1
|
Ganuza M, Morales-Hernández A, Van Huizen A, Chabot A, Hall T, Caprio C, Finkelstein D, Kilimann MW, McKinney-Freeman S. Neurobeachin regulates hematopoietic progenitor differentiation and survival by modulating Notch activity. Blood Adv 2024; 8:4129-4143. [PMID: 38905595 PMCID: PMC11345395 DOI: 10.1182/bloodadvances.2023012426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024] Open
Abstract
ABSTRACT Hematopoietic stem cells (HSCs) can generate all blood cells. This ability is exploited in HSC transplantation (HSCT) to treat hematologic disease. A clear understanding of the molecular mechanisms that regulate HSCT is necessary to continue improving transplant protocols. We identified the Beige and Chediak-Higashi domain-containing protein (BDCP), Neurobeachin (NBEA), as a putative regulator of HSCT. Here, we demonstrated that NBEA and related BDCPs, including LPS Responsive Beige-Like Anchor Protein (LRBA), Neurobeachin Like 1 (NBEAL1) and Lysosomal Trafficking Regulator (LYST), are required during HSCT to efficiently reconstitute the hematopoietic system of lethally irradiated mice. Nbea knockdown in mouse HSCs induced apoptosis and a differentiation block after transplantation. Nbea deficiency in hematopoietic progenitor cells perturbed the expression of genes implicated in vesicle trafficking and led to changes in NOTCH receptor localization. This resulted in perturbation of the NOTCH transcriptional program, which is required for efficient HSC engraftment. In summary, our findings reveal a novel role for NBEA in the control of NOTCH receptor turnover in hematopoietic cells and supports a model in which BDCP-regulated vesicle trafficking is required for efficient HSCT.
Collapse
Affiliation(s)
- Miguel Ganuza
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Antonio Morales-Hernández
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI
| | - Alanna Van Huizen
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ashley Chabot
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Trent Hall
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Claire Caprio
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Manfred W. Kilimann
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | |
Collapse
|
2
|
Pretzsch CM, Ecker C. Structural neuroimaging phenotypes and associated molecular and genomic underpinnings in autism: a review. Front Neurosci 2023; 17:1172779. [PMID: 37457001 PMCID: PMC10347684 DOI: 10.3389/fnins.2023.1172779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Autism has been associated with differences in the developmental trajectories of multiple neuroanatomical features, including cortical thickness, surface area, cortical volume, measures of gyrification, and the gray-white matter tissue contrast. These neuroimaging features have been proposed as intermediate phenotypes on the gradient from genomic variation to behavioral symptoms. Hence, examining what these proxy markers represent, i.e., disentangling their associated molecular and genomic underpinnings, could provide crucial insights into the etiology and pathophysiology of autism. In line with this, an increasing number of studies are exploring the association between neuroanatomical, cellular/molecular, and (epi)genetic variation in autism, both indirectly and directly in vivo and across age. In this review, we aim to summarize the existing literature in autism (and neurotypicals) to chart a putative pathway from (i) imaging-derived neuroanatomical cortical phenotypes to (ii) underlying (neuropathological) biological processes, and (iii) associated genomic variation.
Collapse
Affiliation(s)
- Charlotte M. Pretzsch
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Christine Ecker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|
3
|
Liu Y, Wu Z, Feng Y, Gao J, Wang B, Lian C, Diao B. Integration analysis of single-cell and spatial transcriptomics reveal the cellular heterogeneity landscape in glioblastoma and establish a polygenic risk model. Front Oncol 2023; 13:1109037. [PMID: 37397378 PMCID: PMC10308022 DOI: 10.3389/fonc.2023.1109037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Background Glioblastoma (GBM) is adults' most common and fatally malignant brain tumor. The heterogeneity is the leading cause of treatment failure. However, the relationship between cellular heterogeneity, tumor microenvironment, and GBM progression is still elusive. Methods Integrated analysis of single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing (stRNA-seq) of GBM were conducted to analyze the spatial tumor microenvironment. We investigated the subpopulation heterogeneity of malignant cells through gene set enrichment analyses, cell communications analyses, and pseudotime analyses. Significantly changed genes of the pseudotime analysis were screened to create a tumor progress-related gene risk score (TPRGRS) using Cox regression algorithms in the bulkRNA-sequencing(bulkRNA-seq) dataset. We combined the TPRGRS and clinical characteristics to predict the prognosis of patients with GBM. Furthermore, functional analysis was applied to uncover the underlying mechanisms of the TPRGRS. Results GBM cells were accurately charted to their spatial locations and uncovered their spatial colocalization. The malignant cells were divided into five clusters with transcriptional and functional heterogeneity, including unclassified malignant cells and astrocyte-like, mesenchymal-like, oligodendrocytes-progenitor-like, and neural-progenitor-like malignant cells. Cell-cell communications analysis in scRNA-seq and stRNA-seq identified ligand-receptor pairs of the CXCL, EGF, FGF, and MIF signaling pathways as bridges implying that tumor microenvironment may cause malignant cells' transcriptomic adaptability and disease progression. Pseudotime analysis showed the differentiation trajectory of GBM cells from proneural to mesenchymal transition and identified genes or pathways that affect cell differentiation. TPRGRS could successfully divide patients with GBM in three datasets into high- and low-risk groups, which was proved to be a prognostic factor independent of routine clinicopathological characteristics. Functional analysis revealed the TPRGRS associated with growth factor binding, cytokine activity, signaling receptor activator activity functions, and oncogenic pathways. Further analysis revealed the association of the TPRGRS with gene mutations and immunity in GBM. Finally, the external datasets and qRT-PCR verified high expressions of the TPRGRS mRNAs in GBM cells. Conclusion Our study provides novel insights into heterogeneity in GBM based on scRNA-seq and stRNA-seq data. Moreover, our study proposed a malignant cell transition-based TPRGRS through integrated analysis of bulkRNA-seq and scRNA-seq data, combined with the routine clinicopathological evaluation of tumors, which may provide more personalized drug regimens for GBM patients.
Collapse
Affiliation(s)
- Yaxuan Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- Department of Basic Medicine, General Hospital of Central Theatre Command, Wuhan, Hubei, China
| | - Zhenyu Wu
- Department of Urology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Yueyuan Feng
- Cancer Hospital, The First People's Hospital of Foshan, Foshan, Foshan, Guangdong, China
| | - Jiawei Gao
- College of Medicine, JiShou University, Xiangxi, Hunan, China
| | - Bo Wang
- College of Medicine, JiShou University, Xiangxi, Hunan, China
| | - Changlin Lian
- Cancer Hospital, The First People's Hospital of Foshan, Foshan, Foshan, Guangdong, China
| | - Bo Diao
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- Department of Basic Medicine, General Hospital of Central Theatre Command, Wuhan, Hubei, China
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Command and Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei, China
| |
Collapse
|
4
|
Zhao Q, Liu R, Chen H, Yang X, Dong J, Bai M, Lu Y, Leng Y. Transcriptome-wide association study reveals novel susceptibility genes for coronary atherosclerosis. Front Cardiovasc Med 2023; 10:1149113. [PMID: 37351287 PMCID: PMC10282549 DOI: 10.3389/fcvm.2023.1149113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/23/2023] [Indexed: 06/24/2023] Open
Abstract
Background Genetic risk factors substantially contributed to the development of coronary atherosclerosis. Genome-wide association study (GWAS) has identified many risk loci for coronary atherosclerosis, but the translation of these loci into therapeutic targets is limited for their location in non-coding regions. Here, we aimed to screen the potential coronary atherosclerosis pathogenic genes expressed though TWAS (transcriptome wide association study) and explore the underlying mechanism association. Methods Four TWAS approaches (PrediXcan, JTI, UTMOST, and FUSION) were used to screen genes associated with coronary atherosclerosis. Enrichment analysis of TWAS-identified genes was applied through the Metascape website. The summary-data-based Mendelian randomization (SMR) analysis was conducted to provide the evidence of causal relationship between the candidate genes and coronary atherosclerosis. At last, the cell type-specific expression of the intersection genes was examined by using human coronary artery single-cell RNA-seq, interrogating the immune microenvironment of human coronary atherosclerotic plaque at different stages of maturity. Results We identified 19 genes by at least three approaches and 1 gene (NBEAL1) by four approaches. Enrichment analysis enriching the genes identified at least by two TWAS approaches, suggesting that these genes were markedly enriched in asthma and leukocyte mediated immunity reaction. Further, the summary-data-based Mendelian randomization (SMR) analysis provided the evidence of causal relationship between NBEAL1 gene and coronary atherosclerosis, confirming the protecting effects of NBEAL1 gene and coronary atherosclerosis. At last, the single cell cluster analysis demonstrated that NBEAL1 gene has differential expressions in macrophages, plasma cells and endothelial cells. Conclusion Our study identified the novel genes associated with coronary atherosclerosis and suggested the potential biological function for these genes, providing insightful guidance for further biological investigation and therapeutic approaches development in atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Qiuping Zhao
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Rongmei Liu
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Hui Chen
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Xiaomo Yang
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Jiajia Dong
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Minfu Bai
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Yao Lu
- School of Life Course Sciences, King’s College London, London, United Kingdom
| | - Yiming Leng
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Bhagat R, Marini S, Romero JR. Genetic considerations in cerebral small vessel diseases. Front Neurol 2023; 14:1080168. [PMID: 37168667 PMCID: PMC10164974 DOI: 10.3389/fneur.2023.1080168] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/04/2023] [Indexed: 05/13/2023] Open
Abstract
Cerebral small vessel disease (CSVD) encompasses a broad clinical spectrum united by pathology of the small vessels of the brain. CSVD is commonly identified using brain magnetic resonance imaging with well characterized markers including covert infarcts, white matter hyperintensities, enlarged perivascular spaces, and cerebral microbleeds. The pathophysiology of CSVD is complex involving genetic determinants, environmental factors, and their interactions. While the role of vascular risk factors in CSVD is well known and its management is pivotal in mitigating the clinical effects, recent research has identified novel genetic factors involved in CSVD. Delineating genetic determinants can promote the understanding of the disease and suggest effective treatments and preventive measures of CSVD at the individual level. Here we review CSVD focusing on recent advances in the genetics of CSVD. The knowledge gained has advanced understanding of the pathophysiology of CSVD, offered promising early results that may improve subtype identification of small vessel strokes, has led to additional identification of mendelian forms of small vessel strokes, and is getting closer to influencing clinical care through pharmacogenetic studies.
Collapse
Affiliation(s)
- Riwaj Bhagat
- Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - Sandro Marini
- Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - José R. Romero
- Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
- NHLBI’s Framingham Heart Study, Framingham, MA, United States
- *Correspondence: José R. Romero,
| |
Collapse
|
6
|
Pluthero FG, Kahr WHA. Gray platelet syndrome: NBEAL2 mutations are associated with pathology beyond megakaryocyte and platelet function defects. J Thromb Haemost 2021; 19:318-322. [PMID: 33300270 DOI: 10.1111/jth.15177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 01/13/2023]
Affiliation(s)
- Fred G Pluthero
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Walter H A Kahr
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Division of Haematology/Oncology, Department of Paediatrics, University of Toronto and the Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Bindesbøll C, Aas A, Ogmundsdottir MH, Pankiv S, Reine T, Zoncu R, Simonsen A. NBEAL1 controls SREBP2 processing and cholesterol metabolism and is a susceptibility locus for coronary artery disease. Sci Rep 2020; 10:4528. [PMID: 32161285 PMCID: PMC7066131 DOI: 10.1038/s41598-020-61352-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/21/2020] [Indexed: 01/24/2023] Open
Abstract
Dysregulated cholesterol homeostasis promotes the pathology of atherosclerosis, myocardial infarction and strokes. Cellular cholesterol is mainly regulated at the transcriptional level by SREBP2, but also through uptake of extracellular cholesterol from low density lipoproteins (LDL) via expression of LDL receptors (LDLR) at the cell surface. Identification of the mechanisms involved in regulation of these processes are thus key to understand the pathology of coronary artery disease. Here, we identify the large and poorly characterized BEACH domain protein Neurobeachin-like (NBEAL) 1 as a Golgi- associated protein required for regulation of cholesterol metabolism. NBEAL1 is most abundantly expressed in arteries. Genetic variants in NBEAL1 are associated with decreased expression of NBEAL1 in arteries and increased risk of coronary artery disease in humans. We show that NBEAL1 regulates cholesterol metabolism by modulating LDLR expression in a mechanism involving interaction with SCAP and PAQR3 and subsequent SREBP2-processing. Thus, low expression of NBEAL1 may lead to increased risk of coronary artery disease by downregulation of LDLR levels.
Collapse
Affiliation(s)
- Christian Bindesbøll
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.
| | - Aleksander Aas
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Margret Helga Ogmundsdottir
- Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101, Reykjavik, Iceland
| | - Serhiy Pankiv
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Trine Reine
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.,Section for Interphase genetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.
| |
Collapse
|
8
|
Glentis S, Dimopoulos AC, Rouskas K, Ntritsos G, Evangelou E, Narod SA, Mes-Masson AM, Foulkes WD, Rivera B, Tonin PN, Ragoussis J, Dimas AS. Exome Sequencing in BRCA1- and BRCA2-Negative Greek Families Identifies MDM1 and NBEAL1 as Candidate Risk Genes for Hereditary Breast Cancer. Front Genet 2019; 10:1005. [PMID: 31681433 PMCID: PMC6813924 DOI: 10.3389/fgene.2019.01005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/20/2019] [Indexed: 12/30/2022] Open
Abstract
Approximately 10% of breast cancer (BC) cases are hereditary BC (HBC), with HBC most commonly encountered in the context of hereditary breast and ovarian cancer (HBOC) syndrome. Although thousands of loss-of-function (LoF) alleles in over 20 genes have been associated with HBC susceptibility, the genetic etiology of approximately 50% of cases remains unexplained, even when polygenic risk models are considered. We focused on one of the least-studied European populations and applied whole-exome sequencing (WES) to 52 individuals from 17 Greek HBOC families, in which at least one patient was negative for known HBC risk variants. Initial screening revealed pathogenic variants in known cancer genes, including BARD1:p.Trp91* detected in a cancer-free individual, and MEN1:p.Glu260Lys detected in a BC patient. Gene- and variant-based approaches were applied to exome data to identify candidate risk variants outside of known risk genes. Findings were verified in a collection of Canadian HBOC patients of European ancestry (FBRCAX), in an independent group of Canadian BC patients (CHUM-BC) and controls (CARTaGENE), as well as in individuals from The Cancer Genome Atlas (TCGA) and the UK Biobank (UKB). Rare LoF variants were uncovered in MDM1 and NBEAL1 in Greek and Canadian HBOC patients. We also report prioritized missense variants SETBP1:c.4129G > C and C7orf34:c.248C > T. These variants comprise promising candidates whose role in cancer pathogenicity needs to be explored further.
Collapse
Affiliation(s)
- Stavros Glentis
- Division of Molecular Biology and Genetics, Biomedical Sciences Research Center Al. Fleming, Vari, Greece
| | - Alexandros C Dimopoulos
- Division of Molecular Biology and Genetics, Biomedical Sciences Research Center Al. Fleming, Vari, Greece
| | - Konstantinos Rouskas
- Division of Molecular Biology and Genetics, Biomedical Sciences Research Center Al. Fleming, Vari, Greece
| | - George Ntritsos
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece.,Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Steven A Narod
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, QC, Canada
| | - William D Foulkes
- Department of Oncology, McGill University, Montreal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Department of Medical Genetics, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Barbara Rivera
- Department of Oncology, McGill University, Montreal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Patricia N Tonin
- Department of Medicine, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada.,Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jiannis Ragoussis
- Department of Oncology, McGill University, Montreal, QC, Canada.,McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Antigone S Dimas
- Division of Molecular Biology and Genetics, Biomedical Sciences Research Center Al. Fleming, Vari, Greece
| |
Collapse
|
9
|
Jian X, Satizabal CL, Smith AV, Wittfeld K, Bis JC, Smith JA, Hsu FC, Nho K, Hofer E, Hagenaars SP, Nyquist PA, Mishra A, Adams HHH, Li S, Teumer A, Zhao W, Freedman BI, Saba Y, Yanek LR, Chauhan G, van Buchem MA, Cushman M, Royle NA, Bryan RN, Niessen WJ, Windham BG, DeStefano AL, Habes M, Heckbert SR, Palmer ND, Lewis CE, Eiriksdottir G, Maillard P, Mathias RA, Homuth G, Valdés-Hernández MDC, Divers J, Beiser AS, Langner S, Rice KM, Bastin ME, Yang Q, Maldjian JA, Starr JM, Sidney S, Risacher SL, Uitterlinden AG, Gudnason VG, Nauck M, Rotter JI, Schreiner PJ, Boerwinkle E, van Duijn CM, Mazoyer B, von Sarnowski B, Gottesman RF, Levy D, Sigurdsson S, Vernooij MW, Turner ST, Schmidt R, Wardlaw JM, Psaty BM, Mosley TH, DeCarli CS, Saykin AJ, Bowden DW, Becker DM, Deary IJ, Schmidt H, Kardia SLR, Ikram MA, Debette S, Grabe HJ, Longstreth WT, Seshadri S, Launer LJ, Fornage M. Exome Chip Analysis Identifies Low-Frequency and Rare Variants in MRPL38 for White Matter Hyperintensities on Brain Magnetic Resonance Imaging. Stroke 2019; 49:1812-1819. [PMID: 30002152 DOI: 10.1161/strokeaha.118.020689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background and Purpose- White matter hyperintensities (WMH) on brain magnetic resonance imaging are typical signs of cerebral small vessel disease and may indicate various preclinical, age-related neurological disorders, such as stroke. Though WMH are highly heritable, known common variants explain a small proportion of the WMH variance. The contribution of low-frequency/rare coding variants to WMH burden has not been explored. Methods- In the discovery sample we recruited 20 719 stroke/dementia-free adults from 13 population-based cohort studies within the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium, among which 17 790 were of European ancestry and 2929 of African ancestry. We genotyped these participants at ≈250 000 mostly exonic variants with Illumina HumanExome BeadChip arrays. We performed ethnicity-specific linear regression on rank-normalized WMH in each study separately, which were then combined in meta-analyses to test for association with single variants and genes aggregating the effects of putatively functional low-frequency/rare variants. We then sought replication of the top findings in 1192 adults (European ancestry) with whole exome/genome sequencing data from 2 independent studies. Results- At 17q25, we confirmed the association of multiple common variants in TRIM65, FBF1, and ACOX1 ( P<6×10-7). We also identified a novel association with 2 low-frequency nonsynonymous variants in MRPL38 (lead, rs34136221; PEA=4.5×10-8) partially independent of known common signal ( PEA(conditional)=1.4×10-3). We further identified a locus at 2q33 containing common variants in NBEAL1, CARF, and WDR12 (lead, rs2351524; Pall=1.9×10-10). Although our novel findings were not replicated because of limited power and possible differences in study design, meta-analysis of the discovery and replication samples yielded stronger association for the 2 low-frequency MRPL38 variants ( Prs34136221=2.8×10-8). Conclusions- Both common and low-frequency/rare functional variants influence WMH. Larger replication and experimental follow-up are essential to confirm our findings and uncover the biological causal mechanisms of age-related WMH.
Collapse
Affiliation(s)
- Xueqiu Jian
- From the Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (M.F., X.J.)
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, MA (C.L.S., S. Seshadri)
| | - Albert V Smith
- Icelandic Heart Association, Kópavogur, Iceland (A.V.S., G.E., S. Sigurdsson, V.G.G.)
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases, Site Rostock/Greifswald, Germany (K.W.)
| | - Joshua C Bis
- Cardiovascular Health Research Unit (B.M.P., J.C.B., S.R.H.)
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor (J.A.S., S.L.R.K., W.Z.)
| | - Fang-Chi Hsu
- Division of Public Health Sciences (F.-C.H., J.D.)
| | - Kwangsik Nho
- Center for Neuroimaging, Indiana University School of Medicine, Indianapolis (K.N., S.L.R.)
| | | | - Saskia P Hagenaars
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | - Paul A Nyquist
- Department of Neurology and Neurosurgery (P.A.N., R.F.G.)
| | - Aniket Mishra
- Bordeaux Population Health Research Centre U1219, Inserm, France (A.M., G.C., S.D.)
| | | | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, MA (A.S.B., A.L.D., Q.Y., S.L.)
| | | | - Wei Zhao
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor (J.A.S., S.L.R.K., W.Z.)
| | | | - Yasaman Saba
- Institute of Molecular Biology and Biochemistry (H.S., Y.S.), Medical University of Graz, Austria
| | - Lisa R Yanek
- Department of Medicine (D.M.B., L.R.Y., R.A.M.), Johns Hopkins School of Medicine, Baltimore, MD
| | - Ganesh Chauhan
- Bordeaux Population Health Research Centre U1219, Inserm, France (A.M., G.C., S.D.)
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, the Netherlands (M.A.v.B.)
| | - Mary Cushman
- Department of Medicine, The University of Vermont Larner College of Medicine, Burlington (M.C.)
| | - Natalie A Royle
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | - R Nick Bryan
- Department of Diagnostic Medicine, Dell Medical School at The University of Texas at Austin (R.N.B.)
| | - Wiro J Niessen
- Departments of Radiology and Medical Informatics (W.J.N.).,Department of Medicine, The University of Mississippi School of Medicine, Jackson (W.J.N.)
| | | | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, MA (A.S.B., A.L.D., Q.Y., S.L.)
| | - Mohamad Habes
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia (M.H.)
| | | | - Nicholette D Palmer
- Department of Biochemistry (D.W.B., N.D.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Cora E Lewis
- Department of Epidemiology, The University of Alabama at Birmingham School of Public Health (C.E.L.)
| | - Gudny Eiriksdottir
- Icelandic Heart Association, Kópavogur, Iceland (A.V.S., G.E., S. Sigurdsson, V.G.G.)
| | - Pauline Maillard
- Department of Neurology, UC Davis School of Medicine (C.S.D., P.M.), CA
| | - Rasika A Mathias
- Department of Medicine (D.M.B., L.R.Y., R.A.M.), Johns Hopkins School of Medicine, Baltimore, MD
| | - Georg Homuth
- Institute of Genetics and Functional Genomics, University of Greifswald, Germany (G.H.)
| | - Maria Del C Valdés-Hernández
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | | | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, MA (A.S.B., A.L.D., Q.Y., S.L.)
| | - Sönke Langner
- Institute for Diagnostic Radiology and Neuroradiology (S.L.)
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington School of Public Health, Seattle (K.M.R.)
| | - Mark E Bastin
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, MA (A.S.B., A.L.D., Q.Y., S.L.)
| | - Joseph A Maldjian
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas (J.A.M.)
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | - Stephen Sidney
- Division of Research, Kaiser Permanente Northern California, Oakland (S. Sidney)
| | - Shannon L Risacher
- Center for Neuroimaging, Indiana University School of Medicine, Indianapolis (K.N., S.L.R.)
| | | | - Vilmundur G Gudnason
- Icelandic Heart Association, Kópavogur, Iceland (A.V.S., G.E., S. Sigurdsson, V.G.G.)
| | - Matthias Nauck
- Institute for Clinical Chemistry and Laboratory Medicine (M.N.)
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, CA (J.I.R.)
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis (P.J.S.)
| | - Eric Boerwinkle
- Human Genetics Center, The University of Texas Health Science Center at Houston School of Public Health (E.B.)
| | | | - Bernard Mazoyer
- Neurodegeneratives Diseases Institute-CNRS UMR 5293 (B.M.), University of Bordeaux, France
| | | | | | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD (D.L.)
| | - Sigurdur Sigurdsson
- Icelandic Heart Association, Kópavogur, Iceland (A.V.S., G.E., S. Sigurdsson, V.G.G.)
| | | | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (S.T.T.)
| | | | - Joanna M Wardlaw
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | - Bruce M Psaty
- Cardiovascular Health Research Unit (B.M.P., J.C.B., S.R.H.)
| | | | - Charles S DeCarli
- Department of Neurology, UC Davis School of Medicine (C.S.D., P.M.), CA
| | | | - Donald W Bowden
- Department of Biochemistry (D.W.B., N.D.P.), Wake Forest School of Medicine, Winston-Salem, NC
| | - Diane M Becker
- Department of Medicine (D.M.B., L.R.Y., R.A.M.), Johns Hopkins School of Medicine, Baltimore, MD
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, United Kingdom (I.J.D., J.M.W., J.M.S., M.d.C.V.-H., M.E.B., N.A.R., S.P.H.)
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry (H.S., Y.S.), Medical University of Graz, Austria
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor (J.A.S., S.L.R.K., W.Z.)
| | - M Arfan Ikram
- Departments of Epidemiology, Radiology and Neurology (M.A.I.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stéphanie Debette
- Bordeaux Population Health Research Centre U1219, Inserm, France (A.M., G.C., S.D.)
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy (H.J.G.), University Medicine Greifswald, Germany
| | - W T Longstreth
- Departments of Neurology and Epidemiology (W.T.L.), University of Washington, Seattle, WA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, MA (C.L.S., S. Seshadri)
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, MD (L.J.L.)
| | - Myriam Fornage
- From the Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (M.F., X.J.)
| | | |
Collapse
|
10
|
Kessler MD, Bateman NW, Conrads TP, Maxwell GL, Dunning Hotopp JC, O’Connor TD. Ancestral characterization of 1018 cancer cell lines highlights disparities and reveals gene expression and mutational differences. Cancer 2019; 125:2076-2088. [PMID: 30865299 PMCID: PMC6541501 DOI: 10.1002/cncr.32020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
Abstract
Background Although cell lines are an essential resource for studying cancer biology, many are of unknown ancestral origin, and their use may not be optimal for evaluating the biology of all patient populations. Methods An admixture analysis was performed using genome‐wide chip data from the Catalogue of Somatic Mutations in Cancer (COSMIC) Cell Lines Project to calculate genetic ancestry estimates for 1018 cancer cell lines. After stratifying the analyses by tissue and histology types, linear models were used to evaluate the influence of ancestry on gene expression and somatic mutation frequency. Results For the 701 cell lines with unreported ancestry, 215 were of East Asian origin, 30 were of African or African American origin, and 453 were of European origin. Notable imbalances were observed in ancestral representation across tissue type, with the majority of analyzed tissue types having few cell lines of African American ancestral origin, and with Hispanic and South Asian ancestry being almost entirely absent across all cell lines. In evaluating gene expression across these cell lines, expression levels of the genes neurobeachin line 1 (NBEAL1), solute carrier family 6 member 19 (SLC6A19), HEAT repeat containing 6 (HEATR6), and epithelial cell transforming 2 like (ECT2L) were associated with ancestry. Significant differences were also observed in the proportions of somatic mutation types across cell lines with varying ancestral proportions. Conclusions By estimating genetic ancestry for 1018 cancer cell lines, the authors have produced a resource that cancer researchers can use to ensure that their cell lines are ancestrally representative of the populations they intend to affect. Furthermore, the novel ancestry‐specific signal identified underscores the importance of ancestral awareness when studying cancer. Preclinical cancer cell line research is often conducted without an awareness of ancestral background, which results in incongruities between the genetic backgrounds of used cell lines and the patient populations they are intended to represent. By calculating genetic ancestry for 1018 common cancer cell lines and identifying ancestry‐specific expression and somatic mutation patterns, the importance of ancestral awareness is emphasized, and a resource is provided that can be used by cancer researchers to ensure that their cell lines are ancestrally representative of the populations they aim to impact.
Collapse
Affiliation(s)
- Michael D. Kessler
- Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMaryland
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMaryland
- Program in Personalized and Genomic MedicineUniversity of Maryland School of MedicineBaltimoreMaryland
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterBaltimoreMaryland
| | - Nicholas W. Bateman
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology and the John P. Murtha Cancer CenterUniformed Services University of the Health Sciences and Walter Reed National Military Medical CenterBethesdaMaryland
- Inova Schar Cancer Institute, Inova Center for Personalized HealthFairfaxVirginia
| | - Thomas P. Conrads
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology and the John P. Murtha Cancer CenterUniformed Services University of the Health Sciences and Walter Reed National Military Medical CenterBethesdaMaryland
- Inova Schar Cancer Institute, Inova Center for Personalized HealthFairfaxVirginia
- Department of Obstetrics and GynecologyInova Fairfax Medical CampusFalls ChurchVirginia
| | - George L. Maxwell
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology and the John P. Murtha Cancer CenterUniformed Services University of the Health Sciences and Walter Reed National Military Medical CenterBethesdaMaryland
- Inova Schar Cancer Institute, Inova Center for Personalized HealthFairfaxVirginia
- Department of Obstetrics and GynecologyInova Fairfax Medical CampusFalls ChurchVirginia
| | - Julie C. Dunning Hotopp
- Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMaryland
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterBaltimoreMaryland
- Department of Microbiology and ImmunologyUniversity of Maryland School of MedicineBaltimoreMaryland
| | - Timothy D. O’Connor
- Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMaryland
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMaryland
- Program in Personalized and Genomic MedicineUniversity of Maryland School of MedicineBaltimoreMaryland
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterBaltimoreMaryland
| |
Collapse
|
11
|
Meng Q, Valentini D, Rao M, Moro CF, Paraschoudi G, Jäger E, Dodoo E, Rangelova E, Del Chiaro M, Maeurer M. Neoepitope targets of tumour-infiltrating lymphocytes from patients with pancreatic cancer. Br J Cancer 2019; 120:97-108. [PMID: 30377343 PMCID: PMC6325142 DOI: 10.1038/s41416-018-0262-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/05/2018] [Accepted: 08/23/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pancreatic cancer exhibits a poor prognosis and often presents with metastasis at diagnosis. Immunotherapeutic approaches targeting private cancer mutations (neoantigens) are a clinically viable option to improve clinical outcomes. METHODS 3/40 TIL lines (PanTT26, PanTT39, PanTT77) were more closely examined for neoantigen recognition. Whole-exome sequencing was performed to identify non-synonymous somatic mutations. Mutant peptides were synthesised and assessed for antigen-specific IFN-γ production and specific tumour killing in a standard Cr51 assay. TIL phenotype was tested by flow cytometry. Lymphocytes and HLA molecules in tumour tissue were visualised by immunohistochemistry. RESULTS PanTT26 and PanTT39 TILs recognised and killed the autologous tumour cells. PanTT26 TIL recognised the KRASG12v mutation, while a PanTT39 CD4+ TIL clone recognised the neoepitope (GLLRYWRTERLF) from an aquaporin 1-like protein (gene: K7N7A8). Repeated stimulation of TILs with the autologous tumour cells line lead to focused recognition of several mutated targets, based on IFN-γ production. TILs and corresponding PBMCs from PanTT77 showed shared as well as mutually exclusively tumour epitope recognition (TIL-responsive or PBMC-responsive). CONCLUSION This study provides methods to robustly screen T-cell targets for pancreatic cancer. Pancreatic cancer is immunogenic and immunotherapeutic approaches can be used to develop improved, targeted therapies.
Collapse
Affiliation(s)
- Qingda Meng
- Department of Laboratory Medicine (LABMED), Division of Therapeutic Immunology (TIM), Karolinska Institutet, Stockholm, Sweden
| | - Davide Valentini
- Department of Laboratory Medicine (LABMED), Division of Therapeutic Immunology (TIM), Karolinska Institutet, Stockholm, Sweden
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Martin Rao
- Department of Laboratory Medicine (LABMED), Division of Therapeutic Immunology (TIM), Karolinska Institutet, Stockholm, Sweden
| | - Carlos Fernández Moro
- Department of Laboratory Medicine (LABMED), Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Georgia Paraschoudi
- Department of Laboratory Medicine (LABMED), Division of Therapeutic Immunology (TIM), Karolinska Institutet, Stockholm, Sweden
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Elke Jäger
- Krankenhaus Nordwest, Division of Oncology and Haematology, Frankfurt, Germany
| | - Ernest Dodoo
- Department of Laboratory Medicine (LABMED), Division of Therapeutic Immunology (TIM), Karolinska Institutet, Stockholm, Sweden
| | - Elena Rangelova
- Department of Clinical Science, Pancreatic Surgery Unit, Division of Surgery, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Marco Del Chiaro
- Department of Clinical Science, Pancreatic Surgery Unit, Division of Surgery, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Markus Maeurer
- Department of Laboratory Medicine (LABMED), Division of Therapeutic Immunology (TIM), Karolinska Institutet, Stockholm, Sweden.
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
- Department of Oncology/Haematology, KHNW, Frankfurt, Germany & ImmunoSurgery Unit, Champalimaud Foundation, Lisbon, Portugal.
| |
Collapse
|
12
|
Sanglard LP, Nascimento M, Moriel P, Sommer J, Ashwell M, Poore MH, Duarte MDS, Serão NVL. Impact of energy restriction during late gestation on the muscle and blood transcriptome of beef calves after preconditioning. BMC Genomics 2018; 19:702. [PMID: 30253751 PMCID: PMC6156876 DOI: 10.1186/s12864-018-5089-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 09/19/2018] [Indexed: 02/03/2023] Open
Abstract
Background Maternal nutrition has been highlighted as one of the main factors affecting intra-uterine environment. The increase in nutritional requirements by beef cows during late gestation can cause nutritional deficiency in the fetus and impact the fetal regulation of genes associated with myogenesis and immune response. Methods Forty days before the expected calving date, cows were assigned to one of two diets: 100% (control) or 70% (restricted group) of the daily energy requirement. Muscle samples were collected from 12 heifers and 12 steers, and blood samples were collected from 12 steers. The objective of this work was to identify and to assess the biological relevance of differentially expressed genes (DEG) in the skeletal muscle and blood of beef calves born from cows that experienced [or not] a 30% energy restriction during the last 40 days of gestation. Results A total of 160, 164, and 346 DEG (q-value< 0.05) were identified in the skeletal muscle for the effects of diet, sex, and diet-by-sex interaction, respectively. For blood, 452, 1392, and 155 DEG were identified for the effects of diet, time, and diet-by-time interaction, respectively. For skeletal muscle, results based on diet identified genes involved in muscle metabolism. In muscle, from the 10 most DEG down-regulated in the energy-restricted group (REST), we identified 5 genes associated with muscle metabolism and development: SLCO3A1, ATP6V0D1, SLC2A1, GPC4, and RASD2. In blood, among the 10 most DEG, we found genes related to response to stress up-regulated in the REST after weaning, such as SOD3 and INO80D, and to immune response down-regulated in the REST after vaccination, such as OASL, KLRF1, and LOC104968634. Conclusion In conclusion, maternal energy restriction during late gestation may limit the expression of genes in the muscle and increase expression in the blood of calves. In addition, enrichment analysis showed that a short-term maternal energy restriction during pregnancy affects the expression of genes related to energy metabolism and muscle contraction, and immunity and stress response in the blood. Therefore, alterations in the intra-uterine environment can modify prenatal development with lasting consequences to adult life. Electronic supplementary material The online version of this article (10.1186/s12864-018-5089-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leticia P Sanglard
- Department of Animal Science, Iowa State University, Ames, 50011, USA.,Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Moysés Nascimento
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA.,Department of Statistics, Universidade Federal de Viçosa, Viçosa, 36570-000, Brazil
| | - Philipe Moriel
- Range Cattle Research and Education Center, University of Florida, Ona, Florida, 33865, USA
| | - Jeffrey Sommer
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Melissa Ashwell
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Matthew H Poore
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Márcio de S Duarte
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, 36570-000, Brazil.,Instituto Nacional de Ciência e Tecnologia - Ciência Animal, Viçosa, 36570-000, Brazil
| | - Nick V L Serão
- Department of Animal Science, Iowa State University, Ames, 50011, USA. .,Department of Animal Science, North Carolina State University, Raleigh, 27695, USA.
| |
Collapse
|
13
|
Szlavicz E, Olah P, Szabo K, Pagani F, Bata-Csorgo Z, Kemeny L, Szell M. Analysis of psoriasis-relevant gene expression and exon usage alterations after silencing of SR-rich splicing regulators. Exp Dermatol 2018. [DOI: 10.1111/exd.13530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Eszter Szlavicz
- Faculty of Medicine; Department of Dermatology and Allergology; University of Szeged; Szeged Hungary
- Faculty of Medicine; Department of Dermatology, Venereology and Oncodermatology; University of Pécs; Pécs Hungary
| | - Peter Olah
- Faculty of Medicine; Department of Dermatology, Venereology and Oncodermatology; University of Pécs; Pécs Hungary
- Department of Dermatology; University Hospital Düsseldorf; Düsseldorf Germany
| | - Kornélia Szabo
- Faculty of Medicine; Department of Dermatology and Allergology; University of Szeged; Szeged Hungary
- MTA-SZTE Dermatological Research Group; University of Szeged; Szeged Hungary
| | - Franco Pagani
- International Centre for Genetic Engineering and Biotechnology; Trieste Italy
| | - Zsuzsanna Bata-Csorgo
- Faculty of Medicine; Department of Dermatology and Allergology; University of Szeged; Szeged Hungary
- MTA-SZTE Dermatological Research Group; University of Szeged; Szeged Hungary
| | - Lajos Kemeny
- Faculty of Medicine; Department of Dermatology and Allergology; University of Szeged; Szeged Hungary
- MTA-SZTE Dermatological Research Group; University of Szeged; Szeged Hungary
| | - Márta Szell
- MTA-SZTE Dermatological Research Group; University of Szeged; Szeged Hungary
- Faculty of Medicine; Department of Medical Genetics; University of Szeged; Szeged Hungary
| |
Collapse
|
14
|
Münzel T, Sørensen M, Schmidt F, Schmidt E, Steven S, Kröller-Schön S, Daiber A. The Adverse Effects of Environmental Noise Exposure on Oxidative Stress and Cardiovascular Risk. Antioxid Redox Signal 2018; 28:873-908. [PMID: 29350061 PMCID: PMC5898791 DOI: 10.1089/ars.2017.7118] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 12/29/2022]
Abstract
Epidemiological studies have provided evidence that traffic noise exposure is linked to cardiovascular diseases such as arterial hypertension, myocardial infarction, and stroke. Noise is a nonspecific stressor that activates the autonomous nervous system and endocrine signaling. According to the noise reaction model introduced by Babisch and colleagues, chronic low levels of noise can cause so-called nonauditory effects, such as disturbances of activity, sleep, and communication, which can trigger a number of emotional responses, including annoyance and subsequent stress. Chronic stress in turn is associated with cardiovascular risk factors, comprising increased blood pressure and dyslipidemia, increased blood viscosity and blood glucose, and activation of blood clotting factors, in animal models and humans. Persistent chronic noise exposure increases the risk of cardiometabolic diseases, including arterial hypertension, coronary artery disease, diabetes mellitus type 2, and stroke. Recently, we demonstrated that aircraft noise exposure during nighttime can induce endothelial dysfunction in healthy subjects and is even more pronounced in coronary artery disease patients. Importantly, impaired endothelial function was ameliorated by acute oral treatment with the antioxidant vitamin C, suggesting that excessive production of reactive oxygen species contributes to this phenomenon. More recently, we introduced a novel animal model of aircraft noise exposure characterizing the underlying molecular mechanisms leading to noise-dependent adverse oxidative stress-related effects on the vasculature. With the present review, we want to provide an overview of epidemiological, translational clinical, and preclinical noise research addressing the nonauditory, adverse effects of noise exposure with focus on oxidative stress. Antioxid. Redox Signal. 28, 873-908.
Collapse
Affiliation(s)
- Thomas Münzel
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Mette Sørensen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Frank Schmidt
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Erwin Schmidt
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Sebastian Steven
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Swenja Kröller-Schön
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Andreas Daiber
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
15
|
Hixson JE, Jun G, Shimmin LC, Wang Y, Yu G, Mao C, Warren AS, Howard TD, Heide RSV, Van Eyk J, Wang Y, Herrington DM. Whole Exome Sequencing to Identify Genetic Variants Associated with Raised Atherosclerotic Lesions in Young Persons. Sci Rep 2017. [PMID: 28642624 PMCID: PMC5481334 DOI: 10.1038/s41598-017-04433-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We investigated the influence of genetic variants on atherosclerosis using whole exome sequencing in cases and controls from the autopsy study “Pathobiological Determinants of Atherosclerosis in Youth (PDAY)”. We identified a PDAY case group with the highest total amounts of raised lesions (n = 359) for comparisons with a control group with no detectable raised lesions (n = 626). In addition to the standard exome capture, we included genome-wide proximal promoter regions that contain sequences that regulate gene expression. Our statistical analyses included single variant analysis for common variants (MAF > 0.01) and rare variant analysis for low frequency and rare variants (MAF < 0.05). In addition, we investigated known CAD genes previously identified by meta-analysis of GWAS studies. We did not identify individual common variants that reached exome-wide significance using single variant analysis. In analysis limited to 60 CAD genes, we detected strong associations with COL4A2/COL4A1 that also previously showed associations with myocardial infarction and arterial stiffness, as well as coronary artery calcification. Likewise, rare variant analysis did not identify genes that reached exome-wide significance. Among the 60 CAD genes, the strongest association was with NBEAL1 that was also identified in gene-based analysis of whole exome sequencing for early onset myocardial infarction.
Collapse
Affiliation(s)
- James E Hixson
- Human Genetics Center, UTHealth School of Public Health, Houston, TX, 77030, USA.
| | - Goo Jun
- Human Genetics Center, UTHealth School of Public Health, Houston, TX, 77030, USA
| | - Lawrence C Shimmin
- Human Genetics Center, UTHealth School of Public Health, Houston, TX, 77030, USA
| | - Yizhi Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Guoqiang Yu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Chunhong Mao
- Biocomplexity Institute of Virginia Tech, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Andrew S Warren
- Biocomplexity Institute of Virginia Tech, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Timothy D Howard
- Center for Genomics & Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Richard S Vander Heide
- Department of Pathology, Louisiana State University Health Science Center, New Orleans, LA, 70112, USA
| | - Jennifer Van Eyk
- Advanced Clinical BioSystems Research Institute, Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - David M Herrington
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| |
Collapse
|
16
|
Bariana TK, Ouwehand WH, Guerrero JA, Gomez K. Dawning of the age of genomics for platelet granule disorders: improving insight, diagnosis and management. Br J Haematol 2016; 176:705-720. [PMID: 27984638 DOI: 10.1111/bjh.14471] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Inherited disorders of platelet granules are clinically heterogeneous and their prevalence is underestimated because most patients do not undergo a complete diagnostic work-up. The lack of a genetic diagnosis limits the ability to tailor management, screen family members, aid with family planning, predict clinical progression and detect serious consequences, such as myelofibrosis, lung fibrosis and malignancy, in a timely manner. This is set to change with the introduction of high throughput sequencing (HTS) as a routine clinical diagnostic test. HTS diagnostic tests are now available, affordable and allow parallel screening of DNA samples for variants in all of the 80 known bleeding, thrombotic and platelet genes. Increased genetic diagnosis and curation of variants is, in turn, improving our understanding of the pathobiology and clinical course of inherited platelet disorders. Our understanding of the genetic causes of platelet granule disorders and the regulation of granule biogenesis is a work in progress and has been significantly enhanced by recent genomic discoveries from high-powered genome-wide association studies and genome sequencing projects. In the era of whole genome and epigenome sequencing, new strategies are required to integrate multiple sources of big data in the search for elusive, novel genes underlying granule disorders.
Collapse
Affiliation(s)
- Tadbir K Bariana
- Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London, UK.,Department of Haematology, University College London Cancer Institute, London, UK.,Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.,NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK.,Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Jose A Guerrero
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Keith Gomez
- Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London, UK
| | | |
Collapse
|
17
|
Silencing of PNPLA6, the neuropathy target esterase (NTE) codifying gene, alters neurodifferentiation of human embryonal carcinoma stem cells (NT2). Neuroscience 2014; 281:54-67. [DOI: 10.1016/j.neuroscience.2014.08.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 12/30/2022]
|
18
|
Cullinane AR, Schäffer AA, Huizing M. The BEACH is hot: a LYST of emerging roles for BEACH-domain containing proteins in human disease. Traffic 2013; 14:749-66. [PMID: 23521701 DOI: 10.1111/tra.12069] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/18/2013] [Accepted: 03/23/2013] [Indexed: 12/19/2022]
Abstract
BEACH (named after 'Beige and Chediak-Higashi') is a conserved ∼280 residue domain, present in nine human BEACH domain containing proteins (BDCPs). Most BDCPs are large, containing a PH-like domain for membrane association preceding their BEACH domain, and containing WD40 and other domains for ligand binding. Recent studies found that mutations in individual BDCPs cause several human diseases. BDCP alterations affect lysosome size (LYST and NSMAF), apoptosis (NSMAF), autophagy (LYST, WDFY3, LRBA), granule size (LYST, NBEAL2, NBEA) or synapse formation (NBEA). However, the roles of each BDCP in these membrane events remain controversial. After reviewing studies on individual BDCPs, we propose a unifying hypothesis that BDCPs act as scaffolding proteins that facilitate membrane events, including both fission and fusion, determined by their binding partners. BDCPs may also bind each other, enabling fusion or fission of vesicles that are not necessarily of the same type. Such mechanisms explain why different BDCPs may have roles in autophagy; each BDCP is specific for the cell type or the cargo, but not necessarily specific for attaching to the autophagosome. Further elucidation of these mechanisms, preferably carrying out the same experiment on multiple BDCPs, and possibly using patients' cells, may identify potential targets for therapy.
Collapse
Affiliation(s)
- Andrew R Cullinane
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
19
|
Xia Y, Tang L, Yao L, Wan B, Yang X, Yu L. Literature and patent analysis of the cloning and identification of human functional genes in China. SCIENCE CHINA. LIFE SCIENCES 2012; 55:268-282. [PMID: 22527523 DOI: 10.1007/s11427-012-4299-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2011] [Accepted: 10/13/2011] [Indexed: 05/31/2023]
Abstract
The Human Genome Project was launched at the end of the 1980s. Since then, the cloning and identification of functional genes has been a major focus of research across the world. In China too, the potentially profound impact of such studies on the life sciences and on human health was realized, and relevant studies were initiated in the 1990s. To advance China's involvement in the Human Genome Project, in the mid-1990s, Committee of Experts in Biology from National High Technology Research and Development Program of China (863 Program) proposed the "two 1%" goal. This goal envisaged China contributing 1% of the total sequencing work, and cloning and identifying 1% of the total human functional genes. Over the past 20 years, tremendous achievement has been accomplished by Chinese scientists. It is well known that scientists in China finished the 1% of sequencing work of the Human Genome Project, whereas, there is no comprehensive report about "whether China had finished cloning and identifying 1% of human functional genes". In the present study, the GenBank database at the National Center of Biotechnology Information, the PubMed search tool, and the patent database of the State Intellectual Property Office, China, were used to retrieve entries based on two screening standards: (i) Were the newly cloned and identified genes first reported by Chinese scientists? (ii) Were the Chinese scientists awarded the gene sequence patent? Entries were retrieved from the databases up to the cut-off date of 30 June 2011 and the obtained data were analyzed further. The results showed that 589 new human functional genes were first reported by Chinese scientists and 159 gene sequences were patented (http://gene.fudan.sh.cn/introduction/database/chinagene/chinagene.html). This study systematically summarizes China's contributions to human functional genomics research and answers the question "has China finished cloning and identifying 1% of human functional genes?" in the affirmative.
Collapse
Affiliation(s)
- Yan Xia
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | | | | | | | | | | |
Collapse
|
20
|
Niesmann K, Breuer D, Brockhaus J, Born G, Wolff I, Reissner C, Kilimann MW, Rohlmann A, Missler M. Dendritic spine formation and synaptic function require neurobeachin. Nat Commun 2011; 2:557. [PMID: 22109531 PMCID: PMC3482631 DOI: 10.1038/ncomms1565] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/26/2011] [Indexed: 01/11/2023] Open
Abstract
A challenge in neuroscience is to understand the mechanisms underlying synapse
formation. Most excitatory synapses in the brain are built on spines, which are
actin-rich protrusions from dendrites. Spines are a major substrate of brain
plasticity, and spine pathologies are observed in various mental illnesses. Here we
investigate the role of neurobeachin (Nbea), a multidomain protein previously linked
to cases of autism, in synaptogenesis. We show that deletion of Nbea leads to
reduced numbers of spinous synapses in cultured neurons from complete knockouts and
in cortical tissue from heterozygous mice, accompanied by altered miniature
postsynaptic currents. In addition, excitatory synapses terminate mostly at
dendritic shafts instead of spine heads in Nbea mutants, and actin becomes less
enriched synaptically. As actin and synaptopodin, a spine-associated protein with
actin-bundling activity, accumulate ectopically near the Golgi apparatus of mutant
neurons, a role emerges for Nbea in trafficking important cargo to pre- and
postsynaptic compartments. Most excitatory synapses in the brain are found on dendritic
spines, but the mechanisms underlying synapse formation are poorly understood. Niesmann
et al. investigate the role of neurobeachin in synaptogenesis, and find that
its deletion leads to fewer spinous synapses and altered postsynaptic
currents.
Collapse
Affiliation(s)
- Katharina Niesmann
- Department of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, Münster 48149, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Folkersen L, van't Hooft F, Chernogubova E, Agardh HE, Hansson GK, Hedin U, Liska J, Syvänen AC, Paulsson-Berne G, Paulssson-Berne G, Franco-Cereceda A, Hamsten A, Gabrielsen A, Eriksson P. Association of genetic risk variants with expression of proximal genes identifies novel susceptibility genes for cardiovascular disease. ACTA ACUST UNITED AC 2010; 3:365-73. [PMID: 20562444 DOI: 10.1161/circgenetics.110.948935] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Population-based genome-wide association studies have identified several single nucleotide polymorphisms (SNPs) associated with cardiovascular disease or its risk factors. Genes in close proximity to these risk-SNPs are often thought to be pathogenetically important based on their location alone. However, the actual connections between SNPs and disease mechanisms remain largely unknown. METHODS AND RESULTS To identify novel susceptibility genes, we investigated how 166 SNPs previously found to be associated with increased cardiovascular risk and/or predisposing metabolic traits relate to the expression of nearby genes. Gene expression in 577 samples of aorta, liver, mammary artery, and carotid atherosclerotic plaque was measured using expression arrays. For 47 SNPs, the expression levels of proximal genes (located within 200 kb) were affected (P<0.005). More than 20 of these genes had not previously been identified as candidate genes for cardiovascular or related metabolic traits. SNP-associated gene effects were tissue-specific and the tissue specificity was phenotype-dependent. CONCLUSIONS This study demonstrates several instances of association between risk-SNPs and genes immediately adjacent to them. It also demonstrates instances in which the associated gene is not the immediately proximal and obvious candidate gene for disease. This shows the necessity of careful studies of genetic marker data as a first step toward application of genome-wide association studies findings in a clinical setting.
Collapse
Affiliation(s)
- Lasse Folkersen
- Atherosclerosis Research Unit, Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|