychiatry Res, 112, 251-6.

Page 187: Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease · 2015. 8. 13. · ii Mutation in VMAT2 Causing a Pediatric Neurotransmitter Disease Jennifer Rilstone Doctor of Philosophy

164

ZUCKER, M., VALEVSKI, A., WEIZMAN, A. & REHAVI, M. 2002b. Increased platelet vesicular monoamine transporter density in adult schizophrenia patients. Eur Neuropsychopharmacol, 12, 343-7.