Network


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

Hotspot


Dive into the research topics where Cristin McCabe is active.

Publication


Featured researches published by Cristin McCabe.


Nature Neuroscience | 2014

Alzheimer's disease: early alterations in brain DNA methylation at ANK1 , BIN1 , RHBDF2 and other loci

Philip L. De Jager; Gyan Srivastava; Katie Lunnon; Jeremy D. Burgess; Leonard C. Schalkwyk; Lei Yu; Matthew L. Eaton; Brendan T. Keenan; Jason Ernst; Cristin McCabe; Anna Tang; Towfique Raj; Joseph M. Replogle; Wendy Brodeur; Stacey Gabriel; High Seng Chai; Curtis S. Younkin; Steven G. Younkin; Fanggeng Zou; Moshe Szyf; Charles B. Epstein; Julie A. Schneider; Bradley E. Bernstein; Alexander Meissner; Nilufer Ertekin-Taner; Lori B. Chibnik; Manolis Kellis; Jonathan Mill; David A. Bennett

We used a collection of 708 prospectively collected autopsied brains to assess the methylation state of the brains DNA in relation to Alzheimers disease (AD). We found that the level of methylation at 71 of the 415,848 interrogated CpGs was significantly associated with the burden of AD pathology, including CpGs in the ABCA7 and BIN1 regions, which harbor known AD susceptibility variants. We validated 11 of the differentially methylated regions in an independent set of 117 subjects. Furthermore, we functionally validated these CpG associations and identified the nearby genes whose RNA expression was altered in AD: ANK1, CDH23, DIP2A, RHBDF2, RPL13, SERPINF1 and SERPINF2. Our analyses suggest that these DNA methylation changes may have a role in the onset of AD given that we observed them in presymptomatic subjects and that six of the validated genes connect to a known AD susceptibility gene network.


Science | 2014

Common genetic variants modulate pathogen-sensing responses in human dendritic cells.

Mark Lee; Chun Ye; Alexandra-Chloé Villani; Towfique Raj; Weibo Li; Thomas Eisenhaure; Selina Imboywa; Portia Chipendo; F. Ann Ran; Kamil Slowikowski; Lucas D. Ward; Cristin McCabe; Michelle Lee; Irene Y. Frohlich; David A. Hafler; Manolis Kellis; Soumya Raychaudhuri; Feng Zhang; Barbara E. Stranger; Christophe Benoist; Philip L. De Jager; Aviv Regev; Nir Hacohen

Introduction Variation in an individual’s response to environmental factors is likely to influence susceptibility to complex human diseases. The genetic basis of such variation is poorly understood. Here, we identify natural genetic variants that underlie variation in the host innate immune response to infection and analyze the mechanisms by which such variants alter these responses. Identifying the genetic basis of variability in the host response to pathogens. A cohort of 534 individuals donated blood for (a) genotyping of common DNA variants and (b) isolation of immune DCs. DCs were stimulated with viral and bacterial components, and the variability in individuals’ gene expression responses was mapped to specific DNA variants, which were then shown to affect binding of particular transcription factors. Methods We derived dendritic cells (DCs) from peripheral blood monocytes of healthy individuals (295 Caucasians, 122 African Americans, 117 East Asians) and stimulated them with Escherichia coli lipopolysaccharide (LPS), influenza virus, or the cytokine interferon-β (IFN-β) to generate 1598 transcriptional profiles. We genotyped each of these individuals at sites of common genetic variation and identified the genetic variants that best explain variation in gene expression and gene induction between individuals. We then tested mechanistic predictions from these associations using synthetic promoter constructs and genome engineering. Results We identified 264 loci containing genetic variants associated with variation in absolute gene expression in human DCs, of which 121 loci were associated with variation in the induction of gene expression by one or more stimuli. Fine-mapping identified candidate causal single-nucleotide polymorphisms (SNPs) associated with expression variance, and deeper functional experiments localized three of these SNPs to the binding sites of stimulus-activated transcription factors. We also identified a cis variant in the transcription factor, IRF7, associated in trans with the induction of a module of antiviral genes in response to influenza infection. Of the identified genetic variants, 35 were also associated with autoimmune or infectious disease loci found by genome-wide association studies. Discussion The genetic variants we uncover and the molecular basis for their action provide mechanistic explanations and principles for how the innate immune response to pathogens and cytokines varies across individuals. Our results also link disease-associated variants to specific immune pathways in DCs, which provides greater insight into mechanisms underlying complex human phenotypes. Extending our approach to many immune cell types and pathways will provide a global map linking human genetic variants to specific immunological processes. Immune Variation It is difficult to determine the mechanistic consequences of context-dependent genetic variants, some of which may be related to disease (see the Perspective by Gregersen). Two studies now report on the effects of stimulating immunological monocytes and dendritic cells with proteins that can elicit a response to bacterial or viral infection and assess the functional links between genetic variants and profiles of gene expression. M. N. Lee et al. (10.1126/science.1246980) analyzed the expression of more than 400 genes, in dendritic cells from 534 healthy subjects, which revealed how expression quantitative trait loci (eQTLs) affect gene expression within the interferon-β and the Toll-like receptor 3 and 4 pathways. Fairfax et al. (10.1126/science.1246949) performed a genome-wide analysis to show that many eQTLs affected monocyte gene expression in a stimulus- or time-specific manner. Mapping of human host-pathogen gene-by-environment interactions identifies pathogen-specific loci. [Also see Perspective by Gregersen] Little is known about how human genetic variation affects the responses to environmental stimuli in the context of complex diseases. Experimental and computational approaches were applied to determine the effects of genetic variation on the induction of pathogen-responsive genes in human dendritic cells. We identified 121 common genetic variants associated in cis with variation in expression responses to Escherichia coli lipopolysaccharide, influenza, or interferon-β (IFN-β). We localized and validated causal variants to binding sites of pathogen-activated STAT (signal transducer and activator of transcription) and IRF (IFN-regulatory factor) transcription factors. We also identified a common variant in IRF7 that is associated in trans with type I IFN induction in response to influenza infection. Our results reveal common alleles that explain interindividual variation in pathogen sensing and provide functional annotation for genetic variants that alter susceptibility to inflammatory diseases.


JAMA Neurology | 2013

Genetic susceptibility for Alzheimer disease neuritic plaque pathology.

Joshua M. Shulman; Kewei Chen; Brendan T. Keenan; Lori B. Chibnik; Adam S. Fleisher; Pradeep Thiyyagura; Auttawut Roontiva; Cristin McCabe; Nikolaos A. Patsopoulos; Jason J. Corneveaux; Lei Yu; Matthew J. Huentelman; Denis A. Evans; Julie A. Schneider; Eric M. Reiman; Philip L. De Jager; David A. Bennett

IMPORTANCE While numerous genetic susceptibility loci have been identified for clinical Alzheimer disease (AD), it is important to establish whether these variants are risk factors for the underlying disease pathology, including neuritic plaques. OBJECTIVES To investigate whether AD susceptibility loci from genome-wide association studies affect neuritic plaque pathology and to additionally identify novel risk loci for this trait. DESIGN, SETTING, AND PARTICIPANTS Candidate analysis of single-nucleotide polymorphisms and genome-wide association study in a joint clinicopathologic cohort, including 725 deceased subjects from the Religious Orders Study and the Rush Memory and Aging Project (2 prospective, community-based studies), followed by targeted validation in an independent neuroimaging cohort, including 114 subjects from multiple clinical and research centers. MAIN OUTCOMES AND MEASURES A quantitative measure of neuritic plaque pathologic burden, based on assessments of silver-stained tissue averaged from multiple brain regions. Validation based on β-amyloid load by immunocytochemistry, and replication with fibrillar β-amyloid positron emission tomographic imaging with Pittsburgh Compound B or florbetapir. RESULTS Besides the previously reported APOE and CR1 loci, we found that the ABCA7 (rs3764650; P = .02) and CD2AP (rs9349407; P = .03) AD susceptibility loci are associated with neuritic plaque burden. In addition, among the top results of our genome-wide association study, we discovered a novel variant near the amyloid precursor protein gene (APP, rs2829887) that is associated with neuritic plaques (P = 3.3 × 10-6). This polymorphism was associated with postmortem β-amyloid load as well as fibrillar β-amyloid in 2 independent cohorts of adults with normal cognition. CONCLUSIONS AND RELEVANCE These findings enhance understanding of AD risk factors by relating validated susceptibility alleles to increased neuritic plaque pathology and implicate common genetic variation at the APP locus in the earliest, presymptomatic stages of AD.


PLOS Biology | 2016

NMNAT2:HSP90 Complex Mediates Proteostasis in Proteinopathies.

Yousuf O. Ali; Hunter M. Allen; Lei Yu; David Li-Kroeger; Dena Bakhshizadehmahmoudi; Asante Hatcher; Cristin McCabe; Jishu Xu; Nicole L. Bjorklund; Giulio Taglialatela; David A. Bennett; Philip L. De Jager; Joshua M. Shulman; Hugo J. Bellen; Hui-Chen Lu

Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) is neuroprotective in numerous preclinical models of neurodegeneration. Here, we show that brain nmnat2 mRNA levels correlate positively with global cognitive function and negatively with AD pathology. In AD brains, NMNAT2 mRNA and protein levels are reduced. NMNAT2 shifts its solubility and colocalizes with aggregated Tau in AD brains, similar to chaperones, which aid in the clearance or refolding of misfolded proteins. Investigating the mechanism of this observation, we discover a novel chaperone function of NMNAT2, independent from its enzymatic activity. NMNAT2 complexes with heat shock protein 90 (HSP90) to refold aggregated protein substrates. NMNAT2’s refoldase activity requires a unique C-terminal ATP site, activated in the presence of HSP90. Furthermore, deleting NMNAT2 function increases the vulnerability of cortical neurons to proteotoxic stress and excitotoxicity. Interestingly, NMNAT2 acts as a chaperone to reduce proteotoxic stress, while its enzymatic activity protects neurons from excitotoxicity. Taken together, our data indicate that NMNAT2 exerts its chaperone or enzymatic function in a context-dependent manner to maintain neuronal health.


Nature Neuroscience | 2017

An xQTL map integrates the genetic architecture of the human brain's transcriptome and epigenome

Bernard Ng; Charles C. White; Hans-Ulrich Klein; Solveig K. Sieberts; Cristin McCabe; Ellis Patrick; Jishu Xu; Lei Yu; Chris Gaiteri; David A. Bennett; Philip L. De Jager

We report a multi-omic resource generated by applying quantitative trait locus (xQTL) analyses to RNA sequence, DNA methylation and histone acetylation data from the dorsolateral prefrontal cortex of 411 older adults who have all three data types. We identify SNPs significantly associated with gene expression, DNA methylation and histone modification levels. Many of these SNPs influence multiple molecular features, and we demonstrate that SNP effects on RNA expression are fully mediated by epigenetic features in 9% of these loci. Further, we illustrate the utility of our new resource, xQTL Serve, by using it to prioritize the cell type(s) most affected by an xQTL. We also reanalyze published genome wide association studies using an xQTL-weighted analysis approach and identify 18 new schizophrenia and 2 new bipolar susceptibility variants, which is more than double the number of loci that can be discovered with a larger blood-based expression eQTL resource.


Alzheimers & Dementia | 2016

Methylation profiles in peripheral blood CD4+ lymphocytes versus brain: The relation to Alzheimer's disease pathology

Lei Yu; Lori B. Chibnik; Jingyun Yang; Cristin McCabe; Jishu Xu; Julie A. Schneider; Philip L. De Jager; David A. Bennett

We investigated the change in DNA methylation in peripheral blood CD4+ lymphocytes over time, examined the relation between CD4+ lymphocytes and brain methylation, and compared their associations with AD pathology.


Annals of Neurology | 2016

Genes and Environment in Multiple Sclerosis project: A platform to investigate multiple sclerosis risk

Zongqi Xia; Charles C. White; Emily K. Owen; Alina Von Korff; Sarah R. Clarkson; Cristin McCabe; Maria Cimpean; Phoebe Winn; Ashley Hoesing; Sonya Steele; Irene Cortese; Tanuja Chitnis; Howard L. Weiner; Daniel S. Reich; Lori B. Chibnik; Philip L. De Jager

The Genes and Environment in Multiple Sclerosis project establishes a platform to investigate the events leading to multiple sclerosis (MS) in at‐risk individuals. It has recruited 2,632 first‐degree relatives from across the USA. Using an integrated genetic and environmental risk score, we identified subjects with twice the MS risk when compared to the average family member, and we report an initial incidence rate in these subjects that is 30 times greater than that of sporadic MS. We discuss the feasibility of large‐scale studies of asymptomatic at‐risk subjects that leverage modern tools of subject recruitment to execute collaborative projects. Ann Neurol 2016;79:178–189


Nature Neuroscience | 2018

A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer’s disease

Chris Gaiteri; Sarah E. Sullivan; Charles C. White; Shinya Tasaki; Jishu Xu; Mariko Taga; Hans-Ulrich Klein; Ellis Patrick; Vitalina Komashko; Cristin McCabe; Robert J. Smith; Elizabeth M. Bradshaw; David E. Root; Aviv Regev; Lei Yu; Lori B. Chibnik; Julie A. Schneider; Tracy L. Young-Pearse; David A. Bennett; Philip L. De Jager

There is a need for new therapeutic targets with which to prevent Alzheimer’s disease (AD), a major contributor to aging-related cognitive decline. Here we report the construction and validation of a molecular network of the aging human frontal cortex. Using RNA sequence data from 478 individuals, we first build a molecular network using modules of coexpressed genes and then relate these modules to AD and its neuropathologic and cognitive endophenotypes. We confirm these associations in two independent AD datasets. We also illustrate the use of the network in prioritizing amyloid- and cognition-associated genes for in vitro validation in human neurons and astrocytes. These analyses based on unique cohorts enable us to resolve the role of distinct cortical modules that have a direct effect on the accumulation of AD pathology from those that have a direct effect on cognitive decline, exemplifying a network approach to complex diseases.The authors constructed and validated a molecular network of the aging human cortex from RNA sequencing data from 478 individuals and identified genes that affect cognitive decline or neuropathology in Alzheimer’s disease.


bioRxiv | 2018

Epigenome-wide study uncovers tau pathology-driven changes of chromatin organization in the aging human brain

Hans-Ulrich Klein; Cristin McCabe; Elizabeta Gjoneska; Sarah E. Sullivan; Belinda J. Kaskow; Anna Tang; Robert V Smith; Jishu Xu; Andreas R. Pfenning; Bradley E. Bernstein; Alexander Meissner; Julie A. Schneider; Li-Huei Tsai; Tracy L. Young-Pearse; David A. Bennett; Philip L. De Jager

Accumulation of tau and amyloid-β are two pathologic hallmarks of Alzheimer’s disease (AD). Here, we conducted an epigenome-wide association study using the H3K9 acetylation (H3K9Ac) mark in 669 aged human prefrontal cortices: in contrast to amyloid-β, tau protein burden had a broad effect on the epigenome, affecting 5,590 out of 26,384 H3K9Ac domains. Tau-related alterations aggregated in large genomic segments reflecting spatial chromatin organization, and the magnitude of these effects correlated with the segment’s nuclear lamina association. We confirmed the functional relevance of these chromatin changes by demonstrating (1) consistent transcriptional changes in three independent datasets and (2) similar findings in two AD mouse models. Finally, we found that tau overexpression in iPSC-derived neurons disrupted chromatin organization and that these effects could be blocked by a small molecule predicted to reverse the tau effect. Thus, we report large-scale tau-driven chromatin rearrangements in the aging human brain that may be reversible with HSP90 inhibitors.


Scientific Data | 2018

A multi-omic atlas of the human frontal cortex for aging and Alzheimer’s disease research

Philip L. De Jager; Yiyi Ma; Cristin McCabe; Jishu Xu; Badri N. Vardarajan; Daniel Felsky; Hans-Ulrich Klein; Charles C. White; Mette A. Peters; Ben Lodgson; Parham Nejad; Anna Tang; Lara M. Mangravite; Lei Yu; Chris Gaiteri; Julie A. Schneider; David A. Bennett

We initiated the systematic profiling of the dorsolateral prefrontal cortex obtained from a subset of autopsied individuals enrolled in the Religious Orders Study (ROS) or the Rush Memory and Aging Project (MAP), which are jointly designed prospective studies of aging and dementia with detailed, longitudinal cognitive phenotyping during life and a quantitative, structured neuropathologic examination after death. They include over 3,322 subjects. Here, we outline the first generation of data including genome-wide genotypes (n=2,090), whole genome sequencing (n=1,179), DNA methylation (n=740), chromatin immunoprecipitation with sequencing using an anti-Histone 3 Lysine 9 acetylation (H3K9Ac) antibody (n=712), RNA sequencing (n=638), and miRNA profile (n=702). Generation of other omic data including ATACseq, proteomic and metabolomics profiles is ongoing. Thanks to its prospective design and recruitment of older, non-demented individuals, these data can be repurposed to investigate a large number of syndromic and quantitative neuroscience phenotypes. The many subjects that are cognitively non-impaired at death also offer insights into the biology of the human brain in older non-impaired individuals.

Collaboration


Dive into the Cristin McCabe's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Bennett

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Yu

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Towfique Raj

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aviv Regev

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Selina Imboywa

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge