2,833 results on '"Geschwind, Daniel H"'
Search Results
2. Cross-ancestry atlas of gene, isoform, and splicing regulation in the developing human brain
- Author
-
Wen, Cindy, Margolis, Michael, Dai, Rujia, Zhang, Pan, Przytycki, Pawel F, Vo, Daniel D, Bhattacharya, Arjun, Matoba, Nana, Tang, Miao, Jiao, Chuan, Kim, Minsoo, Tsai, Ellen, Hoh, Celine, Aygün, Nil, Walker, Rebecca L, Chatzinakos, Christos, Clarke, Declan, Pratt, Henry, Peters, Mette A, Gerstein, Mark, Daskalakis, Nikolaos P, Weng, Zhiping, Jaffe, Andrew E, Kleinman, Joel E, Hyde, Thomas M, Weinberger, Daniel R, Bray, Nicholas J, Sestan, Nenad, Geschwind, Daniel H, Roeder, Kathryn, Gusev, Alexander, Pasaniuc, Bogdan, Stein, Jason L, Love, Michael I, Pollard, Katherine S, Liu, Chunyu, Gandal, Michael J, Akbarian, Schahram, Abyzov, Alexej, Ahituv, Nadav, Arasappan, Dhivya, Almagro Armenteros, Jose Juan, Beliveau, Brian J, Bendl, Jaroslav, Berretta, Sabina, Bharadwaj, Rahul A, Bicks, Lucy, Brennand, Kristen, Capauto, Davide, Champagne, Frances A, Chatterjee, Tanima, Chatzinakos, Chris, Chen, Yuhang, Chen, H Isaac, Cheng, Yuyan, Cheng, Lijun, Chess, Andrew, Chien, Jo-fan, Chu, Zhiyuan, Clement, Ashley, Collado-Torres, Leonardo, Cooper, Gregory M, Crawford, Gregory E, Davila-Velderrain, Jose, Deep-Soboslay, Amy, Deng, Chengyu, DiPietro, Christopher P, Dracheva, Stella, Drusinsky, Shiron, Duan, Ziheng, Duong, Duc, Dursun, Cagatay, Eagles, Nicholas J, Edelstein, Jonathan, Emani, Prashant S, Fullard, John F, Galani, Kiki, Galeev, Timur, Gaynor, Sophia, Girdhar, Kiran, Goes, Fernando S, Greenleaf, William, Grundman, Jennifer, Guo, Hanmin, Guo, Qiuyu, Gupta, Chirag, Hadas, Yoav, Hallmayer, Joachim, Han, Xikun, Haroutunian, Vahram, Hawken, Natalie, He, Chuan, Henry, Ella, Hicks, Stephanie C, Ho, Marcus, Ho, Li-Lun, Hoffman, Gabriel E, Huang, Yiling, Huuki-Myers, Louise A, and Hwang, Ahyeon
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Biological Psychology ,Psychology ,Mental Illness ,Mental Health ,Human Genome ,Neurosciences ,Brain Disorders ,Mental health ,Humans ,Alternative Splicing ,Atlases as Topic ,Autism Spectrum Disorder ,Brain ,Gene Expression Regulation ,Developmental ,Gene Regulatory Networks ,Genome-Wide Association Study ,Protein Isoforms ,Quantitative Trait Loci ,Schizophrenia ,Transcriptome ,Mental Disorders ,PsychENCODE Consortium† ,PsychENCODE Consortium ,General Science & Technology - Abstract
Neuropsychiatric genome-wide association studies (GWASs), including those for autism spectrum disorder and schizophrenia, show strong enrichment for regulatory elements in the developing brain. However, prioritizing risk genes and mechanisms is challenging without a unified regulatory atlas. Across 672 diverse developing human brains, we identified 15,752 genes harboring gene, isoform, and/or splicing quantitative trait loci, mapping 3739 to cellular contexts. Gene expression heritability drops during development, likely reflecting both increasing cellular heterogeneity and the intrinsic properties of neuronal maturation. Isoform-level regulation, particularly in the second trimester, mediated the largest proportion of GWAS heritability. Through colocalization, we prioritized mechanisms for about 60% of GWAS loci across five disorders, exceeding adult brain findings. Finally, we contextualized results within gene and isoform coexpression networks, revealing the comprehensive landscape of transcriptome regulation in development and disease.
- Published
- 2024
3. Massively parallel characterization of regulatory elements in the developing human cortex
- Author
-
Deng, Chengyu, Whalen, Sean, Steyert, Marilyn, Ziffra, Ryan, Przytycki, Pawel F, Inoue, Fumitaka, Pereira, Daniela A, Capauto, Davide, Norton, Scott, Vaccarino, Flora M, Pollen, Alex A, Nowakowski, Tomasz J, Ahituv, Nadav, Pollard, Katherine S, Akbarian, Schahram, Abyzov, Alexej, Arasappan, Dhivya, Almagro Armenteros, Jose Juan, Beliveau, Brian J, Bendl, Jaroslav, Berretta, Sabina, Bharadwaj, Rahul A, Bhattacharya, Arjun, Bicks, Lucy, Brennand, Kristen, Champagne, Frances A, Chatterjee, Tanima, Chatzinakos, Chris, Chen, Yuhang, Chen, H Isaac, Cheng, Yuyan, Cheng, Lijun, Chess, Andrew, Chien, Jo-fan, Chu, Zhiyuan, Clarke, Declan, Clement, Ashley, Collado-Torres, Leonardo, Cooper, Gregory M, Crawford, Gregory E, Dai, Rujia, Daskalakis, Nikolaos P, Davila-Velderrain, Jose, Deep-Soboslay, Amy, DiPietro, Christopher P, Dracheva, Stella, Drusinsky, Shiron, Duan, Ziheng, Duong, Duc, Dursun, Cagatay, Eagles, Nicholas J, Edelstein, Jonathan, Emani, Prashant S, Fullard, John F, Galani, Kiki, Galeev, Timur, Gandal, Michael J, Gaynor, Sophia, Gerstein, Mark, Geschwind, Daniel H, Girdhar, Kiran, Goes, Fernando S, Greenleaf, William, Grundman, Jennifer, Guo, Hanmin, Guo, Qiuyu, Gupta, Chirag, Hadas, Yoav, Hallmayer, Joachim, Han, Xikun, Haroutunian, Vahram, Hawken, Natalie, He, Chuan, Henry, Ella, Hicks, Stephanie C, Ho, Marcus, Ho, Li-Lun, Hoffman, Gabriel E, Huang, Yiling, Huuki-Myers, Louise A, Hwang, Ahyeon, Hyde, Thomas M, Iatrou, Artemis, Jajoo, Aarti, Jensen, Matthew, Jiang, Lihua, Jin, Peng, Jin, Ting, Jops, Connor, Jourdon, Alexandre, Kawaguchi, Riki, Kellis, Manolis, Khullar, Saniya, Kleinman, Joel E, Kleopoulos, Steven P, and Kozlenkov, Alex
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biomedical and Clinical Sciences ,Stem Cell Research - Embryonic - Human ,Stem Cell Research ,Human Genome ,Genetics ,Neurosciences ,Underpinning research ,Aetiology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Neurological ,Humans ,Cerebral Cortex ,Chromatin ,Deep Learning ,Enhancer Elements ,Genetic ,Gene Expression Regulation ,Developmental ,Neurogenesis ,Neurons ,Organoids ,Regulatory Sequences ,Nucleic Acid ,Promoter Regions ,Genetic ,Regulatory Elements ,Transcriptional ,PsychENCODE Consortium‡ ,PsychENCODE Consortium ,General Science & Technology - Abstract
Nucleotide changes in gene regulatory elements are important determinants of neuronal development and diseases. Using massively parallel reporter assays in primary human cells from mid-gestation cortex and cerebral organoids, we interrogated the cis-regulatory activity of 102,767 open chromatin regions, including thousands of sequences with cell type-specific accessibility and variants associated with brain gene regulation. In primary cells, we identified 46,802 active enhancer sequences and 164 variants that alter enhancer activity. Activity was comparable in organoids and primary cells, suggesting that organoids provide an adequate model for the developing cortex. Using deep learning we decoded the sequence basis and upstream regulators of enhancer activity. This work establishes a comprehensive catalog of functional gene regulatory elements and variants in human neuronal development.
- Published
- 2024
4. Whole genome‐wide sequence analysis of long‐lived families (Long‐Life Family Study) identifies MTUS2 gene associated with late‐onset Alzheimer's disease
- Author
-
Xicota, Laura, Cosentino, Stephanie, Vardarajan, Badri, Mayeux, Richard, Perls, Thomas T, Andersen, Stacy L, Zmuda, Joseph M, Thyagarajan, Bharat, Yashin, Anatoli, Wojczynski, Mary K, Krinsky‐McHale, Sharon, Handen, Benjamin L, Christian, Bradley T, Head, Elizabeth, Mapstone, Mark E, Schupf, Nicole, Lee, Joseph H, Barral, Sandra, Study, the Long‐Life Family, Abner, Erin, Adams, Perrie M, Aguirre, Alyssa, Albert, Marilyn S, Albin, Roger L, Allen, Mariet, Alvarez, Lisa, Andrews, Howard, Apostolova, Liana G, Arnold, Steven E, Asthana, Sanjay, Atwood, Craig S, Ayres, Gayle, Barber, Robert C, Barnes, Lisa L, Bartlett, Jackie, Beach, Thomas G, Becker, James T, Beecham, Gary W, Benchek, Penelope, Bennett, David A, Bertelson, John, Biber, Sarah A, Bird, Thomas D, Blacker, Deborah, Boeve, Bradley F, Bowen, James D, Boxer, Adam, Brewer, James B, Burke, James R, Burns, Jeffrey M, Bush, William S, Buxbaum, Joseph D, Byrd, Goldie, Cantwell, Laura B, Cao, Chuanhai, Carlsson, Cynthia M, Carrasquillo, Minerva M, Chan, Kwun C, Chasse, Scott, Chen, Yen‐Chi, Chesselet, Marie‐Francoise, Chin, Nathaniel A, Chui, Helena C, Chung, Jaeyoon, Craft, Suzanne, Crane, Paul K, Cranney, Marissa, Cruchaga, Carlos, Cuccaro, Michael L, Culhane, Jessica, Cullum, C Munro, Darby, Eveleen, Davis, Barbara, De Jager, Philip L, DeCarli, Charles, DeToledo, John C, Dickson, Dennis W, Dobbins, Nic, Duara, Ranjan, Ertekin‐Taner, Nilufer, Evans, Denis A, Faber, Kelley M, Fairchild, Thomas J, Fallin, Daniele, Fallon, Kenneth B, Fardo, David W, Farlow, Martin R, Farrell, John, Farrer, Lindsay A, Fernandez‐Hernandez, Victoria, Foroud, Tatiana M, Frosch, Matthew P, Galasko, Douglas R, Gamboa, Adriana, Gauthreaux, Kathryn M, Gefen, Tamar, Geschwind, Daniel H, Ghetti, Bernardino, and Gilbert, John R
- Subjects
Biomedical and Clinical Sciences ,Biological Psychology ,Clinical Sciences ,Neurosciences ,Psychology ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Acquired Cognitive Impairment ,Neurodegenerative ,Brain Disorders ,Dementia ,Alzheimer's Disease ,Human Genome ,Biotechnology ,Aging ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Humans ,Alzheimer Disease ,Amyloid beta-Peptides ,Genetic Predisposition to Disease ,Genome-Wide Association Study ,Microtubule-Associated Proteins ,Polymorphism ,Single Nucleotide ,Sequence Analysis ,genetic risk ,late-onset Alzheimer's disease ,microtubule protein ,MTUS2 gene ,whole genome sequence ,Long‐Life Family Study ,Alzheimer's Disease Genetic Consortium ,Alzheimer's Biomarkers Consortium‐Down Syndrome ,late‐onset Alzheimer's disease ,Geriatrics ,Clinical sciences ,Biological psychology - Abstract
IntroductionLate-onset Alzheimer's disease (LOAD) has a strong genetic component. Participants in Long-Life Family Study (LLFS) exhibit delayed onset of dementia, offering a unique opportunity to investigate LOAD genetics.MethodsWe conducted a whole genome sequence analysis of 3475 LLFS members. Genetic associations were examined in six independent studies (N = 14,260) with a wide range of LOAD risk. Association analysis in a sub-sample of the LLFS cohort (N = 1739) evaluated the association of LOAD variants with beta amyloid (Aβ) levels.ResultsWe identified several single nucleotide polymorphisms (SNPs) in tight linkage disequilibrium within the MTUS2 gene associated with LOAD (rs73154407, p = 7.6 × 10-9). Association of MTUS2 variants with LOAD was observed in the five independent studies and was significantly stronger within high levels of Aβ42/40 ratio compared to lower amyloid.DiscussionMTUS2 encodes a microtubule associated protein implicated in the development and function of the nervous system, making it a plausible candidate to investigate LOAD biology.HighlightsLong-Life Family Study (LLFS) families may harbor late onset Alzheimer's dementia (LOAD) variants. LLFS whole genome sequence analysis identified MTUS2 gene variants associated with LOAD. The observed LLFS variants generalized to cohorts with wide range of LOAD risk. The association of MTUS2 with LOAD was stronger within high levels of beta amyloid. Our results provide evidence for MTUS2 gene as a novel LOAD candidate locus.
- Published
- 2024
5. Polygenic profiles define aspects of clinical heterogeneity in attention deficit hyperactivity disorder
- Author
-
LaBianca, Sonja, Brikell, Isabell, Helenius, Dorte, Loughnan, Robert, Mefford, Joel, Palmer, Clare E, Walker, Rebecca, Gådin, Jesper R, Krebs, Morten, Appadurai, Vivek, Vaez, Morteza, Agerbo, Esben, Pedersen, Marianne Giørtz, Børglum, Anders D, Hougaard, David M, Mors, Ole, Nordentoft, Merete, Mortensen, Preben Bo, Kendler, Kenneth S, Jernigan, Terry L, Geschwind, Daniel H, Ingason, Andrés, Dahl, Andrew W, Zaitlen, Noah, Dalsgaard, Søren, Werge, Thomas M, and Schork, Andrew J
- Subjects
Biological Sciences ,Genetics ,Pediatric ,Behavioral and Social Science ,Autism ,Human Genome ,Mental Illness ,Mental Health ,Prevention ,Attention Deficit Hyperactivity Disorder (ADHD) ,Clinical Research ,Intellectual and Developmental Disabilities (IDD) ,Brain Disorders ,2.1 Biological and endogenous factors ,2.3 Psychological ,social and economic factors ,Mental health ,Good Health and Well Being ,Humans ,Autism Spectrum Disorder ,Attention Deficit Disorder with Hyperactivity ,Multifactorial Inheritance ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Attention deficit hyperactivity disorder (ADHD) is a complex disorder that manifests variability in long-term outcomes and clinical presentations. The genetic contributions to such heterogeneity are not well understood. Here we show several genetic links to clinical heterogeneity in ADHD in a case-only study of 14,084 diagnosed individuals. First, we identify one genome-wide significant locus by comparing cases with ADHD and autism spectrum disorder (ASD) to cases with ADHD but not ASD. Second, we show that cases with ASD and ADHD, substance use disorder and ADHD, or first diagnosed with ADHD in adulthood have unique polygenic score (PGS) profiles that distinguish them from complementary case subgroups and controls. Finally, a PGS for an ASD diagnosis in ADHD cases predicted cognitive performance in an independent developmental cohort. Our approach uncovered evidence of genetic heterogeneity in ADHD, helping us to understand its etiology and providing a model for studies of other disorders.
- Published
- 2024
6. Whole-genome sequencing analysis reveals new susceptibility loci and structural variants associated with progressive supranuclear palsy
- Author
-
Wang, Hui, Chang, Timothy S, Dombroski, Beth A, Cheng, Po-Liang, Patil, Vishakha, Valiente-Banuet, Leopoldo, Farrell, Kurt, Mclean, Catriona, Molina-Porcel, Laura, Rajput, Alex, De Deyn, Peter Paul, Le Bastard, Nathalie, Gearing, Marla, Kaat, Laura Donker, Van Swieten, John C, Dopper, Elise, Ghetti, Bernardino F, Newell, Kathy L, Troakes, Claire, de Yébenes, Justo G, Rábano-Gutierrez, Alberto, Meller, Tina, Oertel, Wolfgang H, Respondek, Gesine, Stamelou, Maria, Arzberger, Thomas, Roeber, Sigrun, Müller, Ulrich, Hopfner, Franziska, Pastor, Pau, Brice, Alexis, Durr, Alexandra, Le Ber, Isabelle, Beach, Thomas G, Serrano, Geidy E, Hazrati, Lili-Naz, Litvan, Irene, Rademakers, Rosa, Ross, Owen A, Galasko, Douglas, Boxer, Adam L, Miller, Bruce L, Seeley, Willian W, Van Deerlin, Vivanna M, Lee, Edward B, White, Charles L, Morris, Huw, de Silva, Rohan, Crary, John F, Goate, Alison M, Friedman, Jeffrey S, Leung, Yuk Yee, Coppola, Giovanni, Naj, Adam C, Wang, Li-San, Dalgard, Clifton, Dickson, Dennis W, Höglinger, Günter U, Schellenberg, Gerard D, Geschwind, Daniel H, and Lee, Wan-Ping
- Subjects
Biological Sciences ,Genetics ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Neurodegenerative ,Acquired Cognitive Impairment ,Frontotemporal Dementia (FTD) ,Rare Diseases ,Neurosciences ,Dementia ,Human Genome ,Brain Disorders ,Biotechnology ,Aging ,Alzheimer's Disease Related Dementias (ADRD) ,Alzheimer's Disease ,2.1 Biological and endogenous factors ,Neurological ,Progressive Supranuclear Palsy ,Whole-Genome Sequencing ,Genome-Wide Association Study ,Structural Variants ,Apolipoprotein E ,P. S. P. genetics study group ,Humans ,Supranuclear Palsy ,Progressive ,Genetic Predisposition to Disease ,Polymorphism ,Single Nucleotide ,Aged ,Aged ,80 and over ,Middle Aged ,Female ,Male ,Whole Genome Sequencing ,Clinical Sciences ,Neurology & Neurosurgery ,Biochemistry and cell biology - Abstract
BackgroundProgressive supranuclear palsy (PSP) is a rare neurodegenerative disease characterized by the accumulation of aggregated tau proteins in astrocytes, neurons, and oligodendrocytes. Previous genome-wide association studies for PSP were based on genotype array, therefore, were inadequate for the analysis of rare variants as well as larger mutations, such as small insertions/deletions (indels) and structural variants (SVs).MethodIn this study, we performed whole genome sequencing (WGS) and conducted association analysis for single nucleotide variants (SNVs), indels, and SVs, in a cohort of 1,718 cases and 2,944 controls of European ancestry. Of the 1,718 PSP individuals, 1,441 were autopsy-confirmed and 277 were clinically diagnosed.ResultsOur analysis of common SNVs and indels confirmed known genetic loci at MAPT, MOBP, STX6, SLCO1A2, DUSP10, and SP1, and further uncovered novel signals in APOE, FCHO1/MAP1S, KIF13A, TRIM24, TNXB, and ELOVL1. Notably, in contrast to Alzheimer's disease (AD), we observed the APOE ε2 allele to be the risk allele in PSP. Analysis of rare SNVs and indels identified significant association in ZNF592 and further gene network analysis identified a module of neuronal genes dysregulated in PSP. Moreover, seven common SVs associated with PSP were observed in the H1/H2 haplotype region (17q21.31) and other loci, including IGH, PCMT1, CYP2A13, and SMCP. In the H1/H2 haplotype region, there is a burden of rare deletions and duplications (P = 6.73 × 10-3) in PSP.ConclusionsThrough WGS, we significantly enhanced our understanding of the genetic basis of PSP, providing new targets for exploring disease mechanisms and therapeutic interventions.
- Published
- 2024
7. Single-nucleus expression characterization of non-enhancing region of recurrent high-grade glioma
- Author
-
Patel, Kunal S, Tessema, Kaleab K, Kawaguchi, Riki, Dudley, Lindsey, Alvarado, Alvaro G, Muthukrishnan, Sree Deepthi, Perryman, Travis, Hagiwara, Akifumi, Swarup, Vivek, Liau, Linda M, Wang, Anthony C, Yong, William, Geschwind, Daniel H, Nakano, Ichiro, Goldman, Steven A, Everson, Richard G, Ellingson, Benjamin M, and Kornblum, Harley I
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Clinical Research ,Stem Cell Research - Nonembryonic - Human ,Cancer ,Human Genome ,Stem Cell Research ,Genetics ,Brain Cancer ,Neurosciences ,Brain Disorders ,Rare Diseases ,contrast enhancing ,glioblastoma ,magnetic resonance imaging ,non-enhancing - Abstract
BackgroundNon-enhancing (NE) infiltrating tumor cells beyond the contrast-enhancing (CE) bulk of tumor are potential propagators of recurrence after gross total resection of high-grade glioma.MethodsWe leveraged single-nucleus RNA sequencing on 15 specimens from recurrent high-grade gliomas (n = 5) to compare prospectively identified biopsy specimens acquired from CE and NE regions. Additionally, 24 CE and 22 NE biopsies had immunohistochemical staining to validate RNA findings.ResultsTumor cells in NE regions are enriched in neural progenitor cell-like cellular states, while CE regions are enriched in mesenchymal-like states. NE glioma cells have similar proportions of proliferative and putative glioma stem cells relative to CE regions, without significant differences in % Ki-67 staining. Tumor cells in NE regions exhibit upregulation of genes previously associated with lower grade gliomas. Our findings in recurrent GBM paralleled some of the findings in a re-analysis of a dataset from primary GBM. Cell-, gene-, and pathway-level analyses of the tumor microenvironment in the NE region reveal relative downregulation of tumor-mediated neovascularization and cell-mediated immune response, but increased glioma-to-nonpathological cell interactions.ConclusionsThis comprehensive analysis illustrates differing tumor and nontumor landscapes of CE and NE regions in high-grade gliomas, highlighting the NE region as an area harboring likely initiators of recurrence in a pro-tumor microenvironment and identifying possible targets for future design of NE-specific adjuvant therapy. These findings also support the aggressive approach to resection of tumor-bearing NE regions.
- Published
- 2024
8. Extracellular free water elevations are associated with brain volume and maternal cytokine response in a longitudinal nonhuman primate maternal immune activation model
- Author
-
Lesh, Tyler A, Iosif, Ana-Maria, Tanase, Costin, Vlasova, Roza M, Ryan, Amy M, Bennett, Jeffrey, Hogrefe, Casey E, Maddock, Richard J, Geschwind, Daniel H, Van de Water, Judy, McAllister, A Kimberley, Styner, Martin A, Bauman, Melissa D, and Carter, Cameron S
- Subjects
Biological Psychology ,Biomedical and Clinical Sciences ,Psychology ,Mental Illness ,Women's Health ,Mental Health ,Prevention ,Biomedical Imaging ,Neurosciences ,Brain Disorders ,Pediatric ,Reproductive health and childbirth ,Neurological ,Mental health ,Female ,Animals ,Humans ,Male ,Cytokines ,Prenatal Exposure Delayed Effects ,Brain ,Schizophrenia ,Disease Models ,Animal ,Primates ,Behavior ,Animal ,Biological Sciences ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Psychiatry ,Clinical sciences ,Biological psychology ,Clinical and health psychology - Abstract
Maternal infection has emerged as an important environmental risk factor for neurodevelopmental disorders, including schizophrenia and autism spectrum disorders. Animal model systems of maternal immune activation (MIA) suggest that the maternal immune response plays a significant role in the offspring's neurodevelopment and behavioral outcomes. Extracellular free water is a measure of freely diffusing water in the brain that may be associated with neuroinflammation and impacted by MIA. The present study evaluates the brain diffusion characteristics of male rhesus monkeys (Macaca mulatta) born to MIA-exposed dams (n = 14) treated with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the end of the first trimester (n = 10) or were untreated (n = 4). Offspring underwent diffusion MRI scans at 6, 12, 24, 36, and 45 months. Offspring born to MIA-exposed dams showed significantly increased extracellular free water in cingulate cortex gray matter starting as early as 6 months of age and persisting through 45 months. In addition, offspring gray matter free water in this region was significantly correlated with the magnitude of the maternal IL-6 response in the MIA-exposed dams. Significant correlations between brain volume and extracellular free water in the MIA-exposed offspring also indicate converging, multimodal evidence of the impact of MIA on brain development. These findings provide strong evidence for the construct validity of the nonhuman primate MIA model as a system of relevance for investigating the pathophysiology of human neurodevelopmental psychiatric disorders. Elevated free water in individuals exposed to immune activation in utero could represent an early marker of a perturbed or vulnerable neurodevelopmental trajectory.
- Published
- 2023
9. Network Connectivity Alterations across the MAPT Mutation Clinical Spectrum
- Author
-
Zhang, Liwen, Flagan, Taru M, Häkkinen, Suvi, Chu, Stephanie A, Brown, Jesse A, Lee, Alex J, Pasquini, Lorenzo, Mandelli, Maria Luisa, Gorno‐Tempini, Maria Luisa, Sturm, Virginia E, Yokoyama, Jennifer S, Appleby, Brian S, Cobigo, Yann, Dickerson, Bradford C, Domoto‐Reilly, Kimiko, Geschwind, Daniel H, Ghoshal, Nupur, Graff‐Radford, Neill R, Grossman, Murray, Hsiung, Ging‐Yuek Robin, Huey, Edward D, Kantarci, Kejal, Lago, Argentina Lario, Litvan, Irene, Mackenzie, Ian R, Mendez, Mario F, Onyike, Chiadi U, Ramos, Eliana Marisa, Roberson, Erik D, Tartaglia, Maria Carmela, Toga, Arthur W, Weintraub, Sandra, Wszolek, Zbigniew K, Forsberg, Leah K, Heuer, Hilary W, Boeve, Bradley F, Boxer, Adam L, Rosen, Howard J, Miller, Bruce L, Seeley, William W, Lee, Suzee E, and Consortia, the ARTFL LEFFTDS ALLFTD
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Clinical Sciences ,Dementia ,Brain Disorders ,Alzheimer's Disease Related Dementias (ADRD) ,Clinical Research ,Frontotemporal Dementia (FTD) ,Mental Health ,Alzheimer's Disease ,Rare Diseases ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Aging ,Acquired Cognitive Impairment ,Neurodegenerative ,Biomedical Imaging ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Humans ,Cross-Sectional Studies ,tau Proteins ,Brain ,Mutation ,Gray Matter ,Magnetic Resonance Imaging ,Frontotemporal Dementia ,Biomarkers ,ARTFL/LEFFTDS/ALLFTD Consortia ,Neurology & Neurosurgery ,Clinical sciences - Abstract
ObjectiveMicrotubule-associated protein tau (MAPT) mutations cause frontotemporal lobar degeneration, and novel biomarkers are urgently needed for early disease detection. We used task-free functional magnetic resonance imaging (fMRI) mapping, a promising biomarker, to analyze network connectivity in symptomatic and presymptomatic MAPT mutation carriers.MethodsWe compared cross-sectional fMRI data between 17 symptomatic and 39 presymptomatic carriers and 81 controls with (1) seed-based analyses to examine connectivity within networks associated with the 4 most common MAPT-associated clinical syndromes (ie, salience, corticobasal syndrome, progressive supranuclear palsy syndrome, and default mode networks) and (2) whole-brain connectivity analyses. We applied K-means clustering to explore connectivity heterogeneity in presymptomatic carriers at baseline. Neuropsychological measures, plasma neurofilament light chain, and gray matter volume were compared at baseline and longitudinally between the presymptomatic subgroups defined by their baseline whole-brain connectivity profiles.ResultsSymptomatic and presymptomatic carriers had connectivity disruptions within MAPT-syndromic networks. Compared to controls, presymptomatic carriers showed regions of connectivity alterations with age. Two presymptomatic subgroups were identified by clustering analysis, exhibiting predominantly either whole-brain hypoconnectivity or hyperconnectivity at baseline. At baseline, these two presymptomatic subgroups did not differ in neuropsychological measures, although the hypoconnectivity subgroup had greater plasma neurofilament light chain levels than controls. Longitudinally, both subgroups showed visual memory decline (vs controls), yet the subgroup with baseline hypoconnectivity also had worsening verbal memory and neuropsychiatric symptoms, and extensive bilateral mesial temporal gray matter decline.InterpretationNetwork connectivity alterations arise as early as the presymptomatic phase. Future studies will determine whether presymptomatic carriers' baseline connectivity profiles predict symptomatic conversion. ANN NEUROL 2023;94:632-646.
- Published
- 2023
10. Author Correction: Genetic, transcriptomic, histological, and biochemical analysis of progressive supranuclear palsy implicates glial activation and novel risk genes
- Author
-
Farrell, Kurt, Humphrey, Jack, Chang, Timothy, Zhao, Yi, Leung, Yuk Yee, Kuksa, Pavel P., Patil, Vishakha, Lee, Wan-Ping, Kuzma, Amanda B., Valladares, Otto, Cantwell, Laura B., Wang, Hui, Ravi, Ashvin, De Sanctis, Claudia, Han, Natalia, Christie, Thomas D., Afzal, Robina, Kandoi, Shrishtee, Whitney, Kristen, Krassner, Margaret M., Ressler, Hadley, Kim, SoongHo, Dangoor, Diana, Iida, Megan A., Casella, Alicia, Walker, Ruth H., Nirenberg, Melissa J., Renton, Alan E., Babrowicz, Bergan, Coppola, Giovanni, Raj, Towfique, Höglinger, Günter U., Müller, Ulrich, Golbe, Lawrence I., Morris, Huw R., Hardy, John, Revesz, Tamas, Warner, Tom T., Jaunmuktane, Zane, Mok, Kin Y., Rademakers, Rosa, Dickson, Dennis W., Ross, Owen A., Wang, Li-San, Goate, Alison, Schellenberg, Gerard, Geschwind, Daniel H., Crary, John F., and Naj, Adam
- Published
- 2024
- Full Text
- View/download PDF
11. Correction: Whole-genome sequencing analysis reveals new susceptibility loci and structural variants associated with progressive supranuclear palsy
- Author
-
Wang, Hui, Chang, Timothy S., Dombroski, Beth A., Cheng, Po-Liang, Patil, Vishakha, Valiente-Banuet, Leopoldo, Farrell, Kurt, Mclean, Catriona, Molina-Porcel, Laura, Rajput, Alex, De Deyn, Peter Paul, Le Bastard, Nathalie, Gearing, Marla, Kaat, Laura Donker, Van Swieten, John C., Dopper, Elise, Ghetti, Bernardino F., Newell, Kathy L., Troakes, Claire, de Yébenes, Justo G., Rábano-Gutierrez, Alberto, Meller, Tina, Oertel, Wolfgang H., Respondek, Gesine, Stamelou, Maria, Arzberger, Thomas, Roeber, Sigrun, Müller, Ulrich, Hopfner, Franziska, Pastor, Pau, Brice, Alexis, Durr, Alexandra, Le Ber, Isabelle, Beach, Thomas G., Serrano, Geidy E., Hazrati, Lili-Naz, Litvan, Irene, Rademakers, Rosa, Ross, Owen A., Galasko, Douglas, Boxer, Adam L., Miller, Bruce L., Seeley, Willian W., Van Deerlin, Vivanna M., Lee, Edward B., White, III, Charles L., Morris, Huw, de Silva, Rohan, Crary, John F., Goate, Alison M., Friedman, Jeffrey S., Leung, Yuk Yee, Coppola, Giovanni, Naj, Adam C., Wang, Li-San, Dalgard, Clifton, Dickson, Dennis W., Höglinger, Günter U., Schellenberg, Gerard D., Geschwind, Daniel H., and Lee, Wan-Ping
- Published
- 2024
- Full Text
- View/download PDF
12. Genetic, transcriptomic, histological, and biochemical analysis of progressive supranuclear palsy implicates glial activation and novel risk genes
- Author
-
Farrell, Kurt, Humphrey, Jack, Chang, Timothy, Zhao, Yi, Leung, Yuk Yee, Kuksa, Pavel P., Patil, Vishakha, Lee, Wan-Ping, Kuzma, Amanda B., Valladares, Otto, Cantwell, Laura B., Wang, Hui, Ravi, Ashvin, De Sanctis, Claudia, Han, Natalia, Christie, Thomas D., Afzal, Robina, Kandoi, Shrishtee, Whitney, Kristen, Krassner, Margaret M., Ressler, Hadley, Kim, SoongHo, Dangoor, Diana, Iida, Megan A., Casella, Alicia, Walker, Ruth H., Nirenberg, Melissa J., Renton, Alan E., Babrowicz, Bergan, Coppola, Giovanni, Raj, Towfique, Höglinger, Günter U., Müller, Ulrich, Golbe, Lawrence I., Morris, Huw R., Hardy, John, Revesz, Tamas, Warner, Tom T., Jaunmuktane, Zane, Mok, Kin Y., Rademakers, Rosa, Dickson, Dennis W., Ross, Owen A., Wang, Li-San, Goate, Alison, Schellenberg, Gerard, Geschwind, Daniel H., Crary, John F., and Naj, Adam
- Published
- 2024
- Full Text
- View/download PDF
13. LATE-NC risk alleles (in TMEM106B, GRN, and ABCC9 genes) among persons with African ancestry.
- Author
-
Katsumata, Yuriko, Fardo, David W, Shade, Lincoln MP, Bowen, James D, Crane, Paul K, Jarvik, Gail P, Keene, C Dirk, Larson, Eric B, McCormick, Wayne C, McCurry, Susan M, Mukherjee, Shubhabrata, Kowall, Neil W, McKee, Ann C, Honig, Robert A, Lawrence, S, Vonsattel, Jean Paul, Williamson, Jennifer, Small, Scott, Burke, James R, Hulette, Christine M, Welsh-Bohmer, Kathleen A, Gearing, Marla, Lah, James J, Levey, Allan I, Wingo, Thomas S, Apostolova, Liana G, Farlow, Martin R, Ghetti, Bernardino, Saykin, Andrew J, Spina, Salvatore, Albert, Marilyn S, Lyketsos, Constantine G, Troncoso, Juan C, Frosch, Matthew P, Green, Robert C, Growdon, John H, Hyman, Bradley T, Tanzi, Rudolph E, Potter, Huntington, Dickson, Dennis W, Ertekin-Taner, Nilufer, Graff-Radford, Neill R, Parisi, Joseph E, Petersen, Ronald C, Duara, Ranjan, Buxbaum, Joseph D, Goate, Alison M, Sano, Mary, Masurkar, Arjun V, Wisniewski, Thomas, Bigio, Eileen H, Mesulam, Marsel, Weintraub, Sandra, Vassar, Robert, Kaye, Jeffrey A, Quinn, Joseph F, Woltjer, Randall L, Barnes, Lisa L, Bennett, David A, Schneider, Julie A, Yu, Lei, Henderson, Victor, Fallon, Kenneth B, Harrell, Lindy E, Marson, Daniel C, Roberson, Erik D, DeCarli, Charles, Jin, Lee-Way, Olichney, John M, Kim, Ronald, LaFerla, Frank M, Monuki, Edwin, Head, Elizabeth, Sultzer, David, Geschwind, Daniel H, Vinters, Harry V, Chesselet, Marie-Francoise, Galasko, Douglas R, Brewer, James B, Boxer, Adam, Karydas, Anna, Kramer, Joel H, Miller, Bruce L, Rosen, Howard J, Seeley, William W, Burns, Jeffrey M, Swerdlow, Russell H, Abner, Erin, Van Eldik, Linda J, Albin, Roger L, Lieberman, Andrew P, Paulson, Henry L, Arnold, Steven E, Trojanowski, John Q, Van Deerlin, Vivianna M, Hamilton, Ronald L, Kamboh, M Ilyas, Lopez, Oscar L, and Becker, James T
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Clinical Sciences ,Brain Disorders ,Neurodegenerative ,Alzheimer's Disease ,Acquired Cognitive Impairment ,Prevention ,Aging ,Minority Health ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Dementia ,Health Disparities ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Humans ,Alleles ,Polymorphism ,Single Nucleotide ,Alzheimer Disease ,TDP-43 Proteinopathies ,Progranulins ,Membrane Proteins ,Nerve Tissue Proteins ,Sulfonylurea Receptors ,Alzheimer’s Disease Genetics Consortium ,KCNMB2 ,Diversity ,Epidemiology ,FTLD ,Genome-Wide Association Studies ,KATP ,Neurology & Neurosurgery ,Clinical sciences - Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) affects approximately one-third of older individuals and is associated with cognitive impairment. However, there is a highly incomplete understanding of the genetic determinants of LATE neuropathologic changes (LATE-NC) in diverse populations. The defining neuropathologic feature of LATE-NC is TDP-43 proteinopathy, often with comorbid hippocampal sclerosis (HS). In terms of genetic risk factors, LATE-NC and/or HS are associated with single nucleotide variants (SNVs) in 3 genes-TMEM106B (rs1990622), GRN (rs5848), and ABCC9 (rs1914361 and rs701478). We evaluated these 3 genes in convenience samples of individuals of African ancestry. The allele frequencies of the LATE-associated alleles were significantly different between persons of primarily African (versus European) ancestry: In persons of African ancestry, the risk-associated alleles for TMEM106B and ABCC9 were less frequent, whereas the risk allele in GRN was more frequent. We performed an exploratory analysis of data from African-American subjects processed by the Alzheimer's Disease Genomics Consortium, with a subset of African-American participants (n = 166) having corroborating neuropathologic data through the National Alzheimer's Coordinating Center (NACC). In this limited-size sample, the ABCC9/rs1914361 SNV was associated with HS pathology. More work is required concerning the genetic factors influencing non-Alzheimer disease pathology such as LATE-NC in diverse cohorts.
- Published
- 2023
14. Increased Striatal Presynaptic Dopamine in a Nonhuman Primate Model of Maternal Immune Activation: A Longitudinal Neurodevelopmental Positron Emission Tomography Study With Implications for Schizophrenia
- Author
-
Smucny, Jason, Vlasova, Roza M, Lesh, Tyler A, Rowland, Douglas J, Wang, Guobao, Chaudhari, Abhijit J, Chen, Shuai, Iosif, Ana-Maria, Hogrefe, Casey E, Bennett, Jeffrey L, Shumann, Cynthia M, Van de Water, Judy A, Maddock, Richard J, Styner, Martin A, Geschwind, Daniel H, McAllister, A Kimberley, Bauman, Melissa D, and Carter, Cameron S
- Subjects
Biological Psychology ,Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Psychology ,Neurosciences ,Pediatric ,Women's Health ,Serious Mental Illness ,Brain Disorders ,Mental Health ,Mental Illness ,Schizophrenia ,Reproductive health and childbirth ,Pregnancy ,Animals ,Female ,Humans ,Male ,Dopamine ,Cross-Sectional Studies ,Longitudinal Studies ,Prospective Studies ,Prenatal Exposure Delayed Effects ,Positron-Emission Tomography ,Primates ,Caudate ,Dopaminergic ,Inflammation ,Macaque ,Putamen ,Striatum ,Biological psychology ,Clinical and health psychology - Abstract
BackgroundEpidemiological studies suggest that maternal immune activation (MIA) is a significant risk factor for future neurodevelopmental disorders, including schizophrenia (SZ), in offspring. Consistent with findings in SZ research and work in rodent systems, preliminary cross-sectional findings in nonhuman primates suggest that MIA is associated with dopaminergic hyperfunction in young adult offspring.MethodsIn this unique prospective longitudinal study, we used [18F]fluoro-l-m-tyrosine positron emission tomography to examine the developmental time course of striatal presynaptic dopamine synthesis in male rhesus monkeys born to dams (n = 13) injected with a modified form of the inflammatory viral mimic, polyinosinic:polycytidylic acid [poly(I:C)], in the late first trimester. Striatal (caudate, putamen, and nucleus accumbens) dopamine from these animals was compared with that of control offspring born to dams that received saline (n = 10) or no injection (n = 4). Dopamine was measured at 15, 26, 38, and 48 months of age. Prior work with this cohort found decreased prefrontal gray matter volume in MIA offspring versus controls between 6 and 45 months of age. Based on theories of the etiology and development of SZ-related pathology, we hypothesized that there would be a delayed (relative to the gray matter decrease) increase in striatal fluoro-l-m-tyrosine signal in the MIA group versus controls.Results[18F]fluoro-l-m-tyrosine signal showed developmental increases in both groups in the caudate and putamen. Group comparisons revealed significantly greater caudate dopaminergic signal in the MIA group at 26 months.ConclusionsThese findings are highly relevant to the known pathophysiology of SZ and highlight the translational relevance of the MIA model in understanding mechanisms by which MIA during pregnancy increases risk for later illness in offspring.
- Published
- 2023
15. A transcriptomic taxonomy of mouse brain-wide spinal projecting neurons
- Author
-
Winter, Carla C., Jacobi, Anne, Su, Junfeng, Chung, Leeyup, van Velthoven, Cindy T. J., Yao, Zizhen, Lee, Changkyu, Zhang, Zicong, Yu, Shuguang, Gao, Kun, Duque Salazar, Geraldine, Kegeles, Evgenii, Zhang, Yu, Tomihiro, Makenzie C., Zhang, Yiming, Yang, Zhiyun, Zhu, Junjie, Tang, Jing, Song, Xuan, Donahue, Ryan J., Wang, Qing, McMillen, Delissa, Kunst, Michael, Wang, Ning, Smith, Kimberly A., Romero, Gabriel E., Frank, Michelle M., Krol, Alexandra, Kawaguchi, Riki, Geschwind, Daniel H., Feng, Guoping, Goodrich, Lisa V., Liu, Yuanyuan, Tasic, Bosiljka, Zeng, Hongkui, and He, Zhigang
- Published
- 2023
- Full Text
- View/download PDF
16. Distinct Patterns of Gene Expression Changes in the Colon and Striatum of Young Mice Overexpressing Alpha-Synuclein Support Parkinson’s Disease as a Multi-System Process
- Author
-
Videlock, Elizabeth J, Hatami, Asa, Zhu, Chunni, Kawaguchi, Riki, Chen, Han, Khan, Tasnin, Yehya, Ashwaq Hamid Salem, Stiles, Linsey, Joshi, Swapna, Hoffman, Jill M, Law, Ka Man, Rankin, Carl Robert, Chang, Lin, Maidment, Nigel T, John, Varghese, Geschwind, Daniel H, and Pothoulakis, Charalabos
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Prevention ,Parkinson's Disease ,Aging ,Neurodegenerative ,Brain Disorders ,Digestive Diseases ,Genetics ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Humans ,Mice ,alpha-Synuclein ,Colon ,Disease Models ,Animal ,Gene Expression ,Mice ,Transgenic ,Parkinson Disease ,Parkinson's disease ,brain-gut axis ,gene expression profiling ,alpha-synuclein ,Parkinson’s disease ,Biochemistry and Cell Biology - Abstract
BackgroundEvidence supports a role for the gut-brain axis in Parkinson's disease (PD). Mice overexpressing human wild type α- synuclein (Thy1-haSyn) exhibit slow colonic transit prior to motor deficits, mirroring prodromal constipation in PD. Identifying molecular changes in the gut could provide both biomarkers for early diagnosis and gut-targeted therapies to prevent progression.ObjectiveTo identify early molecular changes in the gut-brain axis in Thy1-haSyn mice through gene expression profiling.MethodsGene expression profiling was performed on gut (colon) and brain (striatal) tissue from Thy1-haSyn and wild-type (WT) mice aged 1 and 3 months using 3' RNA sequencing. Analysis included differential expression, gene set enrichment and weighted gene co-expression network analysis (WGCNA).ResultsAt one month, differential expression (Thy1-haSyn vs. WT) of mitochondrial genes and pathways related to PD was discordant between gut and brain, with negative enrichment in brain (enriched in WT) but positive enrichment in gut. Linear regression of WGCNA modules showed partial independence of gut and brain gene expression changes. Thy1-haSyn-associated WGCNA modules in the gut were enriched for PD risk genes and PD-relevant pathways including inflammation, autophagy, and oxidative stress. Changes in gene expression were modest at 3 months.ConclusionsOverexpression of haSyn acutely disrupts gene expression in the colon. While changes in colon gene expression are highly related to known PD-relevant mechanisms, they are distinct from brain changes, and in some cases, opposite in direction. These findings are in line with the emerging view of PD as a multi-system disease.
- Published
- 2023
17. Tuberous sclerosis complex is associated with a novel human tauopathy
- Author
-
Hwang, Ji-Hye L, Perloff, Olga S, Gaus, Stephanie E, Benitez, Camila, Alquezar, Carolina, Cosme, Celica Q, Nana, Alissa L, Vatsavayai, Sarat C, Ramos, Eliana M, Geschwind, Daniel H, Miller, Bruce L, Kao, Aimee W, and Seeley, William W
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Aging ,Acquired Cognitive Impairment ,Alzheimer's Disease Related Dementias (ADRD) ,Neurodegenerative ,Dementia ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Clinical Research ,Brain Disorders ,Tuberous Sclerosis ,Rare Diseases ,Intellectual and Developmental Disabilities (IDD) ,Epilepsy ,Frontotemporal Dementia (FTD) ,Alzheimer's Disease ,2.1 Biological and endogenous factors ,Neurological ,Adult ,Humans ,Tumor Suppressor Proteins ,Amyloid beta-Peptides ,Mutation ,Tauopathies ,Tuberous sclerosis complex ,Tau ,Acetylation ,Tauopathy ,TSC1 ,TSC2 ,Neurofibrillary tangle ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
Tuberous sclerosis complex (TSC) is a neurogenetic disorder leading to epilepsy, developmental delay, and neurobehavioral dysfunction. The syndrome is caused by pathogenic variants in TSC1 (coding for hamartin) or TSC2 (coding for tuberin). Recently, we reported a progressive frontotemporal dementia-like clinical syndrome in a patient with a mutation in TSC1, but the neuropathological changes seen in adults with TSC with or without dementia have yet to be systematically explored. Here, we examined neuropathological findings in adults with TSC (n = 11) aged 30-58 years and compared them to age-matched patients with epilepsy unrelated to TSC (n = 9) and non-neurological controls (n = 10). In 3 of 11 subjects with TSC, we observed a neurofibrillary tangle-predominant "TSC tauopathy" not seen in epilepsy or non-neurological controls. This tauopathy was observed in the absence of pathological amyloid beta, TDP-43, or alpha-synuclein deposition. The neurofibrillary tangles in TSC tauopathy showed a unique pattern of post-translational modifications, with apparent differences between TSC1 and TSC2 mutation carriers. Tau acetylation (K274, K343) was prominent in both TSC1 and TSC2, whereas tau phosphorylation at a common phospho-epitope (S202) was observed only in TSC2. TSC tauopathy was observed in selected neocortical, limbic, subcortical, and brainstem sites and showed a 3-repeat greater than 4-repeat tau isoform pattern in both TSC1 and TSC2 mutation carriers, but no tangles were immunolabeled with MC1 or p62 antibodies. The findings suggest that individuals with TSC are at risk for a unique tauopathy in mid-life and that tauopathy pathogenesis may involve TSC1, TSC2, and related molecular pathways.
- Published
- 2023
18. The UCLA ATLAS Community Health Initiative: Promoting precision health research in a diverse biobank
- Author
-
Johnson, Ruth, Ding, Yi, Bhattacharya, Arjun, Knyazev, Sergey, Chiu, Alec, Lajonchere, Clara, Geschwind, Daniel H, and Pasaniuc, Bogdan
- Subjects
Biological Sciences ,Genetics ,Epidemiology ,Health Services and Systems ,Health Sciences ,Networking and Information Technology R&D (NITRD) ,Mental Health ,Biotechnology ,Patient Safety ,Human Genome ,Aetiology ,2.6 Resources and infrastructure (aetiology) ,Generic health relevance ,Good Health and Well Being ,GWAS ,Global-Biobank Meta-analysis Initiative ,PheWAS ,biobanks ,electronic health records ,multi-ancestry - Abstract
The UCLA ATLAS Community Health Initiative (ATLAS) has an initial target to recruit 150,000 participants from across the UCLA Health system with the goal of creating a genomic database to accelerate precision medicine efforts in California. This initiative includes a biobank embedded within the UCLA Health system that comprises de-identified genomic data linked to electronic health records (EHRs). The first freeze of data from September 2020 contains 27,987 genotyped samples imputed to 7.9 million SNPs across the genome and is linked with de-identified versions of the EHRs from UCLA Health. Here, we describe a centralized repository of the genotype data and provide tools and pipelines to perform genome- and phenome-wide association studies across a wide range of EHR-derived phenotypes and genetic ancestry groups. We demonstrate the utility of this resource through the analysis of 7 well-studied traits and recapitulate many previous genetic and phenotypic associations.
- Published
- 2023
19. Broad transcriptomic dysregulation occurs across the cerebral cortex in ASD
- Author
-
Gandal, Michael J, Haney, Jillian R, Wamsley, Brie, Yap, Chloe X, Parhami, Sepideh, Emani, Prashant S, Chang, Nathan, Chen, George T, Hoftman, Gil D, de Alba, Diego, Ramaswami, Gokul, Hartl, Christopher L, Bhattacharya, Arjun, Luo, Chongyuan, Jin, Ting, Wang, Daifeng, Kawaguchi, Riki, Quintero, Diana, Ou, Jing, Wu, Ye Emily, Parikshak, Neelroop N, Swarup, Vivek, Belgard, T Grant, Gerstein, Mark, Pasaniuc, Bogdan, and Geschwind, Daniel H
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Human Genome ,Autism ,Mental Health ,Eye Disease and Disorders of Vision ,Neurosciences ,Brain Disorders ,Pediatric ,Intellectual and Developmental Disabilities (IDD) ,Aetiology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Neurological ,Mental health ,Humans ,Autism Spectrum Disorder ,Cerebral Cortex ,Neurons ,RNA ,Transcriptome ,Autopsy ,Sequence Analysis ,RNA ,Primary Visual Cortex ,Neuroglia ,Genetic Variation ,General Science & Technology - Abstract
Neuropsychiatric disorders classically lack defining brain pathologies, but recent work has demonstrated dysregulation at the molecular level, characterized by transcriptomic and epigenetic alterations1-3. In autism spectrum disorder (ASD), this molecular pathology involves the upregulation of microglial, astrocyte and neural-immune genes, the downregulation of synaptic genes, and attenuation of gene-expression gradients in cortex1,2,4-6. However, whether these changes are limited to cortical association regions or are more widespread remains unknown. To address this issue, we performed RNA-sequencing analysis of 725 brain samples spanning 11 cortical areas from 112 post-mortem samples from individuals with ASD and neurotypical controls. We find widespread transcriptomic changes across the cortex in ASD, exhibiting an anterior-to-posterior gradient, with the greatest differences in primary visual cortex, coincident with an attenuation of the typical transcriptomic differences between cortical regions. Single-nucleus RNA-sequencing and methylation profiling demonstrate that this robust molecular signature reflects changes in cell-type-specific gene expression, particularly affecting excitatory neurons and glia. Both rare and common ASD-associated genetic variation converge within a downregulated co-expression module involving synaptic signalling, and common variation alone is enriched within a module of upregulated protein chaperone genes. These results highlight widespread molecular changes across the cerebral cortex in ASD, extending beyond association cortex to broadly involve primary sensory regions.
- Published
- 2022
20. Pathway-based approach reveals differential sensitivity to E2F1 inhibition in glioblastoma
- Author
-
Alvarado, Alvaro G, Tessema, Kaleab, Muthukrishnan, Sree Deepthi, Sober, Mackenzie, Kawaguchi, Riki, Laks, Dan R, Bhaduri, Aparna, Swarup, Vivek, Nathanson, David A, Geschwind, Daniel H, Goldman, Steven A, and Kornblum, Harley I
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Brain Cancer ,Stem Cell Research ,Rare Diseases ,Cancer ,Brain Disorders ,Genetics ,Neurosciences ,Biotechnology ,Aetiology ,2.1 Biological and endogenous factors ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Humans ,Glioblastoma ,Cell Line ,Tumor ,Transcription Factors ,Cell Proliferation ,E2F1 Transcription Factor ,CIP2A ,E2F1 ,Molecular targets ,Tumoral heterogeneity - Abstract
Analysis of tumor gene expression is an important approach for the classification and identification of therapeutic vulnerabilities. However, targeting glioblastoma (GBM) based on molecular subtyping has not yet translated into successful therapies. Here, we present an integrative approach based on molecular pathways to expose new potentially actionable targets. We used gene set enrichment analysis (GSEA) to conduct an unsupervised clustering analysis to condense the gene expression data from bulk patient samples and patient-derived gliomasphere lines into new gene signatures. We identified key targets that are predicted to be differentially activated between tumors and were functionally validated in a library of gliomasphere cultures. Resultant cluster-specific gene signatures associated not only with hallmarks of cell cycle and stemness gene expression, but also with cell-type specific markers and different cellular states of GBM. Several upstream regulators, such as PIK3R1 and EBF1 were differentially enriched in cells bearing stem cell like signatures and bear further investigation. We identified the transcription factor E2F1 as a key regulator of tumor cell proliferation and self-renewal in only a subset of gliomasphere cultures predicted to be E2F1 signaling dependent. Our in vivo work also validated the functional significance of E2F1 in tumor formation capacity in the predicted samples. E2F1 inhibition also differentially sensitized E2F1-dependent gliomasphere cultures to radiation treatment. Our findings indicate that this novel approach exploring cancer pathways highlights key therapeutic vulnerabilities for targeting GBM.
- Published
- 2022
21. Differences in Motor Features of C9orf72, MAPT, or GRN Variant Carriers With Familial Frontotemporal Lobar Degeneration.
- Author
-
Tipton, Philip Wade, Deutschlaender, Angela B, Savica, Rodolfo, Heckman, Michael G, Brushaber, Danielle E, Dickerson, Bradford C, Gavrilova, Ralitza H, Geschwind, Daniel H, Ghoshal, Nupur, Graff-Radford, Jonathan, Graff-Radford, Neill R, Grossman, Murray, Hsiung, Ging-Yuek R, Huey, Edward D, Irwin, David John, Jones, David T, Knopman, David S, McGinnis, Scott M, Rademakers, Rosa, Ramos, Eliana Marisa, Forsberg, Leah K, Heuer, Hilary W, Onyike, Chiadi, Tartaglia, Carmela, Domoto-Reilly, Kimiko, Roberson, Erik D, Mendez, Mario F, Litvan, Irene, Appleby, Brian S, Grant, Ian, Kaufer, Daniel, Boxer, Adam L, Rosen, Howard J, Boeve, Brad F, and Wszolek, Zbigniew K
- Subjects
C9orf72 Protein: genetics ,Frontotemporal Dementia: diagnosis ,genetics ,Frontotemporal Lobar Degeneration: genetics ,Granulins: genetics ,Humans ,Mutation: genetics ,Progranulins: genetics ,Quality of Life ,Supranuclear Palsy ,Progressive ,tau Proteins: genetics - Abstract
Familial frontotemporal lobar degeneration (f-FTLD) is a phenotypically heterogeneous spectrum of neurodegenerative disorders most often caused by variants within chromosome 9 open reading frame 72 (C9orf72), microtubule-associated protein tau (MAPT), or granulin (GRN). The phenotypic association with each of these genes is incompletely understood. We hypothesized that the frequency of specific clinical features would correspond with different genes.We screened the Advancing Research and Treatment in Frontotemporal Lobar Degeneration (ARTFL)/Longitudinal Evaluation of Familial Frontotemporal Dementia Subjects (LEFFTDS)/ARTFL LEFFTDS Longitudinal Frontotemporal Lobar Degeneration Consortium for symptomatic carriers of pathogenic variants in C9orf72, MAPT, or GRN. We assessed for clinical differences among these 3 groups based on data recorded as part of a detailed neurologic examination, the Progressive Supranuclear Palsy Rating Scale, Progressive Supranuclear Palsy-Quality of Life Rating Scale, Unified Parkinson's Disease Rating Scale Part III (motor items), and the Amyotrophic Lateral Sclerosis Functional Rating Scale, revised version. Data were analyzed using Kruskal-Wallis and Wilcoxon rank-sum tests and Fisher exact test.We identified 184 symptomatic participants who had a single pathogenic variant in C9orf72 (n = 88), MAPT (n = 53), or GRN (n = 43). Motor symptom age at onset was earliest in the MAPT participants followed by C9orf72, whereas the GRN pathogenic variant carriers developed symptoms later. C9orf72 participants more often had fasciculations, muscle atrophy, and weakness, whereas parkinsonism was less frequent. Vertical oculomotor abnormalities were more common in the MAPT cohort, whereas apraxia and focal limb dystonia occurred more often in participants with GRN variants.We present a large comparative study of motor features in C9orf72, MAPT, and GRN pathogenic variant carriers with symptomatic f-FTLD. Our findings demonstrate characteristic phenotypic differences corresponding with specific gene variants that increase our understanding of the genotype-phenotype relationship in this complex spectrum of neurodegenerative disorders.NCT02365922, NCT02372773, and NCT04363684.
- Published
- 2022
22. Integrating de novo and inherited variants in 42,607 autism cases identifies mutations in new moderate-risk genes
- Author
-
Zhou, Xueya, Feliciano, Pamela, Shu, Chang, Wang, Tianyun, Astrovskaya, Irina, Hall, Jacob B, Obiajulu, Joseph U, Wright, Jessica R, Murali, Shwetha C, Xu, Simon Xuming, Brueggeman, Leo, Thomas, Taylor R, Marchenko, Olena, Fleisch, Christopher, Barns, Sarah D, Snyder, LeeAnne Green, Han, Bing, Chang, Timothy S, Turner, Tychele N, Harvey, William T, Nishida, Andrew, O’Roak, Brian J, Geschwind, Daniel H, Michaelson, Jacob J, Volfovsky, Natalia, Eichler, Evan E, Shen, Yufeng, and Chung, Wendy K
- Subjects
Pediatric Research Initiative ,Genetics ,Intellectual and Developmental Disabilities (IDD) ,Autism ,Clinical Research ,Genetic Testing ,Pediatric ,Mental Health ,Brain Disorders ,2.1 Biological and endogenous factors ,Aetiology ,Autism Spectrum Disorder ,Autistic Disorder ,Exome ,Forkhead Transcription Factors ,Genetic Predisposition to Disease ,Humans ,Mutation ,Repressor Proteins ,Exome Sequencing ,SPARK Consortium ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
To capture the full spectrum of genetic risk for autism, we performed a two-stage analysis of rare de novo and inherited coding variants in 42,607 autism cases, including 35,130 new cases recruited online by SPARK. We identified 60 genes with exome-wide significance (P
- Published
- 2022
23. Cross-disorder and disease-specific pathways in dementia revealed by single-cell genomics
- Author
-
Rexach, Jessica E., Cheng, Yuyan, Chen, Lawrence, Polioudakis, Damon, Lin, Li-Chun, Mitri, Vivianne, Elkins, Andrew, Han, Xia, Yamakawa, Mai, Yin, Anna, Calini, Daniela, Kawaguchi, Riki, Ou, Jing, Huang, Jerry, Williams, Christopher, Robinson, John, Gaus, Stephanie E., Spina, Salvatore, Lee, Edward B., Grinberg, Lea T., Vinters, Harry, Trojanowski, John Q., Seeley, William W., Malhotra, Dheeraj, and Geschwind, Daniel H.
- Published
- 2024
- Full Text
- View/download PDF
24. Altered brain metabolites in male nonhuman primate offspring exposed to maternal immune activation
- Author
-
Maddock, Richard J., Vlasova, Roza M., Chen, Shuai, Iosif, Ana-Maria, Bennett, Jeffrey, Tanase, Costin, Ryan, Amy M., Murai, Takeshi, Hogrefe, Casey E., Schumann, Cynthia D., Geschwind, Daniel H., Van de Water, Judy, Amaral, David G., Lesh, Tyler A., Styner, Martin A., Kimberley McAllister, A., Carter, Cameron S., and Bauman, Melissa D.
- Published
- 2024
- Full Text
- View/download PDF
25. Innate immune training restores pro-reparative myeloid functions to promote remyelination in the aged central nervous system
- Author
-
Tiwari, Vini, Prajapati, Bharat, Asare, Yaw, Damkou, Alkmini, Ji, Hao, Liu, Lu, Naser, Nawraa, Gouna, Garyfallia, Leszczyńska, Katarzyna B., Mieczkowski, Jakub, Dichgans, Martin, Wang, Qing, Kawaguchi, Riki, Shi, Zechuan, Swarup, Vivek, Geschwind, Daniel H., Prinz, Marco, Gokce, Ozgun, and Simons, Mikael
- Published
- 2024
- Full Text
- View/download PDF
26. Using Organoids to Model Sex Differences in the Human Brain
- Author
-
Baron-Cohen, Simon, Allison, Carrie, Warrier, Varun, Tsompanidis, Alex, Adhya, Dwaipayan, Holt, Rosie, Smith, Paula, Parsons, Tracey, Davis, Joanna, Hassall, Matthew, Geschwind, Daniel H., Heazell, Alexander EP., Mill, Jonathan, Franklin, Alice, Bamford, Rosie, Davies, Jonathan, Hurles, Matthew E., Martin, Hilary C., Mousa, Mahmoud, Rowitch, David H., Niakan, Kathy K., Burton, Graham J., Ghafari, Fateneh, Srivastava, Deepak P., Dutan-Polit, Lucia, Pavlinek, Adam, Lancaster, Madeline A., Chiaradia, Ilaria, Biron-Shental, Tal, Gabis, Lidia V., Vernon, Anthony C., and Lancaster, Madeline
- Published
- 2024
- Full Text
- View/download PDF
27. Integrative analyses highlight functional regulatory variants associated with neuropsychiatric diseases
- Author
-
Guo, Margaret G., Reynolds, David L., Ang, Cheen E., Liu, Yingfei, Zhao, Yang, Donohue, Laura K. H., Siprashvili, Zurab, Yang, Xue, Yoo, Yongjin, Mondal, Smarajit, Hong, Audrey, Kain, Jessica, Meservey, Lindsey, Fabo, Tania, Elfaki, Ibtihal, Kellman, Laura N., Abell, Nathan S., Pershad, Yash, Bayat, Vafa, Etminani, Payam, Holodniy, Mark, Geschwind, Daniel H., Montgomery, Stephen B., Duncan, Laramie E., Urban, Alexander E., Altman, Russ B., Wernig, Marius, and Khavari, Paul A.
- Published
- 2023
- Full Text
- View/download PDF
28. Genetic insights into human cortical organization and development through genome-wide analyses of 2,347 neuroimaging phenotypes
- Author
-
Warrier, Varun, Stauffer, Eva-Maria, Huang, Qin Qin, Wigdor, Emilie M., Slob, Eric A. W., Seidlitz, Jakob, Ronan, Lisa, Valk, Sofie L., Mallard, Travis T., Grotzinger, Andrew D., Romero-Garcia, Rafael, Baron-Cohen, Simon, Geschwind, Daniel H., Lancaster, Madeline A., Murray, Graham K., Gandal, Michael J., Alexander-Bloch, Aaron, Won, Hyejung, Martin, Hilary C., Bullmore, Edward T., and Bethlehem, Richard A. I.
- Published
- 2023
- Full Text
- View/download PDF
29. Impact of autism genetic risk on brain connectivity: a mechanism for the female protective effect.
- Author
-
Lawrence, Katherine E, Hernandez, Leanna M, Fuster, Emily, Padgaonkar, Namita T, Patterson, Genevieve, Jung, Jiwon, Okada, Nana J, Lowe, Jennifer K, Hoekstra, Jackson N, Jack, Allison, Aylward, Elizabeth, Gaab, Nadine, Van Horn, John D, Bernier, Raphael A, McPartland, James C, Webb, Sara J, Pelphrey, Kevin A, Green, Shulamite A, Bookheimer, Susan Y, Geschwind, Daniel H, Dapretto, Mirella, Nelson, Charles A, Ankenman, Katy, Corrigan, Sarah, Depedro-Mercier, Dianna, Guilford, Desiree, Gupta, Abha R, Jacokes, Zachary, Jeste, Shafali, Keifer, Cara M, Libsack, Erin, Kresse, Anna, MacDonnell, Erin, McDonald, Nicole, Naples, Adam, Neuhaus, Emily, Sullivan, Catherine AW, Tsapelas, Heidi, Torgerson, Carinna M, Ventola, Pamela, Welker, Olivia, and Wolf, Julie
- Subjects
Pediatric ,Neurosciences ,Autism ,Intellectual and Developmental Disabilities (IDD) ,Mental Health ,Genetics ,Serious Mental Illness ,Behavioral and Social Science ,Brain Disorders ,Prevention ,Aetiology ,2.1 Biological and endogenous factors ,2.3 Psychological ,social and economic factors ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Mental health ,Adolescent ,Autism Spectrum Disorder ,Autistic Disorder ,Brain ,Brain Mapping ,Child ,Female ,Humans ,Magnetic Resonance Imaging ,Male ,autism spectrum disorder ,female protective effect ,functional connectivity ,imaging genetics ,polygenic risk ,GENDAAR Consortium ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
The biological mechanisms underlying the greater prevalence of autism spectrum disorder in males than females remain poorly understood. One hypothesis posits that this female protective effect arises from genetic load for autism spectrum disorder differentially impacting male and female brains. To test this hypothesis, we investigated the impact of cumulative genetic risk for autism spectrum disorder on functional brain connectivity in a balanced sample of boys and girls with autism spectrum disorder and typically developing boys and girls (127 youth, ages 8-17). Brain connectivity analyses focused on the salience network, a core intrinsic functional connectivity network which has previously been implicated in autism spectrum disorder. The effects of polygenic risk on salience network functional connectivity were significantly modulated by participant sex, with genetic load for autism spectrum disorder influencing functional connectivity in boys with and without autism spectrum disorder but not girls. These findings support the hypothesis that autism spectrum disorder risk genes interact with sex differential processes, thereby contributing to the male bias in autism prevalence and proposing an underlying neurobiological mechanism for the female protective effect.
- Published
- 2022
30. Oxytocin normalizes altered circuit connectivity for social rescue of the Cntnap2 knockout mouse
- Author
-
Choe, Katrina Y, Bethlehem, Richard AI, Safrin, Martin, Dong, Hongmei, Salman, Elena, Li, Ying, Grinevich, Valery, Golshani, Peyman, DeNardo, Laura A, Peñagarikano, Olga, Harris, Neil G, and Geschwind, Daniel H
- Subjects
Biological Psychology ,Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Psychology ,Basic Behavioral and Social Science ,Neurosciences ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Mental Health ,Autism ,Behavioral and Social Science ,Underpinning research ,1.2 Psychological and socioeconomic processes ,Aetiology ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Neurological ,Mental health ,Animals ,Autism Spectrum Disorder ,Brain ,Membrane Proteins ,Mice ,Mice ,Knockout ,Nerve Tissue Proteins ,Oxytocin ,Receptors ,Oxytocin ,Social Behavior ,autism ,brain network ,fMRI ,functional connectivity ,iDISCO ,mouse model ,nucleus accumbens ,oxytocin ,paraventricular nucleus ,social behavior ,Cognitive Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
The neural basis of abnormal social behavior in autism spectrum disorders (ASDs) remains incompletely understood. Here we used two complementary but independent brain-wide mapping approaches, mouse resting-state fMRI and c-Fos-iDISCO+ imaging, to construct brain-wide activity and connectivity maps of the Cntnap2 knockout (KO) mouse model of ASD. At the macroscale level, we detected reduced functional coupling across social brain regions despite general patterns of hyperconnectivity across major brain structures. Oxytocin administration, which rescues social deficits in KO mice, strongly stimulated many brain areas and normalized connectivity patterns. Notably, chemogenetically triggered release of endogenous oxytocin strongly stimulated the nucleus accumbens (NAc), a forebrain nucleus implicated in social reward. Furthermore, NAc-targeted approaches to activate local oxytocin receptors sufficiently rescued their social deficits. Our findings establish circuit- and systems-level mechanisms of social deficits in Cntnap2 KO mice and reveal the NAc as a region that can be modulated by oxytocin to promote social interactions.
- Published
- 2022
31. Neutrophil-inflicted vasculature damage suppresses immune-mediated optic nerve regeneration
- Author
-
Passino, Ryan, Finneran, Matthew C., Hafner, Hannah, Feng, Qian, Huffman, Lucas D., Zhao, Xiao-Feng, Johnson, Craig N., Kawaguchi, Riki, Oses-Prieto, Juan A., Burlingame, Alma L., Geschwind, Daniel H., Benowitz, Larry I., and Giger, Roman J.
- Published
- 2024
- Full Text
- View/download PDF
32. A gut-derived metabolite alters brain activity and anxiety behaviour in mice
- Author
-
Needham, Brittany D, Funabashi, Masanori, Adame, Mark D, Wang, Zhuo, Boktor, Joseph C, Haney, Jillian, Wu, Wei-Li, Rabut, Claire, Ladinsky, Mark S, Hwang, Son-Jong, Guo, Yumei, Zhu, Qiyun, Griffiths, Jessica A, Knight, Rob, Bjorkman, Pamela J, Shapiro, Mikhail G, Geschwind, Daniel H, Holschneider, Daniel P, Fischbach, Michael A, and Mazmanian, Sarkis K
- Subjects
Neurosciences ,Genetics ,Autoimmune Disease ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Underpinning research ,1.1 Normal biological development and functioning ,Oral and gastrointestinal ,Mental health ,Neurological ,Animals ,Anxiety ,Bacteria ,Brain ,Gastrointestinal Microbiome ,Mice ,Mice ,Inbred C57BL ,Microbiota ,Myelin Sheath ,Phenols ,General Science & Technology - Abstract
Integration of sensory and molecular inputs from the environment shapes animal behaviour. A major site of exposure to environmental molecules is the gastrointestinal tract, in which dietary components are chemically transformed by the microbiota1 and gut-derived metabolites are disseminated to all organs, including the brain2. In mice, the gut microbiota impacts behaviour3, modulates neurotransmitter production in the gut and brain4,5, and influences brain development and myelination patterns6,7. The mechanisms that mediate the gut-brain interactions remain poorly defined, although they broadly involve humoral or neuronal connections. We previously reported that the levels of the microbial metabolite 4-ethylphenyl sulfate (4EPS) were increased in a mouse model of atypical neurodevelopment8. Here we identified biosynthetic genes from the gut microbiome that mediate the conversion of dietary tyrosine to 4-ethylphenol (4EP), and bioengineered gut bacteria to selectively produce 4EPS in mice. 4EPS entered the brain and was associated with changes in region-specific activity and functional connectivity. Gene expression signatures revealed altered oligodendrocyte function in the brain, and 4EPS impaired oligodendrocyte maturation in mice and decreased oligodendrocyte-neuron interactions in ex vivo brain cultures. Mice colonized with 4EP-producing bacteria exhibited reduced myelination of neuronal axons. Altered myelination dynamics in the brain have been associated with behavioural outcomes7,9-14. Accordingly, we observed that mice exposed to 4EPS displayed anxiety-like behaviours, and pharmacological treatments that promote oligodendrocyte differentiation prevented the behavioural effects of 4EPS. These findings reveal that a gut-derived molecule influences complex behaviours in mice through effects on oligodendrocyte function and myelin patterning in the brain.
- Published
- 2022
33. A comprehensive map of genetic relationships among diagnostic categories based on 48.6 million relative pairs from the Danish genealogy
- Author
-
Athanasiadis, Georgios, Meijsen, Joeri J, Helenius, Dorte, Schork, Andrew J, Ingason, Andrés, Thompson, Wesley K, Geschwind, Daniel H, Werge, Thomas, and Buil, Alfonso
- Subjects
Genetics ,Good Health and Well Being ,Denmark ,Genetic Diseases ,Inborn ,Genetic Predisposition to Disease ,Health Services Research ,Humans ,Mental Disorders ,heritability ,genetic correlation ,human disease ,register data - Abstract
For more than half a century, Denmark has maintained population-wide demographic, health care, and socioeconomic registers that provide detailed information on the interaction between all residents and the extensive national social services system. We leverage this resource to reconstruct the genealogy of the entire nation based on all individuals legally residing in Denmark since 1968. We cross-reference 6,691,426 individuals with nationwide health care registers to estimate heritability and genetic correlations of 10 broad diagnostic categories involving all major organs and systems. Heritability estimates for mental disorders were consistently the highest across demographic cohorts (average h2 = 0.406, 95% CI = [0.403, 0.408]), whereas estimates for cancers were the lowest (average h2 = 0.130, 95% CI = [0.125, 0.134]). The average genetic correlation of each of the 10 diagnostic categories with the other nine was highest for gastrointestinal conditions (average rg = 0.567, 95% CI = [0.566, 0.567]) and lowest for urogenital conditions (average rg = 0.386, 95% CI = [0.385, 0.388]). Mental, pulmonary, gastrointestinal, and neurological conditions had similar genetic correlation profiles.
- Published
- 2022
34. Tau interactome maps synaptic and mitochondrial processes associated with neurodegeneration
- Author
-
Tracy, Tara E, Madero-Pérez, Jesus, Swaney, Danielle L, Chang, Timothy S, Moritz, Michelle, Konrad, Csaba, Ward, Michael E, Stevenson, Erica, Hüttenhain, Ruth, Kauwe, Grant, Mercedes, Maria, Sweetland-Martin, Lauren, Chen, Xu, Mok, Sue-Ann, Wong, Man Ying, Telpoukhovskaia, Maria, Min, Sang-Won, Wang, Chao, Sohn, Peter Dongmin, Martin, Jordie, Zhou, Yungui, Luo, Wenjie, Trojanowski, John Q, Lee, Virginia MY, Gong, Shiaoching, Manfredi, Giovanni, Coppola, Giovanni, Krogan, Nevan J, Geschwind, Daniel H, and Gan, Li
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Alzheimer's Disease Related Dementias (ADRD) ,Brain Disorders ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Dementia ,Aging ,Frontotemporal Dementia (FTD) ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research ,Acquired Cognitive Impairment ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Neurodegenerative ,Alzheimer's Disease ,Neurosciences ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Alzheimer Disease ,Amino Acids ,Biotinylation ,Brain ,Cell Nucleus ,Disease Progression ,Energy Metabolism ,Frontotemporal Dementia ,Humans ,Induced Pluripotent Stem Cells ,Mitochondria ,Mutant Proteins ,Mutation ,Nerve Degeneration ,Neurons ,Protein Binding ,Protein Domains ,Protein Interaction Maps ,Proteomics ,Severity of Illness Index ,Subcellular Fractions ,Synapses ,Tauopathies ,tau Proteins ,APEX ,Tau ,Tau secretion ,affinity purification mass spectrometry ,interactome ,mitochondria ,neurodegeneration ,protein-protein interaction ,synapse ,tauopathies ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Tau (MAPT) drives neuronal dysfunction in Alzheimer disease (AD) and other tauopathies. To dissect the underlying mechanisms, we combined an engineered ascorbic acid peroxidase (APEX) approach with quantitative affinity purification mass spectrometry (AP-MS) followed by proximity ligation assay (PLA) to characterize Tau interactomes modified by neuronal activity and mutations that cause frontotemporal dementia (FTD) in human induced pluripotent stem cell (iPSC)-derived neurons. We established interactions of Tau with presynaptic vesicle proteins during activity-dependent Tau secretion and mapped the Tau-binding sites to the cytosolic domains of integral synaptic vesicle proteins. We showed that FTD mutations impair bioenergetics and markedly diminished Tau's interaction with mitochondria proteins, which were downregulated in AD brains of multiple cohorts and correlated with disease severity. These multimodal and dynamic Tau interactomes with exquisite spatial resolution shed light on Tau's role in neuronal function and disease and highlight potential therapeutic targets to block Tau-mediated pathogenesis.
- Published
- 2022
35. Leveraging genomic diversity for discovery in an electronic health record linked biobank: the UCLA ATLAS Community Health Initiative
- Author
-
Johnson, Ruth, Ding, Yi, Venkateswaran, Vidhya, Bhattacharya, Arjun, Boulier, Kristin, Chiu, Alec, Knyazev, Sergey, Schwarz, Tommer, Freund, Malika, Zhan, Lingyu, Burch, Kathryn S, Caggiano, Christa, Hill, Brian, Rakocz, Nadav, Balliu, Brunilda, Denny, Christopher T, Sul, Jae Hoon, Zaitlen, Noah, Arboleda, Valerie A, Halperin, Eran, Sankararaman, Sriram, Butte, Manish J, Lajonchere, Clara, Geschwind, Daniel H, and Pasaniuc, Bogdan
- Subjects
Biological Sciences ,Genetics ,Patient Safety ,Clinical Research ,Human Genome ,Good Health and Well Being ,Asian People ,Biological Specimen Banks ,Electronic Health Records ,Genomics ,Humans ,Public Health ,Electronic health records ,Biobank ,Genetic ancestry ,Genome-wide association studies ,Phenome-wide association studies ,UCLA Precision Health Data Discovery Repository Working Group ,UCLA Precision Health ATLAS Working Group ,Clinical Sciences - Abstract
BackgroundLarge medical centers in urban areas, like Los Angeles, care for a diverse patient population and offer the potential to study the interplay between genetic ancestry and social determinants of health. Here, we explore the implications of genetic ancestry within the University of California, Los Angeles (UCLA) ATLAS Community Health Initiative-an ancestrally diverse biobank of genomic data linked with de-identified electronic health records (EHRs) of UCLA Health patients (N=36,736).MethodsWe quantify the extensive continental and subcontinental genetic diversity within the ATLAS data through principal component analysis, identity-by-descent, and genetic admixture. We assess the relationship between genetically inferred ancestry (GIA) and >1500 EHR-derived phenotypes (phecodes). Finally, we demonstrate the utility of genetic data linked with EHR to perform ancestry-specific and multi-ancestry genome and phenome-wide scans across a broad set of disease phenotypes.ResultsWe identify 5 continental-scale GIA clusters including European American (EA), African American (AA), Hispanic Latino American (HL), South Asian American (SAA) and East Asian American (EAA) individuals and 7 subcontinental GIA clusters within the EAA GIA corresponding to Chinese American, Vietnamese American, and Japanese American individuals. Although we broadly find that self-identified race/ethnicity (SIRE) is highly correlated with GIA, we still observe marked differences between the two, emphasizing that the populations defined by these two criteria are not analogous. We find a total of 259 significant associations between continental GIA and phecodes even after accounting for individuals' SIRE, demonstrating that for some phenotypes, GIA provides information not already captured by SIRE. GWAS identifies significant associations for liver disease in the 22q13.31 locus across the HL and EAA GIA groups (HL p-value=2.32×10-16, EAA p-value=6.73×10-11). A subsequent PheWAS at the top SNP reveals significant associations with neurologic and neoplastic phenotypes specifically within the HL GIA group.ConclusionsOverall, our results explore the interplay between SIRE and GIA within a disease context and underscore the utility of studying the genomes of diverse individuals through biobank-scale genotyping linked with EHR-based phenotyping.
- Published
- 2022
36. P300 promotes tumor recurrence by regulating radiation-induced conversion of glioma stem cells to vascular-like cells
- Author
-
Muthukrishnan, Sree Deepthi, Kawaguchi, Riki, Nair, Pooja, Prasad, Rachna, Qin, Yue, Johnson, Maverick, Wang, Qing, VanderVeer-Harris, Nathan, Pham, Amy, Alvarado, Alvaro G, Condro, Michael C, Gao, Fuying, Gau, Raymond, Castro, Maria G, Lowenstein, Pedro R, Deb, Arjun, Hinman, Jason D, Pajonk, Frank, Burns, Terry C, Goldman, Steven A, Geschwind, Daniel H, and Kornblum, Harley I
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Genetics ,Neurosciences ,Rare Diseases ,Brain Cancer ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Brain Disorders ,Cancer ,Humans ,Neoplasm Recurrence ,Local ,Glioma ,Neoplastic Stem Cells ,Chromatin ,Histone Acetyltransferases - Abstract
Glioma stem cells (GSC) exhibit plasticity in response to environmental and therapeutic stress leading to tumor recurrence, but the underlying mechanisms remain largely unknown. Here, we employ single-cell and whole transcriptomic analyses to uncover that radiation induces a dynamic shift in functional states of glioma cells allowing for acquisition of vascular endothelial-like and pericyte-like cell phenotypes. These vascular-like cells provide trophic support to promote proliferation of tumor cells, and their selective depletion results in reduced tumor growth post-treatment in vivo. Mechanistically, the acquisition of vascular-like phenotype is driven by increased chromatin accessibility and H3K27 acetylation in specific vascular genes allowing for their increased expression post-treatment. Blocking P300 histone acetyltransferase activity reverses the epigenetic changes induced by radiation and inhibits the adaptive conversion of GSC into vascular-like cells and tumor growth. Our findings highlight a role for P300 in radiation-induced stress response, suggesting a therapeutic approach to prevent glioma recurrence.
- Published
- 2022
37. Case Report: Novel CSF1R Variant in a Patient With Behavioral Variant Frontotemporal Dementia Syndrome With Prodromal Repetitive Scratching Behavior
- Author
-
Friedberg, Adit, Ramos, Eliana Marisa, Yang, Zhongan, Bonham, Luke W, Yokoyama, Jennifer S, Ljubenkov, Peter A, Younes, Kyan, Geschwind, Daniel H, and Miller, Bruce L
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Alzheimer's Disease Related Dementias (ADRD) ,Dementia ,Brain Disorders ,Neurodegenerative ,Clinical Research ,Rare Diseases ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Acquired Cognitive Impairment ,Neurosciences ,Behavioral and Social Science ,Aging ,CSF1R-related leukoencephalopathy ,frontotemporal dementia ,repetitive behaviors ,early symptoms ,scratching ,Psychology ,Clinical sciences ,Biological psychology - Abstract
CSF1R-related leukoencephalopathy is an autosomal dominant neurodegenerative disease caused by mutations in the tyrosine kinase domain of the colony stimulating factor 1 receptor (CSF1R). Several studies have found that hematogenic stem cell transplantation is an effective disease modifying therapy however the literature regarding prodromal and early symptoms CSF1R-related leukoencephalopathy is limited. We describe a 63-year-old patient with 4 years of repetitive scratching and skin picking behavior followed by 10 years of progressive behavioral, cognitive, and motor decline in a pattern suggesting behavioral variant of frontotemporal dementia. Brain MRI demonstrated prominent frontal and parietal atrophy accompanied by underlying bilateral patchy white matter hyperintensities sparing the U fibers and cavum septum pellucidum. Whole-exome sequencing revealed a novel, predicted deleterious missense variant in a highly conserved amino acid in the tyrosine kinase domain of CSF1R (p.Gly872Arg). Given this evidence and the characteristic clinical and radiological findings this novel variant was classified as likely pathogenic according to the American College of Medical Genetics standard guidelines. Detailed description of the prodromal scratching and skin picking behavior and possible underlying mechanisms in this case furthers knowledge about early manifestations of CSF1R-related leukoencephalopathy with the hope that early detection and timely administration of disease modifying therapies becomes possible.
- Published
- 2022
38. Transcription factor network analysis identifies REST/NRSF as an intrinsic regulator of CNS regeneration in mice
- Author
-
Cheng, Yuyan, Yin, Yuqin, Zhang, Alice, Bernstein, Alexander M, Kawaguchi, Riki, Gao, Kun, Potter, Kyra, Gilbert, Hui-Ya, Ao, Yan, Ou, Jing, Fricano-Kugler, Catherine J, Goldberg, Jeffrey L, He, Zhigang, Woolf, Clifford J, Sofroniew, Michael V, Benowitz, Larry I, and Geschwind, Daniel H
- Subjects
Biotechnology ,Genetics ,Neurosciences ,Neurodegenerative ,Regenerative Medicine ,Spinal Cord Injury ,Traumatic Head and Spine Injury ,Physical Injury - Accidents and Adverse Effects ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Animals ,Axons ,Mice ,Nerve Regeneration ,Repressor Proteins ,Retinal Ganglion Cells ,Spinal Cord Injuries ,Transcription Factors - Abstract
The inability of neurons to regenerate long axons within the CNS is a major impediment to improving outcome after spinal cord injury, stroke, and other CNS insults. Recent advances have uncovered an intrinsic program that involves coordinate regulation by multiple transcription factors that can be manipulated to enhance growth in the peripheral nervous system. Here, we use a systems genomics approach to characterize regulatory relationships of regeneration-associated transcription factors, identifying RE1-Silencing Transcription Factor (REST; Neuron-Restrictive Silencer Factor, NRSF) as a predicted upstream suppressor of a pro-regenerative gene program associated with axon regeneration in the CNS. We validate our predictions using multiple paradigms, showing that mature mice bearing cell type-specific deletions of REST or expressing dominant-negative mutant REST show improved regeneration of the corticospinal tract and optic nerve after spinal cord injury and optic nerve crush, which is accompanied by upregulation of regeneration-associated genes in cortical motor neurons and retinal ganglion cells, respectively. These analyses identify a role for REST as an upstream suppressor of the intrinsic regenerative program in the CNS and demonstrate the utility of a systems biology approach involving integrative genomics and bio-informatics to prioritize hypotheses relevant to CNS repair.
- Published
- 2022
39. Maternal Immune Activation during Pregnancy Alters Postnatal Brain Growth and Cognitive Development in Nonhuman Primate Offspring
- Author
-
Vlasova, Roza M, Iosif, Ana-Maria, Ryan, Amy M, Funk, Lucy H, Murai, Takeshi, Chen, Shuai, Lesh, Tyler A, Rowland, Douglas J, Bennett, Jeffrey, Hogrefe, Casey E, Maddock, Richard J, Gandal, Michael J, Geschwind, Daniel H, Schumann, Cynthia M, Van de Water, Judy, McAllister, A Kimberley, Carter, Cameron S, Styner, Martin A, Amaral, David G, and Bauman, Melissa D
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Prevention ,Perinatal Period - Conditions Originating in Perinatal Period ,Basic Behavioral and Social Science ,Brain Disorders ,Infectious Diseases ,Mental Illness ,Mental Health ,Pediatric ,Women's Health ,Behavioral and Social Science ,Pregnancy ,Biomedical Imaging ,2.2 Factors relating to the physical environment ,Mental health ,Reproductive health and childbirth ,Neurological ,Animals ,Brain ,Disease Models ,Animal ,Female ,Interferon Inducers ,Macaca mulatta ,Male ,Neurodevelopmental Disorders ,Neurogenesis ,Poly I-C ,Pregnancy Complications ,Infectious ,Prenatal Exposure Delayed Effects ,animal model ,autism ,MRI ,neuroimmunology ,rhesus monkey ,schizophrenia ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.
- Published
- 2021
40. Evolutionary conservation and divergence of the human brain transcriptome
- Author
-
Pembroke, William G, Hartl, Christopher L, and Geschwind, Daniel H
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Brain Disorders ,Neurosciences ,Neurodegenerative ,Biotechnology ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Animals ,Astrocytes ,Brain ,Catechol O-Methyltransferase ,Cerebral Cortex ,Evolution ,Molecular ,Gene Expression Profiling ,Humans ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Mice ,Microfilament Proteins ,Nerve Tissue Proteins ,Neurodegenerative Diseases ,Neurons ,Presenilin-1 ,Primates ,Transcriptome ,alpha-Synuclein ,Evolution ,Genomics ,Co-expression ,Neuroscience ,Disease ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
BackgroundMouse models have allowed for the direct interrogation of genetic effects on molecular, physiological, and behavioral brain phenotypes. However, it is unknown to what extent neurological or psychiatric traits may be human- or primate-specific and therefore which components can be faithfully recapitulated in mouse models.ResultsWe compare conservation of co-expression in 116 independent data sets derived from human, mouse, and non-human primate representing more than 15,000 total samples. We observe greater changes occurring on the human lineage than mouse, and substantial regional variation that highlights cerebral cortex as the most diverged region. Glia, notably microglia, astrocytes, and oligodendrocytes are the most divergent cell type, three times more on average than neurons. We show that cis-regulatory sequence divergence explains a significant fraction of co-expression divergence. Moreover, protein coding sequence constraint parallels co-expression conservation, such that genes with loss of function intolerance are enriched in neuronal, rather than glial modules. We identify dozens of human neuropsychiatric and neurodegenerative disease risk genes, such as COMT, PSEN-1, LRRK2, SHANK3, and SNCA, with highly divergent co-expression between mouse and human and show that 3D human brain organoids recapitulate in vivo co-expression modules representing several human cell types.ConclusionsWe identify robust co-expression modules reflecting whole-brain and regional patterns of gene expression. Compared with those that represent basic metabolic processes, cell-type-specific modules, most prominently glial modules, are the most divergent between species. These data and analyses serve as a foundational resource to guide human disease modeling and its interpretation.
- Published
- 2021
41. Demographic and psychosocial factors associated with the decision to learn mutation status in familial frontotemporal dementia and the impact of disclosure on mood
- Author
-
Bajorek, Lynn P, Kiekhofer, Rachel, Hall, Matthew, Taylor, Joanne, Lucente, Diane E, Brushaber, Danielle, Appleby, Brian, Coppolla, Giovanni, Bordelon, Yvette M, Botha, Hugo, Dickerson, Brad C, Dickson, Dennis W, Domoto‐Reilly, Kimiko, Fagan, Anne M, Fields, Julie A, Fong, Jamie C, Foroud, Tatiana M, Forsberg, Leah K, Galasko, Doug R, Gavrilova, Ralitza H, Geschwind, Daniel H, Ghoshal, Nupur, Goldman, Jill, Graff‐Radford, Neill R, Graff‐Radford, Jonathan, Grant, Ian, Grossman, Murray, Heuer, Hilary W, Hsiung, Ging‐Yuek Robin, Huang, Eric J, Huey, Edward D, Irwin, David J, Jones, David T, Kantarci, Kejal, Kornak, John, Kremers, Walter K, Lapid, Maria I, Leger, Gabriel C, Litvan, Irene, Ljubenkov, Peter A, Mackenzie, Ian R, Masdeu, Joseph C, McMillan, Corey, Mendez, Mario, Miller, Bruce L, Miyagawa, Toji, Onyike, Chiadi U, Pascual, Belen, Pedraza, Otto, Petrucelli, Leonard, Rademakers, Rosa, Ramos, Eliana Marisa, Rankin, Katherine P, Rascovsky, Katya, Rexach, Jessica E, Ritter, Aaron, Roberson, Erik D, Savica, Rodolfo, Rojas, Julio C, Seeley, William W, Tartaglia, Maria Carmela, Toga, Arthur W, Weintraub, Sandra, Wong, Bonnie, Wszolek, Zbigniew, Vandevrede, Lawren, Boeve, Bradley F, Boxer, Adam L, Rosen, Howard J, Staffaroni, Adam M, and Consortium, ALLFTD
- Subjects
Clinical Trials and Supportive Activities ,Aging ,Dementia ,Genetics ,Neurodegenerative ,Clinical Research ,Prevention ,Acquired Cognitive Impairment ,Mental Health ,Depression ,Brain Disorders ,Clinical Sciences ,Neurosciences ,Geriatrics - Abstract
BACKGROUND: Up to 30% of frontotemporal dementia (FTD) cases are due to known pathogenic mutations (f-FTD). Little is known about the factors that predict who will choose to learn their results. Upcoming clinical trials in f-FTD may require disclosure prior to enrollment, even before symptom onset, and thus characterizing this sample is important. Furthermore, understanding the mood impacts of genetic disclosure may guide genetic counseling practice. METHOD: F-FTD participants (n=568) from families with a known pathogenic mutation (MAPT, C9orf72, GRN) were enrolled through the ARTFL/LEFFTDS Longitudinal FTD Study (ALLFTD) and provided the opportunity for disclosure. Independent-sample t-tests compared demographic and psychosocial factors between participants who did and did not receive their results. In participants who were asymptomatic at baseline and follow up (n=199,177 with follow-up), linear mixed effects modeling was used to investigate pre- to post-disclosure changes in the 15-item Geriatric Depression Scale (GDS). RESULT: Of participants from families with a known pathogenic genetic mutation, 47% received genetic disclosure. Of the asymptomatic subset (n=386), 36% know their mutation status. Of these asymptomatic learners, 46% received disclosure through the study, and the remainder learned their genetic status prior to study enrollment. None of the analyzed demographic or psychosocial factors (i.e., sex, age, education, having children) differed between learners and non-learners (p's > 0.05). In the longitudinal analysis of asymptomatic participants, learners showed a pre- to post-increase of 0.31 GDS points/year (95%CI: -0.08, 0.69, p = 0.12), whereas non-learners showed a slight decline (-0.15 points/year, 95%CI: -0.36, 0.06, p = 0.16). This difference between slopes was statistically significant (0.46, 95%CI: 0.02, 0.89, p=0.04) but represents a small clinical effect. In asymptomatic learners, slopes did not differ based on mutation status (0.28, 95%CI: -0.66, 1.20, p=0.55). Conclusions were based on the estimates and full range of confidence intervals. CONCLUSION: The majority of asymptomatic research participants do not know their genetic status, which will be a consideration for clinical trials that require disclosure. No considered demographic factors were strongly associated with the decision to receive disclosure. The findings suggest that disclosure in asymptomatic participants has minimal impact on depressive symptoms regardless of genetic results.
- Published
- 2021
42. Gearing up for the future: Exploring facilitators and barriers to inform clinical trial design in frontotemporal lobar degeneration
- Author
-
Banga, Yasmin B, Lai, Yujung, Kim, Priscilla, Boeve, Bradley F, Boxer, Adam L, Rosen, Howard J, Forsberg, Leah K, Heuer, Hilary W, Brushaber, Danielle, Appleby, Brian, Biernacka, Joanna M, Bordelon, Yvette M, Botha, Hugo, Bozoki, Andrea C, Brannelly, Patrick, Dickerson, Brad C, Dickinson, Susan, Dickson, Dennis W, Domoto‐Reilly, Kimiko, Faber, Kelley, Fagan, Anne M, Fields, Julie A, Fishman, Ann, Foroud, Tatiana M, Galasko, Doug R, Gavrilova, Ralitza H, Gendron, Tania F, Geschwind, Daniel H, Ghoshal, Nupur, Goldman, Jill, Graff‐Radford, Jonathan, Graff‐Radford, Neill R, Grant, Ian, Grossman, Murray, Hsiung, Ging‐Yuek Robin, Huang, Eric J, Huey, Edward D, Irwin, David J, Jones, David T, Kantarci, Kejal, Karydas, Anna M, Kaufer, Daniel, Knopman, David S, Kramer, Joel H, Kremers, Walter K, Kornak, John, Kukull, Walter A, Lagone, Emma, Leger, Gabriel C, Litvan, Irene, Ljubenkov, Peter A, Lucente, Diane E, Mackenzie, Ian R, Manoochehri, Masood, Masdeu, Joseph C, McGinnis, Scott, Mendez, Mario F, Miller, Bruce L, Miyagawa, Toji, Nelson, Kevin M, Onyike, Chiadi U, Pantelyat, Alex, Pascual, Belen, Pearlman, Rodney, Petrucelli, Leonard, Pottier, Cyril P, Rademakers, Rosa, Ramos, Eliana Marisa, Rankin, Katherine P, Rascovsky, Katya, Rexach, Jessica E, Ritter, Aaron, Roberson, Erik D, Rojas, Julio C, Sabbagh, Marwan N, Salmon, David P, Savica, Rodolfo, Seeley, William W, Staffaroni, Adam M, Syrjanen, Jeremy A, Tartaglia, Maria Carmela, Tatton, Nadine, Taylor, Jack C, Toga, Arthur W, Weintraub, Sandra, Wheaton, Diana, Wong, Benjamin, Wszolek, Zbigniew, and Consortium, ALLFTD
- Subjects
Frontotemporal Dementia (FTD) ,Rare Diseases ,Behavioral and Social Science ,Neurodegenerative ,Acquired Cognitive Impairment ,Basic Behavioral and Social Science ,Brain Disorders ,Clinical Research ,Dementia ,Aging ,Biomedical Imaging ,Clinical Trials and Supportive Activities ,Neurological ,Clinical Sciences ,Neurosciences ,Geriatrics - Abstract
BACKGROUND: Frontotemporal lobar degeneration (FTLD) refers to a group of neurodegenerative conditions, affecting the frontal and/or temporal lobes. Ongoing research has provided insight into developing clinical trials for FTLD and key clinical measures such as structural MRI. To inform clinical trial design and optimize participation, it is imperative to explore facilitators and barriers for potential candidates. OBJECTIVE: The objective of this study is to explore facilitators and barriers to participating in future clinical trials for FTLD. METHODS: Advancing Research and Treatment for Frontotemporal Lobar Degeneration (ARTFL) and Longitudinal Evaluation of Familial Frontotemporal Dementia Subjects (LEFFTDS) are observational studies focused on characterizing FTLD syndromes in preparation for clinical trials. The 584 participants enrolled across 18 research sites in the United States and Canada completed a survey assessing interest in clinical trial participation. RESULTS: 29% of respondents self-reported as patients (63±10 years), 26% self-reported as caregivers answering on behalf of patients (65±10 years), and 45% self-reported as healthy but at risk for FTLD (48±14 years). Travel reimbursement was the most common factor reported to positively influence participation (≧66%), with the healthy but at risk group showing the strongest endorsement (83%). Cost and time involved in travel were possible barriers for about half of the patients (48%) and healthy but at risk respondents (53%). The respondents value receiving feedback on the study findings (≧80%) and being informed of their individual disease progression (≧75%). Particularly, keeping participation confidential was very important for the healthy but at risk group (62%). In regard to research assessments, most participants demonstrated a high interest in physical and neurological exams at a research center (≧87%) whereas only half were interested in doing more invasive procedures such as the lumbar puncture (≧52%). Overall, respondents showed a positive attitude and support for research participation (≧77%) and trusted that their health information would remain confidential in a clinical trial (≧53%). CONCLUSIONS: Favorable attitudes and interest towards medical research exist among participants. To optimize participation, clinical trials should allocate funding for travel and involve participants in feedback about study results and their disease progression. Alternatives to invasive assessments may increase participation.
- Published
- 2021
43. Presymptomatic and symptomatic MAPT mutation carriers feature functional connectivity alterations
- Author
-
Zhang, Liwen, Flagan, Taru M, Chu, Stephanie A, Häkkinen, Suvi, Brown, Jesse A, Lee, Alex Jihun, Pasquini, Lorenzo, Mandelli, Maria Luisa, Tempini, Maria Luisa Gorno, Appleby, Brian, Dickerson, Brad C, Domoto‐Reilly, Kimiko, Geschwind, Daniel H, Ghoshal, Nupur, Graff‐Radford, Neill R, Grossman, Murray, Hsiung, Ging‐Yuek Robin, Huey, Edward D, Kantarci, Kejal, Karydas, Anna M, Kaufer, Daniel, Knopman, David S, Litvan, Irene, Mackenzie, Ian R, Mendez, Mario, Onyike, Chiadi U, Ramos, Eliana Marisa, Roberson, Erik D, Trataglia, Maria Carmela, Toga, Arthur W, Weintraub, Sandra, Forsberg, Leah K, Heuer, Hilary W, Boeve, Bradley F, Boxer, Adam L, Rosen, Howard J, Miller, Bruce L, Seeley, William W, Lee, Suzee E, and Consortia, ARTFL LEFFTDS
- Subjects
Brain Disorders ,Clinical Research ,Neurodegenerative ,Biomedical Imaging ,Rare Diseases ,Neurosciences ,Dementia ,Acquired Cognitive Impairment ,Aging ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Neurological ,Clinical Sciences ,Geriatrics - Published
- 2021
44. Defining the nature of human pluripotent stem cell-derived interneurons via single-cell analysis.
- Author
-
Allison, Thomas, Langerman, Justin, Sabri, Shan, Otero-Garcia, Marcos, Lund, Andrew, Huang, John, Wei, Xiaofei, Samarasinghe, Ranmal A, Polioudakis, Damon, Mody, Istvan, Cobos, Inma, Novitch, Bennett G, Geschwind, Daniel H, Plath, Kathrin, and Lowry, William E
- Subjects
human brain interneuron ,neuronal specification ,pluripotent stem cell ,single nuclei transcriptomics ,transcriptional factor programming ,Stem Cell Research ,Neurosciences ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Nonembryonic - Human ,Regenerative Medicine ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Embryonic - Human ,1.1 Normal biological development and functioning ,Neurological ,Biochemistry and Cell Biology ,Clinical Sciences - Abstract
The specification of inhibitory neurons has been described for the mouse and human brain, and many studies have shown that pluripotent stem cells (PSCs) can be used to create interneurons in vitro. It is unclear whether in vitro methods to produce human interneurons generate all the subtypes found in brain, and how similar in vitro and in vivo interneurons are. We applied single-nuclei and single-cell transcriptomics to model interneuron development from human cortex and interneurons derived from PSCs. We provide a direct comparison of various in vitro interneuron derivation methods to determine the homogeneity achieved. We find that PSC-derived interneurons capture stages of development prior to mid-gestation, and represent a minority of potential subtypes found in brain. Comparison with those found in fetal or adult brain highlighted decreased expression of synapse-related genes. These analyses highlight the potential to tailor the method of generation to drive formation of particular subtypes.
- Published
- 2021
45. Postnatal immune activation causes social deficits in a mouse model of tuberous sclerosis: Role of microglia and clinical implications.
- Author
-
López-Aranda, Manuel F, Chattopadhyay, Ishanu, Boxx, Gayle M, Fraley, Elizabeth R, Silva, Tawnie K, Zhou, Miou, Phan, Miranda, Herrera, Isaiah, Taloma, Sunrae, Mandanas, Rochelle, Bach, Karen, Gandal, Michael, Geschwind, Daniel H, Cheng, Genhong, Rzhetsky, Andrey, White, Stephanie A, and Silva, Alcino J
- Abstract
[Figure: see text].
- Published
- 2021
46. Coexpression network architecture reveals the brain-wide and multiregional basis of disease susceptibility
- Author
-
Hartl, Christopher L, Ramaswami, Gokul, Pembroke, William G, Muller, Sandrine, Pintacuda, Greta, Saha, Ashis, Parsana, Princy, Battle, Alexis, Lage, Kasper, and Geschwind, Daniel H
- Subjects
Mental Health ,Brain Disorders ,Neurosciences ,Genetics ,Aetiology ,Underpinning research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Neurological ,Mental health ,Good Health and Well Being ,Brain ,Gene Expression Profiling ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Humans ,Mental Disorders ,Transcriptome ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Gene networks have yielded numerous neurobiological insights, yet an integrated view across brain regions is lacking. We leverage RNA sequencing in 864 samples representing 12 brain regions to robustly identify 12 brain-wide, 50 cross-regional and 114 region-specific coexpression modules. Nearly 40% of genes fall into brain-wide modules, while 25% comprise region-specific modules reflecting regional biology, such as oxytocin signaling in the hypothalamus, or addiction pathways in the nucleus accumbens. Schizophrenia and autism genetic risk are enriched in brain-wide and multiregional modules, indicative of broad impact; these modules implicate neuronal proliferation and activity-dependent processes, including endocytosis and splicing, in disease pathophysiology. We find that cell-type-specific long noncoding RNA and gene isoforms contribute substantially to regional synaptic diversity and that constrained, mutation-intolerant genes are primarily enriched in neurons. We leverage these data using an omnigenic-inspired network framework to characterize how coexpression and gene regulatory networks reflect neuropsychiatric disease risk, supporting polygenic models.
- Published
- 2021
47. Neurite-based white matter alterations in MAPT mutation carriers: A multi-shell diffusion MRI study in the ALLFTD consortium
- Author
-
Corriveau-Lecavalier, Nick, Tosakulwong, Nirubol, Lesnick, Timothy G., Fought, Angela J., Reid, Robert I., Schwarz, Christopher G., Senjem, Matthew L., Jack, Clifford R., Jr., Jones, David T., Vemuri, Prashanthi, Rademakers, Rosa, Ramos, Eliana Marisa, Geschwind, Daniel H., Knopman, David S., Botha, Hugo, Savica, Rodolfo, Graff-Radford, Jonathan, Ramanan, Vijay K., Fields, Julie A., Graff-Radford, Neill, Wszolek, Zbigniew, Forsberg, Leah K., Petersen, Ronald C., Heuer, Hilary W., Boxer, Adam L., Rosen, Howard J., Boeve, Bradley F., and Kantarci, Kejal
- Published
- 2024
- Full Text
- View/download PDF
48. Abnormal sleep physiology in children with 15q11.2-13.1 duplication (Dup15q) syndrome.
- Author
-
Saravanapandian, Vidya, Nadkarni, Divya, Hsu, Sheng-Hsiou, Hussain, Shaun A, Maski, Kiran, Golshani, Peyman, Colwell, Christopher S, Balasubramanian, Saravanavel, Dixon, Amos, Geschwind, Daniel H, and Jeste, Shafali S
- Subjects
Humans ,Seizures ,Electroencephalography ,Sleep ,Child ,Intellectual Disability ,Autism Spectrum Disorder ,Autism ,Biomarkers ,Dup15q syndrome ,EEG ,GABAAR ,Slow wave sleep ,Spindles ,UBE3A ,Neurosciences ,Intellectual and Developmental Disabilities (IDD) ,Genetics ,Epilepsy ,Neurodegenerative ,Mental Health ,Sleep Research ,Pediatric ,Brain Disorders ,Behavioral and Social Science ,Clinical Research ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Mental health ,GABA(A)R ,Clinical Sciences - Abstract
BackgroundSleep disturbances in autism spectrum disorder (ASD) represent a common and vexing comorbidity. Clinical heterogeneity amongst these warrants studies of the mechanisms associated with specific genetic etiologies. Duplications of 15q11.2-13.1 (Dup15q syndrome) are highly penetrant for neurodevelopmental disorders (NDDs) such as intellectual disability and ASD, as well as sleep disturbances. Genes in the 15q region, particularly UBE3A and a cluster of GABAA receptor genes, are critical for neural development, synaptic protein synthesis and degradation, and inhibitory neurotransmission. During awake electroencephalography (EEG), children with Dup15q syndrome demonstrate increased beta band oscillations (12-30 Hz) that likely reflect aberrant GABAergic neurotransmission. Healthy sleep rhythms, necessary for robust cognitive development, are also highly dependent on GABAergic neurotransmission. We therefore hypothesized that sleep physiology would be abnormal in children with Dup15q syndrome.MethodsTo test the hypothesis that elevated beta oscillations persist in sleep in Dup15q syndrome and that NREM sleep rhythms would be disrupted, we computed: (1) beta power, (2) spindle density, and (3) percentage of slow-wave sleep (SWS) in overnight sleep EEG recordings from a cohort of children with Dup15q syndrome (n = 15) and compared them to age-matched neurotypical children (n = 12).ResultsChildren with Dup15q syndrome showed abnormal sleep physiology with elevated beta power, reduced spindle density, and reduced or absent SWS compared to age-matched neurotypical controls.LimitationsThis study relied on clinical EEG where sleep staging was not available. However, considering that clinical polysomnograms are challenging to collect in this population, the ability to quantify these biomarkers on clinical EEG-routinely ordered for epilepsy monitoring-opens the door for larger-scale studies. While comparable to other human studies in rare genetic disorders, a larger sample would allow for examination of the role of seizure severity, medications, and developmental age that may impact sleep physiology.ConclusionsWe have identified three quantitative EEG biomarkers of sleep disruption in Dup15q syndrome, a genetic condition highly penetrant for ASD. Insights from this study not only promote a greater mechanistic understanding of the pathophysiology defining Dup15q syndrome, but also lay the foundation for studies that investigate the association between sleep and cognition. Abnormal sleep physiology may undermine healthy cognitive development and may serve as a quantifiable and modifiable target for behavioral and pharmacological interventions.
- Published
- 2021
49. A neurogenetic analysis of female autism
- Author
-
Jack, Allison, Sullivan, Catherine AW, Aylward, Elizabeth, Bookheimer, Susan Y, Dapretto, Mirella, Gaab, Nadine, Van Horn, John D, Eilbott, Jeffrey, Jacokes, Zachary, Torgerson, Carinna M, Bernier, Raphael A, Geschwind, Daniel H, McPartland, James C, Nelson, Charles A, Webb, Sara J, Pelphrey, Kevin A, Gupta, Abha R, Ventola, Pamela, Kresse, Anna, Neuhaus, Emily, Corrigan, Sarah, Wolf, Julie, McDonald, Nicole, Ankenman, Katy, Jeste, Shafali, Naples, Adam, Libsack, Erin, Guilford, Desiree, Torgerson, Carinna, Welker, Olivia, Lowe, Jennifer K, MacDonnell, Erin, Tsapelas, Heidi, Depedro-Mercier, Dianna, and Keifer, Cara M
- Subjects
Pediatric ,Intellectual and Developmental Disabilities (IDD) ,Neurosciences ,Genetics ,Clinical Research ,Brain Disorders ,Autism ,Human Genome ,Mental Health ,Mental health ,Adolescent ,Autism Spectrum Disorder ,Child ,Corpus Striatum ,DNA Copy Number Variations ,Female ,Genotype ,Humans ,Magnetic Resonance Imaging ,Male ,Neuroimaging ,Sex Characteristics ,autism spectrum disorder ,functional MRI ,genetics ,striatum ,social perception ,GENDAAR Consortium ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Females versus males are less frequently diagnosed with autism spectrum disorder (ASD), and while understanding sex differences is critical to delineating the systems biology of the condition, female ASD is understudied. We integrated functional MRI and genetic data in a sex-balanced sample of ASD and typically developing youth (8-17 years old) to characterize female-specific pathways of ASD risk. Our primary objectives were to: (i) characterize female ASD (n = 45) brain response to human motion, relative to matched typically developing female youth (n = 45); and (ii) evaluate whether genetic data could provide further insight into the potential relevance of these brain functional differences. For our first objective we found that ASD females showed markedly reduced response versus typically developing females, particularly in sensorimotor, striatal, and frontal regions. This difference between ASD and typically developing females does not resemble differences between ASD (n = 47) and typically developing males (n = 47), even though neural response did not significantly differ between female and male ASD. For our second objective, we found that ASD females (n = 61), versus males (n = 66), showed larger median size of rare copy number variants containing gene(s) expressed in early life (10 postconceptual weeks to 2 years) in regions implicated by the typically developing female > female functional MRI contrast. Post hoc analyses suggested this difference was primarily driven by copy number variants containing gene(s) expressed in striatum. This striatal finding was reproducible among n = 2075 probands (291 female) from an independent cohort. Together, our findings suggest that striatal impacts may contribute to pathways of risk in female ASD and advocate caution in drawing conclusions regarding female ASD based on male-predominant cohorts.
- Published
- 2021
50. C9orf72 deficiency promotes microglial-mediated synaptic loss in aging and amyloid accumulation
- Author
-
Lall, Deepti, Lorenzini, Ileana, Mota, Thomas A, Bell, Shaughn, Mahan, Thomas E, Ulrich, Jason D, Davtyan, Hayk, Rexach, Jessica E, Muhammad, AKM Ghulam, Shelest, Oksana, Landeros, Jesse, Vazquez, Michael, Kim, Junwon, Ghaffari, Layla, O'Rourke, Jacqueline Gire, Geschwind, Daniel H, Blurton-Jones, Mathew, Holtzman, David M, Sattler, Rita, and Baloh, Robert H
- Subjects
Genetics ,Aging ,Alzheimer's Disease ,Frontotemporal Dementia (FTD) ,Alzheimer's Disease Related Dementias (ADRD) ,Brain Disorders ,Neurodegenerative ,Rare Diseases ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Dementia ,ALS ,Neurosciences ,Acquired Cognitive Impairment ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Amyloid ,Animals ,C9orf72 Protein ,DNA Repeat Expansion ,Disease Models ,Animal ,Lysosomes ,Mice ,Mice ,Knockout ,Microglia ,Synapses ,Alzheimer’s disease ,C9orf72 ,amyotrophic lateral sclerosis ,frontotemporal dementia ,microglia ,neurodegeneration ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
C9orf72 repeat expansions cause inherited amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD) and result in both loss of C9orf72 protein expression and production of potentially toxic RNA and dipeptide repeat proteins. In addition to ALS/FTD, C9orf72 repeat expansions have been reported in a broad array of neurodegenerative syndromes, including Alzheimer's disease. Here we show that C9orf72 deficiency promotes a change in the homeostatic signature in microglia and a transition to an inflammatory state characterized by an enhanced type I IFN signature. Furthermore, C9orf72-depleted microglia trigger age-dependent neuronal defects, in particular enhanced cortical synaptic pruning, leading to altered learning and memory behaviors in mice. Interestingly, C9orf72-deficient microglia promote enhanced synapse loss and neuronal deficits in a mouse model of amyloid accumulation while paradoxically improving plaque clearance. These findings suggest that altered microglial function due to decreased C9orf72 expression directly contributes to neurodegeneration in repeat expansion carriers independent of gain-of-function toxicities.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.