CENTRE FOR MOTOR NEURONEDISEASE RESEARCH

Faculty of Medicine, Health and Human Sciences

3rd Macquarie Neurodegeneration Meeting

28-29 OCTOBER 2020 AN EVENT FOR AUSTRALIAN NEUROSCIENTISTS TO SHOWCASE THEIR RESEARCH AND TO STIMULATE CONVERSATION AND FOSTER COLLABORATION TO DEVELOP TREATMENTS FOR DISEASES INCLUDING MOTOR NEURON DISEASE, ALZHEIMER’S DISEASE, FRONTOTEMPORAL DEMENTIA, PARKINSON’S DISEASE AND OTHER

DEGENERATIVE BRAIN DISORDERS.

Our Sponsor

We are incredibly thankful to our sponsor for their support of the Macquarie Neurodegeneration Meeting Gain a new perspective on the nervous system and accelerate discovery. Explore 10x Genomics Solutions for neuroscience in this short video.

2 Our Sponsor

3 Our Sponsor

4 Welcome

The Macquarie Neurodegeneration Meeting is an annual event hosted by the Centre for Motor Neuron Disease Research, Macquarie University. The aim of this event is for Australian neuroscientists to showcase their research and to stimulate conversation and foster collaboration to develop treatments for diseases including motor neuron disease, Alzheimer’s disease, frontal temporal dementia, Parkinson’s disease and other degenerative brain disorders.

We welcome your involvement and hope the day provides inspiration and assists in fostering collaboration and connections in the neurodegeneration research community.

Yours Sincerely, The Conference Organising Committee

COMMITTEE MEMBERS

Centre for Motor Neuron Disease Research

Professor Julie Atkin Co-Director Professor Ian Blair Co-Director Christina Cassidy Centre Administrator Flora Cheng Research Assistant Dr Lyndal Henden Postdoctoral Research Fellow Research Dr Emily McCaan Postdoctoral Research Fellow Research Dr Prachi Mehta Research Officer Dr Sonam Parakh Postdoctoral Research Fellow

5 Program

Conference Program WEDNESDAY OCTOBER 28TH 2020

Opening

2:00 pm – 2:05 pm Webinar Opening Welcome remarks by Professor Julie Atkin Co-Director Centre for Motor Neuron Disease Research, Macquarie University

Session 1 Chair – Dr Emily Don

2:05 pm – 2:35 pm Professor Neil Cashman The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada TDP-43 Misfolding-Specific Antibodies in Treatment of TDP-43 Mediated Diseases (30min)

2:35 pm – 2:50pm Dr Sophia Luikinga The Florey Institute of Neuroscience and Mental Health Ambroxol as novel therapeutic intervention to improve motor function in a mouse model of ALS (15min)

2:50 pm – 3:20 pm Professor Gilles Guillemin Centre for Motor Neuron Disease Research, Macquarie University Overview of the involvement of the kynurenine pathway in neuroinflammatory diseases. Applications for prognosis and treatment (30min)

3: 20 pm – 3:35 pm Dr Emily McCann Centre for Motor Neuron Disease Research, Macquarie University Implicating novel genetic variation in ALS through the analysis of disease discordant monozygotic twins (15min)

Break

3:35 pm –4:20 pm Time to view Posters https://www.mq.edu.au/research/research-centres-groups-and- facilities/healthy-people/centres/macquarie-university-centre- for-motor-neuron-disease-research/conference

6 Program

Session 2 Chair – Dr Alison Hogan

4:20 pm – 4:35 pm Ms Sarah El-Wahsh Faculty of Medicine and Health, Speech Pathology Department, The University of Perspectives from the patient: communication symptoms, their impact, and speech pathology services in multiple sclerosis (15 min)

4:35 pm – 4:50 pm Dr Léonie Borne Systems Neuroscience Group, School of Psychology, Faculty of Science, University of Newcastle Modes of covariation between cortical anatomy and multi- domain cognitive function in healthy mid-life adults predict mild cognitive impairment and Alzheimer’s dementia (5min)

4:50 pm – 4.55 pm Dr Cindy Maurel Centre for Motor Neuron Disease Research, Macquarie University The unexplored effects of SUMOylation on TDP-43 aggregation and sub-cellular localization (5 min)

4:55 pm – 5:00 pm Dr Lyndal Henden Centre for Motor Neuron Disease Research, Macquarie University Identity by descent analysis links SOD1 familial and sporadic ALS cases (5 min)

Professor Flaviano Giorgini 5.00 pm – 5.30 pm Department of Genetics and Genome Biology, University of Leicester, UK The kynurenine pathway and Huntington’s disease: from mechanisms to therapeutics (30 min)

Professor Ammar Al-Chalabi 5.30 pm – 6.00 pm Kings College London, UK Understanding the causes of motor neuron disease (30 min)

End Session – Reminder Posters available to view overnight on our website https://www.mq.edu.au/research/research-centres-groups-and-facilities/healthy- people/centres/macquarie-university-centre-for-motor-neuron-disease- research/conference

7 Program

Conference Program THUSDAY OCTOBER 29TH 2020

Session 3 Chair – Dr Shelley Forrest

9:00 am – 9:05 am Welcome by Professor Ian Blair Co-Director - Centre for Motor Neuron Disease Research, Macquarie University

9:05 am – 9:20 am Dr Rachel Atkinson from Wicking Dementia Research and Education Centre, University of New model approaches for understanding axon degeneration in ALS (15min)

9:20 am – 9:35am Dr Mouna Haidar from The Florey Institute of Neuroscience and Mental Health, University of Melbourne Modelling cortical hyperexcitability in amyotrophic lateral sclerosis using chemogenetics (15min)

9:35 am – 9: 50 am Ms Katherine Jane Robinson from Centre for Motor Neuron Disease Research, Macquarie University Pathological expansion of ATXN3 alters memory and anxiety-like behaviours in a mouse model of Machado Joseph Disease/Spinocerebellar Ataxia Type 3 (15min)

9:50 am – 10:20 am Dr Arne Ittner from Dementia Research Centre, Macquarie University Protein kinases and tau phosphorylation in memory (30min)

10:20 am – 10:25 am Mrs Barbora Fulopova from University of Tasmania Differential patterns of cortical connectivity and beta amyloid deposition in APP/PS1 mice relative to ageing and the effects of midlife environmental enrichment (5min)

Break

10:25 am – 11:00 am Time to view Posters https://www.mq.edu.au/research/research-centres-groups-and- facilities/healthy-people/centres/macquarie-university-centre- for-motor-neuron-disease-research/conference

8 Program

Session 4 Chair – Dr Jennifer Fifita

11:00 am – 11:30 am Professor Anna King from University of Tasmania Blood biomarkers of neurodegeneration (30min)

11:30 am – 11:45 am Dr Lisa Oyston from Brain and Mind Centre, Determining the pathogenicity of TBK1 missense variants in FTD and ALS (15min)

11:45 am – 11:50 am Ms Marina Ulanova from Centre for Healthy Brain Ageing, UNSW Magnetic nanoparticles as MRI contrast agents for the diagnosis of Alzheimer’s Disease (5min)

11:50 am – 11:55 am Associate Professor Alison Canty from Wicking Dementia Research and Education Centre, University of Tasmania In vivo imaging of injured cortical axons reveals a rapid onset form of Wallerian degeneration (5min)

11:55 am – 12:00 pm Mr Andres Vidal-Itriago from Centre for Motor Neuron Disease Research, Macquarie University The missing link: Investigating the physiological traits of microglia during neurodegeneration (5min)

12:00 pm – 12:30 pm Professor Colin L Masters Laurette Professor of Dementia Research from The Florey Institute, The University of Melbourne The molecular origins of Alzheimer’s disease: when does it start and what strategies for primary prevention? (30min)

Close of Presentations

12:30 pm – 12:35 pm Closing remarks by Professor Patrick McNeil Deputy Vice-Chancellor (Medicine and Health) and Executive Dean of Faculty of Medicine, Health and Human Science, Macquarie University

12:35 pm – 12:45 pm Prize Presentation

9 Invited Speakers

Professor Ammar Al-Chalabi

PROFESSOR OF NEUROLOGY King's College London

Ammar Al-Chalabi is professor of neurology and complex disease genetics at King’s College London, consultant neurologist at King’s College Hospital and Deputy Editor, Brain. His research focuses on understanding the causes and modifiers of motor neuron disease and how these can be used to find effective treatments.

Professor Neil Cashman

PROFESSOR OF MEDICINE University of British Columbia

Dr. Neil Cashman is Professor of Medicine at the University of British Columbia, where his basic research is focused on the role of protein misfolding in Alzheimer’s and Parkinson’s disease, and amyotrophic lateral sclerosis (ALS). He serves as a Director of the ALS Centre Clinic at G.F. Strong Hospital. He is a neurologist-neuroscientist- entrepreneur who holds leadership positions in these three career domains. Two central ideas inform his scientific and translational work: first, that neurodegenerative diseases are due to prion-like propagated protein misfolding, and second, that antibodies are well suited to specifically bind to misfolded proteins while sparing normal proteins from autoimmune attack. He originally applied these ideas to prion disease itself (Cashman et al, Cell 1990; Paramithiotis et al, Nat Med 2003). He has applied the prion concept to developing novel immunotherapies for amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD). For ALS, he discovered that misfolded Cu/Zn superoxide dismutase (SOD1) acquires the prion-like ability to template misfolding of healthy SOD1 molecules (Grad et al, PNAS 2011), and that prion-like transmission of SOD1 misfolding can be

10 Invited Speakers

propagated between cells by protein aggregates and exosomes (Grad et al, PNAS 2014). For AD, He discovered Aβ oligomer-specific epitopes; which will enable a safe and effective AD treatment (Silverman et al, ACS Chem Neurosci 2018, Gibbs et al Sci Rep 2019). He is the Scientific Founder of three Canadian biotech companies, including ProMIS Neurosciences, where he currently serves as Chief Scientific Officer. Distinctions and awards for his research discoveries include citation by the CIHR for a "Medical Milestone" in 2003 for identifying a prion-specific epitope, Canada Research Chair Tier-1 Awards in 2005 and 2011, Fellowship of the Canadian Academy of Health Sciences in 2008, and his tenure as Scientific Director of PrioNET Canada (2005-2012) a Canadian Network of Centres of Excellence.

Professor Flaviano Giorgini

PROFESSOR OF NEUROGENETICS DEPARTMENT OF GENETICS AND GENOME BIOLOGY- COLLEGE OF LIFE SCIENCES University of Leicester

Prof Giorgini is a native of Indiana (USA) and pursued a BSc degree in Biological Sciences (with an emphasis in Genetics) at Purdue University. He obtained a MA degree in Molecular Genetics at Washington University in Saint Louis and a PhD in Genetics at the University of Washington in Seattle where he studied germ cell biology in the mouse. His interest in neurodegeneration research began as a Senior Fellow in the Department of Pharmacology (University of Washington), where he employed genetics and genomics approaches in yeast, mammalian cells and mice to identify and study genetic modifiers of Huntington’s disease. His work during this time, combined with his background in genetics and model organism biology, formed the basis for his current research in neurogenetics at the University of Leicester, where his laboratory studies the mechanisms underlying Huntington’s, Parkinson’s and Alzheimer’s. He began as a Lecturer in the Department of Genetics in 2006, was promoted to a Readership in 2012, and has been Professor of Neurogenetics since 2015. During this time he has been funded by the Medical Research Council and several charitable foundations. Prof Giorgini is currently a member of the College of Experts for Parkinson’s UK, and has previously served as the Chair for the Scientific and Bioethics Advisory Committee of the European Huntington’s Disease Network (EHDN).

11 Invited Speakers

Professor Gilles Guillemin

CENTRE FOR MOTOR NEURON DISEASE RESEARCH Macquarie University

Professor Gilles Guillemin has been working in the fields of Neuroimmunology and tryptophan metabolism for more than 25 years. He is the Co-founder of the MND and Neurodegenerative Diseases Research Centre, The Co-founder of the Macquarie University Neurodegenerative Diseases Biobank and the Head of the Neuroinflammation group at Macquarie University. Prof Guillemin’s team is one of the world’s leading research groups working on the involvement of the tryptophan catabolism (via the kynurenine pathway) in human neurodegenerative diseases. His team has demonstrated the importance of the KP in multiple sclerosis, Alzheimer's disease and motor neuron disease, which not only has opened numerous very promising research directions but has significant diagnostic, prognostic and therapeutic potential. With his microbiologist/virologist background, Prof Guillemin has recently been involved in several COVID-19 projects and research on the involvement of microorganisms various neurological diseases (PANDIS). He is the President of the International Society for Tryptophan Research and Editor-in-Chief of the International Journal for Tryptophan Research. He has published more than 250 peer reviewed scientific articles

Dr Arne Ittner

DEMENTIA RESEARCH CENTRE Macquarie University Arne graduated in Molecular Biology and Biochemistry from Swiss Federal Institute of Technology Zurich (ETH Zurich), Switzerland, in 2006 and received his PhD from ETH Zurich in 2010 for work on signal transduction by protein kinases. Arne moved then to University of Sydney, , to work as post- doctoral fellow at the Brain and Mind Research Institute from 2011 to 2013. In 2013, he continued his post-doctoral work at University of (UNSW Sydney), where he started his own research team on signal transduction and phosphorylation events in neurons in 2017. Arne was a post- doctoral fellow with the Alzheimer’s Australia Dementia Research Foundation in 2016. Arne’s team is currently supported by ARC and NHMRC. Arne is a NHMRC Emerging Leadership (EL2) fellow.

12 Invited Speakers

Professor Anna King

WICKING DEMENTIA RESEARCH & EDUCATION CENTRE University of Tasmania

Professor Anna King is a neuroscientist, NHMRC boosting dementia research leadership fellow and Associate Director (research) at the Wicking Dementia Research and Education Centre at the University of Tasmania where she leads a team of researchers, technical staff and students. Prof King received training in molecular biology and biochemistry at Durham University (UK) and the (Australia), before completing her PhD in neuropathology of ALS at the University of Tasmania in 2008. Her research interests span cell to human studies and lie in understanding, detecting and preventing the adverse neuronal changes that result in the clinical symptoms of neurodegenerative disease with a focus on vulnerable structures such as the axon and synapse.

Professor Colin L Masters

LAUREATE PROFESSOR OF DEMENTIA RESEARCH - THE FLOREY INSTITUTE The University of Melbourne

Colin Masters has focused his career on research in Alzheimer's disease and other neurodegenerative diseases, including Creutzfeldt-Jakob disease. His work over the last 35 years is widely acknowledged as having had a major influence on Alzheimer’s disease research world-wide, particularly the collaborative studies conducted with Konrad Beyreuther in which they discovered the proteolytic neuronal origin of the Aβ amyloid protein which causes Alzheimer’s disease. This work has led to the continued development of diagnostics and therapeutic strategies. More recently, his focus has been on describing the natural history of Alzheimer’s disease as a necessary preparatory step for therapeutic disease modification. Professor Masters is a Laureate Professor of Dementia Research at the Florey Institute, University of Melbourne and a consultant at the Royal Melbourne Hospital. His achievements have been recognised by the receipt of many international awards.

13 Abstract Lists

Invited Speaker Abstracts

Understanding the causes of motor neuron Professor Ammar Al-Chalabi disease (30 min) 20 Kings College London, UK

Professor Neil Cashman TDP-43 Misfolding-Specific Antibodies in The Djavad Mowafaghian Centre for Brain Treatment of TDP-43 Mediated Diseases (30min) 21 Health, University of British Columbia, Canada

The kynurenine pathway and Huntington’s Professor Flaviano Giorgini disease: from mechanisms to therapeutics (30 min) Department of Genetics and Genome 22 Biology, University of Leicester, UK

Overview of the involvement of the kynurenine Professor Gilles Guillemin pathway in neuroinflammatory diseases. 23 Centre for Motor Neuron Disease Research, Applications for prognosis and treatment (30min) Macquarie University

Dr Arne Ittner Protein kinases and tau phosphorylation in 24 Dementia Research Centre, Macquarie memory (30min) University

Professor Anna King Blood biomarkers of neurodegeneration (30min) 25 University of Tasmania

Professor Colin L Masters The molecular origins of Alzheimer’s disease: when does it start and what strategies for primary Laurette Professor of Dementia Research 26 from The Florey Institute, The University of prevention? (30min) Melbourne

14 Abstract Lists

Abstracts (15 min)

Dr Rachel Atkinson Wicking New model approaches for understanding axon Dementia Research and Education degeneration in ALS (15min) Centre, University of Tasmania 28

Dr Léonie Borne Modes of covariation between cortical anatomy Systems Neuroscience Group, School of and multi-domain cognitive function in healthy Psychology, Faculty of Science, mid-life adults predict mild cognitive impairment 29 University of Newcastle and Alzheimer’s dementia (5min)

Ms Sarah El-Wahsh Perspectives from the patient: communication Faculty of Medicine and Health, Speech symptoms, their impact, and speech pathology Pathology Department, The University services in multiple sclerosis (15 min) 31 of Sydney

Dr Mouna Haidar Modelling cortical hyperexcitability in The Florey Institute of Neuroscience and amyotrophic lateral sclerosis using chemogenetics 32 Mental Health, University of Melbourne (15min)

Dr Sophia Luikinga Ambroxol as novel therapeutic intervention to The Florey Institute of Neuroscience and improve motor function in a mouse model of ALS 33 Mental Health (15min)

Dr Emily McCann Implicating novel genetic variation in ALS Centre for Motor Neuron Disease through the analysis of disease discordant 34 Research, Macquarie University monozygotic twins (15min)

Dr Lisa Oyston Determining the pathogenicity of TBK1 missense Brain and Mind Centre, University of variants in FTD and ALS (15min) 35 Sydney

Ms Katherine Jane Robinson Pathological expansion of ATXN3 alters memory and anxiety-like behaviours in a mouse model of Centre for Motor Neuron Disease Machado Joseph Disease/Spinocerebellar Ataxia 36 Research, Macquarie University Type 3 (15min)

15 Abstract Lists

Abstracts (5 min)

Associate Professor Alison Canty In vivo imaging of injured cortical axons reveals a Wicking Dementia Research and rapid onset form of Wallerian degeneration Education Centre, University of (5min) 38 Tasmania

Differential patterns of cortical connectivity and Mrs Barbora Fulopova beta amyloid deposition in APP/PS1 mice relative University of Tasmania to ageing and the effects of midlife environmental 39 enrichment (5min)

Dr Lyndal Henden Identity by descent analysis links SOD1 familial Centre for Motor Neuron Disease and sporadic ALS cases (5 min) 40 Research, Macquarie University

Dr Cindy Maurel The unexplored effects of SUMOylation on TDP- Centre for Motor Neuron Disease 43 aggregation and sub-cellular localization (5 41 Research, Macquarie University min)

Ms Marina Ulanova Magnetic nanoparticles as MRI contrast agents for Centre for Healthy Brain Ageing, UNSW the diagnosis of Alzheimer’s Disease (5min) 42

Mr Andres Vidal-Itriago Centre for The missing link: Investigating the physiological Motor Neuron Disease Research, traits of microglia during neurodegeneration Macquarie University (5min) 43

16 Abstract Lists

Posters

Empirical evidence supports an aetiological role Mr Amr Abdeen for changes in biometal pathways in parkinson’s 45 The University of Sydney disease

Ms Azin Amin Development of autophagy-inducing peptides as a The Florey Institute of Neuroscience & potential therapy for motor neurone disease 46 Mental Health

Miss Sandrine Chan Moi Fat Analysis of GLT8D1 and ARPP21 in Australian Centre for MND Research familial and sporadic MND cases 47

Neuropeptide Y suppresses hyperexcitability of Miss Courtney Clark cortical SOD1G93A mouse neurons in vitro. 48 Menzies Institute for Medical Research Medium dose chronic CBD treatment reverses Miss Madilyn Coles object recognition memory deficits of 49 Western Sydney University APPSwe/PS1ΔE9 transgenic female mice

Is there a role for ketogenic therapies in the Ms Lauren Dewsbury clinical management of neurodegenerative 50 NICM Health Research Institute disease? A systematic review.

Dr Emily Don Utilising molecular imaging to investigate protein Centre for MND Research aggregate formation in vivo 51

Amyotrophic lateral sclerosis associated mislocalisation of TDP-43 to the cytoplasm causes Mr Marcus Dyer cortical hyperexcitability and reduced excitatory 52 Menzies Institute Medical Research neurotransmission in the motor cortex.

Dr Shelley Forrest Coexisting Lewy body disease and clinical Dementia Research Centre parkinsonism in amyotrophic lateral sclerosis 53

Dr Sarah Furlong Macquarie University Neurodegenerative Disease Centre for MND Research Biobank 54

Miss Jasmin Galper Lipid profiles of clinical and pre-clinical The University of Sydney Parkinson’s disease 55

A high-throughput flow cytometry drug screen to Dr Nicholas Geraghty discover new treatments for motor neurone 56 The University of Wollongong disease.

Losing track of the heart: Impaired cardiac interoceptive accuracy in behavioural-variant Ms Jessica Hazelton frontotemporal dementia and Alzheimer’s disease 57 The University of Sydney in an international cohort.

Dr Alison Hogan SFPQ pathology is a feature of ALS patient central Centre for MND Research nervous system tissue 58

Ms Bahar Kavyani Investigation of the temporal development of Tau Centre for MND Research pathology in TAU58/2 transgenic mice 59

17 Abstract Lists Vitamin D intake enhances Vitamin D receptor Dr Pragya Komal expression in the striatum and rescues memory Birla Institute of Technology and and motor dysfunction in mouse model 60 Sciences Hyderabad Huntington’s disease

Cannabidiol (CBD) treatment improves spatial Dr Fabian Kreilaus memory in 14-month-old female TAU58/2 61 Western Sydney University transgenic mice

Miss Yi Ling Low The involvement of fatty acid-binding proteins in Monash University docosahexaenoic acid uptake in microglia 62

Dr Nirma Perera Cleaning house: monitoring the flux of cellular The Florey Institute of Neuroscience & housekeeping autophagy pathway in amyotrophic 63 Mental Health lateral sclerosis (ALS) mice

Cellular changes in the substantia nigra and Dr Asheeta Prasad subthalamic nucleus in Parkinson’s disease 64 The University of NSW following deep brain stimulation

Robust astrocytic leptin and leptin receptor Anishchal Pratap expression in the 5XFAD mouse model of 65 The University of Sydney Alzheimer’s disease.

Can localized stimulation of the motor cortex Ms Laura Reale through TMS paradigms rescue the disease 66 Menzies Institute for Medical Research phenotype in a familial mouse model of ALS?

Mr Monokesh Kumer Sen Histological and proteomic investigations in the The University of Western Sydney visual pathway of cuprizone-fed mice 67

Dr Stephanie Shepheard Urinary Neopterin as a Potential Biomarker for Flinders University Motor Neurone Disease 68

Dr Momo Vuyisich Viome - Systems Biology Platform Viome, Inc 69

Miss Sharlynn Shin Lin Wu Elucidating the potential pathogenicity of novel Centre for MND Research MND candidate variants in vitro 70

Dr Fatemeh Zanganeh Gene expression profiling identifies excitotoxic The Florey Institute of Neuroscience & mechanisms initiated very early in motor neurons 71 Mental Health of the SOD1G93A mouse model of ALS

18 Abstracts

INVITED SPEAKERS

19 Abstracts

PROFESSOR AMMAR AL-CHALABI Professor of Neurology King's College London [email protected]

Understanding the causes of motor neuron disease

Motor neuron disease is a term that in fact covers many different conditions, all with a similar disease trajectory of relentless and relatively rapid loss of upper and lower motor neurons, resulting in progressive paralysis and ultimately death from respiratory failure. Through advances in genetics, statistics and epidemiology, we are beginning to identify causes, with the subsequent possibility of targeted treatments. Gene variations with a large effect explain motor neuron disease in about 15% of affected people. Finding the cause in the remaining 85% or a common pathway amenable to treatment is vital.

20 Abstracts

PROFESSOR NEIL CASHMAN The Djavad Mowafaghian Centre for Brain Health - University of British Columbia - Canada [email protected]

TDP-43 Misfolding-Specific Antibodies in Treatment of TDP-43 Mediated Diseases

Neil Cashman1,2, Sarah Louadi1, Andrei Roman1, Ebrima Gibbs1, Anke Dijkstra3, Johanne Kaplan2 1. The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver British Columbia Canada 2. ProMIS Neurosciences, Toronto Ontario, CANADA 3. Amsterdam University Medical Center, Amsterdam, Netherlands

Misfolded, aggregated TDP-43 has been implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTLD) through direct toxicity, loss of function of nuclear TDP-43, induction of misfolding of other neuronal proteins, and prion-like, cell-to-cell propagation of disease. We generated antibodies selectively targeting the misfolded, pathogenic form of TDP-43 while sparing physiological forms of TDP-43 important for normal cell function. Mice and rabbits were immunized with an unfolded N-terminal domain (NTD) linear epitope predicted to become exposed in cytosolically mislocalized, aggregated TDP-43, but otherwise buried in natively folded TDP-43. Monoclonal antibodies (mAbs) clones displayed pM affinity for the NTD epitope by surface plasmon resonance. ICC showed mAb reactivity with cytoplasmic aggregates of DNLS-TDP-43 but not wild-type nuclear TDP-43. Antibodies did not recognize

TDP-43 in stress granules in HEK-293FT cells. IHC of ALS and FTLD CNS sections confirmed selective immunoreactivity of mAbs with pathogenic wild-typeTDP-43. In cell culture, mAbs inhibited transmission of misfolding TDP-43 from the conditioned medium of donor HEK293FT cells transfected with dNLS-TDP-43 to recipient cells. Flow cytometry of brain-derived extracellular vesicles revealed surface decoration by misfolded TDP-43. Intrabodies constructed from the mAbs demonstrated 30-50-% reduction of TDP-43 aggregates in cells transfected with dNLS-TDP-43 and the selected intrabody constructs. We conclude that misfolded TDP-43 are selective for pathogenic vs physiologically important forms of TDP-43. Such antibodies may be able to: 1) neutralize extracellular transmission of misfolded TDP-43; and 2) decrease pathological TDP-43 intracellularly mediated by the IgG1-FCR-TRIM-21 degradation pathway. Alternately, 3) recombinant intrabodies may be able to alleviate intracellular pathogenic TDP-43 when expressed through viral transduction of CNS cells.

21 Abstracts

PROFESSOR FLAVIANO GIORGINI Department of Genetics and Genome Biology - University of Leicester - UK [email protected]

The kynurenine pathway and Huntington’s disease: from mechanisms to therapeutics

My research team employs genetic and genomic technologies in model organisms to elucidate the mechanisms underlying neurodegeneration and provide insight into novel therapeutic approaches for treating neurodegenerative disorders. Using models of protein misfolding as a paradigm for these disorders, we have identified and characterised genetic modifiers of proteotoxicity as “windows” into the cellular mechanisms underlying pathogenesis, with a particular focus on Huntington’s disease – a fatal, inherited neurodegenerative disorder. Our studies in simple model organisms, including the baker’s yeast S. cerevisiae and the fruit fly D. melanogaster, coupled with studies in mammalian cells and mice have formed a “pipeline” for the identification, validation and characterisation of relevant genes and pathways. This research has led to novel insights into the pathogenic roles of several cellular mechanisms and pathways which modulate neurodegeneration, including studies exploring the kynurenine pathway of tryptophan degradation. My talk will focus on this pathway and our efforts to therapeutically target a key pathway regulator kynurenine 3- monooxygenase (KMO) in Huntington’s disease.

22 Abstracts

PROFESSOR GILLES GUILLEMIN Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Overview of the involvement of the kynurenine pathway in neuroinflammatory diseases. Applications for prognosis and treatment.

Gilles J. Guillemin and the Macquarie University Neuroinflammation Team Neuroinflammation research group, Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109 Australia. Email: [email protected]

The kynurenine pathway (KP) of tryptophan metabolism is one of the major regulatory mechanisms of the immune response. Activation of the KP is implicated in the pathogenesis of a wide range of neuroinflammatory diseases. Several pro-inflammatory mediators can activate indoleamine 2,3 dioxygenase (IDO-1) one of the first and regulatory enzymes of the KP. A prolonged activation of the KP leads to production and accumulation of several neuroactive metabolites including the potent excitotoxin quinolinic acid (QUIN). Every brain cell type appears to express differently the KP enzymes and producing different KP metabolites. Over the last decade, together with our collaborators, we have shown that the KP is activated and QUIN level increases in most of the major neurodegenerative diseases (amyotrophic lateral sclerosis, multiple sclerosis, Alzheimer’s disease, Parkinson’s disease…), neuropsychiatric disorders (suicidality, schizophrenia, autism…) and cancers (glioblastomas, breast cancers, liver cancers…). We demonstrated that some KP metabolites can be used as accurate biomarkers for the prognosis and the rate of progression of several diseases. Designing new, sensitive, accessible and low-cost tools is the critical next step to be able to use these markers in clinical, biological and hospital laboratories. The identification of these molecules involved in the neuropathogenesis of brain diseases has also allowed us to design new therapeutic approaches.

23 Abstracts

DR ARNE ITTNER Dementia Research Centre - Faculty of Health and Medical Sciences - Macquarie University [email protected]

Protein kinases and tau phosphorylation in memory

The microtubule-associated protein tau is prominently involved in the pathogenesis of Alzheimer’s disease (AD). Tau is a target of protein kinases and is progressively hyper-phosphorylated at multiple sites, contributing to the formation of neurofibrillary tangles and cognitive decline in AD. My team’s work builds on the discovery that a member of the p38 mitogen-activated protein (MAP) kinase family – p38γ – targets tau, thereby inhibiting toxic signals downstream of tau. This site- specific phosphorylation confers an unprecedented neuroprotective function of tau. We extend this work now to therapeutic concepts as well as to a broad approach to understand how site-specific and hyper-phosphorylation of tau are connected. Furthermore, we explore the physiologic role of tau phosphorylation by p38γ in memory. We believe that this work will provide a new understanding of the molecular basis of memory function involving protein kinases and the tau protein.

Biography: Arne graduated in Molecular Biology and Biochemistry from Swiss Federal Institute of Technology Zurich (ETH Zurich), Switzerland, in 2006 and received his PhD from ETH Zurich in 2010 for work on signal transduction by protein kinases. Arne moved then to University of Sydney, Australia, to work as post-doctoral fellow at the Brain and Mind Research Institute from 2011 to 2013. In 2013, he continued his post-doctoral work at University of New South Wales (UNSW Sydney), where he started his own research team on signal transduction and phosphorylation events in neurons in 2017. Arne was a post-doctoral fellow with the Alzheimer’s Australia Dementia Research Foundation in 2016. Arne’s team is currently supported by ARC and NHMRC. Arne is a NHMRC Emerging Leadership (EL2) fellow.

24 Abstracts

PROFESSOR ANNA KING University of Tasmania [email protected]

Blood biomarkers of neurodegeneration

Jessica Collins, Nina Daniels, Anissuzaman Chowdury, Jenna Ziebell, Yasmine Doust, Ellie Bucher, Sharn Perry, Rachel Atkinson, Aidan Bindoff, David Gell, Sarah Shigdar, Michael Breadmore, James Vickers, Anna King

Early detection of brain changes is critical to strategies to prevent the onset or delay the progression of neurodegenerative diseases. The brain is a difficult organ to monitor, due to its inaccessible location within the skull, therefore biomarkers are needed as surrogate markers of brain changes, in order to monitor brain health. Over the last few years there has been substantial progress in the development of fluid biomarkers for monitoring brain changes that are implicated in the development and progression of neurodegenerative disease. Blood biomarkers in particular may offer opportunities for wide-scale screening of brain health. In spite of this, these fluid biomarkers are not yet well characterized and their relationship to specific brain changes and neurodegeneration mechanisms is not clear. We are asking whether we can use blood biomarkers to precisely detect changes in the brain, such as synapse, axon and neuron loss. Taking a multi- disciplinary approach, we are characterizing how biomarkers change in ageing and how these changes correlate with cognition, in healthy individuals. We are using animal models to determine the relationship between biomarkers and pathology as well as mechanisms of neurodegeneration and are taking a biochemical approach to biosensing for detection of blood proteins at picomole levels for future diagnostic or prognostic devices. This presentation will focus on a key biomarker of neurodegeneration, neurofilament light chain.

25 Abstracts

PROFESSOR COLIN L MASTERS

Laurette Professor of Dementia Research The Florey Institute - The University of Melbourne [email protected]

The molecular origins of Alzheimer’s disease: when does it start and what strategies for primary prevention?

The etiology of Alzheimer’s disease (AD) is best understood through the deposition of Aβ-amyloid (Aβ). There are two basic forms of AD. The common (>95%) form is sporadic and is caused by the failure to clear Aβ (mean age at onset 80 years). The rare (< 5%) autosomal dominant familial form is caused by the over-production of Aβ42, also on a background of failure to clear (mean age at onset 45 years). In both forms, the kinetics of Aβ accumulation are similar, taking about 30 years to accumulate a total of approximately 7mg of Aβ. Thus, we estimate that sporadic AD starts about the age of 50 years and the autosomal dominant form starts about 15 years of age. The advent of validated biomarkers (PET/CSF Aβ and tau) now provides us with unprecedented opportunities for preclinical diagnosis, enabling the development of primary and secondary prevention strategies. Predictive algorithms utilizing age, biomarkers, polygenic and vascular risk scores are now being developed from longitudinal cohort studies to estimate times of onset and rates of cognitive decline. Applications of biomarker screens (blood, CSF, PET) to subjects who are about to cross the lower cutpoint threshold will define a population who may be suitable for primary prevention clinical trials.

Therapeutic targeting the Aβ pathway remains the principal strategy for delaying onset of AD. There are many molecular targets in this pathway, and no single one is likely to prove efficacious on its own. Therefore, a combination of strategies needs to be developed and applied.

26 Abstracts

15-MINUTE SPEAKERS

27 Abstracts

DR RACHEL ATKINSON

Wicking Dementia Research and Education Centre - University of Tasmania, Australia [email protected]

New model approaches for understanding axon degeneration in ALS

Rachel Atkinson1, James Bender1, Jacqueline Leung1, James Vickers1, Anna King1 1. Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, TAS, 7000

Axon degeneration is a key feature of amyotrophic lateral sclerosis (ALS), however the mechanisms which drive these cellular changes are unclear. Currently, appropriate models which recapitulate axon degeneration are lacking. We have developed a novel visual system model which allows introduction of genetically altered proteins, such as those observed in disease, or drugs to mimic disease processes and the effects of these to be studied in the retinal ganglion cell (RGC) bodies, axons and synapses. The first pathologic process investigated was mislocalisation of TDP-43, which occurs in 90% of ALS cases. TDP-43 with a defective nuclear localisation sequence (TDP-NLS) or wildtype TDP-43 (TDP-WT) were packaged into adeno-associated virus 2 (AAV2) and, along with PBS vehicle, were delivered intravitreally to C57Bl6 adult mice. After 3 months, tissue was harvested and analysis demonstrated that TDP-WT resulted in synaptic alterations in the retina, while TDP-NLS resulted in neurofilament alterations in the retina and optic nerve. Excitotoxicity, a pathologic process known to occur in ALS, was also examined by intravitreal injection of kainic acid (KA) in a separate cohort of C57Bl6 mice. Tissue analysis 7 days after treatment revealed significant axonal loss and alterations to axonal structure following KA treatment. We are currently using this model to determine the effects of SARM1 knockout, key mediator of Wallerian degeneration. This novel visual system model is a positive step forward for recapitulating disease processes in an in vivo setting, and allows for rapid screening of therapeutic compounds and examination of the effects at the cellular level.

28 Abstracts

DR LÉONIE BORNE

Systems Neuroscience Group - School of Psychology - Faculty of Science - University of Newcastle [email protected]

Modes of covariation between cortical anatomy and multi-domain cognitive function in healthy mid-life adults predict mild cognitive impairment and Alzheimer’s dementia

Léonie Borne1, Michelle K Lupton2, Philip Mosley2, Robert Adam3, Gail Robinson4, Michael Breakspear1 1. University of Newcastle, Callaghan, NSW, Australia; 2. QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; 3. University of , St Lucia, QLD, Australia; 4. Royal Brisbane & Women’sHospital / UQCCR, Herston, QLD, Australia

Neurodegeneration in Alzheimer's Disease (AD) commences decades before cognitive and functional impairment [1]. The detection of the first symptoms is compounded by the variability across healthy adults in cognitive function and brain anatomy. Here, we invert this problem and ask whether common patterns of brain-cognitive covariation in health predicts cognitive dysfunction and cortical atrophy in mild cognitive impairment (MCI) and AD.

As part of the Prospective Imaging Study of Ageing [2], neurocognition was assessed through a battery of in-person multidomain neurocognitive tests. T1-weighted MRI scans were analyzed using the Morphologist/Brainvisa pipeline [3], allowing the automatic sulcal width measurement of 64 sulci. We trained a multivariate linear model on common patterns of brain-cognitive covariation in 172 healthy adults (61.5 +/- 5.4 yrs, 46M) using partial least squares. Robustness of the ensuing modes was tested using resampling methods. We tested the ability of this model to predict unseen data from age-matched adults with MCI or AD recruited from memory clinics (16MCI, 22AD, 63.5 +/- 6.5 yrs, 16M).

We observed a single robust mode linking variation in cognitive function and multi-areal cortical sulci width (Figure 1). The model loaded heavily onto all major cognitive domains and encompassed all cortical regions. Although this mode was trained only on healthy adults, it disambiguates the clinical cohort with high accuracy (80.8%). Hence, common patterns of co- variations between brain anatomy and cognitive function in healthy adults may be exaggerated in MCI and AD in a manner that is only weakly correlated with age.

29 Abstracts

References: [1] J. C. Morris, “Early-stage and preclinical Alzheimer disease,” Alzheimer Dis. Assoc. Disord., vol. 19, no. 3, pp. 163–165, Sep. 2005, doi: 10.1097/01.wad.0000184005.22611.cc. [2] M. K. Lupton et al., “A prospective cohort study of prodromal Alzheimer′s disease: Prospective Imaging Study of Ageing: Genes, Brain and Behaviour (PISA),” medRxiv, p. 2020.05.04.20091140, Jun. 2020, doi: 10.1101/2020.05.04.20091140. [3] L. Borne, D. Rivière, M. Mancip, and J.-F. Mangin, “Automatic labeling of cortical sulci using patch- or CNN-based segmentation techniques combined with bottom-up geometric constraints,” Med. Image Anal., vol. 62, p. 101651, May 2020, doi: 10.1016/j.media.2020.101651.

30 Abstracts

MS SARAH EL-WAHSH

Speech Pathology Department - Faculty of Medicine and Health - The University of Sydney [email protected]

Perspectives from the patient: communication symptoms, their impact, and speech pathology services in multiple sclerosis

Sarah El-Wahsh1, Claire Layfield4, Hans Bogaardt1, Fiona Kumfor2,3, & Kirrie Ballard 1,3 1Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; 2School of Psychology, The University of Sydney, Sydney, NSW, Australia; 3Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia; 4 Faculty Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia E-mail address of presenter: [email protected] Background: Communication symptoms in multiple sclerosis (MS) are often overlooked. Persons with MS (PwMS) are experts in their condition and can play a valuable role in informing clinicians and researchers about their condition and the impact of its symptoms. Objective: To investigate the insider’s perspective on (a) communication symptoms, their impact on quality of life, and strategies used to manage these symptoms, and (b) whether PwMS with communication symptoms access speech pathology (SP) services. Methods: Two-hundred and sixty PwMS were recruited internationally and completed an online survey. A content analysis by two independent researchers was used to analyse open-ended questions. Descriptive statistics were used to report on quantitative data. Results: One-hundred and ninety-seven (75.8%) participants reported communication symptoms. Communication symptoms experienced by participants were categorised into four themes: language, social-communication, cognitive, and other spoken difficulties. These symptoms were identified to have impacts across four key domains: psychological wellbeing, interpersonal relationships, work activities, and tertiary studies and aspiration. Nine themes were identified indicating the range of strategies that PwMS employ to facilitate their communication: managing personal factors, communication strategies, language and cognitive activities, preparation, enhancing the communicative environment, alternative and augmentative communication, medication, physical activity, and self-disclosure. Only 11% (n=22) of participants who reported communication symptoms accessed SP services. Conclusion: PwMS can experience communication symptoms, which can impact on relationships, careers, study, and psychological wellbeing. Results suggest that PwMS employ a variety of strategies to help manage these symptoms; however, very few accessed SP services. There is a need to raise awareness of the role of SP in MS to include management of communication symptoms.

31 Abstracts

DR MOUNA HAIDAR

The Florey Institute of Neuroscience and Mental Health - University of Melbourne [email protected]

Modelling cortical hyperexcitability in amyotrophic lateral sclerosis using chemogenetics

Mouna Haidar1, Aida Viden1, Brittany Cuic1, Taide Wang1, Valeria Rytova1, Sophia Luikinga1, Bradley Turner1 1. Florey Institute of Neuroscience and Mental Health, Florey Department of Neuroscience and Mental Health, Melbourne, , Australia. Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no available treatment. In ALS, hyperexcitability of brain (upper motor neurons, UMNs) and spinal cord (lower motor neurons, LMNs) are linked to excitotoxicity and cell death. Both UMN and LMN hyperexcitability occurs early in ALS, however the sequence and contribution of motor cortex and spinal neuronal hyperexcitability and dysfunction to ALS remains contentious. Elucidating the neuroanatomical site of disease initiation is critical to understanding the processes leading to cell death in ALS. The primary aim of this study is to determine the contribution of UMNs in disease initiation and progression in ALS by modelling chronic cortical excitability using DREADDs. We stereotaxically injected a viral vector encoding the excitatory DREADD hM3Dq bilaterally into the motor cortex, and hM3Dq expressing mice were treated with clozapine-N-oxide (CNO) or saline. Motor coordination (Rotarod) and muscle force (grip strength) were tested. For histological analyses, mice were perfused, and brain, spinal cord and hind limb muscle tissue collected. We observed transduction of Layer V pyramidal neurons, and activation of hM3Dq expressing UMNs, relative to saline-treated controls. CNO-treated hM3Dq mice displayed motor deficits and neuropathology including; (i) abnormal hind limb posture (clasping), (ii) reduced Rotarod performance, (iii) reduced hind limb grip strength; (iv) and cytoplasmic phosphorylated TDP-43 aggregates (a hallmark of ALS) within LMNs and UMNs, relative to saline-treated controls. These data support a ‘dying-forward’ hypothesis in ALS and demonstrate the first exciting evidence of ALS-like symptoms in a novel mouse model recapitulating sporadic ALS.

32 Abstracts

DR SOPHIA LUIKINGA

The Florey Institute of Neuroscience and Mental Health [email protected]

Ambroxol as novel therapeutic intervention to improve motor function in a mouse model of ALS

Luikinga SJ1, Henriques A2,3, Ngo ST4, Loeffler JP2, Spedding M3, Rapasinghe T5 Turner BJ1 1. Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia [email protected] 2. University of Strasbourg, Strasbourg, France 3. Spedding Research Solutions SAS, Le Vesinet, France 4. Australian Institute for Bioengineering and Nanotechnology, University of Queensland 5. University of Melbourne, Melbourne, Australia Amyotrophic lateral sclerosis (ALS) is characterized by selective degeneration of motor neurons, with death occurring 3-5 years after diagnosis. Lipid metabolism dysfunction has been observed in ALS patients as well as familial mouse models. With 90 percent of ALS cases being sporadic, it is important to understand lipid metabolism dysfunction in sporadic ALS. Here we used a mouse with the human TDP-43Q331K mutation, representing key features of sporadic ALS and aim to improve motor functioning through manipulating glycosphingolipid metabolism. Ambroxol is a selective Glucocerebrosidase 2 (GBA2) inhibitor and in turn will improve lipid metabolism as shown in figure 1.

UGCG MN preservation Cer GlcCer GSL NMJ stability Lipid metabolism

GBA2

Ambroxol

We treated TDP-43Q331K mice with 3mM Ambroxol ad libitum in their drinking water from postnatal day (P) 60 – P300. Forced motor activity on a rotating rod was tested weekly, where gait analysis and voluntary locomotion was tested monthly. Neuromuscular junction integrity and lumbar spinal motor neurone count were analysed at P300. Lipidomic analysis of lumbar cord and gastrocnemius muscle was performed at P210. Here, we show a significant improvement of motor functioning, gait and voluntary locomotion of TDP-43Q331K mice after Ambroxol treatment. Furthermore, Ambroxol prevented motor neurone loss in the lumbar spinal cord, as well as denervation of the neuromuscular junction through preservation of gangliosides. Lipidomic analysis revealed significant glycosphingolipid changes in both the muscle and lumbar spinal cord of TDP43Q331K mice indicating sphingomyelin, ganglioside GM3 and Acylcarnitine as key role players in lipid dysfunction. Together, these results indicate Ambroxol as a potential novel therapeutic to improve symptoms of ALS.

33 Abstracts

DR EMILY MCCANN

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Implicating novel genetic variation in ALS through the analysis of disease discordant monozygotic twins

Emily P. McCann1, Natalie A. Twine1,2, Dennis C. Bauer2, Natalie Grima1, Jennifer A. Fifita1, Garth A. Nicholson1,3,4,5, Dominic B. Rowe1, Ian P. Blair1, Kelly L. Williams1

1. Macquarie University Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia; 2. Transformational Bioinformatics, Commonwealth Scientific and Industrial Research Organisation, Sydney, New South Wales, Australia; 3. Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, New South Wales, Australia; 4. , University of Sydney, Sydney, New South Wales, Australia; 5. Molecular Medicine Laboratory, Concord Hospital, Concord, New South Wales, Australia

Amyotrophic lateral sclerosis (ALS) is genetically and phenotypically heterogenous. Though onset typically occurs at ~50-60 years, this can range anywhere from 20-90 years of age. To-date, gene mutations remain the only known cause of ALS, yet despite heritability estimates of 40-60% for all forms of ALS, >90% of cases have an unknown genetic basis of disease. Using monozygotic twins discordant for ALS, we aim to uncover novel nucleotide level and/or structural variation contributing to the clinical manifestation of ALS. This includes ALS causal and/or risk variants in twin pairs where one twin is affected by sporadic ALS (n=2), as well as phenotypic modifier variants influencing age of disease onset in twin/triplet sets with a family history of ALS and carrying causal mutations in SOD1 (n=1), C9orf72 (n=1) or an unidentified ALS gene (n=1). By applying custom bioinformatics pipelines combining various genomic analysis tools to whole genome sequencing data, we have identified between 1-9 nucleotide level, and 59-120 structural putative somatic variants in each twin/triplet set. Wet-lab validation of these putative somatic variants is currently underway, and all those validated will be further screened through an additional >700 ALS cases to evaluate the extent of their contribution to ALS pathogenesis. The somatic variants implicated in ALS by this work will further implicate novel genes and molecular pathways in disease pathogenesis, which will act as the foundation for functional investigations to tease apart ALS disease mechanisms as well as for the development of therapeutics to delay or prevent the onset of ALS.

34 Abstracts

DR LISA OYSTON

Brain and Mind Centre, University of Sydney [email protected] Determining the pathogenicity of TBK1 missense variants in FTD and ALS

Lisa J. Oyston1, Lauren Boccanfuso1, Johnny Zhang1, Lauren Fitzpatrick1, Marianne Hallupp1, John B. Kwok1,2, Carol Dobson-Stone1,2 1. Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Camperdown, NSW 2006, Australia School of Medical Sciences, University of New South Wales, Kensington, NSW 2052, Australia Frontotemporal dementia (FTD) is one of the most common forms of presenile dementia. Increasing genetic evidence and clinical observations have linked FTD to amyotrophic lateral sclerosis (ALS), a rapidly progressive neurodegenerative disorder that causes muscle weakness and paralysis. Mutations in TANK-binding kinase 1 (TBK1) have been identified in 1-2% of FTD/ALS patients1. TBK1 has known roles in inflammation and autophagy and interacts with other FTD/ALS proteins such as optineurin (OPTN)2. To date, >90 TBK1 rare variants have been identified: >50% of these are missense variants of unknown significance (VUS)3. We investigated the functional effect of eight TBK1 VUS with in silico evidence of pathogenicity, together with known pathogenic and benign TBK1 variants. Using a dual luciferase assay to measure NF-κB activity and coimmunoprecipitation to assess OPTN binding, we demonstrate that several TBK1 missense variants have similar effects to TBK1 loss-of-function mutations. Mislocalisation of neuronal TDP- 43 from the nucleus to the cytoplasm is a key neuropathological feature of ~95% ALS and ~50% of FTD cases4, including those with TBK1 mutations5. Using a high-throughput, unbiased method for the in vitro quantification of cytoplasmic TDP-43 we also show several TBK1 missense variants increase cytoplasmic TDP-43. By identifying pathogenic missense variants in TBK1 we hope to identify key functions of TBK1 that are involved in the pathogenesis of FTD/ALS. References: 1. R. Cui, M. Tuo, P. Li, and C. Zhou, “Association between TBK1 mutations and risk of amyotrophic lateral sclerosis/frontotemporal dementia spectrum: a meta-analysis,” Neurol. Sci., vol. 39, no. 5, pp. 811–820, 2018. 2. J. A. Oakes, M. C. Davies, and M. O. Collins, “TBK1: a new player in ALS linking autophagy and neuroinflammation,” Mol. Brain, vol. 10, no. 1, pp. 1–10, 2017. 3. Y. A. Abramzon, P. Fratta, B. J. Traynor, and R. Chia, “The Overlapping Genetics of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia,” Front. Neurosci., vol. 14, no. February, pp. 1–10, 2020. 4. A. Prasad, V. Bharathi, V. Sivalingam, A. Girdhar, and B. K. Patel, “Molecular mechanisms of TDP-43 misfolding and pathology in amyotrophic lateral sclerosis,” Front. Mol. Neurosci., vol. 12, no. February, pp. 1–36, 2019. 5. A. Freischmidt et al., “Haploinsufficiency of TBK1 causes familial ALS and fronto- temporal dementia,” Nat. Neurosci., vol. 18, no. 5, pp. 631–636, 2015.

35 Abstracts

MS KATHERINE JANE ROBINSON

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Pathological expansion of ATXN3 alters memory and anxiety-like behaviours in a mouse model of Machado Joseph Disease/Spinocerebellar Ataxia Type 3

Katherine Jane Robinson, Luan Luu, Kristy Yuan, Angela S Laird 1. Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University

Machado Joseph Disease (MJD) is a fatal neurodegenerative disease resulting in motor impairments, progressive neurodegeneration and eventual death. MJD is caused by inheritance of an expanded trinucleotide repeat (CAG) within the ATXN3/MJD1 gene, encoding an expanded polyglutamine repeat region. MJD patients have also been reported to possess cognitive impairments, presenting with impaired memory, increased anxiety and depression. It is debated within clinical literature if these cognitive changes are a direct consequence of pathologically expanded ATXN3 or due to anticipation of declining health. Transgenic mice expressing human expanded ATXN3 perform significantly worse when in accelerated rotarod (p<0.001), grip strength (p<0.001) and balance beam testing (p<0.001) when compared to wildtype littermate controls. Further analysis of movement within an open field revealed decreased movement and decreased time spent in the centre zone (p<0.05). These findings were surprising, as anxiety-like behaviour has not previously been reported in animal models of MJD. These findings were confirmed by analysis of behaviour in an elevated plus maze, a second measure of anxiety-like behaviour. EPM findings revealed decreased exploration within the open arms in MJD mice (p=0.042) when compared to littermate controls, aligning with our open field findings. Findings from the novel object recognition task revealed suggested impaired visual memory in MJD animals, as mice were unable to distinguish between a familiar or novel stimulus. Future immunohistochemical experiments will shed light on if mutant ATXN3 drives neuropathology within the hippocampus and amygdala, areas of the brain functionally involved in memory and anxiety.

36 Abstracts

5-MINUTE SPEAKERS

37 Abstracts

ASSOCIATE PROFESSOR ALISON CANTY

Wicking Dementia Research and Education Centre - University of Tasmania. [email protected]

In vivo imaging of injured cortical axons reveals a rapid onset form of Wallerian degeneration

Alison Canty1, Johanna Jackson2, Lieven Huang3, Antonio Trabalza3, Cher Bass3, Graham Little3, Maria Tortora3, Shabana Khan3, Vincenzo De Paola3 4 1. Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia; 2. Dementia Research Institute at Imperial College, Department of Brain Sciences, Imperial College London, London W12 0NN, UK; 3. Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK; 4. Medical Research Council London Institute of Medical Sciences, London W12 0NN, UK.

Despite the widespread occurrence of axon and synaptic loss in the injured and diseased nervous system, the cellular and molecular mechanisms of these key degenerative processes remain incompletely understood. Wallerian degeneration (WD) is a tightly regulated form of axon loss after injury, intensively studied in large myelinated fibre tracts of the spinal cord, optic nerve and peripheral nervous system. Few studies have focused on WD in the complex neuronal circuits of the mammalian brain, and the exact sequence of degenerative events remains undefined, as does the contribution of axon arborization and synaptic architecture on the degeneration process. To gain a comprehensive picture of the spatiotemporal dynamics and synaptic mechanisms of WD in the nervous system, we identify the factors that regulate WD within the mouse cerebral cortex. We combined single-axon-resolution multiphoton imaging with laser microsurgery through a cranial window and a fluorescent membrane reporter. Longitudinal imaging of > 150 individually injured excitatory cortical axons revealed a threshold length below which injured axons consistently underwent a rapid-onset form of WD (roWD). roWD started 10 times earlier and was executed 4 times slower than WD described in other regions of the nervous system. Cortical axon WD and roWD were dependent on synaptic density, but independent of axon complexity. Our data illustrate how in vivo time-lapse imaging can provide new insights into the spatiotemporal dynamics and synaptic mechanisms of axon loss and assess therapeutic interventions in the injured mammalian brain. Canty, A.J.*, Jackson, J.S.*, Huang, L., Trabalza, A., Bass, C., Little, G., Tortora, M., Khan, S. and De Paola, V. (2020). In vivo imaging of injured cortical axons reveals a rapid onset form of Wallerian degeneration. BMC Biology. Accepted for publication September 2020.

38 Abstracts

MRS BARBORA FULOPOVA

University of Tasmania [email protected]

Differential patterns of cortical connectivity and beta amyloid deposition in APP/PS1 mice relative to ageing and the effects of midlife environmental enrichment.

Fulopova B., Stuart K., Bennett W., Bindoff A., King A.E., Vickers J.C., Canty A.J. Wicking Dementia Research and Education Centre, University of Tasmania; [email protected]

Experience-dependent lifestyle factors were identified as potentially modifiable targets for delaying/preventing the onset of dementia. Environmental enrichment (EE) is a model of experience-dependent lifestyle stimulation in laboratory mice. While EE housing can increase performance on cognitive tasks in amyloidosis APP/PS1 mice, the potential effect of EE on disease modification in terms of pathological change are inconclusive. We hypothesised that previous contrasting findings may be attributable to differences in susceptibility to amyloid beta (Aβ) plaque deposition in cortical regions functionally associated with EE. We characterised fibrillar plaque deposition in 6, 12, and 18-22 months old APP/PS1 mice in the prefrontal (PFC), somatosensory (SS2) and primary motor cortex (M1). We found a significant increase in plaque load between 6 and 12 months in all regions. In animals over 12 months, only the PFC continued to significantly accumulate plaques. Additionally, 12 months old animals subjected to 6 months of EE showed improved spatial navigation and had significantly fewer plaques in M1 and SS2, but not in the PFC. Density of excitatory pre-synaptic outputs was unchanged across age, genotype, and housing conditions. However, PFC had significantly higher pre-synaptic density compare to SS2 and M1. These findings suggest that PFC is selectively susceptible to Aβ deposition and less responsive to the attenuating effects of EE, while maintaining overall higher density of pre-synaptic outputs compare to SS2 and M1. In contrast, M1 and SS2 plateau with respect to Aβ deposition by 12 months of age and are susceptible to amyloid pathology modification by midlife EE.

39 Abstracts

DR LYNDAL HENDEN

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Identity by descent analysis links SOD1 familial and sporadic ALS cases

Lyndal Henden1, Natalie A. Twine2, Piotr Szul3, Emily P. McCann1, Garth A. Nicholson4, Dominic B. Rowe1,5, Matthew Kiernan6, Denis C. Bauer2, Ian P. Blair1 and Kelly L. Williams1 1. Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; 2. Transformational Bioinformatics, CSIRO, Sydney, NSW 2113, Australia; 3. Data61, CSIRO, Dutton Park, QLD 4102, Australia; 4. Concord Clinical School, ANZAC Research Institute, Concord Repatriation Hospital, Sydney, NSW 2139, Australia; 5. Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2019, Australia; 6. Brain and Mind Institute, The University of Sydney, Sydney, NSW 2050, Australia Email of presenting author: [email protected]

Approximately 10% of motor neuron disease (MND) cases have a family history of disease, while the remaining cases present as apparently sporadic. Heritability studies suggest a significant genetic component to sporadic MND, and although most sporadic cases have an unknown genetic etiology1, mutations that are present in familial MND cases have also been found in sporadic cases2. This suggests that some sporadic cases may actually be unrecognised familial cases with reduced penetrance. Identifying a familial basis of disease in apparently sporadic MND cases has important genetic counselling implications for immediate relatives, including a 50% chance of inheriting the mutation and a significantly increased chance of developing MND. Identity-by- descent (IBD) analysis detects genomic regions that have been inherited from a common ancestor3 and is a powerful strategy to uncover a familial basis in apparently sporadic cases.

We performed IBD analysis on 83 Australian familial MND cases from 25 families and three sporadic MND cases, each carrying one of three common SOD1 mutations in Australia (p.I114T, p.V149G and p.E101G). We identified five unique 350-SNP haplotypes that carry these mutations in our cohort, indicative of five founder events. This included two different haplotypes carrying SOD1 p.I114T, linking familial and sporadic cases. We found that SOD1 p.E101G arose independently in each family that carries this mutation and linked two families carrying SOD1 p.V149G. The age of disease onset differed between cases that carried each SOD1 p.I114T haplotype, while the rate of disease progression differed between cases that carried each SOD1 p.E101G haplotype.

References: 1. Renton AE, Chio A and Traynor BJ (2014). State of play in amyotrophic lateral sclerosis genetics. Nat Neurosci. 17(1): 17-23 2. Jones CT, Swingler RJ, Simpson SA and Brock DJH (1995). Superoxide dismutase mutations in an unselected cohort of Scottish amyotrophic lateral sclerosis patients. J Med Genet. 32: 290-292 3. Albrechtsen A, et al. (2009). Relatedness mapping and tracts of relatedness for genome-wide data in the presence of linkage disequilibrium. Genet Epidemiol. 33: 266-274

40 Abstracts

DR CINDY MAUREL

Macquarie Centre for Motor Neuron Disease Research - Macquarie University

[email protected] The unexplored effects of SUMOylation on TDP-43 aggregation and sub- cellular localization Cindy Maurel1, Natalie Scherer1, Andrés Vidal-Itriago1, Emily Don1, Tyler Chapman1, Albert Lee1, Roger Chung1, Patrick Vourc’h2, Marco Morsch1 Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; 2. UMR 1253, iBRAIN, Université de Tours, Inserm, Tours, France.

TDP-43 localizes predominantly in the nucleus, rearranging itself in highly dynamic granules due to a mechanism called liquid-liquid phase separation. Moreover, TDP-43 actively shuttles between the nucleus and the cytoplasm [1]. The mechanisms underlying how TDP-43 is compartmentalized in the cell remain elusive and crucial. We provide novel evidence the post-translational modification (PTM) SUMOylation can affect TDP-43 localization and aggregation in vitro and in vivo [2].

We developed an ALS in vitro model that presents TDP-43 positive cytoplasmic aggregation by over-expressing GFP-TDP-43WT in cell lines. Inhibition of the first step of the SUMOylation pathway in these cells significantly reduced the presence of TDP-43 aggregates and improved neuritogenesis and cell viability. Similarly, over-expressing TDP-43 that was mutated at the putative SUMOylation site lysine 136, GFP-TDP-43K136R, also rescued neurite outgrowth and increased toxicity that was associated with the over-expression of TDP-43WT. Notably, these GFP- TDP-43K136R cells presented with nuclear aggregates. Our in vivo results are in line with these findings. We found a significant increase in nuclear localisation of eGFP-TDP43K136R compared to eGFP-TDP43WT. We also found that zebrafish motor neurons expressing non-SUMOylated forms of TDP-43 showed less cytoplasmic aggregates, as well as altered phase separation properties.

Our data demonstrate for the first time the importance of the SUMOylation pathway (and the lysine 136 of TDP-43) in the process of cytoplasmic localization of TDP-43 aggregates in vitro and in vivo. Influencing PTMs such as SUMOylation could be a promising medium-term therapeutic target compared to modifying the basic expression levels of TDP-43.

References 1. Svahn AJ, Don EK, Badrock AP, et al (2018) 136:445–459. 2. Maurel C, Chami AA, Thépault R-A, et al (2020) Mol Neurobiol 57:1361–1373.

41 Abstracts

MS MARINA ULANOVA

Centre for Healthy Brain Ageing -UNSW - Sydney [email protected] Magnetic nanoparticles as MRI contrast agents for the diagnosis of Alzheimer’s Disease Marina Ulanova1, Lucy Gloag1,2, Andre Bongers3,4,5, Justin Gooding1,6,7, Joanna Biazik- Richmond8, Richard Tilley1,9,10, Perminder S. Sachdev11 and Nady Braidy12,

1. University of New South Wales, Sydney, NSW, Australia 2. ARC Centre of Excellence in Convergent Bio-Nano Science and Technolo, Sydney, NSW, Australia 3. Mark Wainwright Analytical Centre, UNSW, Sydney, NSW, Australia 4. National Imaging Facility, Sydney, NSW, Australia 5. Prince of Wales Clinical School, Sydney, NSW, Australia 6. Australian Centre for Nanomedicine, Sydney, NSW, Australia 7. ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Sydney, NSW, Australia 8. UNSW MWAC Electron Microscopy Unit, Sydney, NSW, Australia 9. Mark Wainwright Analytical Centre, Sydney, NSW, Australia 10. Australian Centre for NanoMedicine, Sydney, NSW, Australia 11. Neuropsychiatric Institute (NPI), Euroa Centre, Prince of Wales Hospital, Sydney, NSW, Australia 12. Centre for Healthy Brain Ageing, University of New South Wales, Randwick, NSW, Australia

Nanoparticle-based magnetic contrast agents have opened the potential for Magnetic Resonance Imaging (MRI) to be used for early non-invasive diagnosis of Alzheimer’s disease (AD). Current methods for clinical diagnosis in the early stages of the disease, such as Positron Emission Tomography imaging of amyloid build-up are limited by their availability and cost. The aim of this research is to develop a novel non-toxic amyloid targeted nanoparticle which can successfully permeate the blood brain barrier and bind amyloid plaques, to improve MRI contrast and diagnostic sensitivity. Targeted iron nanoparticles were assessed using a U-251 cell line to determine their in vitro toxicity. Transmission electron microscopy was used to determine the movement of the nanoparticles within the cell and in vitro binding to amyloid fibrils. The novel targeted nanoparticles have demonstrated no significant in vitro toxicity and electron microscopy results show their movement through the endocytic cycle within the cell, demonstrating an effective degradation and clearance pathway. In addition, immunohistochemistry demonstrated nanoparticle co-localisation with plaques on ex vivo brain sections. The present work shows promising preliminary results in the development of a targeted non-invasive method of early AD diagnosis using contrast enhanced MRI.

42 Abstracts

MR ANDRES VIDAL-ITRIAGO

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

The missing link: Investigating the physiological traits of microglia during neurodegeneration Andrés Vidal-Itriago1, Rowan AW Radford1, Cindy Maurel1, Natalie M Scherer1, Emily K Don1, Roger S Chung1, Manuel B Graeber2 and Marco Morsch1 1. Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; 2. Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia Microglia are phagocytes responsible for the homeostasis and immune responses in the central nervous system (CNS). Whether microglia are beneficial or detrimental during onset or progression of neurodegenerative diseases remains controversial. Microgliosis, the increased number of activated microglia, is a pathological hallmark in neurodegenerative disorders [1]. However, it has been extremely difficult to examine the dynamic interactions of microglia with degenerating neurons real-time and over extended periods of time. Peripheral nerve injury (axotomy) models have greatly contributed to our understanding of microglial response to neurodegeneration [2]. Here we established the first axotomy model in zebrafish. Using in vivo confocal imaging we have characterized the long-term dynamics of microglia-neuron interactions in response to peripheral nerve injury. In vivo time-lapse imaging confirmed that peripheral nerve injury triggered a microglial response in the CNS. This response was characterized by an increased number of microglia displaying rod- like morphology and reduced motility. Microglia engaged in prolonged interactions (around 12 hours) with injured neurons, often forming specialized structures that established intimate connections with neuronal somas. These results reveal dynamic and morphological changes in microglia that have not been described before, outlining a novel microglial response to axotomy- induced neurodegeneration. Compelling evidence suggest that microglia are crucial in maintaining CNS homeostasis. However, how microglia contribute to neuron survival or degeneration remains elusive. Our preliminary results identify an unprecedented microglial response to induced neurodegeneration that may affect neuron viability. Understanding the morphophysiological traits of microglia-neuron interactions may provide the fundamental knowledge to harvest their therapeutic capacity in the future. References: [1] Lull, M. E., & Block, M. L. (2010). Microglial activation and chronic neurodegeneration. Neurotherapeutics, 7(4), 354-365. [2]Moran, L. B., & Graeber, M. B. (2004). The facial nerve axotomy model. Brain Research Reviews, 44(2-3), 154-178

43 Abstracts

POSTERS

44 Abstracts

MR AMR ABDEEN

Brain and Mind Centre and Discipline of Pharmacology - School of Medical Sciences - Faculty of Medicine and Health - The University of Sydney, [email protected]

Empirical evidence supports an aetiological role for changes in biometal pathways in parkinson’s disease Amr Abdeen, Benjamin Trist, Kay Lorraine Double 1. Brain and Mind Centre and Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia Iron overload and copper deficiency are characteristic of the degenerating substantia nigra (SN) in Parkinson’s disease brains, however the evidence for causality between these pathological features has not been previously assessed. We gathered empirical evidence of metal changes in the Parkinson’s disease SN using a systematic review, then employed the Bradford Hill model of causation to systematically assess whether the available evidence supports a causal relationship between metal alterations and neuron death in Parkinson’s disease. The systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta- analyses guidelines. An electronic search of articles published up to September 2019 was conducted in PubMed, EMBASE, Central Register of Controlled Trials and Scopus, and 8437 resultant articles were screened for pre-defined inclusion and exclusion criteria restricted to human research. The quality of the included 182 articles was assessed using our published Quality Assessment Scale and the NIH Quality Assessment tools; studies of limited quality were removed, resulting in 156 studies for final analysis. The Bradford Hill model evaluates a potential causal inference of one variable on another by assessing published evidence supporting and opposing each according to a set of nine criteria. Assignment of the final 156 studies to these nine criteria revealed that at least seven criteria supported a causal role for alterations to iron and copper levels in nigral neuron death in Parkinson’s disease patients. This study supports the development and clinical testing of current and future therapeutic interventions targeting metal alterations in Parkinson’s disease.

45 Abstracts

MS AZIN AMIN

The Florey Institute of Neuroscience & Mental Health, University of Melbourne [email protected] Development of autophagy-inducing peptides as a potential therapy for motor neurone disease

Azin Amin1, Nirma Perrera1, Bradley Turner1, Fazel Shabanpoor1 1. The Florey Institute of Neuroscience & Mental Health, University of Melbourne, Victoria 3010, Australia. E-mail address: [email protected]

The pathological hallmark of a range of neurodegenerative diseases, including Motor Neurone Disease (MND) is the presence of protein inclusions and aggregates in affected neurons. Aggregates progress continually from the site of onset to disturb cellular processes, ultimately resulting in neuron degeneration1. The only intracellular degradative pathway that can purge the cells of these misfolded proteins, aggregates, and dysfunctional organelles is autophagy. Since impaired/reduced autophagy may contribute to MND pathogenesis; upregulating autophagy offers a potential therapeutic option. This study aimed to develop and synthesize novel pharmacological agents that have non-toxic autophagy-inducing properties in motor neuronal NSC-34 cells. A series of peptides analogues of the evolutionary conserved domain of Beclin1 protein, the master regulator of autophagy, were designed and synthesized. In order to enhance the cellular uptake and increase the cytosolic bioavailability, the peptides were cyclized, and conjugated to a cell- penetrating peptide domain, and an endosomal escape domain. The autophagy-inducing effect of the peptides was determined by measuring the level of autophagosome-associated LC3-II, a marker of macroautophagy, in NSC-34 cells. We have shown that the peptides are capable of inducing autophagy at 5 micromolar concentration, which is 10-fold more potent than the previously reported Beclin-1 analogues. More importantly, the most potent peptide analogue significantly decreased aggregate-prone MND-associated protein levels and reduced their toxicity. This study provides in vitro evidence for a novel peptide-based approach to enhance autophagy as a unique and safe treatment modality with potential therapeutic benefit in MND and other proteinopathies.

References: 1. Hara, T., et al., 2006. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature, 441(7095), p. 885-889.

46 Abstracts

MISS SANDRINE CHAN MOI FAT

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Analysis of GLT8D1 and ARPP21 in Australian familial and sporadic MND cases Sandrine Chan Moi Fat1, Emily P McCann1, Kelly L Williams1, Lyndal Henden1, Natalie A Twine1,2, Denis Bauer2, Dominic B Rowe1,7, Roger Pamphlett3,4,5, Matthew C Kiernan4,6, Garth A Nicholson1,8, Jennifer A Fifita1, Ian P Blair1 1. Macquarie University Centre for Motor neuron Disease Research, Department of Biomedical sciences, Faculty of Medicine, Health and Human sciences, Macquarie University, Sydney, New South Wales, Australia 2. Transformational Bioinformatics, Commonwealth Scientific and Industrial Research Organisation, sydney, New South Wales, Australia 3. Discipline of Pathology and Department of Neuropathology, The University of Sydney, Sydney, New South Wales, Australia 4. Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia 5. Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, New South Wales 6. Australia Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, New South Wales 7. Department of clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University 8. ANZAC Research Institute, University of Sydney, Concord Hospital, Sydney, New South Wales, Australia Motor Neuron Disease (MND) is a paralytic disorder caused by the death of motor neurons. Recently, genetic sequencing analysis of MND cases of predominantly European ancestry have linked variants in GLT8D1 and ARPP21 to MND (1). We sought to investigate the prevalence of GLT8D1 and ARPP21 variants among Australian patients using next-generation sequencing data from 618 sporadic and 81 familial MND cases, all with unknown MND causative mutations. We developed custom bioinformatics pipelines to perform gene-based burden analysis to determine whether GLT8D1 and/or ARPP21 carried a burden of protein-altering or splicing variants in comparison to three control cohorts. Moreover, we investigated whether any qualifying variants were overrepresented or underrepresented among cases as compared to controls, and potentially acting as risk or protective alleles for MND, respectively. Results identified an absence of any protein-altering or splicing variants in the GLT8D1 gene. Eight such variants were found within ARPP21, and three of these were predicted to be pathogenic by at least 4/7 prediction tools. However, all these variants were also present in healthy controls and none of these variants showed evidence of association with ALS. Gene-based burden analysis of ARPP21 indicated no enrichment of rare protein altering ARPP21 variants in MND cases when compared to each of three control cohorts. These findings suggest that mutations in GLT8D1 and ARPP21 are not a common cause of MND in Australia. Further studies with larger sample size and diverse ancestries will be required to validate the potential contribution of GLT8D1 and/or ARPP21 variants to MND. References

1. Cooper-Knock, J., Moll, T., Ramesh, T., Castelli, L., et al. (2019). Cell reports, 26(9), 2298- 2306.

47 Abstracts

DR COURTNEY CLARK

Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania

[email protected]

Neuropeptide Y suppresses hyperexcitability of cortical SOD1G93A mouse neurons in vitro. Courtney Clark1, Marcus Dyer1, Rosie Clark1, Catherine Blizzard1, Tracey Dickson1 Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS 7000 [email protected] Introduction: Cortical hyperexcitability has been implicated as an early event which contributes to dysfunction in amyotrophic lateral sclerosis (ALS)(1, 2). Therefore, modulating excitability could potentially slow the progression of ALS in the cortex. Neuropeptide Y (NPY) is an inhibitory neuromodulator which has been shown to protect against hyperexcitability in neurological disorders (ie. Epilepsy) (3-5). Objectives: The objective of this study was to investigate whether NPY could modify excitability within the cortex and prevent neuronal hyperexcitability. We hypothesise: exogenous application of NPY suppresses neuronal activity. Methods: Cortical neurons were cultured from Thy1YFP x SOD1G93A (E15.5) (n =10) and wild type (WT) littermates (n= 18). At 14 DIV cultures were treated with 100nM, 1uM NPY or vehicle. Network excitability was assessed by multi-electrode array. Patch clamp electrophysiology assessed the effect of NPY on cortical pyramidal (n = 11) and inhibitory neurons (n = 7). Statistical analysis; Two-Way ANOVA and paired t-test. Results: NPY reduced neuronal firing and spike rate of SOD1G93A (p < 0.05) but not WT cultures (p > 0.05). Neuronal firing (p < 0.01) and spike rate (p < 0.05) of cortical neurons was increased in SOD1G93A cultures compared to WT. NPY increased rheobase of pyramidal neurons (p > 0.05) and decreased rheobase of inhibitory neurons (p < 0.01) Discussion: NPY modulates neuronal excitability by altering the action potential threshold of pyramidal and inhibitory neurons. NPY suppressed increased excitability observed in SOD1G93A cortical cultures. Suggesting it as a therapeutic candidate for preclinical excitatory dysfunction in ALS. References: 1. Menon P, Kiernan MC, Vucic S. Cortical hyperexcitability precedes lower motor neuron dysfunction in ALS. Clin Neurophysiol. 2015;126(4):803-9. 2. Vucic S, Nicholson GA, Kiernan MC. Cortical hyperexcitability may precede the onset of familial amyotrophic lateral sclerosis. Brain. 2008;131(Pt 6):1540-50. 3. El Bahh B, Balosso S, Hamilton T, Herzog H, Beck-Sickinger AG, Sperk G, et al. The anti- epileptic actions of neuropeptide Y in the hippocampus are mediated by Y and not Y receptors. Eur J Neurosci. 2005;22(6):1417-30. 4. Colmers WF, El Bahh B. Neuropeptide Y and Epilepsy. Epilepsy Curr. 2003;3(2):53-8. 5. Wickham J, Ledri M, Bengzon J, Jespersen B, Pinborg LH, Englund E, et al. Inhibition of epileptiform activity by neuropeptide Y in brain tissue from drug-resistant temporal lobe epilepsy patients. Sci Rep. 2019;9(1):19393.

48 Abstracts

MISS MADILYN COLES

Behavioural Neuroscience - School of Medicine - Western Sydney University [email protected] Medium dose chronic CBD treatment reverses object recognition memory deficits of APPSwe/PS1ΔE9 transgenic female mice

Madilyn Coles1*, Georgia Watt1, Fabian Kreilaus1, Tim Karl1,2 1. School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; 2. Neuroscience Research Australia, Randwick, NSW 2031, Australia *Presenting author email address: [email protected]

Alzheimer’s disease (AD) is a neurodegenerative disease that causes behavioural and cognitive impairments. The phytocannabinoid cannabidiol (CBD) has anti-inflammatory, anti-oxidant and neuroprotective properties and in vitro and limited in vivo evidence suggests that CBD possesses therapeutic-like properties for the treatment of AD. Cannabinoids are known to have dose- dependent effects and the therapeutic potential of medium dose CBD for AD transgenic mice has not been assessed in great detail yet. 12-month-old control and APPSwe/PS1ΔE9 (APPxPS1) transgenic female mice were treated daily via intraperitoneal injection with 5 mg/kg bodyweight CBD (or vehicle) commencing three weeks prior to the assessment of behavioural domains including anxiety, exploration and locomotion, motor functions, cognition, and sensorimotor gating. APPxPS1 mice exhibited a hyper-locomotive and anxiogenic-like phenotype and had wild type-like motor and spatial learning abilities, although AD transgenic mice took generally longer to complete the cheeseboard training (due to a lower locomotion speed). All mice displayed intact spatial memory (although this was delayed in AD transgenic mice) and retrieval memory, but APPxPS1 mice showed reduced levels of perseverance in the cheeseboard probe trial. Importantly, vehicle-treated APPxPS1 mice were characterised by object recognition deficits and delayed spatial learning, which were reversed by CBD treatment. Finally, impairments in sensorimotor gating of APPxPS1 mice were not affected by CBD. In conclusion, medium dose CBD appears to have therapeutic value for the treatment of particular behavioural impairments present in AD patients. Future research should consider the molecular mechanisms behind CBD’s beneficial properties for AD transgenic mice.

49 Abstracts

MS LAUREN DEWSBURY

NICM Health Research Institute - Western Sydney University [email protected]

Is there a role for ketogenic therapies in the clinical management of neurodegenerative disease? A systematic review.

Lauren S. Dewsbury1, Chai K. Lim2, Genevieve Z. Steiner1,3

1. NICM Health Research Institute, Western Sydney University, Penrith NSW 2751, Australia 2. Dept. of Biomedical Science, Faculty of Medicine Health and Human Sciences, Macquarie University, Macquarie Park, NSW, 2109, Australia 3. Translational Health Research Institute (THRI), Western Sydney University, Penrith NSW 2751, Australia Email: [email protected]

Introduction: Ketone bodies have potential disease-modifying activity that may represent a novel therapeutic approach for neurodegenerative disease. The aim of this systematic review was to summarise and critically appraise the evidence for the utility of ketogenic therapies (dietary or exogenous ketogenic agents) in the clinical management of neurodegenerative diseases, and provide recommendations to direct future research. Method: Eight databases were electronically searched for controlled-trials (≥ 4 weeks duration) that induced ketosis or elevated serum ketones in people with neurodegenerative disease. Results: Of 4,498 results, 18 articles met the inclusion criteria reporting on interventions in mild cognitive impairment (MCI; n = 5), multiple sclerosis (n = 5), Alzheimer’s disease (n = 5), Parkinson’s disease (n = 1), MCI secondary to Parkinson’s disease (n = 1) and MCI combined with subjective cognitive impairment (n = 1). Most studies used dietary interventions (n = 10), followed by medium-chain triglyceride supplementation (n = 7) and a fasting protocol (n = 1). Type and severity of neurodegenerative disease, interventions implemented, and outcome measures assessed were heterogenous across included studies. The majority of trials involved people with MCI, employed a dietary intervention, were 6-weeks or 6-months in duration, and measured cognition as the primary outcome. Conclusion: Considering compliance and adherence, exogenous ketogenic agents may be more feasible than dietary interventions in neurodegenerative disease populations. Future research should improve exogenous formulations to reduce adverse effects, characterise inter-individual factors affecting response-to-treatment, and establish a ‘minimum required dose’ for clinically meaningful improvements in disease-specific symptoms, such as cognition or motor function, to inform clinical guidelines.

50 Abstracts

DR EMILY DON

Department of Biomedical Sciences - Macquarie University [email protected]

Utilising molecular imaging to investigate protein aggregate formation in vivo Emily K Don1 , Alina Maschirow1 , Rowan A W Radford1 , Natalie M Scherer1 , Andres Vidal- Itriago1 , Alison Hogan1 , Cindy Maurel1 , Isabel Formella1 , Jack J Stoddart1 , Thomas E Hall2 , Albert Lee1 , Bingyang Shi1 , Nicholas J Cole1 , Angela S Laird1 , Andrew P Badrock1* , Roger S Chung1 , Marco Morsch1*

1. Centre for Motor Neuron Disease Research, Faculty of Health and Medical Sciences, Department of Biomedical Science, Macquarie University, Sydney, NSW 2019, Australia. 2. Institute for Molecular Bioscience, The University of Queensland, QLD 4072, Australia.

Protein aggregation of TDP-43 (and other ALS linked proteins) is a pathological hallmark in nearly all ALS patients. However, one aspect that remains unclear is the dynamic process of how these aggregates assemble and how they can form pathological inclusions. Clinical verification of aggregation pathology is limited to histological techniques that provide a static snapshot of the aggregation pattern. This limits the ability to investigate the dynamic molecular mechanisms that are believed to trigger aggregate formation, maturation and mislocalisation. With recent advances in technology, Biomolecular Fluorescence Complementation (BiFC) can be utilised to study aggregate formation in real time. Here, we present the first BiFC study assessing ALS aggregate formation in vivo by combining the power of molecular imaging with the advantages of the zebrafish system. We validate and visualize the formation of TDP-43 and FUS aggregates in real time in a vertebrate model. We report preliminary findings on the dynamic aggregation of the ALS-linked hallmark proteins Fus and TDP-43 in their corresponding nuclear and cytoplasmic compartments using BiFC. Taken together, our results confirm that BiFC can be utilized to study ALS-linked aggregate formation and conceivably spread in vivo. To the best of our knowledge, this BiFC approach may be one of the very few ways to assess how recruitment of mutant or pathological forms of ALS linked aggregating proteins can directly influence their wild-type counterparts. Identification of the mechanistic and molecular underpinnings of these aggregation processes has important implications for ALS and a range of other human proteinopathies.

51 Abstracts

MR MARCUS DYER

Menzies Institute for Medical Research [email protected]

Amyotrophic lateral sclerosis associated mislocalisation of TDP-43 to the cytoplasm causes cortical hyperexcitability and reduced excitatory neurotransmission in the motor cortex. Dyer MS1, Reale LA1, Lewis KE1, Walker AK2, Dickson TC1, Woodhouse A3 and CA Blizzard1 1. Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tas, 7000, Australia 2. Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, University of Queensland, Brisbane, Queensland, 4072, Australia 3. Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tas, 7000, Australia Amyotrophic lateral sclerosis (ALS) is a chronic neurodegenerative disease pathologically characterised by mislocalisation of the RNA binding protein TAR-DNA binding protein 43 (TDP- 43) from the nucleus to the cytoplasm. Changes to neuronal excitability and synapse dysfunction in the motor cortex are early pathological changes occurring in people with ALS and mouse models of disease. To investigate the effect of mislocalised TDP-43 on the function of motor cortex neurons we utilised mouse models that express either human wild-type (TDP-43WT) or nuclear localization sequence-deficient TDP-43 (TDP-43ΔNLS) on an inducible promoter that enriches expression to forebrain neurons. Pathophysiology was investigated through immunohistochemistry, Western blot and whole-cell patch-clamp electrophysiology. Thirty days expression of TDP-43ΔNLS in adult mice did not cause any changes in the number of CTIP2-positive neurons in the motor cortex. However, at this time-point the expression of TDP-43ΔNLS drives intrinsic hyperexcitability in layer V excitatory neurons of the motor cortex. This hyperexcitability occurs concomitantly with a decrease in excitatory synaptic input and fluctuations in both directions of ionotropic glutamate receptors. This pathophysiology is not present with TDP-43WT expression, demonstrating that the localisation of TDP-43 to the cytoplasm is crucial for the altered excitability phenotype. This study has important implications for the mechanisms of toxicity of one of the most notorious proteins linked to ALS, TDP-43. We provide the first evidence that TDP-43 mislocalisation causes aberrant synaptic function and a hyperexcitability phenotype in the motor cortex, linking some of the earliest dysfunctions to arise in people with ALS to mislocalisation of TDP-43.

52 Abstracts

DR SHELLEY FORREST

Dementia Research Centre - Department of Biomedical Sciences - Macquarie University [email protected]

Coexisting Lewy body disease and clinical parkinsonism in amyotrophic lateral sclerosis

Shelley L. Forrest1,2, Jordan Hanxi Kim2, Clair De Sousa2, Rosie Soos2, Daniel R. Crockford2, Donna Sheedy2, Julia Stevens2, Toni McCrossin2, Rachel H. Tan3, Heather McCann4, Claire E. Shepherd4, Dominic Rowe5,6, Matthew C. Kiernan3, Glenda M. Halliday3,4,7, Jillian J. Kril2.

1. Dementia Research Centre, School of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia. 2. Discipline of Pathology and , Faculty of Medicine and Heath, University of Sydney, Australia. 3. Brain and Mind Centre and Central Clinical School, Faculty of Medicine and Health, University of Sydney, Australia. 4. Neuroscience Research Australia, Randwick, Australia. 5. Department of Clinical Medicine, Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, Australia. 6. Centre for MND Research, Department of Biomedical Science, Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, Australia. 7. School of Medical Sciences, The University of New South Wales, Sydney, Australia.

Amyotrophic lateral sclerosis (ALS) is associated with a range of clinical phenotypes with progressive degeneration of upper and lower motor neurons, and pTDP-43 inclusions in motor and non-motor pathways. Parkinsonian features have been reported in up to 30% of ALS patients1 and Lewy bodies, normally associated with Lewy body disorders (LBD), have been reported in a small number of ALS cases, with unknown clinical relevance. This study investigates the prevalence of clinically relevant LBD in a prospectively studied ALS cohort to determine if concomitant pathology contributes to the heterogeneity of clinical features. All ALS cases held by the NSW Brain Tissue Resource Centre and Sydney Brain Bank (n=99) were screened for coexisting LBD (Braak ≥ stage IV), corresponding to clinical parkinsonism. Six cases (5 male) had coexisting LBD Braak ≥ stage IV pathology. The age at symptom onset (69±7) and disease duration (4±3 years) in ALS cases with coexisting LBD did not differ from pure ALS cases. Three patients had lower limb onset, two patients had bulbar onset, and the region of onset was unknown in one patient. One patient developed clinical features of Parkinson’s disease (PD) and multiple system atrophy, but did not receive a secondary clinical diagnosis. One patient also developed clinical features of PD and received a dual diagnosis. All cases had no known relevant family history or genetic abnormalities. The prevalence of clinically relevant LBD in ALS is higher than the general population, which has implications for clinical and neuropathological diagnoses and the identification of biomarkers.

References: Calvo A, Chiò A, Pagani M, et al. Parkinsonian traits in amyotrophic lateral sclerosis (ALS): a prospective population-based study. J Neurol 2019;266:1633-1642.

53 Abstracts

DR SARAH FURLONG

Macquarie Centre for Motor Neuron Disease Research - Macquarie University

[email protected]

Macquarie University Neurodegenerative Disease Biobank

Sarah Furlong1, Kristine Deang, Elisa Cachia1, , Susan D’Silva1,, Julie Atkin1, Roger Chung1, Gilles Guillemin1, Kyle Ratinac1, Dominic B Rowe1,2, Kelly Williams1, Ian P Blair1 1. Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; 2. Department of Clinical Medicine, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia [email protected]

In 2013, Macquarie University’s Centre for MND research and Macquarie Health Neurology clinic joined forces to establish the Neurodegenerative Disease (ND) Biobank. MND patients, family members and controls are invited to participate, by donating blood, urine, hair and skin biopsies. Extensive demographic, clinical and lifestyle data is collected from each participant. This resource has grown rapidly with more than 800 participants: 65 familial patients, 320 sporadic patients, 429 controls and 17 asymptomatic participants. The bank also contains multi- generational participants from large Australian MND families. Biological samples and data are collected longitudinally in order study disease progression. Nearly 2,500 collections of DNA, RNA, plasma, serum, urine and hair have been obtained, thus far. In addition, there is a bank of more than 20 fibroblast cell lines developed from participant skin biopsies. More than 20 research projects have availed of this resource. Most of these projects are large complex, national and international collaborations. Multiple biomarker projects study longitudinal collections of plasma, serum, urine and hair, to identify biomarkers. Fibroblasts are used to investigate protein degradation and generate iPSCs. DNA samples are used to identify new MND genes and to develop new models for investigating MND. RNA samples have been used to study gene expression. Environment projects search for evidence of neurotoxin and pollutant exposure in plasma from sporadic patients. As one of the largest MND biobanks in the world, with a collection of well-characterized samples and extensive clinical and lifestyle data, it is an in valuable resource to investigate MND pathogenesis and treatment strategies. https://www.mq.edu.au/mnd

54 Abstracts MISS JASMIN GALPER

Faculty of Medicine and Health - University of Sydney [email protected]

Lipid profiles of clinical and pre-clinical Parkinson’s disease Jasmin Galper1, Scott Kim1, Russell Pickford2, Glenda Halliday1, Nicolas Dzamko1 1. Brain and Mind Centre, Central clinical school, Faculty of Medicine & Health, University of Sydney, Sydney, NSW 2150, Australia; 2. Bioanalytical Mass Spectrometry Facility, University of NSW, NSW 2052, Australia [email protected] Patients with Parkinson’s disease (PD) experience progressive motor function impairment, the cause of which is largely unknown. PD patients are estimated to have gone undiagnosed for up to 20 years with prodromal symptoms, at which time critical disease pathogenesis and extensive neurological damage is occurring. At the time of patient diagnosis, about 50% of the dopaminergic neurons responsible for controlling movement are lost [1]. This presents a challenge to clinical trials which are attempting to reverse, stop or slow disease pathology that has progressed substantially. Further, the opportunity to research patients at initial disease stages and better understand early pathology is limited by the relatively late diagnosis. Several lipid related genes have been implicated as PD risk factors for a proportion of patients [2-7], highlighting an important role for lipid biology in disease pathogenesis. We investigated cerebrospinal fluid (n = 88) and serum (n = 221) lipidomes from the Michael J Fox Foundation LRRK2 clinical cohort consortium. This cohort included controls, idiopathic PD and clinical and pre-clinical mutation carriers of one of the most common PD risk genes: LRRK2. We used an untargeted approach to screen lipidomes via ultra high-performance liquid chromatography-tandem mass spectrometry. Significant cerebrospinal fluid and serum lipid changes were found in both idiopathic PD and LRRK2 associated PD compared to controls, with some changes detected pre-clinically. Further, lipid markers significantly correlated to both motor and non-motor clinical measures. Our data indicate that lipid alterations are not restricted to those with mutations in lipid related genes and that lipid changes may be involved in early disease processes.

References: 1. Bernheimer, H., et al., Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci, 1973. 20(4): p. 415- 55. 2. Aharon-Peretz, J., H. Rosenbaum, and R. Gershoni-Baruch, Mutations in the Glucocerebrosidase Gene and Parkinson's Disease in Ashkenazi Jews. New England Journal of Medicine, 2004. 351(19): p. 1972-1977. 3. Chang, D., et al., A meta-analysis of genome-wide association studies identifies 17 new Parkinson's disease risk loci. Nat Genet, 2017. 49(10): p. 1511-1516. 4. Gan-Or, Z., et al., The p.L302P mutation in the lysosomal enzyme gene SMPD1 is a risk factor for Parkinson disease. Neurology, 2013. 80(17): p. 1606-1610. 5. Do, C.B., et al., Web-Based Genome-Wide Association Study Identifies Two Novel Loci and a Substantial Genetic Component for Parkinson's Disease. PLOS Genetics, 2011. 7(6): p. e1002141. 6. Simón-Sánchez, J., et al., Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nature Genetics, 2009. 41(12): p. 1308-1312. 7. Simón-Sánchez, J., et al., Genome-wide association study confirms extant PD risk loci among the Dutch. European Journal Of Human Genetics, 2011. 19: p. 655.

55 Abstracts

DR NICHOLAS GERAGHTY

Molecular Horizons and School of Chemistry and Molecular Bioscience - University of Wollongong [email protected]

A high-throughput flow cytometry drug screen to discover new treatments for motor neurone disease. Nicholas Geraghty1,2*, Nicole Miles1,2, Mark Wilson1,2 Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2500, Australia IHMRI, Wollongong, NSW 2500, Australia * [email protected] A pathological hallmark of motor neurone disease (MND) is the accumulation of insoluble misfolded protein aggregates in neurones and glia in the spinal cord and/or brain of affected individuals. TAR DNA-Binding Protein of 43 kDa (TDP-43) is a ubiquitously expressed protein with many proposed functions, and although primarily a nuclear protein, TDP-43 can shuttle between the nucleus and cytoplasm. The accumulation and aggregation of abnormally phosphorylated, ubiquitinated and truncated TDP-43 in the cytoplasm of cells is strongly associated with neurone loss in disease (1). Sporadic MND (sMND) constitutes ~ 90% of total MND cases, and ~ 97% of sMND cases are associated with protein inclusions enriched in TDP-43. Our lab established a novel flow cytometric assay (known as FloIT (2)), which we have since adapted to provide a 96-well plate high-throughput screening platform designed to identify potential new drugs to treat MND. Using this platform, we have screened small molecules from a number of chemical libraries sourced from Compounds Australia, including ~4,000 FDA approved drugs, 10,000 NatureBank fractions (of ~120,000), and 10,000 Open Scaffold compounds (of ~35,000). Five FDA drugs reduced the numbers of inclusions by 30-40%, with three demonstrating a consistent reduction of TDP-43 inclusions across a range of concentrations as low as 1 μM. Three NatureBank fractions reduced the numbers of TDP-43 inclusions by up to 70%, and the active compound(s) are currently being identified. These compounds will subsequently be tested in C. elegans, zebrafish and murine models. This screening platform therefore rapidly identifies potential drug candidates, which can be further tested in a variety of both cell and animal models.

References: 1. Walker AK, Tripathy K, Restrepo CR, Ge G, Xu Y, et al. An insoluble frontotemporal lobar degeneration-associated TDP-43 C-terminal fragment causes neurodegeneration and hippocampus pathology in transgenic mice. Human Molecular Genetics. 2015;24(25):7241-54. 2. Whiten DR, San Gil R, McAlary L, Yerbury JJ, Ecroyd H, et al. Rapid flow cytometric measurement of protein inclusions and nuclear trafficking. Scientific Reports. 2016;6(1):31138.

56 Abstracts

MS JESSICA L. HAZELTON

The School of Psychology/The University of Sydney [email protected]

Losing track of the heart: Impaired cardiac interoceptive accuracy in behavioural-variant frontotemporal dementia and Alzheimer’s disease in an international cohort. Jessica L. Hazelton1,2, Sol Fittipaldi 3,4,5, Matias Fraile-Vazquez3, Agustina Legaz 3,4,5, Indira Garcia Cordero 3,4, Marion Sourty6, Agustin Ibanez 3,4,7,8,9,, Olivier Piguet1,2, Fiona Kumfor 1,2 1. The University of Sydney, School of Psychology 2. The University of Sydney, Brain & Mind Centre 3. Universidad de San Andrés, Buenos Aires, Argentina 4. National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina 5. Universidad Nacional de Córdoba, Facultad de Psicología, ArgentinaThe University of 6. Sydney, Sydney, School of Biomedical Engineering 7. University of California, Global Brain Health Institute, San Francisco, USA 8. Universidad Autónoma del Caribe, Barranquilla, Colombia 9. Universidad Adolfo Ibáñez, Department of Psychology, Center for Social and Cognitive 10. Neuroscience, School of Psychology, Santiago de Chile, Chile

Cardiac interoception, the ability to detect fluctuations in our hearts’ rhythm, influences how we feel. Few studies have investigated cardiac interoception in dementia, with mixed findings to date. These mixed findings may be due to the accuracy indexes used, where attention was not considered. Here, we hypothesised cardiac interoceptive accuracy would be impaired in people with dementia, after accounting for periods of inattention. One-hundred-and-sixty-eight participants (51 Alzheimer’s disease, AD; 40 behavioural-variant frontotemporal dementia, bvFTD; 24 Parkinson’s disease, PD; 53 healthy controls) were recruited from three international research centres (Argentina, Australia, and Chile). All participants completed two 2-minute tasks. Participants responded when they: 1) felt their heartbeat (cardiac- interoception) or 2) heard a recorded heartbeat (exteroception). ECG was simultaneously recorded. Accuracy was calculated by comparing the event frequency (i.e., actual or recorded heartbeat) with response frequency in successive, overlapping 10-second windows. The regularity of responses within each window was used to assess attention. Cardiac interoceptive accuracy was impaired in bvFTD (p = .002) and AD (p = .032) compared to healthy controls. In contrast, PD patients performed similarly to healthy controls (p = .17). No significant difference in exteroception was observed between groups (p = .09). This study provides the first evidence of impaired cardiac interoceptive accuracy in bvFTD and AD patients, after accounting for inattention. Contrarily, cardiac interoceptive accuracy was preserved in PD patients, highlighting the need to consider attention in interoception. Future neuroimaging research is needed to uncover neural mechanisms driving the interoceptive impairments unique to each syndrome.

57 Abstracts

DR ALISON HOGAN

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

SFPQ pathology is a feature of ALS patient central nervous system tissue Alison L. Hogan1, Natalie Grima1, Jennifer A. Fifita1, Emily P. McCann1, Benjamin Heng1, Sandrine Chan Moi Fat1, Ram Maharjan2, Amy K Cain2, Lyndal Henden1, Ingrid Tarr1, Katharine Y. Zang1, Qiongyi Zhao3, Zong-Hong Zhang4, Amanda Wright1, Sharlynn Wu1, Marco Morsch1, Shu Yang1, Kelly L. Williams1†, Ian P. Blair 1 1. Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia; 2. ARC Centre of Excellence in Synthetic Biology, Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, New South Wales, Australia 3. Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia 4. School of Medicine, IMPACT, Bioinformatics Core Research Facility, Deakin University, Geelong, Australia

Presenting author email address: [email protected] Splicing factor proline and glutamine rich (SFPQ) is a RNA-DNA binding protein with roles in key cellular pathways dysregulated in amyotrophic lateral sclerosis (ALS). Altered processing of SFPQ RNA, specifically increased retention of intron nine, and nuclear depletion of the protein have recently been reported as pathological features of ALS1. Importantly, these pathologies have been demonstrated in models based on multiple genetic subtypes of ALS, suggesting that SFPQ dysregulation may be a common pathological feature of this highly heterogeneous disease. To validate SFPQ pathology as a feature of ALS and further investigate its prevalence in genetic subtypes of the disease, we have performed the first comprehensive analysis of SFPQ pathology in large ALS patient cohorts. We examined SFPQ at the genetic, transcript and protein level in ALS patient central nervous system. At the genetic level, we identified two novel, and two rare sequence variants in SFPQ not previously reported, suggesting a potential genetic link between SFPQ and ALS. At the transcript level, we observed a significant downregulation of SFPQ expression in ALS patient brain tissue and significantly increased retention of SFPQ intron nine in ALS patient motor cortex. At the protein level, we observed SFPQ positive ubiquitinated protein aggregates in ALS spinal motor neurons. Our study confirms SFPQ pathology as a feature of ALS patient central nervous system tissue. This pathology implicates SFPQ dysregulation in the biology of ALS and presents a novel target through which to investigate dysfunction of key cellular pathways linked to disease onset and progression.

References 1. Luisier, R. et al. Intron retention and nuclear loss of SFPQ are molecular hallmarks of ALS. Nat. Commun. 9, 2010 (2018).

58 Abstracts

MS BAHAR KAVYANI

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Investigation of the temporal development of Tau pathology in TAU58/2 transgenic mice Bahar Kavyani1, Amanda Wright1, Jia Li1, Stephanie Rayner1, Rowan Radford1, Roger Chung1, Bingyang Shi1 1. Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW 2109, Australia. [email protected] Alzheimer’s disease (AD) is a progressive neurodegenerative disease, which is a major public health burden worldwide [1]. Despite recent progress in understanding the pathology of AD, the exact causes of AD are largely unknown [2]. Two of the hallmark pathologies of AD are the formation of β-amyloid plaques and neurofibrillary tangles (NFTs) [3,4]. NFTs are composed of hyperphosphorylated Tau protein which aggregates and becomes insoluble [5]. TAU58/2, a P301S mutant Tau transgenic mouse line has been developed to identify underlying molecular mechanisms by demonstrating tauopathy [6]. To choose the best treatment timepoint in the TAU58/2 mouse model various characteristics of tauopathy has been investigated in the present study : the expression profile of Tau regarding different phosphorylation sites and isoforms in hippocampus for two timepoints by western blot and compared to their wild type littermates, as well as abnormal Tau distribution and morphology, and its correlation with neuronal loss and neuroinflammation by immunohistochemistry. Tau hyperphosphorylation at Ser202/Thr205, Thr231 phosphorylation sites, and total Tau level were increased in hippocampus both in three and twelve months old, whereas insoluble Tau formation only occurred in 12 months and not at Thr231 phosphorylation site. The pattern of phosphorylation and Astrogliosis has been reported, particularly in the Amygdala nuclei. These results provide important insights into Tauopathy in the TAU58/2 mouse model at two timepoints in hippocampus. Given the role of Tau hyperphosphorylation in severity of the disease, further investigation across other brain regions in TAU58/2 mice would contribute to the understanding of the disease etiology.

1. Livingston, G., et al., Dementia prevention, intervention, and care. The Lancet, 2017. 390(10113): p. 2673-2734. 2. Qiu, C., M. Kivipelto, and E. von Strauss, Epidemiology of Alzheimer's disease: occurrence, determinants, and strategies toward intervention. Dialogues in clinical neuroscience, 2009. 11(2): p. 111. 3. Karch, C.M. and A.M. Goate, Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biological Psychiatry. 77(1): p. 43-51. 4. Trinczek, B., et al., Domains of tau protein, differential phosphorylation, and dynamic instability of microtubules. Molecular biology of the cell, 1995. 6(12): p. 1887-1902. 5. Iqbal, K., et al., Defective brain microtubule assembly in Alzheimer's disease. The Lancet, 1986. 328(8504): p. 421-426. 6. van Ersel, J., et al., Early-onset Axonal Pathology in a Novel P301S-Tau Transgenic Mouse Model of Frontotemporal Dementia. Neuropathology Appl Neurobiol., 2015. 41(7):p. 906- 925.

59 Abstracts

DR PRAGYA KOMAL

BITS-Pilani - Hyderabad [email protected] Vitamin D intake enhances Vitamin D receptor expression in the striatum and rescues memory and motor dysfunction in mouse model Huntington’s disease S K V Manajri1, P G satwik1, Sanjana Srinivas2 and Pragya Komal1 Department of Biological Sciences, Birla Institute of Technology and Sciences, Hyderabad, Telangana, India - 500078; Department of Biotechnology, PES University, Bengaluru, Karnataka, India - 560085

Introduction: A coordinated neuronal network between cortex and the basal ganglia is required for normal motor function, which gets severely impaired in Huntington’s disease (HD). In particular, the selective loss of medium spiny neurons in the striatum is considered as a prime brain region responsible for movement disability observed in HD. In this regard, we explored the effect of high Vitamin D (VD) supplementation in HD animals which were induced with 3- nitropropionic acid (3-NP; 75mg/kg) as described previously (Fernagut et al., 2002). Methods: Male C57BL/6J mice (3-4 months old) were divided following groups: Vehicle (group I), 3-nitropropionic acid (HD induced group, intraperitoneal injection), only VD supplemented (group III, 500IU/kg) and 3-NP + VD supplementation (group IV). Animals were then subjected to various behavioral tests like locomotion, gait analysis, Morris water maze (MWM) and rota-rod analysis. After 30 days, animals were decapitated and striatal tissues were isolated to check the mRNA expression of nerve growth factor (NGF), brain derived neurotrophic factor (BDNF) and Vitamin D receptor (VDR). Results: Our behavior and mRNA data shows that VD intake significantly rescued striatal functions like motor co-ordination, locomotion and memory known to be severely impaired in HD. Our results also indicate that VD mediated downstream neuroprotective pathomolecular pathway involves increase expression of Vitamin D receptor (VDR) and neurotrophic factors like NGF and BDNF in the striatum. Conclusion: Altogether, we show that Vitamin D can be a potential “biodrug” to rescue neurodegeneration as observed in HD.

References: 1. Fernagut, P. O. et al. Subacute systemic 3-nitropropionic acid intoxication induces a distinct motor disorder in adult C57Bl/6 mice: Behavioural and histopathological characterisation. Neuroscience 114, 1005–1017 (2002).

60 Abstracts

DR FABAIN KREILAUS

School of Medicine - Western Sydney University [email protected]

Cannabidiol (CBD) treatment improves spatial memory in 14-month-old female TAU58/2 transgenic mice Fabian Kreilaus1, Magdalena Przybyla2, Arne Ittner2, Lars Ittner2, and Tim Karl1,3,4 # 1. School of Medicine, Western Sydney University, NSW 2560, Australia 2. Dementia Research Centre, Faculty of Medicine and Health Sciences, Macquarie University, NSW 2109, Australia 3. Neuroscience Research Australia (NeuRA), NSW 2031, Australia 4. School of Medical Sciences, University of New South Wales, NSW 2052, Australia

Frontotemporal dementia (FTD) and Alzheimer’s disease (AD) share the pathological hallmark of intracellular neurofibrillary tangles (NFT), which result from the hyperphosphorylation of microtubule associated protein tau (i.e. caused by mutations in the MAPT gene). The P301S mutation in human tau carried by TAU58/2 transgenic mice results in brain pathology and behavioural deficits relevant to FTD and AD. The phytocannabinoid cannabidiol (CBD) exhibits properties potentially beneficial for multiple pathological processes in FTD and AD. We treated 14-month-old female TAU58/2 transgenic and wild type-like (WT) littermates with 100 mg/kg CBD for three weeks via intraperitoneal injection prior to conducting a battery of behavioural tests relevant to FTD and AD. These included the elevated plus maze, motor function tests, the social preference test, the cheeseboard task and fear conditioning. TAU58/2 females had reduced motor function, bodyweight and anxiety compared to WT. The moderate reduction in sociability, and defective spatial recognition memory of vehicle-treated transgenic mice was restored by CBD treatment. Average speed on the cheeseboard was increased in CBD-treated mice although this effect was strongest in TAU58/2 mice. CBD treatment also reduced anxiety-like behaviour and reduce contextual fear-associated freezing in both TAU58/2 and WT mice. Chronic administration of 100 mg/kg CBD ameliorated several dementia-relevant phenotypes in TAU58/2 mice, suggesting this phytocannabinoid may be useful for treating aspects of tauopathy- related neurodegenerative diseases.

61 Abstracts

MISS YI LING LOW

Drug Delivery, Disposition, and Dynamics - Monash Institute of Pharmaceutical Science Faculty of Pharmacy and Pharmaceutical Sciences - Monash University [email protected]

The involvement of fatty acid-binding proteins in docosahexaenoic acid uptake in microglia Yi Ling Low1, Yijun Pan1, Jennifer L. Short2, Joseph A. Nicolazzo1 1. Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia 2. Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia. Email addresses: Yi Ling Low: [email protected], Yijun Pan: [email protected], Jennifer Short: [email protected], Joseph Nicolazzo: [email protected] The overactivation of microglia leads to the excessive release of proinflammatory mediators, which is detrimental to brain health. Docosahexaenoic acid (DHA) has been shown to alleviate neuroinflammation by inhibiting the release of proinflammatory mediators from microglia. Therefore, the uptake of DHA into microglia is essential for reducing neuroinflammation. Cytoplasmic carrier proteins, fatty acid-binding proteins (FABPs), are involved in DHA trafficking in other cell types. Whether FABPs are expressed and involved in DHA trafficking into microglia remains to be investigated. This study focused on screening whether various FABP isoforms are expressed in microglia, and whether they are involved in the uptake of DHA into microglia. Using immortalised mouse microglia (BV-2) cells, RT-qPCR and western blotting were used to quantitatively determine the mRNA and protein levels of the 10 known FABP isoforms. FABP3, FABP4, and FABP5 were expressed at both the mRNA and protein level in BV-2 cells. A genetic knockdown approached was taken to investigate the involvement of these FABP isoforms in the microglial uptake of DHA-d5, a surrogate of DHA. DHA-d5 uptake in BV-2 cells was quantified using an optimised LC-MS/MS technique. Following 77.5-92.3% (at mRNA level) and 45.4-81.7% (at protein level) knockdown of FABP3, FABP4, and FABP5 at 48 hours, no changes in DHA-d5 uptake into microglia was observed at 2 min. This suggested the involvement of other microglial DHA uptake mechanisms, such as the possible involvement of membrane transporters like fatty acid transport proteins. Therefore, further DHA uptake mechanisms present in microglia will need to be further investigated. References: Laye et al. J. Nutr. Health Aging. 2015 Heras-Sandoval et al. J. Neuroinflammation. 2016 Pan et al. Mol. Pharm. 2015 Lee et al. Pharm. Res. 2015 Serafim et al. Molecules. 2019

62 Abstracts

DR NIRMA PERERA

Florey Institute of Neuroscience [email protected]

Cleaning house: monitoring the flux of cellular housekeeping autophagy pathway in amyotrophic lateral sclerosis (ALS) mice Nirma Perera1, Fatemeh Zanganeh1, Mouna Haidar1, Victoria McLeod1, Brittany Cuic1, Doris Tomas1, Bradley Turner1 1Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia

Autophagy pathway targets protein aggregates for clearance by packaging them into autophagosomes and delivering to lysosomes for degradation. Autophagy impairment is central to pathology of amyotrophic lateral sclerosis (ALS). However, it is unclear whether activation or inhibition of autophagy would be beneficial. Measuring autophagic flux, the key measure of autophagic degradation activity quantified by ratio of autolysosomes to autophagosomes will allow therapeutic modulation of autophagy in ALS. We monitored autophagic flux in motor neurons (MNs) of ALS SOD1G93A mice, using a novel autophagy reporter mouse model. SOD1G93A mice were crossbred with LC3 autophagy reporter mice. Brains and lumbar spinal cords were analysed with microscopy and IMARIS 3D image analysis software. Spinal MNs had a much higher rate of autophagic flux than corticospinal MNs. For example, an adult spinal α-MN contained ~700 autolysosomes and 6 autophagosomes whereas a corticospinal MN, ~7 autolysosomes and 30 autophagosomes, representing differential basal autophagy rates in varying MN populations. In SOD1 mice, spinal MNs a had significant 68% reduction in autophagic flux compared to controls from pre-symptomatic 90 days of age, persisting to end stage (p<0.05), due to accumulating autophagosomes. Corticospinal MN flux impairment preceded the spinal impairment, starting at 60 days of age with a 44% reduction also continuing to end stage (p<0.05). Our novel mouse model resolves that autophagic flux is chronically compromised in vulnerable motor neurons of SOD1G93A mice. Our findings rationalize the use of autophagy enhancers particularly those targeting later stages of autophagy pathway, with early treatment in ALS disease process.

63 Abstracts

DR ASHEETA PRASAD

School of Psychology - UNSW [email protected]

Cellular changes in the substantia nigra and subthalamic nucleus in Parkinson’s disease following deep brain stimulation

Asheeta A Prasad*1, Srestha Mazumder 1, Anita Y Bahar2, Claire E Shepherd*2,3 1School of Psychology, University of New South Wales 2Neuroscience Research Australia 3School of Medicine, University of New South Wales *Joint senior author

Background: Parkinson’s disease (PD) is a progressive neurodegenerative disorder; pathologically hallmarked by the loss of dopamine neurons in the substantia nigra (SN) and alpha- synuclein aggregation. Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is a common target to treat the motor symptoms in PD. However, we have little understanding of the cellular changes in the STN during PD and our knowledge concerning the impact of DBS on the STN and SN is limited. Objective: To examine cellular changes in the SN and STN in PD patients with and without STN- DBS treatment. Method: Post-mortem brain tissue from 7 PD non STN-DBS patients, 5 PD STN-DBS and 6 age- matched controls were stained with markers for neuroinflammation (astrocytes and microglia) and neurodegeneration (tyrosine hydroxylase, alpha-synuclein and neuronal loss). Changes were assessed using quantitative and semi-quantitative microscopy techniques. Results: This study confirms previous findings of significant neuronal loss (p < 0.001), increased neuroinflammation (p < 0.018) and increased alpha-synuclein (p < 0.001) in the SN in PD patients compared to controls. Significant neuronal loss or neuroinflammation was not seen in the STN in PD (p > 0.494). Deep brain stimulation did not alter neuronal loss or neuroinflammation in either the SN or STN in PD (p > 0.639 for all variables). However, increased alpha-synuclein deposition was observed in the STN following STN-DBS (p = 0.030). Conclusion: This study does not identify any change in inflammation or neuronal loss in the STN or SN following STN-DBS. However, significant alpha-synuclein pathology was present in the STN in all PD cases and was significantly increased in cases with STN-DBS.

References Hardman CD, McRitchie DA, Halliday GM, Morris JGL. The Subthalamic Nucleus in Parkinson's Disease and Progressive Supranuclear Palsy. Journal of Neuropathology & Experimental Neurology 1997;56(2):132-142. Kordower JH, Olanow CW, Dodiya HB, et al. Disease duration and the integrity of the nigrostriatal system in Parkinson’s disease. Brain 2013;136(8):2419-2431.

64 Abstracts

ANISHCHAL PRATAP

Discipline of Pathology - The University of Sydney [email protected]

Robust astrocytic leptin and leptin receptor expression in the 5XFAD mouse model of Alzheimer’s disease. Anishchal Pratap1,2 and R. M. Damian Holsinger1,2 1. Laboratory of Molecular Neuroscience and Dementia, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, NSW, Australia 2. Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, Australia [email protected] [email protected] Alzheimer’s disease (AD) is a chronic neurodegenerative disorder, characterized by the deposition of amyloid plaques and neurofibrillary tangles. Obesity and type 2 diabetes are metabolic syndromes that share similar pathological features with AD including cortical atrophy and chronic neuroinflammation. Leptin, an adipocytokine secreted by adipocytes, regulates satiety and energy metabolism via its receptor, LepR. To determine the role of leptin and LepR in metabolic impairment in the AD brain, we examined the expression of these two proteins in the brains of the 5XFAD mouse model of AD. Cortical brain tissue acquired from young (10-12-weeks; n=6) and old (48-52-weeks; n=6) transgenic (Tg), and age-matched wild-type (WT) controls for both age groups (young-WT, n=6; old-WT, n=6), were analyzed employing immunofluorescent staining. Leptin and LepR expression in neurons and endothelia were observed in cortical tissue of young-Tg, young- WT and old-WT mice. Marked diminution of neuronal leptin and LepR expression were observed in old-Tg mice. Conversely, robust astrocytic leptin and LepR expression was observed in old-Tg mice compared to young-Tg animals. Our findings indicate that LepR signalling may regulate the reactive state of astrocytes and aid in the removal and degradation of amyloid in the AD brain. Overall, increased astrocytic and decreased neuronal expression of leptin and LepR in the 5XFAD brain provides further evidence of metabolic impairment as a pathological characteristic of AD.

65 Abstracts

MS LAURA REALE

Menzies Institute for Medical Research - University of Tasmania [email protected]

Can localized stimulation of the motor cortex through TMS paradigms rescue the disease phenotype in a familial mouse model of ALS? Laura Reale1, Carlie Cullen1, Kaylene Young1, Tracey Dickson1, Catherine Blizzard1 1. Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS 7000 [email protected] Introduction: Excitability alterations in the motor cortex are among the earliest detectable clinical changes in amyotrophic lateral sclerosis (ALS) (1-2). It is unknown whether increased or decreased excitation causes neurodegeneration or if manipulating excitability may be an effective therapeutic strategy. To address this question, we applied transcranial magnetic stimulation (TMS) to safely and non-invasively increase or decrease neuronal firing (3). Objectives: This research aimed to determine whether altering the excitability of the motor cortex, by applying excitatory or inhibitory patterns of TMS could improve outcomes for mice that develop an ALS-like disease phenotype. Methods: This study utilised a familial mouse model of ALS expressing mutant TDP-43A315T. Low intensity TMS was applied to the motor cortex using a custom built rodent TMS coil. It was administered as an intermittent (inducing excitation) or continuous (inducing inhibition) theta burst (iTBS/cTBS) pattern or sham stimulation, 5 days a week across 38 days. The effect of TMS was determined by evaluating rotarod performance, hindlimb clinical score and survival. Results: Preliminary analysis revealed that iTBS (n=9) improved rotarod performance by week 2 (p=0.038) and week 3 (p=0.040) of TMS. Furthermore, iTBS improved clinical score relative to sham (n=7; p<0.0001) and cTBS (n=9; p<0.01) mice but did not alter survival. cTBS did not alter rotarod performance or clinical score, but decreased survival (p<0.05). Discussion: Altering cortical excitability through different TMS paradigms alleviated motor phenotypes in this model and altered survival. Therefore, selectively and differentially enhancing excitability may be a feasible therapeutic strategy to stop ALS progression. References: 1. Menon, P., Higashihara, M., van den Bos, M., Geevasinga, N., Kiernan, M.C. and Vucic, S., 2020. Cortical hyperexcitability evolves with disease progression in ALS. Annals of Clinical and Translational Neurology, 7(5), pp.733-741. 2. Vucic, S., Nicholson, G.A. and Kiernan, M.C., 2008. Cortical hyperexcitability may precede the onset of familial amyotrophic lateral sclerosis. Brain, 131(6), pp.1540-1550. 3. Tang, A., Thickbroom, G. and Rodger, J., 2017. Repetitive transcranial magnetic stimulation of the brain: mechanisms from animal and experimental models. The Neuroscientist, 23(1), pp.82- 94.

66 Abstracts

MR MONOKESH KUMER SEN

School of Medicine - Western Sydney University [email protected]

Histological and proteomic investigations in the visual pathway of cuprizone- fed mice

Mohammed SM. Almuslehi1,2¥, Monokesh K. Sen1¥, Peter J. Shortland3, David A. Mahns1* and Jens R. Coorssen4*

2. School of Medicine, Western Sydney University, Penrith, NSW, Australia. 3. Department of Physiology, College of Veterinary Medicine, University of Diyala, Diyala, Iraq. 4. School of Science, Western Sydney University, Penrith, NSW, Australia. 5. Department of Health Sciences, Faculty of Applied Health Sciences, and Department of Biological Sciences, Faculty of Mathematics and Science, Brock University, St. Catharines, Ontario, ON L2S 3A1, Canada. ¥Contributed equally *Co-corresponding authors

Changes in visual perception are frequently used as early diagnostic markers of multiple sclerosis (MS). Using cuprizone (CPZ) ingestion in mice as a model of central nervous system demyelination, this study investigated components of the visual pathway using histology and proteomics following 0.1% CPZ feeding of young adult mice for 12 weeks. Histological analysis of the optic nerve, pretectal nucleus, visual cortex, lateral geniculate nucleus, and superior colliculus found limited changes in myelination and gliosis. However, top-down proteomic analysis (2D gels coupled with liquid chromatography and tandem mass spectrometry) identified 75 proteoforms that changed in abundance. Literature mining confirmed the relevance of the identified proteoforms with regard to proteins previously identified in MS and animal models of the disorder. Moreover, bioinformatic analysis revealed the involvement of the identified proteoforms in cytoskeleton organization, metabolic dysregulation, protein aggregation and axonal support. Collectively, slow low-dose CPZ-feeding revealed marked proteomic alternations that may precede histological changes in visual pathway.

Key words: Optic neuritis, multiple sclerosis, demyelination, top-down proteomics, bioinformatics, proteoforms

67 Abstracts

DR STEPHANIE SHEPHEARD

College of Medicine & Public Health – Flinders University [email protected]

Urinary Neopterin as a Potential Biomarker for Motor Neurone Disease Karnaros V.1, Shepheard S.R.1, Dubowsky, M.1, Beben, B.2, Malaspina, A.4, Schultz, D.3, Rogers, M-L.1 1. College of Medicine and Public Health, Flinders Health and Medical Research Institute and Flinders University, Adelaide, , Australia; 2. University of South Australia and South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; 3. Flinders Medical Centre, Bedford Park, South Australia, Australia. 4. National Hospital for Neurology & Neurosurgery, University College London, London, United Kingdom. Objective markers of disease progression and prognosis are advantageous in assessing treatment effectiveness in clinical trials. Our laboratory discovered urinary p75ECD as a biochemical marker that changes as disease progresses1 and can be used to test treatment efficacy2 in Motor Neuron Disease (MND). Using p75ECD in a urinary biomarker panel with additional candidates will help to subgroup patients in trials, improve efficacy detection, and increase understanding of disease processes. With immune dysfunction known to occur in MND, one such biomarker candidate is neopterin, a marker of cell mediated immune response which is secreted in urine. We aimed to analyse neopterin as a prognostic and progression biomarker of MND by measuring urinary neopterin levels at first visit to neurologist (baseline) and over disease progression using enzyme-linked immunoassays and comparing these values to clinical data including ALSFRS-r scores, age, gender, disease onset; p75ECD levels in people with MND; and healthy individuals. To date, results show urinary neopterin levels are higher in people with MND (n=37), than healthy individuals at first clinic visit (n=18, 162.9±12.9μmol Neopterin/mol creatinine vs 85.3±8.9μmol Neopterin/mol creatinine, unpaired t test, p=0.0003), and urinary neopterin and p75ECD levels are correlated (baseline, r=0.52, p=0.0009). The analysis of prognostic and progression value of neopterin is ongoing. Neopterin shows promise as an additional urinary biomarker of MND, and may be useful as an immune response marker. References: 1. Shepheard, et al., Neurology 2017;88:1137-1143. 2. Gold, et al., ALS & FTD 2019;63:1-10. 3. Sussmuth, et al., Curr Med Chem. 2008;15(18):1788–801.

68 Abstracts

DR MOMO VUYISICH

Viome Inc [email protected]

Viome is a private company that was created out of frustration for the lack of understanding, preventing, and curing chronic diseases and cancer. Over the last four years, we have built a powerful systems biology platform that can be used to identify the root causes of chronic diseases, develop diagnostics and companion diagnostics, and develop novel preventative and curative therapeutic avenues. The platform includes: • Collection and ambient temperature transportation of any clinical sample. Stool, saliva, whole blood and vaginal swabs can be collected at home. Viome provides all collection kits and includes preservation reagents. • Highly accurate RNA sequencing analysis that are fully automated and performed in a clinical laboratory. • Highly accurate cloud-based bioinformatic analysis that perform strain-level taxonomic classification and quantitative gene expression. • An advanced machine learning team with some of the leading data scientists. • Ability to rapidly convert the knowledge from clinical research into a commercial product. Viome has already released several products and has plans for more. This systems biology platform is now available to anyone, worldwide, via Viome Research Institute Grants Program. This presentation will describe the details of the platform and several products developed so far.

69 Abstracts

MS SHARLYNN WU

Macquarie Centre for Motor Neuron Disease Research - Macquarie University [email protected]

Elucidating the potential pathogenicity of novel MND candidate variants in vitro Sharlynn Wu, Jennifer Fifita, Emily McCann, Natalie Grima, Dominic Rowe, Ian Blair and Shu Yang 1. Macquarie University Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia

Disease gene discoveries drive our understanding of disease mechanisms underlying amyotrophic lateral sclerosis (ALS). Despite recent novel ALS gene discoveries, 40% of Australian ALS families remain unsolved because they often do not exhibit classical Mendelian inheritance patterns and are challenging to solve via traditional genetic analyses alone. Thus, in vitro and in vivo studies are critical in assessing the potential pathogenicity of novel candidate genes. In this study, we developed and validated the utility of an in vitro functional pipeline for the prioritisation of novel ALS candidate genes based on their capacity to induce ALS-like pathologies in cell lines. The pipeline consists of a panel of cell-based assays that include immunocytochemistry to examine localisation, morphology, TDP-43 interaction and inclusion formation, flow cytometry to assess toxicity, and western blotting to study aberrant protein degradation and solubility changes. We applied this pipeline to five candidate genes from an ALS family with reduced disease penetrance and that was negative for known ALS genes. Two candidates were prioritised based on their higher toxicity than wild type, the formation of detergent-insoluble inclusions and co-localisation with TDP-43. Interestingly, the genomic proximity of these genes suggest that they were likely co- inherited. Thus, it was prudent to also investigate the compound effect of both candidate on potential pathogenicity using the in vitro pipeline. Preliminary results suggest that inclusion formation was exacerbated in co-transfected cells that co-expressed both candidate genes. Overall, we have demonstrated the utility of a functional prioritization pipeline and successfully prioritised two novel candidate ALS genes.

70 Abstracts

DR FATEMEH ZANGANEH

University of Melbourne Fatemeh [email protected]

Gene expression profiling identifies excitotoxic mechanisms initiated very early in motor neurons of the SOD1G93A mouse model of ALS

Fatemeh Zanganeh, Samantha K. Barton, Christopher R Bye, and Bradley J Turner Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne VIC 3052, Australia

ALS presents in mid- to late-life, although there is increasing evidence from patients and animal models for a long preclinical period of motor neuron susceptibility and dysfunction, before clinical diagnosis. The stage of life at which motor neuron vulnerability is conferred in ALS remains unclear. Thus, deciphering the earliest molecular mechanisms of motor neuron vulnerability are likely to be key to developing disease-modifying treatments. Here, we employed RNA sequencing to determine how early motor neuron vulnerability is conferred in a mouse model of ALS. Transgenic SOD1G93A mice were crossed with HB9:GFP reporter mice to allow unambiguous isolation of motor neurons using fluorescence activated cell sorting (FACS). Gene expression profiling of isolated motor neurons revealed transcriptional dysregulation in SOD1G93A mice as early as embryonic (E) day 12.5 with the majority of differentially expressed genes involved in RNA processing and AMPA receptor signalling. Our findings have the potential to reveal key genes, pathways and biological networks responsible for selective neuronal tolerance, vulnerability and dysfunction in the preclinical period of ALS, in addition to identifying meaningful new drug targets for future intervention studies.

71

Macquarie University is a vibrant hub of intellectual thinkers, all working towards a brighter future for our communities and our planet.

A PLACE OF INSPIRATION Macquarie is uniquely located in the heart of Australia’s largest high-tech precinct, a thriving locale which is predicted to double in size in the next 20 years to become the fourth largest CBD in Australia. Our campus spans 126 hectares, with open green space that gives our community the freedom to think and grow. We are home to fantastic facilities with excellent transport links to the city and suburbs, supported by an on-campus train station.

RENOWNED FOR EXCELLENCE We are ranked among the top two per cent of universities in the world, and with a 5-star QS rating, we are renowned for producing graduates that are among the most sought after professionals in the world.

A PROUD TRADITION OF DISCOVERY Our enviable research efforts are brought to life by renowned researchers whose audacious solutions to issues of global significance are benefiting the world we live in.

BUILDING SUCCESSFUL GRADUATES Our pioneering approach to teaching and learning is built around a connected learning community: our students are considered partners and co-creators in their learning experience.

FIND OUT MORE Macquarie University NSW 2109 Australia T: +61 (2) 9850 7111 mq.edu.au

CRICOS Provider 00002J