Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jennifer A. Fifita is active.

Publication


Featured researches published by Jennifer A. Fifita.


Nature Neuroscience | 2013

Exome sequencing to identify de novo mutations in sporadic ALS trios

Alessandra Chesi; Brett T. Staahl; Ana Jovičić; Julien Couthouis; Maria Fasolino; Alya R. Raphael; Tomohiro Yamazaki; Laura Elias; Meraida Polak; Crystal Kelly; Kelly L. Williams; Jennifer A. Fifita; Nicholas J. Maragakis; Garth A. Nicholson; Oliver D. King; Robin Reed; Gerald R. Crabtree; Ian P. Blair; Jonathan D. Glass; Aaron D. Gitler

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease whose causes are still poorly understood. To identify additional genetic risk factors, we assessed the role of de novo mutations in ALS by sequencing the exomes of 47 ALS patients and both of their unaffected parents (n = 141 exomes). We found that amino acid–altering de novo mutations were enriched in genes encoding chromatin regulators, including the neuronal chromatin remodeling complex (nBAF) component SS18L1 (also known as CREST). CREST mutations inhibited activity-dependent neurite outgrowth in primary neurons, and CREST associated with the ALS protein FUS. These findings expand our understanding of the ALS genetic landscape and provide a resource for future studies into the pathogenic mechanisms contributing to sporadic ALS.


Nature Communications | 2016

CCNF mutations in amyotrophic lateral sclerosis and frontotemporal dementia

Kelly L. Williams; Simon Topp; Shu Yang; Bradley Smith; Jennifer A. Fifita; Sadaf T. Warraich; Katharine Y. Zhang; Natalie E. Farrawell; Caroline Vance; Xun Hu; Alessandra Chesi; Claire S. Leblond; Albert Lee; Stephanie L. Rayner; Vinod Sundaramoorthy; Carol Dobson-Stone; Mark P. Molloy; Marka van Blitterswijk; Dennis W. Dickson; Ronald C. Petersen; Neill R. Graff-Radford; Bradley F. Boeve; Melissa E. Murray; Cyril Pottier; Emily K. Don; Claire Winnick; Emily P. McCann; Alison L. Hogan; Hussein Daoud; Annie Levert

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are overlapping, fatal neurodegenerative disorders in which the molecular and pathogenic basis remains poorly understood. Ubiquitinated protein aggregates, of which TDP-43 is a major component, are a characteristic pathological feature of most ALS and FTD patients. Here we use genome-wide linkage analysis in a large ALS/FTD kindred to identify a novel disease locus on chromosome 16p13.3. Whole-exome sequencing identified a CCNF missense mutation at this locus. Interrogation of international cohorts identified additional novel CCNF variants in familial and sporadic ALS and FTD. Enrichment of rare protein-altering CCNF variants was evident in a large sporadic ALS replication cohort. CCNF encodes cyclin F, a component of an E3 ubiquitin–protein ligase complex (SCFCyclin F). Expression of mutant CCNF in neuronal cells caused abnormal ubiquitination and accumulation of ubiquitinated proteins, including TDP-43 and a SCFCyclin F substrate. This implicates common mechanisms, linked to protein homeostasis, underlying neuronal degeneration.


Human Molecular Genetics | 2015

Defects in optineurin- and myosin VI-mediated cellular trafficking in amyotrophic lateral sclerosis

Vinod Sundaramoorthy; Adam K. Walker; Vanessa Tan; Jennifer A. Fifita; Emily P. McCann; Kelly L. Williams; Ian P. Blair; Gilles J. Guillemin; Manal A. Farg; Julie D. Atkin

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder primarily affecting motor neurons. Mutations in optineurin cause a small proportion of familial ALS cases, and wild-type (WT) optineurin is misfolded and forms inclusions in sporadic ALS patient motor neurons. However, it is unknown how optineurin mutation or misfolding leads to ALS. Optineurin acts an adaptor protein connecting the molecular motor myosin VI to secretory vesicles and autophagosomes. Here, we demonstrate that ALS-linked mutations p.Q398X and p.E478G disrupt the association of optineurin with myosin VI, leading to an abnormal diffuse cytoplasmic distribution, inhibition of secretory protein trafficking, endoplasmic reticulum (ER) stress and Golgi fragmentation in motor neuron-like NSC-34 cells. We also provide further insight into the role of optineurin as an autophagy receptor. WT optineurin associated with lysosomes and promoted autophagosome fusion to lysosomes in neuronal cells, implying that it mediates trafficking of lysosomes during autophagy in association with myosin VI. However, either expression of ALS mutant optineurin or small interfering RNA-mediated knockdown of endogenous optineurin blocked lysosome fusion to autophagosomes, resulting in autophagosome accumulation. Together these results indicate that ALS-linked mutations in optineurin disrupt myosin VI-mediated intracellular trafficking processes. In addition, in control human patient tissues, optineurin displayed its normal vesicular localization, but in sporadic ALS patient tissues, vesicles were present in a significantly decreased proportion of motor neurons. Optineurin binding to myosin VI was also decreased in tissue lysates from sporadic ALS spinal cords. This study therefore links several previously described pathological mechanisms in ALS, including defects in autophagy, fragmentation of the Golgi and induction of ER stress, to disruption of optineurin function. These findings also indicate that optineurin-myosin VI dysfunction is a common feature of both sporadic and familial ALS.


Neurobiology of Aging | 2015

Novel TBK1 truncating mutation in a familial amyotrophic lateral sclerosis patient of Chinese origin

Kelly L. Williams; Emily P. McCann; Jennifer A. Fifita; Katharine Y. Zhang; Emma L. Duncan; Paul Leo; Mhairi Marshall; Dominic B. Rowe; Garth A. Nicholson; Ian P. Blair

Missense and frameshift mutations in TRAF family member-associated NF-kappa-B activator (TANK)-binding kinase 1 (TBK1) have been reported in European sporadic and familial amyotrophic lateral sclerosis (ALS) cohorts. To assess the role of TBK1 in ALS patient cohorts of wider ancestry, we have analyzed whole-exome sequence data from an Australian cohort of familial ALS (FALS) patients and controls. We identified a novel TBK1 deletion (c.1197delC) in a FALS patient of Chinese origin. This frameshift mutation (p.L399fs) likely results in a truncated protein that lacks functional domains required for adapter protein binding, as well as protein activation and structural integrity. No novel or reported TBK1 mutations were identified in FALS patients of European ancestry. This is the first report of a TBK1 mutation in an ALS patient of Asian origin and indicates that sequence variations in TBK1 are a rare cause of FALS in Australia.


Clinical Genetics | 2017

The genotype–phenotype landscape of familial amyotrophic lateral sclerosis in Australia

Emily P. McCann; Kelly L. Williams; Jennifer A. Fifita; Ingrid S. Tarr; Jody O'Connor; Dominic B. Rowe; Garth A. Nicholson; Ian P. Blair

Amyotrophic lateral sclerosis (ALS) is a clinically and genetically heterogeneous fatal neurodegenerative disease. Around 10% of ALS cases are hereditary. ALS gene discoveries have provided most of our understanding of disease pathogenesis. We aimed to describe the genetic landscape of ALS in Australia by assessing 1013 Australian ALS patients for known ALS mutations by direct sequencing, whole exome sequencing or repeat primed polymerase chain reaction. Age of disease onset and disease duration were used for genotype–phenotype correlations. We report 60.8% of Australian ALS families in this cohort harbour a known ALS mutation. Hexanucleotide repeat expansions in C9orf72 accounted for 40.6% of families and 2.9% of sporadic patients. We also report ALS families with mutations in SOD1 (13.7%), FUS (2.4%), TARDBP (1.9%), UBQLN2 (.9%), OPTN (.5%), TBK1 (.5%) and CCNF (.5%). We present genotype–phenotype correlations between these genes as well as between gene mutations. Notably, C9orf72 hexanucleotide repeat expansion positive patients experienced significantly later disease onset than ALS mutation patients. Among SOD1 families, p.I114T positive patients had significantly later onset and longer survival. Our report highlights a unique spectrum of ALS gene frequencies among patients from the Australian population, and further, provides correlations between specific ALS mutations with disease onset and/or duration.


Human Molecular Genetics | 2017

Expression of ALS/FTD-linked mutant CCNF in zebrafish leads to increased cell death in the spinal cord and an aberrant motor phenotype

Alison L. Hogan; Emily K. Don; Stephanie L. Rayner; Albert Lee; Angela S. Laird; Maxinne Watchon; Claire Winnick; Ingrid S. Tarr; Marco Morsch; Jennifer A. Fifita; Serene S. L. Gwee; Isabel Formella; Elinor Hortle; Kristy C. Yuan; Mark P. Molloy; Kelly L. Williams; Garth A. Nicholson; Roger S. Chung; Ian P. Blair; Nicholas J. Cole

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive, fatal neurodegenerative disease characterised by the death of upper and lower motor neurons. Approximately 10% of cases have a known family history of ALS and disease-linked mutations in multiple genes have been identified. ALS-linked mutations in CCNF were recently reported, however the pathogenic mechanisms associated with these mutations are yet to be established. To investigate possible disease mechanisms, we developed in vitro and in vivo models based on an ALS-linked missense mutation in CCNF. Proteomic analysis of the in vitro models identified the disruption of several cellular pathways in the mutant model, including caspase-3 mediated cell death. Transient overexpression of human CCNF in zebrafish embryos supported this finding, with fish expressing the mutant protein found to have increased levels of cleaved (activated) caspase-3 and increased cell death in the spinal cord. The mutant CCNF fish also developed a motor neuron axonopathy consisting of shortened primary motor axons and increased frequency of aberrant axonal branching. Importantly, we demonstrated a significant correlation between the severity of the CCNF-induced axonopathy and a reduced motor response to a light stimulus (photomotor response). This is the first report of an ALS-linked CCNF mutation in vivo and taken together with the in vitro model identifies the disruption of cell death pathways as a significant consequence of this mutation. Additionally, this study presents a valuable new tool for use in ongoing studies investigating the pathobiology of ALS-linked CCNF mutations.


Amyotrophic Lateral Sclerosis | 2017

A novel amyotrophic lateral sclerosis mutation in OPTN induces ER stress and Golgi fragmentation in vitro

Jennifer A. Fifita; Kelly L. Williams; Vinod Sundaramoorthy; Emily P. McCann; Garth A. Nicholson; Julie D. Atkin; Ian P. Blair

Abstract Mutations in the optineurin gene (OPTN) have been identified in a small proportion (<1%) of sporadic and familial ALS cases, and the exact role of optineurin in the pathogenesis of ALS remains unclear. To further examine the role of OPTN in ALS, we sought to identify novel ALS variants in OPTN and examine their potential for pathogenicity in vitro. Whole exome sequence data from 74 familial ALS cases were analysed for the presence of novel OPTN mutations. Pathogenicity was assessed by analysing effects on Golgi fragmentation, endoplasmic reticulum (ER) stress-linked CHOP activation, and cellular localization of optineurin in motor neuron-like NSC-34 cells expressing mutant optineurin. We identified a novel heterozygous missense mutation in OPTN (c.883G > T, p.Val295Phe) in a single familial ALS case. This mutation induced recognized cellular features of ALS pathogenesis including Golgi fragmentation and ER stress in NSC-34 cells. In conclusion, the identification of a novel OPTN mutation in an Australian ALS family, and its capacity to induce ALS-like pathological features in vitro, further strengthens evidence for the role of optineurin in the pathogenesis of ALS.


Neurotoxicity Research | 2015

Evaluation of Skin Fibroblasts from Amyotrophic Lateral Sclerosis Patients for the Rapid Study of Pathological Features

Shu Yang; Katharine Y. Zhang; Ruvini Kariawasam; Monique Bax; Jennifer A. Fifita; Lezanne Ooi; Justin J. Yerbury; Garth A. Nicholson; Ian P. Blair

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterised by the progressive degeneration of brain and spinal cord motor neurons. Ubiquitin–proteasome system (UPS) dysfunction and oxidative stress have been implicated in ALS pathogenesis. However, it is unknown whether the defects in these pathways extend to non-neuronal tissues such as fibroblasts. Fibroblasts, unlike neuronal tissue, are readily available and may hold potential for short-term, rapid diagnostic and prognostic purposes. We investigated whether primary skin fibroblasts from ALS patients share, or can be manipulated to develop, functional and pathological abnormalities seen in affected neuronal cells. We inhibited UPS function and induced oxidative stress in the fibroblasts and found that ALS-related cellular changes, such as aggregate formation and ubiquitination of ALS-associated proteins (TDP-43 and ubiquilin 2), can be reproduced in these cells. Higher levels of TDP-43 ubiquitination, as evident by colocalization between TDP-43 and ubiquitin, were found in all six ALS cases compared to controls following extracellular insults. In contrast, colocalization between ubiquilin 2 and ubiquitin was not markedly different between ALS cases and control. A UPS reporter assay revealed UPS abnormalities in patient fibroblasts. Despite the presence of ALS-related cellular changes in the patient fibroblasts, no elevated toxicity was observed. This suggests that aggregate formation and colocalization of ALS-associated proteins may be insufficient alone to confer toxicity in fibroblasts used in the present study. Chronic exposure to ALS-linked stresses and the ALS-linked cellular pathologies may be necessary to breach an unknown threshold that triggers cell death.


Acta Neuropathologica | 2018

Accumulation of dysfunctional SOD1 protein in Parkinson’s disease is not associated with mutations in the SOD1 gene

Benjamin G. Trist; Jennifer A. Fifita; Sarah Freckleton; Dominic J. Hare; Simon J.G. Lewis; Glenda M. Halliday; Ian P. Blair; Kay L. Double

biochemical pathway contributing to neuron loss in both disorders. This provokes the question of whether mutant SOD1 is a feature of Parkinson’s disease. In the time since our report was published, we have conducted genotyping experiments on the 17 idiopathic Parkinson’s disease cases in which we observed SOD1 dysfunction and aggregation to identify possible mutations in SOD1, using our previously reported methods for genetic profiling of SOD1 in fALS [7]. No sequence variations from wild type SOD1 were identified in any of these cases of Parkinson’s disease. This finding is consistent with the single study reported to date that failed to identify SOD1 mutations in index familial Parkinson’s disease patients representing 23 genealogies [1]. One participant in our Parkinson’s disease cohort possessed a known intronic deletion found in healthy individuals (dbSNP rs398081559, c.573 + 88 del A), but did not represent an outlier within our published datasets for SOD1 misfolding and deposition [10]. The absence of mutations in SOD1 in our Parkinson’s disease cohort indicates that aggregated SOD1 in these cases is wild type protein. This negative result is important, as it demonstrates that wild type, and mutant, SOD1 can express comparable dysfunctional activities and abnormal conformations in Parkinson’s disease and fALS, respectively. These perturbations may represent a common basis for neuronal vulnerability in these disorders through a common molecular pathway that may involve either wild type or mutant SOD1. The formation of a thermally stable SOD1 homodimer is essential for catalytic dismutation of superoxide to hydrogen peroxide and oxygen, mediated by two copper (II) ions. The binding of these copper (II) ions, along with the binding of two zinc (II) ions and the formation of an intramonomeric metal-stabilized disulfide bridge (Cys57-Cys146), affords the protein its exceptional thermodynamic stability. Consequently, reduced copper binding to SOD1 results in a In the first issue of Volume 134 of Acta Neuropathologica, we reported the substantial accumulation of abnormal deposits of superoxide dismutase 1 (SOD1) protein in the idiopathic Parkinson’s disease brain, reflecting the pattern of neuronal loss in this disorder more closely than that of α-synuclein [10]. We presented evidence of catalytically dysfunctional, misfolded conformations of soluble and aggregated SOD1 protein in degenerating Parkinson’s disease brain regions, similar to neurotoxic SOD1 proteinopathy in the spinal cord [8] and substantia nigra pars compacta (SNc) of familial amyotrophic lateral sclerosis (fALS) patients with mutations in the SOD1 gene. Comparable changes in SOD1 structure and function suggest a common


Biology Open | 2018

Neuronal cell culture from transgenic zebrafish models of neurodegenerative disease.

Jamie Rae Acosta; Maxinne Watchon; Kristy C. Yuan; Jennifer A. Fifita; Adam J. Svahn; Emily K. Don; Claire Winnick; Ian P. Blair; Garth A. Nicholson; Nicholas J. Cole; Claire Goldsbury; Angela S. Laird

ABSTRACT We describe a protocol for culturing neurons from transgenic zebrafish embryos to investigate the subcellular distribution and protein aggregation status of neurodegenerative disease-causing proteins. The utility of the protocol was demonstrated on cell cultures from zebrafish that transgenically express disease-causing variants of human fused in sarcoma (FUS) and ataxin-3 proteins, in order to study amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia type-3 (SCA3), respectively. A mixture of neuronal subtypes, including motor neurons, exhibited differentiation and neurite outgrowth in the cultures. As reported previously, mutant human FUS was found to be mislocalized from nuclei to the cytosol, mimicking the pathology seen in human ALS and the zebrafish FUS model. In contrast, neurons cultured from zebrafish expressing human ataxin-3 with disease-associated expanded polyQ repeats did not accumulate within nuclei in a manner often reported to occur in SCA3. Despite this, the subcellular localization of the human ataxin-3 protein seen in cell cultures was similar to that found in the SCA3 zebrafish themselves. The finding of similar protein localization and aggregation status in the neuronal cultures and corresponding transgenic zebrafish models confirms that this cell culture model is a useful tool for investigating the cell biology and proteinopathy signatures of mutant proteins for the study of neurodegenerative disease. Summary: This article describes the optimization and validation of a protocol for culturing of neurons from transgenic zebrafish for the study of neurodegenerative diseases.

Collaboration


Dive into the Jennifer A. Fifita's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shu Yang

Macquarie University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge