Network


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

Hotspot


Dive into the research topics where Owen A. Ross is active.

Publication


Featured researches published by Owen A. Ross.


American Journal of Human Genetics | 2011

VPS35 Mutations in Parkinson Disease

Carles Vilariño-Güell; Christian Wider; Owen A. Ross; Justus C. Dachsel; Jennifer M. Kachergus; Sarah Lincoln; Alexandra I. Soto-Ortolaza; Stephanie A. Cobb; Greggory J. Wilhoite; Justin A. Bacon; Behrouz Bahareh Behrouz; Heather L. Melrose; Emna Hentati; Andreas Puschmann; Daniel M. Evans; Elizabeth Conibear; Wyeth W. Wasserman; Jan O. Aasly; Pierre Burkhard; Ruth Djaldetti; Joseph Ghika; F. Hentati; Anna Krygowska-Wajs; Timothy Lynch; Eldad Melamed; Alex Rajput; Ali H. Rajput; Alessandra Solida; Ruey-Meei Wu; Ryan J. Uitti

The identification of genetic causes for Mendelian disorders has been based on the collection of multi-incident families, linkage analysis, and sequencing of genes in candidate intervals. This study describes the application of next-generation sequencing technologies to a Swiss kindred presenting with autosomal-dominant, late-onset Parkinson disease (PD). The family has tremor-predominant dopa-responsive parkinsonism with a mean onset of 50.6 ± 7.3 years. Exome analysis suggests that an aspartic-acid-to-asparagine mutation within vacuolar protein sorting 35 (VPS35 c.1858G>A; p.Asp620Asn) is the genetic determinant of disease. VPS35 is a central component of the retromer cargo-recognition complex, is critical for endosome-trans-golgi trafficking and membrane-protein recycling, and is evolutionarily highly conserved. VPS35 c.1858G>A was found in all affected members of the Swiss kindred and in three more families and one patient with sporadic PD, but it was not observed in 3,309 controls. Further sequencing of familial affected probands revealed only one other missense variant, VPS35 c.946C>T; (p.Pro316Ser), in a pedigree with one unaffected and two affected carriers, and thus the pathogenicity of this mutation remains uncertain. Retromer-mediated sorting and transport is best characterized for acid hydrolase receptors. However, the complex has many types of cargo and is involved in a diverse array of biologic pathways from developmental Wnt signaling to lysosome biogenesis. Our study implicates disruption of VPS35 and retromer-mediated trans-membrane protein sorting, rescue, and recycling in the neurodegenerative process leading to PD.


Neurology | 2017

Diagnosis and management of dementia with Lewy bodies Fourth consensus report of the DLB Consortium

Ian G. McKeith; Bradley F. Boeve; Dennis W. Dickson; Glenda Halliday; John-Paul Taylor; Daniel Weintraub; Dag Aarsland; James E. Galvin; Johannes Attems; Clive Ballard; Ashley Bayston; Thomas G. Beach; Frédéric Blanc; Nicolaas Bohnen; Laura Bonanni; José Miguel Brás; Patrick Brundin; David Burn; Alice Chen-Plotkin; John E. Duda; Omar M. El-Agnaf; Howard Feldman; Tanis J. Ferman; Dominic ffytche; Hiroshige Fujishiro; Douglas Galasko; Jennifer G. Goldman; Stephen N. Gomperts; Neill R. Graff-Radford; Lawrence S. Honig

The Dementia with Lewy Bodies (DLB) Consortium has refined its recommendations about the clinical and pathologic diagnosis of DLB, updating the previous report, which has been in widespread use for the last decade. The revised DLB consensus criteria now distinguish clearly between clinical features and diagnostic biomarkers, and give guidance about optimal methods to establish and interpret these. Substantial new information has been incorporated about previously reported aspects of DLB, with increased diagnostic weighting given to REM sleep behavior disorder and 123iodine-metaiodobenzylguanidine (MIBG) myocardial scintigraphy. The diagnostic role of other neuroimaging, electrophysiologic, and laboratory investigations is also described. Minor modifications to pathologic methods and criteria are recommended to take account of Alzheimer disease neuropathologic change, to add previously omitted Lewy-related pathology categories, and to include assessments for substantia nigra neuronal loss. Recommendations about clinical management are largely based upon expert opinion since randomized controlled trials in DLB are few. Substantial progress has been made since the previous report in the detection and recognition of DLB as a common and important clinical disorder. During that period it has been incorporated into DSM-5, as major neurocognitive disorder with Lewy bodies. There remains a pressing need to understand the underlying neurobiology and pathophysiology of DLB, to develop and deliver clinical trials with both symptomatic and disease-modifying agents, and to help patients and carers worldwide to inform themselves about the disease, its prognosis, best available treatments, ongoing research, and how to get adequate support.


Lancet Neurology | 2011

Neuropathologically defined subtypes of Alzheimer's disease with distinct clinical characteristics: a retrospective study

Melissa E. Murray; Neill R. Graff-Radford; Owen A. Ross; Ronald C. Petersen; Ranjan Duara; Dennis W. Dickson

BACKGROUND Neurofibrillary pathology has a stereotypical progression in Alzheimers disease (AD) that is encapsulated in the Braak staging scheme; however, some AD cases are atypical and do not fit into this scheme. We aimed to compare clinical and neuropathological features between typical and atypical AD cases. METHODS AD cases with a Braak neurofibrillary tangle stage of more than IV were identified from a brain bank database. By use of thioflavin-S fluorescence microscopy, we assessed the density and the distribution of neurofibrillary tangles in three cortical regions and two hippocampal sectors. These data were used to construct an algorithm to classify AD cases into typical, hippocampal sparing, or limbic predominant. Classified cases were then compared for clinical, demographic, pathological, and genetic characteristics. An independent cohort of AD cases was assessed to validate findings from the initial cohort. FINDINGS 889 cases of AD, 398 men and 491 women with age at death of 37-103 years, were classified with the algorithm as hippocampal sparing (97 cases [11%]), typical (665 [75%]), or limbic predominant (127 [14%]). By comparison with typical AD, neurofibrillary tangle counts per 0.125 mm(2) in hippocampal sparing cases were higher in cortical areas (median 13, IQR 11-16) and lower in the hippocampus (7.5, 5.2-9.5), whereas counts in limbic-predominant cases were lower in cortical areas (4.3, 3.0-5.7) and higher in the hippocampus (27, 22-35). Hippocampal sparing cases had less hippocampal atrophy than did typical and limbic-predominant cases. Patients with hippocampal sparing AD were younger at death (mean 72 years [SD 10]) and a higher proportion of them were men (61 [63%]), whereas those with limbic-predominant AD were older (mean 86 years [SD 6]) and a higher proportion of them were women (87 [69%]). Microtubule-associated protein tau (MAPT) H1H1 genotype was more common in limbic-predominant AD (54 [70%]) than in hippocampal sparing AD (24 [46%]; p=0.011), but did not differ significantly between limbic-predominant and typical AD (204 [59%]; p=0.11). Apolipoprotein E (APOE) ɛ4 allele status differed between AD subtypes only when data were stratified by age at onset. Clinical presentation, age at onset, disease duration, and rate of cognitive decline differed between the AD subtypes. These findings were confirmed in a validation cohort of 113 patients with AD. INTERPRETATION These data support the hypothesis that AD has distinct clinicopathological subtypes. Hippocampal sparing and limbic-predominant AD subtypes might account for about 25% of cases, and hence should be considered when designing clinical, genetic, biomarker, and treatment studies in patients with AD. FUNDING US National Institutes of Health via Mayo Alzheimers Disease Research Center, Mayo Clinic Study on Aging, Florida Alzheimers Disease Research Center, and Einstein Aging Study; and State of Florida Alzheimers Disease Initiative.


Science Translational Medicine | 2012

Pharmacological Rescue of Mitochondrial Deficits in iPSC-Derived Neural Cells from Patients with Familial Parkinson’s Disease

Oliver Cooper; Hyemyung Seo; Shaida A. Andrabi; Cristina Guardia-Laguarta; John Graziotto; Maria Sundberg; Jesse R. McLean; Luis Carrillo-Reid; Zhong Xie; Teresia Osborn; Gunnar Hargus; Michela Deleidi; Tristan Lawson; Helle Bogetofte; Eduardo Perez-Torres; Lorraine N. Clark; Carol Moskowitz; Joseph R. Mazzulli; Li Chen; Laura A. Volpicelli-Daley; Norma Romero; Houbo Jiang; Ryan J. Uitti; Zhigao Huang; Grzegorz Opala; Leslie A. Scarffe; Valina L. Dawson; Christine Klein; Jian Feng; Owen A. Ross

Neural cells derived from induced pluripotent stem cells from patients with genetic forms of Parkinson’s disease provide insights into disease pathogenesis. Understanding Mitochondrial Deficits in Parkinson’s Disease Parkinson’s disease (PD) is a common, progressive neurodegenerative disease characterized by loss of dopaminergic neurons in the nigrostriatal pathway of the brain, resulting in motor and cognitive deficits. Rodent and primate models only partially predict disease mechanisms. In a new study, Cooper et al. set out to make a human cellular model of PD. First, the authors obtained fibroblasts from members of families with genetically defined forms of PD and generated induced pluripotent stem cells (iPSCs) from the fibroblasts. They then induced differentiation of these PD patient–derived iPSCs into neural cells including dopaminergic neurons to study how the genetic mutations influenced the responses of neural cells to various cellular stressors. Mitochondrial dysfunction has already been implicated in the pathogenesis of PD, so the authors decided to treat their iPSC-derived neural cells from patients with rare familial forms of PD with chemical stressors and toxins known to disrupt mitochondrial function. The researchers observed a gradual increase in sensitivity to cellular stress as the cell type analyzed became functionally closer to the vulnerable cell types in the PD brain; that is, fibroblasts taken directly from PD patients were less sensitive to the chemical stressors than iPSC-derived neural cells. Several drugs helped iPSC-derived neural cells to resist the damaging effects of the cellular stressors. These studies with human neural cells from iPSCs from patients with familial PD highlight opportunities to characterize disease pathways and to screen for new therapeutic agents. Parkinson’s disease (PD) is a common neurodegenerative disorder caused by genetic and environmental factors that results in degeneration of the nigrostriatal dopaminergic pathway in the brain. We analyzed neural cells generated from induced pluripotent stem cells (iPSCs) derived from PD patients and presymptomatic individuals carrying mutations in the PINK1 (PTEN-induced putative kinase 1) and LRRK2 (leucine-rich repeat kinase 2) genes, and compared them to those of healthy control subjects. We measured several aspects of mitochondrial responses in the iPSC-derived neural cells including production of reactive oxygen species, mitochondrial respiration, proton leakage, and intraneuronal movement of mitochondria. Cellular vulnerability associated with mitochondrial dysfunction in iPSC-derived neural cells from familial PD patients and at-risk individuals could be rescued with coenzyme Q10, rapamycin, or the LRRK2 kinase inhibitor GW5074. Analysis of mitochondrial responses in iPSC-derived neural cells from PD patients carrying different mutations provides insight into convergence of cellular disease mechanisms between different familial forms of PD and highlights the importance of oxidative stress and mitochondrial dysfunction in this neurodegenerative disease.


Nature Genetics | 2012

Mutations in the colony stimulating factor 1 receptor ( CSF1R ) gene cause hereditary diffuse leukoencephalopathy with spheroids

Rosa Rademakers; Matt Baker; Alexandra M. Nicholson; Nicola J. Rutherford; NiCole Finch; Alexandra I. Soto-Ortolaza; Jennifer Lash; Christian Wider; Aleksandra Wojtas; Mariely DeJesus-Hernandez; Jennifer Adamson; Naomi Kouri; Christina Sundal; Elizabeth A. Shuster; Jan O. Aasly; James MacKenzie; Sigrun Roeber; Hans A. Kretzschmar; Bradley F. Boeve; David S. Knopman; Ronald C. Petersen; Nigel J. Cairns; Bernardino Ghetti; Salvatore Spina; James Garbern; Alexandros Tselis; Ryan J. Uitti; Pritam Das; Jay A. Van Gerpen; James F. Meschia

Hereditary diffuse leukoencephalopathy with spheroids (HDLS) is an autosomal-dominant central nervous system white-matter disease with variable clinical presentations, including personality and behavioral changes, dementia, depression, parkinsonism, seizures and other phenotypes. We combined genome-wide linkage analysis with exome sequencing and identified 14 different mutations affecting the tyrosine kinase domain of the colony stimulating factor 1 receptor (encoded by CSF1R) in 14 families with HDLS. In one kindred, we confirmed the de novo occurrence of the mutation. Follow-up sequencing identified an additional CSF1R mutation in an individual diagnosed with corticobasal syndrome. In vitro, CSF-1 stimulation resulted in rapid autophosphorylation of selected tyrosine residues in the kinase domain of wild-type but not mutant CSF1R, suggesting that HDLS may result from partial loss of CSF1R function. As CSF1R is a crucial mediator of microglial proliferation and differentiation in the brain, our findings suggest an important role for microglial dysfunction in HDLS pathogenesis.


Annals of Neurology | 2006

Lrrk2 and lewy body disease

Owen A. Ross; Mathias Toft; Andrew J. Whittle; Joseph L. Johnson; Spiridon Papapetropoulos; Deborah C. Mash; Irene Litvan; Mark F. Gordon; Zbigniew K. Wszolek; Matthew J. Farrer; Dennis W. Dickson

The Lrrk2 kinase domain G2019S substitution is the most common genetic basis of familial and sporadic parkinsonism. Patients harboring the G2019S substitution usually present with clinical Parkinsons disease.


Annals of Neurology | 2008

Genomic investigation of alpha-synuclein multiplication and parkinsonism.

Owen A. Ross; Adam Braithwaite; Lisa Skipper; Jennifer M. Kachergus; Mary M. Hulihan; Frank A. Middleton; Kenya Nishioka; Julia Fuchs; Thomas Gasser; Demetrius M. Maraganore; Charles H. Adler; Lydie Larvor; Marie Christine Chartier-Harlin; Christer Nilsson; J. William Langston; Katrina Gwinn; Nobutaka Hattori; Matthew J. Farrer

Copy number variation is a common polymorphic phenomenon within the human genome. Although the majority of these events are non‐deleterious they can also be highly pathogenic. Herein we characterize five families with parkinsonism that have been identified to harbor multiplication of the chromosomal 4q21 locus containing the α‐synuclein gene (SNCA).


Nature Genetics | 2009

DCTN1 mutations in Perry syndrome

Matthew J. Farrer; Mary M. Hulihan; Jennifer M. Kachergus; Justus C. Dachsel; A. Jon Stoessl; Linda L. Grantier; Susan Calne; Donald B. Calne; Bernard Lechevalier; Françoise Chapon; Yoshio Tsuboi; Tatsuo Yamada; Ludwig Gutmann; Bulent Elibol; Kailash P. Bhatia; Christian Wider; Carles Vilariño-Güell; Owen A. Ross; L. Brown; Monica Castanedes-Casey; Dennis W. Dickson; Zbigniew K. Wszolek

Perry syndrome consists of early-onset parkinsonism, depression, severe weight loss and hypoventilation, with brain pathology characterized by TDP-43 immunostaining. We carried out genome-wide linkage analysis and identified five disease-segregating mutations affecting the CAP-Gly domain of dynactin (encoded by DCTN1) in eight families with Perry syndrome; these mutations diminish microtubule binding and lead to intracytoplasmic inclusions. Our findings show that DCTN1 mutations, previously associated with motor neuron disease, can underlie the selective vulnerability of other neuronal populations in distinct neurodegenerative disorders.


American Journal of Human Genetics | 2011

Translation Initiator EIF4G1 Mutations in Familial Parkinson Disease

Marie Christine Chartier-Harlin; Justus C. Dachsel; Carles Vilariño-Güell; Sarah Lincoln; Frédéric Leprêtre; Mary M. Hulihan; Jennifer M. Kachergus; Austen J. Milnerwood; Lucia Tapia; Mee Sook Song; Emilie Le Rhun; Eugénie Mutez; Lydie Larvor; Aurélie Duflot; Christel Vanbesien-Mailliot; Alexandre Kreisler; Owen A. Ross; Kenya Nishioka; Alexandra I. Soto-Ortolaza; Stephanie A. Cobb; Heather L. Melrose; Bahareh Behrouz; Brett H. Keeling; Justin A. Bacon; Emna Hentati; Williams L; Akiko Yanagiya; Nahum Sonenberg; Paul J. Lockhart; Abba C. Zubair

Genome-wide analysis of a multi-incident family with autosomal-dominant parkinsonism has implicated a locus on chromosomal region 3q26-q28. Linkage and disease segregation is explained by a missense mutation c.3614G>A (p.Arg1205His) in eukaryotic translation initiation factor 4-gamma (EIF4G1). Subsequent sequence and genotype analysis identified EIF4G1 c.1505C>T (p.Ala502Val), c.2056G>T (p.Gly686Cys), c.3490A>C (p.Ser1164Arg), c.3589C>T (p.Arg1197Trp) and c.3614G>A (p.Arg1205His) substitutions in affected subjects with familial parkinsonism and idiopathic Lewy body disease but not in control subjects. Despite different countries of origin, persons with EIF4G1 c.1505C>T (p.Ala502Val) or c.3614G>A (p.Arg1205His) mutations appear to share haplotypes consistent with ancestral founders. eIF4G1 p.Ala502Val and p.Arg1205His disrupt eIF4E or eIF3e binding, although the wild-type protein does not, and render mutant cells more vulnerable to reactive oxidative species. EIF4G1 mutations implicate mRNA translation initiation in familial parkinsonism and highlight a convergent pathway for monogenic, toxin and perhaps virally-induced Parkinson disease.


Lancet Neurology | 2011

Association of LRRK2 exonic variants with susceptibility to Parkinson's disease: A case-control study

Owen A. Ross; Alexandra I. Soto-Ortolaza; Michael G. Heckman; Jan O. Aasly; Nadine Abahuni; Grazia Annesi; Justin A. Bacon; Soraya Bardien; Maria Bozi; Alexis Brice; Laura Brighina; Christine Van Broeckhoven; Jonathan Carr; Marie Christine Chartier-Harlin; Efthimios Dardiotis; Dennis W. Dickson; Nancy N. Diehl; Alexis Elbaz; Carlo Ferrarese; Alessandro Ferraris; Brian K. Fiske; J. Mark Gibson; Rachel A. Gibson; Georgios M. Hadjigeorgiou; Nobutaka Hattori; John P. A. Ioannidis; Barbara Jasinska-Myga; Beom S. Jeon; Yun Joong Kim; Christine Klein

BACKGROUND Background The leucine-rich repeat kinase 2 gene (LRRK2) harbours highly penetrant mutations that are linked to familial parkinsonism. However, the extent of its polymorphic variability in relation to risk of Parkinsons disease (PD) has not been assessed systematically. We therefore assessed the frequency of LRRK2 exonic variants in individuals with and without PD, to investigate the role of the variants in PD susceptibility. METHODS LRRK2 was genotyped in patients with PD and controls from three series (white, Asian, and Arab-Berber) from sites participating in the Genetic Epidemiology of Parkinsons Disease Consortium. Genotyping was done for exonic variants of LRRK2 that were identified through searches of literature and the personal communications of consortium members. Associations with PD were assessed by use of logistic regression models. For variants that had a minor allele frequency of 0·5% or greater, single variant associations were assessed, whereas for rarer variants information was collapsed across variants. FINDINGS 121 exonic LRRK2 variants were assessed in 15 540 individuals: 6995 white patients with PD and 5595 controls, 1376 Asian patients and 962 controls, and 240 Arab-Berber patients and 372 controls. After exclusion of carriers of known pathogenic mutations, new independent risk associations were identified for polymorphic variants in white individuals (M1646T, odds ratio 1·43, 95% CI 1·15-1·78; p=0·0012) and Asian individuals (A419V, 2·27, 1·35-3·83; p=0·0011). A protective haplotype (N551K-R1398H-K1423K) was noted at a frequency greater than 5% in the white and Asian series, with a similar finding in the Arab-Berber series (combined odds ratio 0·82, 0·72-0·94; p=0·0043). Of the two previously reported Asian risk variants, G2385R was associated with disease (1·73, 1·20-2·49; p=0·0026), but no association was noted for R1628P (0·62, 0·36-1·07; p=0·087). In the Arab-Berber series, Y2189C showed potential evidence of risk association with PD (4·48, 1·33-15·09; p=0·012). INTERPRETATION The results for LRRK2 show that several rare and common genetic variants in the same gene can have independent effects on disease risk. LRRK2, and the pathway in which it functions, is important in the cause and pathogenesis of PD in a greater proportion of patients with this disease than previously believed. These results will help discriminate those patients who will benefit most from therapies targeted at LRRK2 pathogenic activity. FUNDING Michael J Fox Foundation and National Institutes of Health.

Collaboration


Dive into the Owen A. Ross's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew J. Farrer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ryan J. Uitti

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Carles Vilariño-Güell

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Timothy Lynch

Mater Misericordiae University Hospital

View shared research outputs
Top Co-Authors

Avatar

Jan O. Aasly

Norwegian University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge