Network


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

Hotspot


Dive into the research topics where Zbigniew K. Wszolek is active.

Publication


Featured researches published by Zbigniew K. Wszolek.


Neuron | 2011

Expanded GGGGCC Hexanucleotide Repeat in Noncoding Region of C9ORF72 Causes Chromosome 9p-Linked FTD and ALS

Mariely DeJesus-Hernandez; Ian R. Mackenzie; Bradley F. Boeve; Adam L. Boxer; Matt Baker; Nicola J. Rutherford; Alexandra M. Nicholson; NiCole Finch; Heather C. Flynn; Jennifer Adamson; Naomi Kouri; Aleksandra Wojtas; Pheth Sengdy; Ging-Yuek Robin Hsiung; Anna Karydas; William W. Seeley; Keith A. Josephs; Giovanni Coppola; Daniel H. Geschwind; Zbigniew K. Wszolek; Howard Feldman; David S. Knopman; Ronald C. Petersen; Bruce L. Miller; Dennis W. Dickson; Kevin B. Boylan; Neill R. Graff-Radford; Rosa Rademakers

Several families have been reported with autosomal-dominant frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), genetically linked to chromosome 9p21. Here, we report an expansion of a noncoding GGGGCC hexanucleotide repeat in the gene C9ORF72 that is strongly associated with disease in a large FTD/ALS kindred, previously reported to be conclusively linked to chromosome 9p. This same repeat expansion was identified in the majority of our families with a combined FTD/ALS phenotype and TDP-43-based pathology. Analysis of extended clinical series found the C9ORF72 repeat expansion to be the most common genetic abnormality in both familial FTD (11.7%) and familial ALS (23.5%). The repeat expansion leads to the loss of one alternatively spliced C9ORF72 transcript and to formation of nuclear RNA foci, suggesting multiple disease mechanisms. Our findings indicate that repeat expansion in C9ORF72 is a major cause of both FTD and ALS.


Neuron | 2004

Mutations in LRRK2 Cause Autosomal-Dominant Parkinsonism with Pleomorphic Pathology

Alexander Zimprich; Saskia Biskup; Petra Leitner; Peter Lichtner; Matthew J. Farrer; Sarah Lincoln; Jennifer M. Kachergus; Mary M. Hulihan; Ryan J. Uitti; Donald B. Calne; A. Jon Stoessl; Ronald F. Pfeiffer; Nadja Patenge; Iria Carballo Carbajal; Peter Vieregge; Friedrich Asmus; Bertram Müller-Myhsok; Dennis W. Dickson; Thomas Meitinger; Tim M. Strom; Zbigniew K. Wszolek; Thomas Gasser

We have previously linked families with autosomal-dominant, late-onset parkinsonism to chromosome 12p11.2-q13.1 (PARK8). By high-resolution recombination mapping and candidate gene sequencing in 46 families, we have found six disease-segregating mutations (five missense and one putative splice site mutation) in a gene encoding a large, multifunctional protein, LRRK2 (leucine-rich repeat kinase 2). It belongs to the ROCO protein family and includes a protein kinase domain of the MAPKKK class and several other major functional domains. Within affected carriers of families A and D, six post mortem diagnoses reveal brainstem dopaminergic degeneration accompanied by strikingly diverse pathologies. These include abnormalities consistent with Lewy body Parkinsons disease, diffuse Lewy body disease, nigral degeneration without distinctive histopathology, and progressive supranuclear palsy-like pathology. Clinical diagnoses of Parkinsonism with dementia or amyotrophy or both, with their associated pathologies, are also noted. Hence, LRRK2 may be central to the pathogenesis of several major neurodegenerative disorders associated with parkinsonism.


Lancet Neurology | 2008

Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a case-control study

Daniel G. Healy; Mario Falchi; Sean S. O'Sullivan; Vincenzo Bonifati; Alexandra Durr; Susan Bressman; Alexis Brice; Jan O. Aasly; Cyrus P. Zabetian; Stefano Goldwurm; Joaquim J. Ferreira; Eduardo Tolosa; Denise M. Kay; Christine Klein; David R. Williams; Connie Marras; Anthony E. Lang; Zbigniew K. Wszolek; José Berciano; A. H. V. Schapira; Timothy Lynch; Kailash P. Bhatia; Thomas Gasser; Andrew J. Lees; Nicholas W. Wood

Summary Background Mutations in LRRK2, the gene that encodes leucine-rich repeat kinase 2, are a cause of Parkinsons disease (PD). The International LRRK2 Consortium was established to answer three key clinical questions: can LRRK2-associated PD be distinguished from idiopathic PD; which mutations in LRRK2 are pathogenic; and what is the age-specific cumulative risk of PD for individuals who inherit or are at risk of inheriting a deleterious mutation in LRRK2? Methods Researchers from 21 centres across the world collaborated on this study. The frequency of the common LRRK2 Gly2019Ser mutation was estimated on the basis of data from 24 populations worldwide, and the penetrance of the mutation was defined in 1045 people with mutations in LRRK2 from 133 families. The LRRK2 phenotype was defined on the basis of 59 motor and non-motor symptoms in 356 patients with LRRK2-associated PD and compared with the symptoms of 543 patients with pathologically proven idiopathic PD. Findings Six mutations met the consortiums criteria for being proven pathogenic. The frequency of the common LRRK2 Gly2019Ser mutation was 1% of patients with sporadic PD and 4% of patients with hereditary PD; the frequency was highest in the middle east and higher in southern Europe than in northern Europe. The risk of PD for a person who inherits the LRRK2 Gly2019Ser mutation was 28% at age 59 years, 51% at 69 years, and 74% at 79 years. The motor symptoms (eg, disease severity, rate of progression, occurrence of falls, and dyskinesia) and non-motor symptoms (eg, cognition and olfaction) of LRRK2-associated PD were more benign than those of idiopathic PD. Interpretation Mutations in LRRK2 are a clinically relevant cause of PD that merit testing in patients with hereditary PD and in subgroups of patients with PD. However, this knowledge should be applied with caution in the diagnosis and counselling of patients. Funding UK Medical Research Council; UK Parkinsons Disease Society; UK Brain Research Trust; Internationaal Parkinson Fonds; Volkswagen Foundation; National Institutes of Health: National Institute of Neurological Disorders and Stroke and National Institute of Aging; Udall Parkinsons Disease Centre of Excellence; Pacific Alzheimer Research Foundation Centre; Italian Telethon Foundation; Fondazione Grigioni per il Morbo di Parkinson; Michael J Fox Foundation for Parkinsons Research; Safra Global Genetics Consortium; US Department of Veterans Affairs; French Agence Nationale de la Recherche.


Annals of Neurology | 2004

Comparison of kindreds with parkinsonism and α‐synuclein genomic multiplications

Matthew J. Farrer; Jennifer M. Kachergus; Lysia S. Forno; Sarah Lincoln; Deng Shun Wang; Mary M. Hulihan; Demetrius M. Maraganore; Katrina Gwinn-Hardy; Zbigniew K. Wszolek; Dennis W. Dickson; J. William Langston

Genomic triplication of the α‐synuclein gene recently has been associated with familial Parkinsons disease in the Spellman–Muenter kindred. Here, we present an independent family, of Swedish‐American descent, with hereditary early‐onset parkinsonism with dementia due to α‐synuclein triplication. Brain tissue available from affected individuals in both kindreds provided the opportunity to compare their clinical, pathological, and biochemical phenotypes. Of note, studies of brain mRNA and soluble protein levels demonstrate a doubling of α‐synuclein expression, consistent with molecular genetic data. Pathologically, cornu ammonis 2/3 hippocampal neuronal loss appears to be a defining feature of this form of inherited parkinsonism. The profound implications of α‐synuclein overexpression for idiopathic synucleinopathies are discussed.


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.


American Journal of Human Genetics | 2005

Identification of a Novel LRRK2 Mutation Linked to Autosomal Dominant Parkinsonism: Evidence of a Common Founder across European Populations

Jennifer M. Kachergus; Ignacio F. Mata; Mary M. Hulihan; Julie P. Taylor; Sarah Lincoln; Jan O. Aasly; J. Mark Gibson; Owen A Ross; Timothy Lynch; Joseph Wiley; Haydeh Payami; John G. Nutt; Demetrius M. Maraganore; Krzysztof Czyzewski; Maria Styczyńska; Zbigniew K. Wszolek; Matthew J. Farrer; Mathias Toft

Autosomal dominant parkinsonism has been attributed to pathogenic amino acid substitutions in leucine-rich repeat kinase 2 (LRRK2). By sequencing multiplex families consistent with a PARK8 assignment, we identified a novel heterozygous LRRK2 mutation. A referral sample of 248 affected probands from families with autosomal dominant parkinsonism was subsequently assessed; 7 (2.8%) were found to carry a heterozygous LRRK2 6055G-->A transition (G2019S). These seven patients originate from the United States, Norway, Ireland, and Poland. In samples of patients with idiopathic Parkinson disease (PD) from the same populations, further screening identified six more patients with LRRK2 G2019S; no mutations were found in matched control individuals. Subsequently, 42 family members of the 13 probands were examined; 22 have an LRRK2 G2019S substitution, 7 with a diagnosis of PD. Of note, all patients share an ancestral haplotype indicative of a common founder, and, within families, LRRK2 G2019S segregates with disease (multipoint LOD score 2.41). Penetrance is age dependent, increasing from 17% at age 50 years to 85% at age 70 years. In summary, our study demonstrates that LRRK2 G2019S accounts for parkinsonism in several families within Europe and North America. Our work highlights the fact that a proportion of clinically typical, late-onset PD cases have a genetic basis.


Lancet Neurology | 2009

Neuropathological assessment of Parkinson's disease: refining the diagnostic criteria

Dennis W. Dickson; Heiko Braak; John E. Duda; Charles Duyckaerts; Thomas Gasser; Glenda M. Halliday; John Hardy; James B. Leverenz; Kelly Del Tredici; Zbigniew K. Wszolek; Irene Litvan

To date, there have been few systematic attempts to provide a standard operating procedure for the neuropathological diagnosis of Parkinsons disease (PD). Pathological examination cannot classify the clinical syndrome with certainty; therefore, the neuropathological diagnosis is, at best, a probability statement. The neuropathological diagnosis of parkinsonism has become increasingly based on fundamental molecular underpinnings, with recognition that the genetics of parkinsonism is heterogeneous and includes disorders that are associated with and without Lewy bodies. The advent of alpha-synuclein immunohistochemistry has substantially improved the ability to identify Lewy pathology, particularly cortical Lewy bodies and smaller aggregates within processes and the neuropil. In this Review we discuss the diagnostic criteria for the neuropathological assessment of PD. These criteria are provisional and need to be validated through an iterative process that could help with their refinement. Additionally, we suggest future directions for neuropathology research on PD.


The Journal of Neuroscience | 2007

Accumulation of Pathological Tau Species and Memory Loss in a Conditional Model of Tauopathy

Zdenek Berger; Hanno Roder; Amanda Hanna; Vijayaraghavan Rangachari; Mei Yue; Zbigniew K. Wszolek; Karen H. Ashe; Joshua Knight; Dennis W. Dickson; Cathy A. Andorfer; Terrone L. Rosenberry; Jada Lewis; Mike Hutton; Christopher Janus

Neurofibrillary tangles (NFTs) are a pathological hallmark of Alzheimers disease and other tauopathies, but recent studies in a conditional mouse model of tauopathy (rTg4510) have suggested that NFT formation can be dissociated from memory loss and neurodegeneration. This suggests that NFTs are not the major neurotoxic tau species, at least during the early stages of pathogenesis. To identify other neurotoxic tau protein species, we performed biochemical analyses on brain tissues from the rTg4510 mouse model and then correlated the levels of these tau proteins with memory loss. We describe the identification and characterization of two forms of tau multimers (140 and 170 kDa), whose molecular weight suggests an oligomeric aggregate, that accumulate early in the pathogenic cascade in this mouse model. Similar tau multimers were detected in a second mouse model of tauopathy (JNPL3) and in tissue from patients with Alzheimers disease and FTDP-17 (frontotemporal dementia and parkinsonism linked to chromosome 17). Moreover, levels of the tau multimers correlated consistently with memory loss at various ages in the rTg4510 mouse model. Our findings suggest that accumulation of early-stage aggregated tau species, before the formation of NFT, is associated with the development of functional deficits during the pathogenic progression of tauopathy.


PLOS Genetics | 2008

Novel mutations in TARDBP (TDP-43) in patients with familial amyotrophic lateral sclerosis

Nicola J. Rutherford; Yong Jie Zhang; Matt Baker; Jennifer Gass; NiCole Finch; Yafei Xu; Heather Stewart; Brendan J. Kelley; Karen M. Kuntz; Richard Crook; Jemeen Sreedharan; Caroline Vance; Eric J. Sorenson; Carol F. Lippa; Eileen H. Bigio; Daniel H. Geschwind; David S. Knopman; Hiroshi Mitsumoto; Ronald C. Petersen; Neil R. Cashman; Mike Hutton; Christopher Shaw; Kevin B. Boylan; Bradley F. Boeve; Neill R. Graff-Radford; Zbigniew K. Wszolek; Richard J. Caselli; Dennis W. Dickson; Ian R. Mackenzie; Leonard Petrucelli

The TAR DNA-binding protein 43 (TDP-43) has been identified as the major disease protein in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitin inclusions (FTLD-U), defining a novel class of neurodegenerative conditions: the TDP-43 proteinopathies. The first pathogenic mutations in the gene encoding TDP-43 (TARDBP) were recently reported in familial and sporadic ALS patients, supporting a direct role for TDP-43 in neurodegeneration. In this study, we report the identification and functional analyses of two novel and one known mutation in TARDBP that we identified as a result of extensive mutation analyses in a cohort of 296 patients with variable neurodegenerative diseases associated with TDP-43 histopathology. Three different heterozygous missense mutations in exon 6 of TARDBP (p.M337V, p.N345K, and p.I383V) were identified in the analysis of 92 familial ALS patients (3.3%), while no mutations were detected in 24 patients with sporadic ALS or 180 patients with other TDP-43–positive neurodegenerative diseases. The presence of p.M337V, p.N345K, and p.I383V was excluded in 825 controls and 652 additional sporadic ALS patients. All three mutations affect highly conserved amino acid residues in the C-terminal part of TDP-43 known to be involved in protein-protein interactions. Biochemical analysis of TDP-43 in ALS patient cell lines revealed a substantial increase in caspase cleaved fragments, including the ∼25 kDa fragment, compared to control cell lines. Our findings support TARDBP mutations as a cause of ALS. Based on the specific C-terminal location of the mutations and the accumulation of a smaller C-terminal fragment, we speculate that TARDBP mutations may cause a toxic gain of function through novel protein interactions or intracellular accumulation of TDP-43 fragments leading to apoptosis.


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.

Collaboration


Dive into the Zbigniew K. Wszolek's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew J. Farrer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan O. Aasly

Norwegian University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge