scispace - formally typeset
Search or ask a question
Author

Jan Konrad

Bio: Jan Konrad is an academic researcher from Washington University in St. Louis. The author has contributed to research in topics: Genome-wide association study & Exome sequencing. The author has an hindex of 1, co-authored 1 publications receiving 126 citations.

Papers
More filters
Journal ArticleDOI
Joshua C. Bis1, Xueqiu Jian2, Brian W. Kunkle3, Yuning Chen4, Kara L. Hamilton-Nelson3, William S. Bush5, William J Salerno6, Daniel Lancour4, Yiyi Ma4, Alan E. Renton7, Edoardo Marcora7, John J. Farrell4, Yi Zhao8, Liming Qu8, Shahzad Ahmad9, Najaf Amin10, Philippe Amouyel10, Philippe Amouyel11, Philippe Amouyel12, Gary W. Beecham3, Jennifer E. Below13, Dominique Campion10, Laura B. Cantwell8, Camille Charbonnier10, Jaeyoon Chung4, Paul K. Crane1, Carlos Cruchaga14, L. Adrienne Cupples4, Jean-François Dartigues15, Stéphanie Debette15, Jean-François Deleuze, Lucinda Fulton14, Stacey Gabriel16, Emmanuelle Génin10, Richard A. Gibbs6, Alison Goate7, Benjamin Grenier-Boley10, Namrata Gupta16, Jonathan L. Haines5, Aki S. Havulinna17, Aki S. Havulinna18, Seppo Helisalmi19, Mikko Hiltunen19, Daniel P. Howrigan16, Daniel P. Howrigan20, M. Arfan Ikram9, Jaakko Kaprio17, Jan Konrad14, Amanda B. Kuzma8, Eric S. Lander16, Mark Lathrop21, Terho Lehtimäki22, Honghuang Lin4, Kari Mattila22, Richard Mayeux23, Donna M. Muzny6, Waleed Nasser6, Benjamin M. Neale20, Benjamin M. Neale16, Kwangsik Nho24, Gaël Nicolas10, Devanshi Patel4, Margaret A. Pericak-Vance3, Markus Perola17, Markus Perola25, Markus Perola18, Bruce M. Psaty, Olivier Quenez10, Farid Rajabli3, Richard Redon26, Christiane Reitz23, Anne M. Remes19, Anne M. Remes27, Veikko Salomaa18, Chloé Sarnowski4, Helena Schmidt28, Michael A. Schmidt3, Reinhold Schmidt28, Hilkka Soininen19, Timothy A. Thornton1, Giuseppe Tosto23, Christophe Tzourio15, Sven J. van der Lee9, Cornelia M. van Duijn9, Otto Valladares8, Badri N. Vardarajan23, Li-San Wang8, Weixin Wang8, Ellen M. Wijsman1, Richard K. Wilson14, Daniela Witten1, Kim C. Worley6, Xiaoling Zhang4, Alzheimer’s Disease Sequencing16, Alzheimer’s Disease Sequencing20, Alzheimer’s Disease Sequencing17, Céline Bellenguez10, Jean-Charles Lambert10, Mitja I. Kurki20, Mitja I. Kurki16, Mitja I. Kurki17, Aarno Palotie20, Aarno Palotie17, Aarno Palotie16, Mark J. Daly17, Mark J. Daly16, Mark J. Daly20, Eric Boerwinkle6, Eric Boerwinkle2, Kathryn L. Lunetta4, Anita L. DeStefano4, Josée Dupuis4, Eden R. Martin3, Gerard D. Schellenberg8, Sudha Seshadri4, Sudha Seshadri29, Adam C. Naj8, Myriam Fornage2, Lindsay A. Farrer 
TL;DR: The Alzheimer’s Disease Sequencing Project undertook whole exome sequencing in 5,740 late-onset Alzheimer disease cases and 5,096 cognitively normal controls primarily of European ancestry, identifying novel and predicted functional genetic variants in genes previously associated with AD.
Abstract: The Alzheimer’s Disease Sequencing Project (ADSP) undertook whole exome sequencing in 5,740 late-onset Alzheimer disease (AD) cases and 5,096 cognitively normal controls primarily of European ancestry (EA), among whom 218 cases and 177 controls were Caribbean Hispanic (CH). An age-, sex- and APOE based risk score and family history were used to select cases most likely to harbor novel AD risk variants and controls least likely to develop AD by age 85 years. We tested ~1.5 million single nucleotide variants (SNVs) and 50,000 insertion-deletion polymorphisms (indels) for association to AD, using multiple models considering individual variants as well as gene-based tests aggregating rare, predicted functional, and loss of function variants. Sixteen single variants and 19 genes that met criteria for significant or suggestive associations after multiple-testing correction were evaluated for replication in four independent samples; three with whole exome sequencing (2,778 cases, 7,262 controls) and one with genome-wide genotyping imputed to the Haplotype Reference Consortium panel (9,343 cases, 11,527 controls). The top findings in the discovery sample were also followed-up in the ADSP whole-genome sequenced family-based dataset (197 members of 42 EA families and 501 members of 157 CH families). We identified novel and predicted functional genetic variants in genes previously associated with AD. We also detected associations in three novel genes: IGHG3 (p = 9.8 × 10−7), an immunoglobulin gene whose antibodies interact with β-amyloid, a long non-coding RNA AC099552.4 (p = 1.2 × 10−7), and a zinc-finger protein ZNF655 (gene-based p = 5.0 × 10−6). The latter two suggest an important role for transcriptional regulation in AD pathogenesis.

191 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: Pathway analysis implicates immunity, lipid metabolism, tau binding proteins, and amyloid precursor protein (APP) metabolism, showing that genetic variants affecting APP and Aβ processing are associated not only with early-onset autosomal dominant Alzheimer’s disease but also with LOAD.
Abstract: Risk for late-onset Alzheimer’s disease (LOAD), the most prevalent dementia, is partially driven by genetics. To identify LOAD risk loci, we performed a large genome-wide association meta-analysis of clinically diagnosed LOAD (94,437 individuals). We confirm 20 previous LOAD risk loci and identify five new genome-wide loci (IQCK, ACE, ADAM10, ADAMTS1, and WWOX), two of which (ADAM10, ACE) were identified in a recent genome-wide association (GWAS)-by-familial-proxy of Alzheimer’s or dementia. Fine-mapping of the human leukocyte antigen (HLA) region confirms the neurological and immune-mediated disease haplotype HLA-DR15 as a risk factor for LOAD. Pathway analysis implicates immunity, lipid metabolism, tau binding proteins, and amyloid precursor protein (APP) metabolism, showing that genetic variants affecting APP and Aβ processing are associated not only with early-onset autosomal dominant Alzheimer’s disease but also with LOAD. Analyses of risk genes and pathways show enrichment for rare variants (P = 1.32 × 10−7), indicating that additional rare variants remain to be identified. We also identify important genetic correlations between LOAD and traits such as family history of dementia and education.

1,641 citations

Journal ArticleDOI
TL;DR: In insights into the coordinated control of Alzheimer’s disease risk genes and their cell-type-specific contribution to disease susceptibility, single-nucleus RNA sequencing is applied to entorhinal cortex samples from control and Alzheimer's disease brains and identified transcription factor networks predicted to control disease progression in a cell-sub type-specific way.
Abstract: There is currently little information available about how individual cell types contribute to Alzheimer’s disease. Here we applied single-nucleus RNA sequencing to entorhinal cortex samples from control and Alzheimer’s disease brains (n = 6 per group), yielding a total of 13,214 high-quality nuclei. We detail cell-type-specific gene expression patterns, unveiling how transcriptional changes in specific cell subpopulations are associated with Alzheimer’s disease. We report that the Alzheimer’s disease risk gene APOE is specifically repressed in Alzheimer’s disease oligodendrocyte progenitor cells and astrocyte subpopulations and upregulated in an Alzheimer’s disease-specific microglial subopulation. Integrating transcription factor regulatory modules with Alzheimer’s disease risk loci revealed drivers of cell-type-specific state transitions towards Alzheimer’s disease. For example, transcription factor EB, a master regulator of lysosomal function, regulates multiple disease genes in a specific Alzheimer’s disease astrocyte subpopulation. These results provide insights into the coordinated control of Alzheimer’s disease risk genes and their cell-type-specific contribution to disease susceptibility. These results are available at http://adsn.ddnetbio.com. Grubman et al. generated a single-cell transcriptomic atlas of the entorhinal cortex from patients with Alzheimer’s disease and identified transcription factor networks predicted to control disease progression in a cell-subtype-specific way.

495 citations

Journal ArticleDOI
TL;DR: ApoE4 is a promising AD therapeutic target that remains understudied and recent studies are now paving the way for effective apoE4-directed AD treatment approaches.
Abstract: The growing body of evidence indicating the heterogeneity of Alzheimer’s disease (AD), coupled with disappointing clinical studies directed at a fit-for-all therapy, suggest that the development of a single magic cure suitable for all cases may not be possible. This calls for a shift in paradigm where targeted treatment is developed for specific AD subpopulations that share distinct genetic or pathological properties. Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor of AD, is expressed in more than half of AD patients and is thus an important possible AD therapeutic target. This review focuses initially on the pathological effects of apoE4 in AD, as well as on the corresponding cellular and animal models and the suggested cellular and molecular mechanisms which mediate them. The second part of the review focuses on recent apoE4-targeted (from the APOE gene to the apoE protein and its interactors) therapeutic approaches that have been developed in animal models and are ready to be translated to human. Further, the issue of whether the pathological effects of apoE4 are due to loss of protective function or due to gain of toxic function is discussed herein. It is possible that both mechanisms coexist, with certain constituents of the apoE4 molecule and/or its downstream signaling mediating a toxic effect, while others are associated with a loss of protective function. ApoE4 is a promising AD therapeutic target that remains understudied. Recent studies are now paving the way for effective apoE4-directed AD treatment approaches.

258 citations

Journal ArticleDOI
TL;DR: In this paper, the authors systematically review and update the vast state-of-the-art literature of amyloid-β (Aβ) science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics.
Abstract: Breakthroughs in molecular medicine have positioned the amyloid-β (Aβ) pathway at the center of Alzheimer's disease (AD) pathophysiology. While the detailed molecular mechanisms of the pathway and the spatial-temporal dynamics leading to synaptic failure, neurodegeneration, and clinical onset are still under intense investigation, the established biochemical alterations of the Aβ cycle remain the core biological hallmark of AD and are promising targets for the development of disease-modifying therapies. Here, we systematically review and update the vast state-of-the-art literature of Aβ science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics. We discuss the evidence highlighting a differentiated interaction of distinct Aβ species with other AD-related biological mechanisms, such as tau-mediated, neuroimmune and inflammatory changes, as well as a neurochemical imbalance. Through the lens of the latest development of multimodal in vivo biomarkers of AD, this cross-disciplinary review examines the compelling hypothesis- and data-driven rationale for Aβ-targeting therapeutic strategies in development for the early treatment of AD.

251 citations

Journal ArticleDOI
TL;DR: The multiplex model reflects the combination of some, or all, of these model components (genetic and environmental), in a tissue-specific manner, to trigger or sustain a disease cascade, which ultimately results in the cell and synaptic loss observed in AD.
Abstract: Genes play a strong role in Alzheimer's disease (AD), with late-onset AD showing heritability of 58-79% and early-onset AD showing over 90%. Genetic association provides a robust platform to build our understanding of the etiology of this complex disease. Over 50 loci are now implicated for AD, suggesting that AD is a disease of multiple components, as supported by pathway analyses (immunity, endocytosis, cholesterol transport, ubiquitination, amyloid-β and tau processing). Over 50% of late-onset AD heritability has been captured, allowing researchers to calculate the accumulation of AD genetic risk through polygenic risk scores. A polygenic risk score predicts disease with up to 90% accuracy and is an exciting tool in our research armory that could allow selection of those with high polygenic risk scores for clinical trials and precision medicine. It could also allow cellular modelling of the combined risk. Here we propose the multiplex model as a new perspective from which to understand AD. The multiplex model reflects the combination of some, or all, of these model components (genetic and environmental), in a tissue-specific manner, to trigger or sustain a disease cascade, which ultimately results in the cell and synaptic loss observed in AD.

243 citations