Network


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

Hotspot


Dive into the research topics where Jayalakshmi S. Mysore is active.

Publication


Featured researches published by Jayalakshmi S. Mysore.


Cell | 2013

Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease.

Bin Zhang; Chris Gaiteri; Liviu-Gabriel Bodea; Zhi Wang; Joshua McElwee; Alexei Podtelezhnikov; Chunsheng Zhang; Tao Xie; Linh Tran; Radu Dobrin; Eugene M. Fluder; Bruce E. Clurman; Stacey Melquist; Manikandan Narayanan; Christine Suver; Hardik Shah; Milind Mahajan; Tammy Gillis; Jayalakshmi S. Mysore; Marcy E. MacDonald; John Lamb; David A. Bennett; Cliona Molony; David J. Stone; Vilmundur Gudnason; Amanda J. Myers; Eric E. Schadt; Harald Neumann; Jun Zhu; Valur Emilsson

The genetics of complex disease produce alterations in the molecular interactions of cellular pathways whose collective effect may become clear through the organized structure of molecular networks. To characterize molecular systems associated with late-onset Alzheimers disease (LOAD), we constructed gene-regulatory networks in 1,647 postmortem brain tissues from LOAD patients and nondemented subjects, and we demonstrate that LOAD reconfigures specific portions of the molecular interaction structure. Through an integrative network-based approach, we rank-ordered these network structures for relevance to LOAD pathology, highlighting an immune- and microglia-specific module that is dominated by genes involved in pathogen phagocytosis, contains TYROBP as a key regulator, and is upregulated in LOAD. Mouse microglia cells overexpressing intact or truncated TYROBP revealed expression changes that significantly overlapped the human brain TYROBP network. Thus the causal network structure is a useful predictor of response to gene perturbations and presents a framework to test models of disease mechanisms underlying LOAD.


Cell Stem Cell | 2012

Induced Pluripotent Stem Cells from Patients with Huntington’s Disease : Show CAG Repeat-Expansion-Associated Phenotypes

Virginia B. Mattis; Soshana Svendsen; Allison D. Ebert; Clive N. Svendsen; Alvin R. King; Malcolm Casale; Sara T. Winokur; Gayani Batugedara; Marquis P. Vawter; Peter J. Donovan; Leslie F. Lock; Leslie M. Thompson; Yu Zhu; Elisa Fossale; Ranjit S. Atwal; Tammy Gillis; Jayalakshmi S. Mysore; Jian Hong Li; Ihn Sik Seong; Yiping Shen; Xiaoli Chen; Vanessa C. Wheeler; Marcy E. MacDonald; James F. Gusella; Sergey Akimov; Nicolas Arbez; Tarja Juopperi; Tamara Ratovitski; Jason H. Chiang; Woon Roung Kim

Huntingtons disease (HD) is an inherited neurodegenerative disorder caused by an expanded stretch of CAG trinucleotide repeats that results in neuronal dysfunction and death. Here, The HD Consortium reports the generation and characterization of 14 induced pluripotent stem cell (iPSC) lines from HD patients and controls. Microarray profiling revealed CAG-repeat-expansion-associated gene expression patterns that distinguish patient lines from controls, and early onset versus late onset HD. Differentiated HD neural cells showed disease-associated changes in electrophysiology, metabolism, cell adhesion, and ultimately cell death for lines with both medium and longer CAG repeat expansions. The longer repeat lines were however the most vulnerable to cellular stressors and BDNF withdrawal, as assessed using a range of assays across consortium laboratories. The HD iPSC collection represents a unique and well-characterized resource to elucidate disease mechanisms in HD and provides a human stem cell platform for screening new candidate therapeutics.


Neurology | 2012

CAG repeat expansion in Huntington disease determines age at onset in a fully dominant fashion

Jong-Min Lee; Eliana Marisa Ramos; Ji Hyun Lee; Tammy Gillis; Jayalakshmi S. Mysore; Michael R. Hayden; Simon C. Warby; Patrick J. Morrison; Martha Nance; Christopher A. Ross; Russell L. Margolis; Ferdinando Squitieri; S. Orobello; S. Di Donato; Estrella Gomez-Tortosa; Carmen Ayuso; Oksana Suchowersky; Ronald J. Trent; Elizabeth McCusker; Andrea Novelletto; Marina Frontali; Randi Jones; Tetsuo Ashizawa; Samuel Frank; Marie Saint-Hilaire; Steven M. Hersch; H.D. Rosas; Diane Lucente; Madeline Harrison; Andrea Zanko

Objective: Age at onset of diagnostic motor manifestations in Huntington disease (HD) is strongly correlated with an expanded CAG trinucleotide repeat. The length of the normal CAG repeat allele has been reported also to influence age at onset, in interaction with the expanded allele. Due to profound implications for disease mechanism and modification, we tested whether the normal allele, interaction between the expanded and normal alleles, or presence of a second expanded allele affects age at onset of HD motor signs. Methods: We modeled natural log-transformed age at onset as a function of CAG repeat lengths of expanded and normal alleles and their interaction by linear regression. Results: An apparently significant effect of interaction on age at motor onset among 4,068 subjects was dependent on a single outlier data point. A rigorous statistical analysis with a well-behaved dataset that conformed to the fundamental assumptions of linear regression (e.g., constant variance and normally distributed error) revealed significance only for the expanded CAG repeat, with no effect of the normal CAG repeat. Ten subjects with 2 expanded alleles showed an age at motor onset consistent with the length of the larger expanded allele. Conclusions: Normal allele CAG length, interaction between expanded and normal alleles, and presence of a second expanded allele do not influence age at onset of motor manifestations, indicating that the rate of HD pathogenesis leading to motor diagnosis is determined by a completely dominant action of the longest expanded allele and as yet unidentified genetic or environmental factors. Neurology® 2012;78:690–695


Cell | 2015

Identification of Genetic Factors that Modify Clinical Onset of Huntington’s Disease

Jong-Min Lee; Vanessa C. Wheeler; Michael J. Chao; Jean Paul Vonsattel; Ricardo Mouro Pinto; Diane Lucente; Kawther Abu-Elneel; Eliana Marisa Ramos; Jayalakshmi S. Mysore; Tammy Gillis; Marcy E. MacDonald; James F. Gusella; Denise Harold; Timothy Stone; Valentina Escott-Price; Jun Han; Alexey Vedernikov; Peter Holmans; Lesley Jones; Seung Kwak; Mithra Mahmoudi; Michael Orth; G. Bernhard Landwehrmeyer; Jane S. Paulsen; E. Ray Dorsey; Ira Shoulson; Richard H. Myers

As a Mendelian neurodegenerative disorder, the genetic risk of Huntingtons disease (HD) is conferred entirely by an HTT CAG repeat expansion whose length is the primary determinant of the rate of pathogenesis leading to disease onset. To investigate the pathogenic process that precedes disease, we used genome-wide association (GWA) analysis to identify loci harboring genetic variations that alter the age at neurological onset of HD. A chromosome 15 locus displays two independent effects that accelerate or delay onset by 6.1 years and 1.4 years, respectively, whereas a chromosome 8 locus hastens onset by 1.6 years. Association at MLH1 and pathway analysis of the full GWA results support a role for DNA handling and repair mechanisms in altering the course of HD. Our findings demonstrate that HD disease modification in humans occurs in nature and offer a genetic route to identifying in-human validated therapeutic targets in this and other Mendelian disorders.


Archive | 2012

COHORT study oft the HSG. CAG repeat expansion in Huntington disease determines age at onset in al fully dominant fashion

Jong-Min Lee; Eliana Marisa Ramos; Ji Hyun Lee; Tammy Gillis; Jayalakshmi S. Mysore; Hayden; Simon C. Warby; Patrick J. Morrison; Martha Nance; Christopher A. Ross; Russell L. Margolis; Ferdinando Squitieri; S. Orobello; S Di Donato; Estrella Gomez-Tortosa; Carmen Ayuso; Oksana Suchowersky; Ronald J. Trent; Elizabeth McCusker; Andrea Novelletto; Marina Frontali; Randi Jones; Tetsuo Ashizawa; Samuel Frank; Marie-Helene Saint-Hilaire; Steven M. Hersch; H.D. Rosas; Diane Lucente; Madeline Harrison; Andrea Zanko

Objective: Age at onset of diagnostic motor manifestations in Huntington disease (HD) is strongly correlated with an expanded CAG trinucleotide repeat. The length of the normal CAG repeat allele has been reported also to influence age at onset, in interaction with the expanded allele. Due to profound implications for disease mechanism and modification, we tested whether the normal allele, interaction between the expanded and normal alleles, or presence of a second expanded allele affects age at onset of HD motor signs. Methods: We modeled natural log-transformed age at onset as a function of CAG repeat lengths of expanded and normal alleles and their interaction by linear regression. Results: An apparently significant effect of interaction on age at motor onset among 4,068 subjects was dependent on a single outlier data point. A rigorous statistical analysis with a well-behaved dataset that conformed to the fundamental assumptions of linear regression (e.g., constant variance and normally distributed error) revealed significance only for the expanded CAG repeat, with no effect of the normal CAG repeat. Ten subjects with 2 expanded alleles showed an age at motor onset consistent with the length of the larger expanded allele. Conclusions: Normal allele CAG length, interaction between expanded and normal alleles, and presence of a second expanded allele do not influence age at onset of motor manifestations, indicating that the rate of HD pathogenesis leading to motor diagnosis is determined by a completely dominant action of the longest expanded allele and as yet unidentified genetic or environmental factors. Neurology® 2012;78:690–695


American Journal of Human Genetics | 2003

A Genome Scan for Modifiers of Age at Onset in Huntington Disease: The HD MAPS Study

Jian Liang Li; Michael R. Hayden; Elisabeth W. Almqvist; Ryan R. Brinkman; Alexandra Durr; Catherine Dodé; Patrick J. Morrison; Oksana Suchowersky; Christopher A. Ross; Russell L. Margolis; Adam Rosenblatt; Estrella Gomez-Tortosa; David Mayo Cabrero; Andrea Novelletto; Marina Frontali; Martha Nance; Ronald J. Trent; Elizabeth McCusker; Randi Jones; Jane S. Paulsen; Madeline Harrison; Andrea Zanko; Ruth K. Abramson; Ana L. Russ; Beth Knowlton; Luc Djoussé; Jayalakshmi S. Mysore; Suzanne Tariot; Michael F. Gusella; Vanessa C. Wheeler

Huntington disease (HD) is caused by the expansion of a CAG repeat within the coding region of a novel gene on 4p16.3. Although the variation in age at onset is partly explained by the size of the expanded repeat, the unexplained variation in age at onset is strongly heritable (h2=0.56), which suggests that other genes modify the age at onset of HD. To identify these modifier loci, we performed a 10-cM density genomewide scan in 629 affected sibling pairs (295 pedigrees and 695 individuals), using ages at onset adjusted for the expanded and normal CAG repeat sizes. Because all those studied were HD affected, estimates of allele sharing identical by descent at and around the HD locus were adjusted by a positionally weighted method to correct for the increased allele sharing at 4p. Suggestive evidence for linkage was found at 4p16 (LOD=1.93), 6p21-23 (LOD=2.29), and 6q24-26 (LOD=2.28), which may be useful for investigation of genes that modify age at onset of HD.


Human Molecular Genetics | 2009

Somatic expansion of the Huntington's disease CAG repeat in the brain is associated with an earlier age of disease onset

Meera Swami; Audrey E. Hendricks; Tammy Gillis; Tiffany Massood; Jayalakshmi S. Mysore; Richard H. Myers; Vanessa C. Wheeler

The age of onset of Huntingtons disease (HD) is determined primarily by the length of the HD CAG repeat mutation, but is also influenced by other modifying factors. Delineating these modifiers is a critical step towards developing validated therapeutic targets in HD patients. The HD CAG repeat is somatically unstable, undergoing progressive length increases over time, particularly in brain regions that are the targets of neurodegeneration. Here, we have explored the hypothesis that somatic instability of the HD CAG repeat is itself a modifier of disease. Using small-pool PCR, we quantified somatic instability in the cortex region of the brain from a cohort of HD individuals exhibiting phenotypic extremes of young and old disease onset as predicted by the length of their constitutive HD CAG repeat lengths. After accounting for constitutive repeat length, somatic instability was found to be a significant predictor of onset age, with larger repeat length gains associated with earlier disease onset. These data are consistent with the hypothesis that somatic HD CAG repeat length expansions in target tissues contribute to the HD pathogenic process, and support pursuing factors that modify somatic instability as viable therapeutic targets.


BMC Medical Genetics | 2006

Genome-wide significance for a modifier of age at neurological onset in Huntington's Disease at 6q23-24: the HD MAPS study

Jian Liang Li; Michael R. Hayden; Simon C. Warby; Alexandra Durr; Patrick J. Morrison; Martha Nance; Chirstopher A. Ross; Russell L. Margolis; Adam Rosenblatt; Ferdinando Squitieri; Luigi Frati; Estrella Gomez-Tortosa; Carmen Ayuso García; Oksana Suchowersky; Mary Lou Klimek; Ronald J. Trent; Elizabeth McCusker; Andrea Novelletto; Marina Frontali; Jane S. Paulsen; Randi Jones; Tetsuo Ashizawa; Alice Lazzarini; Vanessa C. Wheeler; Ranjana Prakash; Gang Xu; Luc Djoussé; Jayalakshmi S. Mysore; Tammy Gillis; Michael Hakky

BackgroundAge at onset of Huntingtons disease (HD) is correlated with the size of the abnormal CAG repeat expansion in the HD gene; however, several studies have indicated that other genetic factors also contribute to the variability in HD age at onset. To identify modifier genes, we recently reported a whole-genome scan in a sample of 629 affected sibling pairs from 295 pedigrees, in which six genomic regions provided suggestive evidence for quantitative trait loci (QTL), modifying age at onset in HD.MethodsIn order to test the replication of this finding, eighteen microsatellite markers, three from each of the six genomic regions, were genotyped in 102 newly recruited sibling pairs from 69 pedigrees, and data were analyzed, using a multipoint linkage variance component method, in the follow-up sample and the combined sample of 352 pedigrees with 753 sibling pairs.ResultsSuggestive evidence for linkage at 6q23-24 in the follow-up sample (LOD = 1.87, p = 0.002) increased to genome-wide significance for linkage in the combined sample (LOD = 4.05, p = 0.00001), while suggestive evidence for linkage was observed at 18q22, in both the follow-up sample (LOD = 0.79, p = 0.03) and the combined sample (LOD = 1.78, p = 0.002). Epistatic analysis indicated that there is no interaction between 6q23-24 and other loci.ConclusionIn this replication study, linkage for modifier of age at onset in HD was confirmed at 6q23-24. Evidence for linkage was also found at 18q22. The demonstration of statistically significant linkage to a potential modifier locus opens the path to location cloning of a gene capable of altering HD pathogenesis, which could provide a validated target for therapeutic development in the human patient.


Annals of Human Genetics | 2007

The Relationship Between CAG Repeat Length and Age of Onset Differs for Huntington's Disease Patients with Juvenile Onset or Adult Onset

J. Michael Andresen; Javier Gayán; Luc Djoussé; Simone Roberts; Denise Brocklebank; Stacey S. Cherny; Lon R. Cardon; James F. Gusella; Marcy E. MacDonald; Richard H. Myers; David E. Housman; Nancy S. Wexler; Judith Lorimer; Julie Porter; Fidela Gomez; Carol Moskowitz; Kelly Posner Gerstenhaber; Edith Shackell; Karen Marder; Graciela K. Penchaszadeh; Simone A. Roberts; Adam M. Brickman; Jacqueline Gray; Stephen R. Dlouhy; Sandra Wiktorski; Marion E. Hodes; P. Michael Conneally; John B. Penney; Jang Ho Cha; Micheal Irizarry

Age of onset for Huntingtons disease (HD) varies inversely with the length of the disease‐causing CAG repeat expansion in the HD gene. A simple exponential regression model yielded adjusted R‐squared values of 0.728 in a large set of Venezuelan kindreds and 0.642 in a North American, European, and Australian sample (the HD MAPS cohort). We present evidence that a two‐segment exponential regression curve provides a significantly better fit than the simple exponential regression. A plot of natural log‐transformed age of onset against CAG repeat length reveals this segmental relationship. This two‐segment exponential regression on age of onset data increases the adjusted R‐squared values by 0.012 in the Venezuelan kindreds and by 0.035 in the HD MAPS cohort. Although the amount of additional variance explained by the segmental regression approach is modest, the two slopes of the two‐segment regression are significantly different from each other in both the Venezuelan kindreds [F(2, 439) = 11.13, P= 2 × 10−5] and in the HD MAPS cohort [F(2, 688) = 38.27, P= 2 × 10−16]. In both populations, the influence of each CAG repeat on age of onset appears to be stronger in the adult‐onset range of CAG repeats than in the juvenile‐onset range.


Annals of Neurology | 2004

Huntington's disease–like 2 (HDL2) in North America and Japan

Russell L. Margolis; Susan E. Holmes; Adam Rosenblatt; Lisa Gourley; Elizabeth O'Hearn; Christopher A. Ross; William K. Seltzer; Ruth H. Walker; Tetsuo Ashizawa; Astrid Rasmussen; Michael R. Hayden; Elisabeth W. Almqvist; Juliette Harris; Stanley Fahn; Marcy E. MacDonald; Jayalakshmi S. Mysore; Takayoshi Shimohata; Shoji Tsuji; Nicholas T. Potter; Kazuhiro Nakaso; Yoshiki Adachi; Kenji Nakashima; Bird Td; Amanda Krause; Penny Greenstein

Huntingtons Disease–like 2 (HDL2) is a progressive, autosomal dominant, neurodegenerative disorder with marked clinical and pathological similarities to Huntingtons disease (HD). The causal mutation is a CTG/CAG expansion mutation on chromosome 16q24.3, in a variably spliced exon of junctophilin‐3. The frequency of HDL2 was determined in nine independent series of patients referred for HD testing or selected for the presence of an HD‐like phenotype in North America or Japan. The repeat length, ancestry, and age of onset of all North American HDL2 cases were determined. The results show that HDL2 is very rare, with a frequency of 0 to 15% among patients in the nine case series with an HD‐like presentation who do not have the HD mutation. HDL2 is predominantly, and perhaps exclusively, found in individuals of African ancestry. Repeat expansions ranged from 44 to 57 triplets, with length instability in maternal transmission detected in a repeat of 33 triplets. A younger age of onset is correlated with a longer repeat length (r2 = 0.29, p = 0.0098). The results further support the evidence that the repeat expansion at the chromosome 16q24.3 locus is the direct cause of HDL2 and provide preliminary guidelines for the genetic testing of patients with an HD‐like phenotype. Ann Neurol 2004

Collaboration


Dive into the Jayalakshmi S. Mysore'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

Christopher A. Ross

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Russell L. Margolis

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Michael R. Hayden

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Martha Nance

Hennepin County Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge