Network


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

Hotspot


Dive into the research topics where Victoria Zismann is active.

Publication


Featured researches published by Victoria Zismann.


Neuron | 2007

GAB2 Alleles Modify Alzheimer's Risk in APOE ε4 Carriers

Eric M. Reiman; Jennifer A. Webster; Amanda J. Myers; John Hardy; Travis Dunckley; Victoria Zismann; Keta Joshipura; John V. Pearson; Diane Hu-Lince; Matthew J. Huentelman; David Craig; Keith D. Coon; Winnie S. Liang; RiLee H. Herbert; Thomas G. Beach; Kristen Rohrer; Alice S. Zhao; Doris Leung; Leslie Bryden; Lauren Marlowe; Mona Kaleem; Diego Mastroeni; Andrew Grover; Christopher B. Heward; Rivka Ravid; Joseph Rogers; Mike Hutton; Stacey Melquist; R. C. Petersen; Gene E. Alexander

The apolipoprotein E (APOE) epsilon4 allele is the best established genetic risk factor for late-onset Alzheimers disease (LOAD). We conducted genome-wide surveys of 502,627 single-nucleotide polymorphisms (SNPs) to characterize and confirm other LOAD susceptibility genes. In epsilon4 carriers from neuropathologically verified discovery, neuropathologically verified replication, and clinically characterized replication cohorts of 1411 cases and controls, LOAD was associated with six SNPs from the GRB-associated binding protein 2 (GAB2) gene and a common haplotype encompassing the entire GAB2 gene. SNP rs2373115 (p = 9 x 10(-11)) was associated with an odds ratio of 4.06 (confidence interval 2.81-14.69), which interacts with APOE epsilon4 to further modify risk. GAB2 was overexpressed in pathologically vulnerable neurons; the Gab2 protein was detected in neurons, tangle-bearing neurons, and dystrophic neuritis; and interference with GAB2 gene expression increased tau phosphorylation. Our findings suggest that GAB2 modifies LOAD risk in APOE epsilon4 carriers and influences Alzheimers neuropathology.


Nature Genetics | 2007

A survey of genetic human cortical gene expression

Amanda J. Myers; J. Raphael Gibbs; Jennifer A. Webster; Kristen Rohrer; Alice Zhao; Lauren Marlowe; Mona Kaleem; Doris Leung; Leslie Bryden; Priti Nath; Victoria Zismann; Keta Joshipura; Matthew J. Huentelman; Diane Hu-Lince; Keith D. Coon; David Craig; John V. Pearson; Peter Holmans; Christopher B. Heward; Eric M. Reiman; Dietrich A. Stephan; John Hardy

It is widely assumed that genetic differences in gene expression underpin much of the difference among individuals and many of the quantitative traits of interest to geneticists. Despite this, there has been little work on genetic variability in human gene expression and almost none in the human brain, because tools for assessing this genetic variability have not been available. Now, with whole-genome SNP genotyping arrays and whole-transcriptome expression arrays, such experiments have become feasible. We have carried out whole-genome genotyping and expression analysis on a series of 193 neuropathologically normal human brain samples using the Affymetrix GeneChip Human Mapping 500K Array Set and Illumina HumanRefseq-8 Expression BeadChip platforms. Here we present data showing that 58% of the transcriptome is cortically expressed in at least 5% of our samples and that of these cortically expressed transcripts, 21% have expression profiles that correlate with their genotype. These genetic-expression effects should be useful in determining the underlying biology of associations with common diseases of the human brain and in guiding the analysis of the genomic regions involved in the control of normal gene expression.


Nature | 2011

A novel recurrent mutation in MITF predisposes to familial and sporadic melanoma

Satoru Yokoyama; Susan L. Woods; Glen M. Boyle; Lauren G. Aoude; Stuart Macgregor; Victoria Zismann; Michael Gartside; Anne E. Cust; Rizwan Haq; Mark Harland; John C. Taylor; David L. Duffy; Kelly Holohan; Ken Dutton-Regester; Jane M. Palmer; Vanessa F. Bonazzi; Mitchell S. Stark; Judith Symmons; Matthew H. Law; Christopher W. Schmidt; Cathy Lanagan; Linda O’Connor; Elizabeth A. Holland; Helen Schmid; Judith A. Maskiell; Jodie Jetann; Megan Ferguson; Mark A. Jenkins; Richard F. Kefford; Graham G. Giles

So far, two genes associated with familial melanoma have been identified, accounting for a minority of genetic risk in families. Mutations in CDKN2A account for approximately 40% of familial cases, and predisposing mutations in CDK4 have been reported in a very small number of melanoma kindreds. Here we report the whole-genome sequencing of probands from several melanoma families, which we performed in order to identify other genes associated with familial melanoma. We identify one individual carrying a novel germline variant (coding DNA sequence c.G1075A; protein sequence p.E318K; rs149617956) in the melanoma-lineage-specific oncogene microphthalmia-associated transcription factor (MITF). Although the variant co-segregated with melanoma in some but not all cases in the family, linkage analysis of 31 families subsequently identified to carry the variant generated a log of odds (lod) score of 2.7 under a dominant model, indicating E318K as a possible intermediate risk variant. Consistent with this, the E318K variant was significantly associated with melanoma in a large Australian case–control sample. Likewise, it was similarly associated in an independent case–control sample from the United Kingdom. In the Australian sample, the variant allele was significantly over-represented in cases with a family history of melanoma, multiple primary melanomas, or both. The variant allele was also associated with increased naevus count and non-blue eye colour. Functional analysis of E318K showed that MITF encoded by the variant allele had impaired sumoylation and differentially regulated several MITF targets. These data indicate that MITF is a melanoma-predisposition gene and highlight the utility of whole-genome sequencing to identify novel rare variants associated with disease susceptibility.


American Journal of Human Genetics | 2009

Genetic Control of Human Brain Transcript Expression in Alzheimer Disease

Jennifer A. Webster; J. Raphael Gibbs; Jennifer Clarke; Monika Ray; Weixiong Zhang; Peter Holmans; Kristen Rohrer; Alice Zhao; Lauren Marlowe; Mona Kaleem; Donald S. McCorquodale; Cindy Cuello; Doris Leung; Leslie Bryden; Priti Nath; Victoria Zismann; Keta Joshipura; Matthew J. Huentelman; Diane Hu-Lince; Keith D. Coon; David Craig; John V. Pearson; Christopher B. Heward; Eric M. Reiman; Dietrich A. Stephan; John Hardy; Amanda J. Myers

We recently surveyed the relationship between the human brain transcriptome and genome in a series of neuropathologically normal postmortem samples. We have now analyzed additional samples with a confirmed pathologic diagnosis of late-onset Alzheimer disease (LOAD; final n = 188 controls, 176 cases). Nine percent of the cortical transcripts that we analyzed had expression profiles correlated with their genotypes in the combined cohort, and approximately 5% of transcripts had SNP-transcript relationships that could distinguish LOAD samples. Two of these transcripts have been previously implicated in LOAD candidate-gene SNP-expression screens. This study shows how the relationship between common inherited genetic variants and brain transcript expression can be used in the study of human brain disorders. We suggest that studying the transcriptome as a quantitative endo-phenotype has greater power for discovering risk SNPs influencing expression than the use of discrete diagnostic categories such as presence or absence of disease.


Nature Genetics | 2014

Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4

Pilar Ramos; Anthony N. Karnezis; David Craig; Aleksandar Sekulic; Megan Russell; William Hendricks; Jason J. Corneveaux; Michael T. Barrett; Karey Shumansky; Yidong Yang; Sohrab P. Shah; Leah M Prentice; Marco A. Marra; Jeffrey Kiefer; Victoria Zismann; Bodour Salhia; Jaime Prat; Emanuela D'Angelo; Blaise Clarke; Joseph G. Pressey; John H Farley; Stephen P Anthony; Richard Roden; Heather E. Cunliffe; David Huntsman; Jeffrey M. Trent

Small cell carcinoma of the ovary of hypercalcemic type (SCCOHT) is an extremely rare, aggressive cancer affecting children and young women. We identified germline and somatic inactivating mutations in the SWI/SNF chromatin-remodeling gene SMARCA4 in 69% (9/13) of SCCOHT cases in addition to SMARCA4 protein loss in 82% (14/17) of SCCOHT tumors but in only 0.4% (2/485) of other primary ovarian tumors. These data implicate SMARCA4 in SCCOHT oncogenesis.


Neurobiology of Aging | 2010

Evidence for an association between KIBRA and late-onset Alzheimer's disease

Jason J. Corneveaux; Winnie S. Liang; Eric M. Reiman; Jennifer A. Webster; Amanda J. Myers; Victoria Zismann; Keta Joshipura; John V. Pearson; Diane Hu-Lince; David Craig; Keith D. Coon; Travis Dunckley; Daniel Bandy; Wendy Lee; Kewei Chen; Thomas G. Beach; Diego Mastroeni; Andrew Grover; Rivka Ravid; Sigrid Botne Sando; Jan O. Aasly; Reinhard Heun; Frank Jessen; Heike Kölsch; Joseph G. Rogers; Mike Hutton; Stacey Melquist; R. C. Petersen; Gene E. Alexander; Richard J. Caselli

We recently reported evidence for an association between the individual variation in normal human episodic memory and a common variant of the KIBRA gene, KIBRA rs17070145 (T-allele). Since memory impairment is a cardinal clinical feature of Alzheimers disease (AD), we investigated the possibility of an association between the KIBRA gene and AD using data from neuronal gene expression, brain imaging studies, and genetic association tests. KIBRA was significantly over-expressed and three of its four known binding partners under-expressed in AD-affected hippocampal, posterior cingulate and temporal cortex regions (P<0.010, corrected) in a study of laser-capture microdissected neurons. Using positron emission tomography in a cohort of cognitively normal, late-middle-aged persons genotyped for KIBRA rs17070145, KIBRA T non-carriers exhibited lower glucose metabolism than did carriers in posterior cingulate and precuneus brain regions (P<0.001, uncorrected). Lastly, non-carriers of the KIBRA rs17070145 T-allele had increased risk of late-onset AD in an association study of 702 neuropathologically verified expired subjects (P=0.034; OR=1.29) and in a combined analysis of 1026 additional living and expired subjects (P=0.039; OR=1.26). Our findings suggest that KIBRA is associated with both individual variation in normal episodic memory and predisposition to AD.


Neurodegenerative Diseases | 2008

Sorl1 as an Alzheimer’s Disease Predisposition Gene?

Jennifer A. Webster; Amanda J. Myers; John V. Pearson; David Craig; Diane Hu-Lince; Keith D. Coon; Victoria Zismann; Thomas G. Beach; Doris Leung; Leslie Bryden; Rebecca F. Halperin; Lauren Marlowe; Mona Kaleem; Matthew J. Huentelman; Keta Joshipura; Douglas G. Walker; Christopher B. Heward; Rivka Ravid; Joseph Rogers; Andreas Papassotiropoulos; J. Hardy; Eric M. Reiman; Dietrich A. Stephan

Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressively disabling impairments in memory, cognition, and non-cognitive behavioural symptoms. Sporadic AD is multifactorial and genetically complex. While several monogenic mutations cause early-onset AD and gene alleles have been suggested as AD susceptibility factors, the only extensively validated susceptibility gene for late-onset AD is the apolipoprotein E (APOE) Ε4 allele. Alleles of the APOE gene do not account for all of the genetic load calculated to be responsible for AD predisposition. Recently, polymorphisms across the neuronal sortilin-related receptor (SORL1) gene were shown to be significantly associated with AD in several cohorts. Here we present the results of our large case-control whole-genome scan at over 500,000 polymorphisms which presents weak evidence for association and potentially narrows the association interval.


BMC Genomics | 2005

Identification of disease causing loci using an array-based genotyping approach on pooled DNA

David Craig; Matthew J. Huentelman; Diane Hu-Lince; Victoria Zismann; Michael C. Kruer; Anne M. Lee; Erik G. Puffenberger; John M Pearson; Dietrich A. Stephan

BackgroundPooling genomic DNA samples within clinical classes of disease followed by genotyping on whole-genome SNP microarrays, allows for rapid and inexpensive genome-wide association studies. Key to the success of these studies is the accuracy of the allelic frequency calculations, the ability to identify false-positives arising from assay variability and the ability to better resolve association signals through analysis of neighbouring SNPs.ResultsWe report the accuracy of allelic frequency measurements on pooled genomic DNA samples by comparing these measurements to the known allelic frequencies as determined by individual genotyping. We describe modifications to the calculation of k-correction factors from relative allele signal (RAS) values that remove biases and result in more accurate allelic frequency predictions. Our results show that the least accurate SNPs, those most likely to give false-positives in an association study, are identifiable by comparing their frequencies to both those from a known database of individual genotypes and those of the pooled replicates. In a disease with a previously identified genetic mutation, we demonstrate that one can identify the disease locus through the comparison of the predicted allelic frequencies in case and control pools. Furthermore, we demonstrate improved resolution of association signals using the mean of individual test-statistics for consecutive SNPs windowed across the genome. A database of k-correction factors for predicting allelic frequencies for each SNP, derived from several thousand individually genotyped samples, is provided. Lastly, a Perl script for calculating RAS values for the Affymetrix platform is provided.ConclusionOur results illustrate that pooling of DNA samples is an effective initial strategy to identify a genetic locus. However, it is important to eliminate inaccurate SNPs prior to analysis by comparing them to a database of individually genotyped samples as well as by comparing them to replicates of the pool. Lastly, detection of association signals can be improved by incorporating data from neighbouring SNPs.


American Journal of Human Genetics | 2007

Identification of a Novel Risk Locus for Progressive Supranuclear Palsy by a Pooled Genomewide Scan of 500,288 Single-Nucleotide Polymorphisms

Stacey Melquist; David Craig; Matthew J. Huentelman; Richard Crook; John V. Pearson; Matt Baker; Victoria Zismann; Jennifer Gass; Jennifer Adamson; Szabolcs Szelinger; Jason J. Corneveaux; Ashley Cannon; Keith D. Coon; Sarah Lincoln; Charles H. Adler; Paul Tuite; Donald B. Calne; Eileen H. Bigio; Ryan J. Uitti; Zbigniew K. Wszolek; Lawrence I. Golbe; Richard J. Caselli; Neill R. Graff-Radford; Irene Litvan; Matthew J. Farrer; Dennis W. Dickson; Mike Hutton; Dietrich A. Stephan

To date, only the H1 MAPT haplotype has been consistently associated with risk of developing the neurodegenerative disease progressive supranuclear palsy (PSP). We hypothesized that additional genetic loci may be involved in conferring risk of PSP that could be identified through a pooling-based genomewide association study of >500,000 SNPs. Candidate SNPs with large differences in allelic frequency were identified by ranking all SNPs by their probe-intensity difference between cohorts. The MAPT H1 haplotype was strongly detected by this methodology, as was a second major locus on chromosome 11p12-p11 that showed evidence of association at allelic (P<.001), genotypic (P<.001), and haplotypic (P<.001) levels and was narrowed to a single haplotype block containing the DNA damage-binding protein 2 (DDB2) and lysosomal acid phosphatase 2 (ACP2) genes. Since DNA damage and lysosomal dysfunction have been implicated in aging and neurodegenerative processes, both genes are viable candidates for conferring risk of disease.


Molecular Genetics & Genomic Medicine | 2015

Personalized treatment of Sézary syndrome by targeting a novel CTLA4:CD28 fusion

Aleksandar Sekulic; Winnie S. Liang; Waibhav Tembe; Tyler Izatt; Semyon Kruglyak; Jeffrey Kiefer; Lori Cuyugan; Victoria Zismann; Christophe Legendre; Mark R. Pittelkow; John J. Gohmann; Fernando R. De Castro; Jeffrey M. Trent; John D. Carpten; David Craig; Timothy K. McDaniel

Matching molecularly targeted therapies with cancer subtype‐specific gene mutations is revolutionizing oncology care. However, for rare cancers this approach is problematic due to the often poor understanding of the diseases natural history and phenotypic heterogeneity, making treatment of these cancers a particularly unmet medical need in clinical oncology. Advanced Sézary syndrome (SS), an aggressive, exceedingly rare variant of cutaneous T‐cell lymphoma (CTCL) is a prototypical example of a rare cancer. Through whole genome and RNA sequencing (RNA‐seq) of a SS patients tumor we discovered a highly expressed gene fusion between CTLA4 (cytotoxic T lymphocyte antigen 4) and CD28 (cluster of differentiation 28), predicting a novel stimulatory molecule on the surface of tumor T cells. Treatment with the CTLA4 inhibitor ipilimumab resulted in a rapid clinical response. Our findings suggest a novel driver mechanism for SS, and cancer in general, and exemplify an emerging model of cancer treatment using exploratory genomic analysis to identify a personally targeted treatment option when conventional therapies are exhausted.

Collaboration


Dive into the Victoria Zismann's collaboration.

Top Co-Authors

Avatar

David Craig

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dietrich A. Stephan

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Huentelman

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jeffrey M. Trent

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jennifer A. Webster

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Keith D. Coon

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Diane Hu-Lince

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Eric M. Reiman

Missouri State University

View shared research outputs
Top Co-Authors

Avatar

Keta Joshipura

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

William Hendricks

Translational Genomics Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge