1
|
Dieckmann L, Lahti-Pulkkinen M, Cruceanu C, Räikkönen K, Binder EB, Czamara D. Quantitative trait locus mapping in placenta: A comparative study of chorionic villus and birth placenta. HGG ADVANCES 2024; 5:100326. [PMID: 38993113 PMCID: PMC11365441 DOI: 10.1016/j.xhgg.2024.100326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
The placenta, a pivotal player in the prenatal environment, holds crucial insights into early developmental pathways and future health outcomes. In this study, we explored genetic molecular regulation in chorionic villus samples (CVS) from the first trimester and placenta tissue at birth. We assessed quantitative trait locus (QTL) mapping on DNA methylation and gene expression data in a Finnish cohort of 574 individuals. We found more QTLs in birth placenta than in first-trimester placenta. Nevertheless, a substantial amount of associations overlapped in their effects and showed consistent direction in both tissues, with increasing molecular genetic effects from early pregnancy to birth placenta. The identified QTLs in birth placenta were most enriched in genes with placenta-specific expression. Conducting a phenome-wide-association study (PheWAS) on the associated SNPs, we observed numerous overlaps with genome-wide association study (GWAS) hits (spanning 57 distinct traits and 23 SNPs), with notable enrichments for immunological, skeletal, and respiratory traits. The QTL-SNP rs1737028 (chr6:29737993) presented with the highest number of GWAS hits. This SNP was related to HLA-G expression via DNA methylation and was associated with various immune, respiratory, and psychiatric traits. Our findings implicate increasing genetic molecular regulation during the course of pregnancy and support the involvement of placenta gene regulation, particularly in immunological traits. This study presents a framework for understanding placenta-specific gene regulation during pregnancy and its connection to health-related traits.
Collapse
Affiliation(s)
- Linda Dieckmann
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry, 80804 Munich, Germany
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; Finnish Institute for Health and Welfare, 00271 Helsinki, Finland; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Cristiana Cruceanu
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Darina Czamara
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
2
|
Yang P, Corbett R, Daharsh L, Uribe JH, Byrne KA, Loving CL, Tuggle C. Definition of regulatory elements and transcription factors controlling porcine immune cell gene expression at single cell resolution using single nucleus ATAC-seq. Genomics 2024; 116:110944. [PMID: 39326643 DOI: 10.1016/j.ygeno.2024.110944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
The transcriptome of porcine peripheral blood mononuclear cells (PBMC) at single cell (sc) resolution is well described, but little is understood about the cis-regulatory mechanism behind scPBMC gene expression. Here, we profiled the open chromatin landscape of porcine PBMC that define cis-regulatory elements and mechanism contributing to the transcription using single nucleus ATAC sequencing (snATAC-seq). Approximately 22 % of the identified peaks overlapped with annotated transcription start sites (TSS). Using clustering based on open chromatin pattern similarity, we demonstrate that cell type annotations using snATAC-seq are highly concordant to that reported for sc RNA sequencing (scRNA-seq). The differentially accessible peaks (DAPs) for each cell type were characterized and the pattern of accessibility of the DAPs near cell type markers across cell types was similar to that of the average gene expression level of corresponding marker genes. Additionally, we found that peaks identified in snATAC-seq have the potential power to predict the cell type specific transcription starting site (TSS). We identified both transcription factors (TFs) whose binding motif were enriched in cell type DAPs of multiple cell types and cell type specific TFs by conducting transcription factor binding motif (TFBM) analysis. Furthermore, we identified the putative enhancer or promoter regions bound by TFs for each differentially expressed gene (DEG) with a DAP that overlapped with its TSS by generating cis-co-accessibility networks (CCAN). To predict the regulators of such DEGs, TFBM analysis was performed for each CCAN. The regulator TF-target DEG pairs predicted in this way were largely consistent with the results reported in the ENCODE Transcription Factor Targets Dataset (TFTD). This snATAC-seq approach provides insights into the regulation of chromatin accessibility landscape of porcine PBMCs and enables discovery of TFs predicted to control DEG through binding regulatory elements whose chromatin accessibility correlates with the DEG promoter region.
Collapse
Affiliation(s)
- Pengxin Yang
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames, IA 50011, USA
| | - Ryan Corbett
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames, IA 50011, USA
| | - Lance Daharsh
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames, IA 50011, USA
| | - Juber Herrera Uribe
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames, IA 50011, USA
| | | | | | - Christopher Tuggle
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames, IA 50011, USA.
| |
Collapse
|
3
|
Kamble P, Nagar PR, Bhakhar KA, Garg P, Sobhia ME, Naidu S, Bharatam PV. Cancer pharmacoinformatics: Databases and analytical tools. Funct Integr Genomics 2024; 24:166. [PMID: 39294509 DOI: 10.1007/s10142-024-01445-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cancer is a subject of extensive investigation, and the utilization of omics technology has resulted in the generation of substantial volumes of big data in cancer research. Numerous databases are being developed to manage and organize this data effectively. These databases encompass various domains such as genomics, transcriptomics, proteomics, metabolomics, immunology, and drug discovery. The application of computational tools into various core components of pharmaceutical sciences constitutes "Pharmacoinformatics", an emerging paradigm in rational drug discovery. The three major features of pharmacoinformatics include (i) Structure modelling of putative drugs and targets, (ii) Compilation of databases and analysis using statistical approaches, and (iii) Employing artificial intelligence/machine learning algorithms for the discovery of novel therapeutic molecules. The development, updating, and analysis of databases using statistical approaches play a pivotal role in pharmacoinformatics. Multiple software tools are associated with oncoinformatics research. This review catalogs the databases and computational tools related to cancer drug discovery and highlights their potential implications in the pharmacoinformatics of cancer.
Collapse
Affiliation(s)
- Pradnya Kamble
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prinsa R Nagar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Kaushikkumar A Bhakhar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Srivatsava Naidu
- Center of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Prasad V Bharatam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
4
|
George SHL, Medina-Rivera A, Idaghdour Y, Lappalainen T, Gallego Romero I. Increasing diversity of functional genetics studies to advance biological discovery and human health. Am J Hum Genet 2023; 110:1996-2002. [PMID: 37995684 PMCID: PMC10716434 DOI: 10.1016/j.ajhg.2023.10.012] [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: 05/22/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
In this perspective we discuss the current lack of genetic and environmental diversity in functional genomics datasets. There is a well-described Eurocentric bias in genetic and functional genomic research that has a clear impact on the benefit this research can bring to underrepresented populations. Current research focused on genetic variant-to-function experiments aims to identify molecular QTLs, but the lack of data from genetically diverse individuals has limited analyses to mostly populations of European ancestry. Although some efforts have been established to increase diversity in functional genomic studies, much remains to be done to consistently generate data for underrepresented populations from now on. We discuss the major barriers for this continuity and suggest actionable insights, aiming to empower research and researchers from underserved populations.
Collapse
Affiliation(s)
- Sophia H L George
- Department of Obstetrics, Gynecology and Reproductive Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación Sobre El Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Youssef Idaghdour
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, UAE; Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE; Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Tuuli Lappalainen
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden; New York Genome Center, New York, NY, USA.
| | - Irene Gallego Romero
- Melbourne Integrative Genomics and School of BioSciences, University of Melbourne, Parkville, VIC, Australia; Center for Genomics, Evolution and Medicine, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
5
|
Abraham LN, Croll D. Genome-wide expression QTL mapping reveals the highly dynamic regulatory landscape of a major wheat pathogen. BMC Biol 2023; 21:263. [PMID: 37981685 PMCID: PMC10658818 DOI: 10.1186/s12915-023-01763-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND In agricultural ecosystems, outbreaks of diseases are frequent and pose a significant threat to food security. A successful pathogen undergoes a complex and well-timed sequence of regulatory changes to avoid detection by the host immune system; hence, well-tuned gene regulation is essential for survival. However, the extent to which the regulatory polymorphisms in a pathogen population provide an adaptive advantage is poorly understood. RESULTS We used Zymoseptoria tritici, one of the most important pathogens of wheat, to generate a genome-wide map of regulatory polymorphism governing gene expression. We investigated genome-wide transcription levels of 146 strains grown under nutrient starvation and performed expression quantitative trait loci (eQTL) mapping. We identified cis-eQTLs for 65.3% of all genes and the majority of all eQTL loci are within 2kb upstream and downstream of the transcription start site (TSS). We also show that polymorphism in different gene elements contributes disproportionally to gene expression variation. Investigating regulatory polymorphism in gene categories, we found an enrichment of regulatory variants for genes predicted to be important for fungal pathogenesis but with comparatively low effect size, suggesting a separate layer of gene regulation involving epigenetics. We also show that previously reported trait-associated SNPs in pathogen populations are frequently cis-regulatory variants of neighboring genes with implications for the trait architecture. CONCLUSIONS Overall, our study provides extensive evidence that single populations segregate large-scale regulatory variation and are likely to fuel rapid adaptation to resistant hosts and environmental change.
Collapse
Affiliation(s)
- Leen Nanchira Abraham
- Laboratory of Evolutionary Genetics, Institute of Biology, University of Neuchâtel, 2000, Neuchâtel, Switzerland
- Present address: Institute of Plant Sciences, University of Cologne, Cologne, Germany
| | - Daniel Croll
- Laboratory of Evolutionary Genetics, Institute of Biology, University of Neuchâtel, 2000, Neuchâtel, Switzerland.
| |
Collapse
|
6
|
Zhu Z, Chen X, Zhang S, Yu R, Qi C, Cheng L, Zhang X. Leveraging molecular quantitative trait loci to comprehend complex diseases/traits from the omics perspective. Hum Genet 2023; 142:1543-1560. [PMID: 37755483 DOI: 10.1007/s00439-023-02602-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
Comprehending the molecular basis of quantitative genetic variation is a principal goal for complex diseases or traits. Molecular quantitative trait loci (molQTLs) have made it possible to investigate the effects of genetic variants hiding behind large-scale omics data. A deeper understanding of molQTL is urgently required in light of the multi-dimensionalization of omics data to more fully elucidate the pertinent biological mechanisms. Herein, we reviewed molQTLs with the corresponding resource from the omics perspective and further discussed the integrative strategy of GWAS-molQTL to infer their causal effects. Subsequently, we described the opportunities and challenges encountered by molQTL. The case studies showed that molQTL is essential for complex diseases and traits, whether single- or multi-omics QTLs. Overall, we highlighted the functional significance of genetic variants to employ the discovery of molQTL in complex diseases and traits.
Collapse
Affiliation(s)
- Zijun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xinyu Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Sainan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Rui Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Changlu Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150028, Heilongjiang, China.
| | - Xue Zhang
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150028, Heilongjiang, China
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
7
|
Auwerx C, Sadler MC, Woh T, Reymond A, Kutalik Z, Porcu E. Exploiting the mediating role of the metabolome to unravel transcript-to-phenotype associations. eLife 2023; 12:81097. [PMID: 36891970 PMCID: PMC9998083 DOI: 10.7554/elife.81097] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023] Open
Abstract
Despite the success of genome-wide association studies (GWASs) in identifying genetic variants associated with complex traits, understanding the mechanisms behind these statistical associations remains challenging. Several methods that integrate methylation, gene expression, and protein quantitative trait loci (QTLs) with GWAS data to determine their causal role in the path from genotype to phenotype have been proposed. Here, we developed and applied a multi-omics Mendelian randomization (MR) framework to study how metabolites mediate the effect of gene expression on complex traits. We identified 216 transcript-metabolite-trait causal triplets involving 26 medically relevant phenotypes. Among these associations, 58% were missed by classical transcriptome-wide MR, which only uses gene expression and GWAS data. This allowed the identification of biologically relevant pathways, such as between ANKH and calcium levels mediated by citrate levels and SLC6A12 and serum creatinine through modulation of the levels of the renal osmolyte betaine. We show that the signals missed by transcriptome-wide MR are found, thanks to the increase in power conferred by integrating multiple omics layer. Simulation analyses show that with larger molecular QTL studies and in case of mediated effects, our multi-omics MR framework outperforms classical MR approaches designed to detect causal relationships between single molecular traits and complex phenotypes.
Collapse
Affiliation(s)
- Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland.,University Center for Primary Care and Public Health, Lausanne, Switzerland.,Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Marie C Sadler
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.,University Center for Primary Care and Public Health, Lausanne, Switzerland.,Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Tristan Woh
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.,University Center for Primary Care and Public Health, Lausanne, Switzerland.,Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Eleonora Porcu
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland.,University Center for Primary Care and Public Health, Lausanne, Switzerland
| |
Collapse
|
8
|
Prowse-Wilkins CP, Lopdell TJ, Xiang R, Vander Jagt CJ, Littlejohn MD, Chamberlain AJ, Goddard ME. Genetic variation in histone modifications and gene expression identifies regulatory variants in the mammary gland of cattle. BMC Genomics 2022; 23:815. [PMID: 36482302 PMCID: PMC9733386 DOI: 10.1186/s12864-022-09002-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Causal variants for complex traits, such as eQTL are often found in non-coding regions of the genome, where they are hypothesised to influence phenotypes by regulating gene expression. Many regulatory regions are marked by histone modifications, which can be assayed by chromatin immunoprecipitation followed by sequencing (ChIP-seq). Sequence reads from ChIP-seq form peaks at putative regulatory regions, which may reflect the amount of regulatory activity at this region. Therefore, eQTL which are also associated with differences in histone modifications are excellent candidate causal variants. RESULTS We assayed the histone modifications H3K4Me3, H3K4Me1 and H3K27ac and mRNA in the mammary gland of up to 400 animals. We identified QTL for peak height (histone QTL), exon expression (eeQTL), allele specific expression (aseQTL) and allele specific binding (asbQTL). By intersecting these results, we identify variants which may influence gene expression by altering regulatory regions of the genome, and may be causal variants for other traits. Lastly, we find that these variants are found in putative transcription factor binding sites, identifying a mechanism for the effect of many eQTL. CONCLUSIONS We find that allele specific and traditional QTL analysis often identify the same genetic variants and provide evidence that many eQTL are regulatory variants which alter activity at regulatory regions of the bovine genome. Our work provides methodological and biological updates on how regulatory mechanisms interplay at multi-omics levels.
Collapse
Affiliation(s)
- Claire P Prowse-Wilkins
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, Bundoora, Victoria, 3082, Australia.
- Faculty of Veterinary & Agricultural Science, University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Thomas J Lopdell
- Research and Development, Livestock Improvement Corporation, Private Bag 3016, Hamilton, 3240, New Zealand
| | - Ruidong Xiang
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, Bundoora, Victoria, 3082, Australia
- Faculty of Veterinary & Agricultural Science, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Christy J Vander Jagt
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, Bundoora, Victoria, 3082, Australia
| | - Mathew D Littlejohn
- Research and Development, Livestock Improvement Corporation, Private Bag 3016, Hamilton, 3240, New Zealand
| | - Amanda J Chamberlain
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, Bundoora, Victoria, 3082, Australia
| | - Michael E Goddard
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, 5 Ring Road, Bundoora, Victoria, 3082, Australia
- Faculty of Veterinary & Agricultural Science, University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
9
|
Huang D, Feng X, Yang H, Wang J, Zhang W, Fan X, Dong X, Chen K, Yu Y, Ma X, Yi X, Li M. QTLbase2: an enhanced catalog of human quantitative trait loci on extensive molecular phenotypes. Nucleic Acids Res 2022; 51:D1122-D1128. [PMID: 36330927 PMCID: PMC9825467 DOI: 10.1093/nar/gkac1020] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Deciphering the fine-scale molecular mechanisms that shape the genetic effects at disease-associated loci from genome-wide association studies (GWAS) remains challenging. The key avenue is to identify the essential molecular phenotypes that mediate the causal variant and disease under particular biological conditions. Therefore, integrating GWAS signals with context-specific quantitative trait loci (QTLs) (such as different tissue/cell types, disease states, and perturbations) from extensive molecular phenotypes would present important strategies for full understanding of disease genetics. Via persistent curation and systematic data processing of large-scale human molecular trait QTLs (xQTLs), we updated our previous QTLbase database (now QTLbase2, http://mulinlab.org/qtlbase) to comprehensively analyze and visualize context-specific QTLs across 22 molecular phenotypes and over 95 tissue/cell types. Overall, the resource features the following major updates and novel functions: (i) 960 more genome-wide QTL summary statistics from 146 independent studies; (ii) new data for 10 previously uncompiled QTL types; (iii) variant query scope expanded to fit 195 QTL datasets based on whole-genome sequencing; (iv) supports filtering and comparison of QTLs for different biological conditions, such as stimulation types and disease states; (v) a new linkage disequilibrium viewer to facilitate variant prioritization across tissue/cell types and QTL types.
Collapse
Affiliation(s)
| | | | - Hongxi Yang
- Department of Pharmacology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jianhua Wang
- Department of Pharmacology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wenwen Zhang
- Department of Pharmacology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xutong Fan
- Department of Pharmacology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaobao Dong
- Department of Bioinformatics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kexin Chen
- Department of Bioinformatics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xin Ma
- Correspondence may also be addressed to Xin Ma.
| | - Xianfu Yi
- Correspondence may also be addressed to Xianfu Yi.
| | - Mulin Jun Li
- To whom correspondence should be addressed. Tel: +86 22 83336668; Fax: +86 22 83336668;
| |
Collapse
|
10
|
Putscher E, Hecker M, Fitzner B, Boxberger N, Schwartz M, Koczan D, Lorenz P, Zettl UK. Genetic risk variants for multiple sclerosis are linked to differences in alternative pre-mRNA splicing. Front Immunol 2022; 13:931831. [PMID: 36405756 PMCID: PMC9670805 DOI: 10.3389/fimmu.2022.931831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/12/2022] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic immune-mediated disease of the central nervous system to which a genetic predisposition contributes. Over 200 genetic regions have been associated with increased disease risk, but the disease-causing variants and their functional impact at the molecular level are mostly poorly defined. We hypothesized that single-nucleotide polymorphisms (SNPs) have an impact on pre-mRNA splicing in MS. METHODS Our study focused on 10 bioinformatically prioritized SNP-gene pairs, in which the SNP has a high potential to alter alternative splicing events (ASEs). We tested for differential gene expression and differential alternative splicing in B cells from MS patients and healthy controls. We further examined the impact of the SNP genotypes on ASEs and on splice isoform expression levels. Novel genotype-dependent effects on splicing were verified with splicing reporter minigene assays. RESULTS We were able to confirm previously described findings regarding the relation of MS-associated SNPs with the ASEs of the pre-mRNAs from GSDMB and SP140. We also observed an increased IL7R exon 6 skipping when comparing relapsing and progressive MS patients to healthy subjects. Moreover, we found evidence that the MS risk alleles of the SNPs rs3851808 (EFCAB13), rs1131123 (HLA-C), rs10783847 (TSFM), and rs2014886 (TSFM) may contribute to a differential splicing pattern. Of particular interest is the genotype-dependent exon skipping of TSFM due to the SNP rs2014886. The minor allele T creates a donor splice site, resulting in the expression of the exon 3 and 4 of a short TSFM transcript isoform, whereas in the presence of the MS risk allele C, this donor site is absent, and thus the short transcript isoform is not expressed. CONCLUSION In summary, we found that genetic variants from MS risk loci affect pre-mRNA splicing. Our findings substantiate the role of ASEs with respect to the genetics of MS. Further studies on how disease-causing genetic variants may modify the interactions between splicing regulatory sequence elements and RNA-binding proteins can help to deepen our understanding of the genetic susceptibility to MS.
Collapse
Affiliation(s)
- Elena Putscher
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Michael Hecker
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Brit Fitzner
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Nina Boxberger
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Margit Schwartz
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| | - Dirk Koczan
- Rostock University Medical Center, Institute of Immunology, Rostock, Germany
| | - Peter Lorenz
- Rostock University Medical Center, Institute of Immunology, Rostock, Germany
| | - Uwe Klaus Zettl
- Rostock University Medical Center, Department of Neurology, Division of Neuroimmunology, Rostock, Germany
| |
Collapse
|
11
|
Girard D, Vandiedonck C. How dysregulation of the immune system promotes diabetes mellitus and cardiovascular risk complications. Front Cardiovasc Med 2022; 9:991716. [PMID: 36247456 PMCID: PMC9556991 DOI: 10.3389/fcvm.2022.991716] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by persistent hyperglycemia due to insulin resistance or failure to produce insulin. Patients with DM develop microvascular complications that include chronic kidney disease and retinopathy, and macrovascular complications that mainly consist in an accelerated and more severe atherosclerosis compared to the general population, increasing the risk of cardiovascular (CV) events, such as stroke or myocardial infarction by 2- to 4-fold. DM is commonly associated with a low-grade chronic inflammation that is a known causal factor in its development and its complications. Moreover, it is now well-established that inflammation and immune cells play a major role in both atherosclerosis genesis and progression, as well as in CV event occurrence. In this review, after a brief presentation of DM physiopathology and its macrovascular complications, we will describe the immune system dysregulation present in patients with type 1 or type 2 diabetes and discuss its role in DM cardiovascular complications development. More specifically, we will review the metabolic changes and aberrant activation that occur in the immune cells driving the chronic inflammation through cytokine and chemokine secretion, thus promoting atherosclerosis onset and progression in a DM context. Finally, we will discuss how genetics and recent systemic approaches bring new insights into the mechanisms behind these inflammatory dysregulations and pave the way toward precision medicine.
Collapse
Affiliation(s)
- Diane Girard
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, IMMEDIAB Laboratory, Paris, France
- Université Paris Cité, Institut Hors-Mur du Diabète, Faculté de Santé, Paris, France
| | - Claire Vandiedonck
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, IMMEDIAB Laboratory, Paris, France
- Université Paris Cité, Institut Hors-Mur du Diabète, Faculté de Santé, Paris, France
| |
Collapse
|
12
|
Pudjihartono M, Perry JK, Print C, O'Sullivan JM, Schierding W. Interpretation of the role of germline and somatic non-coding mutations in cancer: expression and chromatin conformation informed analysis. Clin Epigenetics 2022; 14:120. [PMID: 36171609 PMCID: PMC9520844 DOI: 10.1186/s13148-022-01342-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There has been extensive scrutiny of cancer driving mutations within the exome (especially amino acid altering mutations) as these are more likely to have a clear impact on protein functions, and thus on cell biology. However, this has come at the neglect of systematic identification of regulatory (non-coding) variants, which have recently been identified as putative somatic drivers and key germline risk factors for cancer development. Comprehensive understanding of non-coding mutations requires understanding their role in the disruption of regulatory elements, which then disrupt key biological functions such as gene expression. MAIN BODY We describe how advancements in sequencing technologies have led to the identification of a large number of non-coding mutations with uncharacterized biological significance. We summarize the strategies that have been developed to interpret and prioritize the biological mechanisms impacted by non-coding mutations, focusing on recent annotation of cancer non-coding variants utilizing chromatin states, eQTLs, and chromatin conformation data. CONCLUSION We believe that a better understanding of how to apply different regulatory data types into the study of non-coding mutations will enhance the discovery of novel mechanisms driving cancer.
Collapse
Affiliation(s)
| | - Jo K Perry
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Cris Print
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Justin M O'Sullivan
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- Australian Parkinson's Mission, Garvan Institute of Medical Research, Sydney, NSW, Australia
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - William Schierding
- Liggins Institute, The University of Auckland, Auckland, New Zealand.
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
13
|
Hecker M, Fitzner B, Putscher E, Schwartz M, Winkelmann A, Meister S, Dudesek A, Koczan D, Lorenz P, Boxberger N, Zettl UK. Implication of genetic variants in primary microRNA processing sites in the risk of multiple sclerosis. EBioMedicine 2022; 80:104052. [PMID: 35561450 PMCID: PMC9111935 DOI: 10.1016/j.ebiom.2022.104052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/01/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system with a well-established genetic contribution to susceptibility. Over 200 genetic regions have been linked to the inherited risk of developing MS, but the disease-causing variants and their functional effects at the molecular level are still largely unresolved. We hypothesised that MS-associated single-nucleotide polymorphisms (SNPs) affect the recognition and enzymatic cleavage of primary microRNAs (pri-miRNAs). Methods Our study focused on 11 pri-miRNAs (9 primate-specific) that are encoded in genetic risk loci for MS. The levels of mature miRNAs and potential isoforms (isomiRs) produced from those pri-miRNAs were measured in B cells obtained from the peripheral blood of 63 MS patients and 28 healthy controls. We tested for associations between SNP genotypes and miRNA expression in cis using quantitative trait locus (cis-miR-eQTL) analyses. Genetic effects on miRNA stem-loop processing efficiency were verified using luciferase reporter assays. Potential direct miRNA target genes were identified by transcriptome profiling and computational binding site assessment. Findings Mature miRNAs and isomiRs from hsa-mir-26a-2, hsa-mir-199a-1, hsa-mir-4304, hsa-mir-4423, hsa-mir-4464 and hsa-mir-4492 could be detected in all B-cell samples. When MS patient subgroups were compared with healthy controls, a significant differential expression was observed for miRNAs from the 5’ and 3’ strands of hsa-mir-26a-2 and hsa-mir-199a-1. The cis-miR-eQTL analyses and reporter assays pointed to a slightly more efficient Drosha-mediated processing of hsa-mir-199a-1 when the MS risk allele T of SNP rs1005039 is present. On the other hand, the MS risk allele A of SNP rs817478, which substitutes the first C in a CNNC sequence motif, was found to cause a markedly lower efficiency in the processing of hsa-mir-4423. Overexpression of hsa-mir-199a-1 inhibited the expression of 60 protein-coding genes, including IRAK2, MIF, TNFRSF12A and TRAF1. The only target gene identified for hsa-mir-4423 was TMEM47. Interpretation We found that MS-associated SNPs in sequence determinants of pri-miRNA processing can affect the expression of mature miRNAs. Our findings complement the existing literature on the dysregulation of miRNAs in MS. Further studies on the maturation and function of miRNAs in different cell types and tissues may help to gain a more detailed functional understanding of the genetic basis of MS. Funding This study was funded by the Rostock University Medical Center (FORUN program, grant: 889002), Sanofi Genzyme (grant: GZ-2016-11560) and Merck Serono GmbH (Darmstadt, Germany, an affiliate of Merck KGaA, CrossRef Funder ID: 10.13039/100009945, grant: 4501860307). NB was supported by the Stiftung der Deutschen Wirtschaft (sdw) and the FAZIT foundation. EP was supported by the Landesgraduiertenförderung Mecklenburg-Vorpommern.
Collapse
|
14
|
Genetic approaches for increasing fitness in endangered species. Trends Ecol Evol 2022; 37:332-345. [PMID: 35027225 DOI: 10.1016/j.tree.2021.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Abstract
The global rate of wildlife extinctions is accelerating, and the persistence of many species requires conservation breeding programs. A central paradigm of these programs is to preserve the genetic diversity of the founder populations. However, this may preserve original characteristics that make them vulnerable to extinction. We introduce targeted genetic intervention (TGI) as an alternative approach that promotes traits that enable species to persist in the face of threats by changing the incidence of alleles that impact on fitness. The TGI toolkit includes methods with established efficacy in model organisms and agriculture but are largely untried for conservation, such as synthetic biology and artificial selection. We explore TGI approaches as a species-restoration tool for intractable threats including infectious disease and climate change.
Collapse
|
15
|
Ramachandran D, Dennis J, Fachal L, Schürmann P, Bousset K, Hülse F, Mao Q, Wang Y, Jentschke M, Böhmer G, Strauß HG, Hirchenhain C, Schmidmayr M, Müller F, Runnebaum I, Hein A, Stübs F, Koch M, Ruebner M, Beckmann MW, Fasching PA, Luyten A, Dürst M, Hillemanns P, Easton DF, Dörk T. OUP accepted manuscript. Hum Mol Genet 2022; 31:2483-2497. [PMID: 35157032 PMCID: PMC9396939 DOI: 10.1093/hmg/ddac031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Dhanya Ramachandran
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Laura Fachal
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Peter Schürmann
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Kristine Bousset
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Fabienne Hülse
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Qianqian Mao
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Yingying Wang
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Matthias Jentschke
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | | | - Hans-Georg Strauß
- Department of Gynaecology, University Clinics, Martin-Luther University, Halle-Wittenberg, Germany
| | - Christine Hirchenhain
- Department of Gynaecology, Clinics Carl Gustav Carus, University of Dresden, Dresden, Germany
| | - Monika Schmidmayr
- Department of Gynaecology, Technische Universität München, Munich, Germany
| | - Florian Müller
- Martin-Luther Hospital, Charite University, Berlin, Germany
| | - Ingo Runnebaum
- Department of Gynaecology, Jena University Hospital, Friedrich-Schiller-University Jena, Jena 07747, Germany
| | - Alexander Hein
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Frederik Stübs
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Martin Koch
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Matthias Ruebner
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Matthias W Beckmann
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Peter A Fasching
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Alexander Luyten
- Dysplasia Unit, Department of Gynaecology and Obstetrics, Mare Klinikum, Kronshagen, Germany
- Department of Gynaecology, Wolfsburg Hospital, Wolfsburg, Germany
| | - Matthias Dürst
- Department of Gynaecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich Alexander University of Erlangen–Nuremberg (FAU), Erlangen 91054, Germany
| | - Peter Hillemanns
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, Hannover 30625, Germany
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Thilo Dörk
- To whom correspondence should be addressed at: Gynaecology Research Unit (OE6411), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany. Tel: +49 5115326075; Fax: +49 5115326081;
| |
Collapse
|
16
|
Hoskins JW, Chung CC, O’Brien A, Zhong J, Connelly K, Collins I, Shi J, Amundadottir LT. Inferred expression regulator activities suggest genes mediating cardiometabolic genetic signals. PLoS Comput Biol 2021; 17:e1009563. [PMID: 34793442 PMCID: PMC8639061 DOI: 10.1371/journal.pcbi.1009563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/02/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Expression QTL (eQTL) analyses have suggested many genes mediating genome-wide association study (GWAS) signals but most GWAS signals still lack compelling explanatory genes. We have leveraged an adipose-specific gene regulatory network to infer expression regulator activities and phenotypic master regulators (MRs), which were used to detect activity QTLs (aQTLs) at cardiometabolic trait GWAS loci. Regulator activities were inferred with the VIPER algorithm that integrates enrichment of expected expression changes among a regulator's target genes with confidence in their regulator-target network interactions and target overlap between different regulators (i.e., pleiotropy). Phenotypic MRs were identified as those regulators whose activities were most important in predicting their respective phenotypes using random forest modeling. While eQTLs were typically more significant than aQTLs in cis, the opposite was true among candidate MRs in trans. Several GWAS loci colocalized with MR trans-eQTLs/aQTLs in the absence of colocalized cis-QTLs. Intriguingly, at the 1p36.1 BMI GWAS locus the EPHB2 cis-aQTL was stronger than its cis-eQTL and colocalized with the GWAS signal and 35 BMI MR trans-aQTLs, suggesting the GWAS signal may be mediated by effects on EPHB2 activity and its downstream effects on a network of BMI MRs. These MR and aQTL analyses represent systems genetic methods that may be broadly applied to supplement standard eQTL analyses for suggesting molecular effects mediating GWAS signals.
Collapse
Affiliation(s)
- Jason W. Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JWH); (LTA)
| | - Charles C. Chung
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Aidan O’Brien
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jun Zhong
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Katelyn Connelly
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Irene Collins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jianxin Shi
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JWH); (LTA)
| |
Collapse
|
17
|
Challenges and Opportunities in Understanding Genetics of Fungal Diseases: Towards a Functional Genomics Approach. Infect Immun 2021; 89:e0000521. [PMID: 34031131 DOI: 10.1128/iai.00005-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Infectious diseases are a leading cause of morbidity and mortality worldwide, and human pathogens have long been recognized as one of the main sources of evolutionary pressure, resulting in a high variable genetic background in immune-related genes. The study of the genetic contribution to infectious diseases has undergone tremendous advances over the last decades. Here, focusing on genetic predisposition to fungal diseases, we provide an overview of the available approaches for studying human genetic susceptibility to infections, reviewing current methodological and practical limitations. We describe how the classical methods available, such as family-based studies and candidate gene studies, have contributed to the discovery of crucial susceptibility factors for fungal infections. We will also discuss the contribution of novel unbiased approaches to the field, highlighting their success but also their limitations for the fungal immunology field. Finally, we show how a systems genomics approach can overcome those limitations and can lead to efficient prioritization and identification of genes and pathways with a critical role in susceptibility to fungal diseases. This knowledge will help to stratify at-risk patient groups and, subsequently, develop early appropriate prophylactic and treatment strategies.
Collapse
|
18
|
Villicaña S, Bell JT. Genetic impacts on DNA methylation: research findings and future perspectives. Genome Biol 2021; 22:127. [PMID: 33931130 PMCID: PMC8086086 DOI: 10.1186/s13059-021-02347-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/09/2021] [Indexed: 12/17/2022] Open
Abstract
Multiple recent studies highlight that genetic variants can have strong impacts on a significant proportion of the human DNA methylome. Methylation quantitative trait loci, or meQTLs, allow for the exploration of biological mechanisms that underlie complex human phenotypes, with potential insights for human disease onset and progression. In this review, we summarize recent milestones in characterizing the human genetic basis of DNA methylation variation over the last decade, including heritability findings and genome-wide identification of meQTLs. We also discuss challenges in this field and future areas of research geared to generate insights into molecular processes underlying human complex traits.
Collapse
Affiliation(s)
- Sergio Villicaña
- Department of Twin Research and Genetic Epidemiology, St. Thomas’ Hospital, King’s College London, 3rd Floor, South Wing, Block D, London, SE1 7EH UK
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, St. Thomas’ Hospital, King’s College London, 3rd Floor, South Wing, Block D, London, SE1 7EH UK
| |
Collapse
|
19
|
Park E, Jiang Y, Hao L, Hui J, Xing Y. Genetic variation and microRNA targeting of A-to-I RNA editing fine tune human tissue transcriptomes. Genome Biol 2021; 22:77. [PMID: 33685485 PMCID: PMC7942016 DOI: 10.1186/s13059-021-02287-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A-to-I RNA editing diversifies the transcriptome and has multiple downstream functional effects. Genetic variation contributes to RNA editing variability between individuals and has the potential to impact phenotypic variability. RESULTS We analyze matched genetic and transcriptomic data in 49 tissues across 437 individuals to identify RNA editing events that are associated with genetic variation. Using an RNA editing quantitative trait loci (edQTL) mapping approach, we identify 3117 unique RNA editing events associated with a cis genetic polymorphism. Fourteen percent of these edQTL events are also associated with genetic variation in their gene expression. A subset of these events are associated with genome-wide association study signals of complex traits or diseases. We determine that tissue-specific levels of ADAR and ADARB1 are able to explain a subset of tissue-specific edQTL events. We find that certain microRNAs are able to differentiate between the edited and unedited isoforms of their targets. Furthermore, microRNAs can generate an expression quantitative trait loci (eQTL) signal from an edQTL locus by microRNA-mediated transcript degradation in an editing-specific manner. By integrative analyses of edQTL, eQTL, and microRNA expression profiles, we computationally discover and experimentally validate edQTL-microRNA pairs for which the microRNA may generate an eQTL signal from an edQTL locus in a tissue-specific manner. CONCLUSIONS Our work suggests a mechanism in which RNA editing variability can influence the phenotypes of complex traits and diseases by altering the stability and steady-state level of critical RNA molecules.
Collapse
Affiliation(s)
- Eddie Park
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Yan Jiang
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Lili Hao
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Jingyi Hui
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yi Xing
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
20
|
Nancy Z, Yan L, Hui S, Paul B, Liye C. From the Genetics of Ankylosing Spondylitis to New Biology and Drug Target Discovery. Front Immunol 2021; 12:624632. [PMID: 33679768 PMCID: PMC7925991 DOI: 10.3389/fimmu.2021.624632] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified 113 single nucleotide polymorphisms (SNPs) affecting the risk of developing ankylosing spondylitis (AS), and an on-going GWAS study will likely identify 100+ new risk loci. The translation of genetic findings to novel disease biology and treatments has been difficult due to the following challenges: (1) difficulties in determining the causal genes regulated by disease-associated SNPs, (2) difficulties in determining the relevant cell-type(s) that causal genes exhibit their function(s), (3) difficulties in determining appropriate cellular contexts to interrogate the functional role of causal genes in disease biology. This review will discuss recent progress and unanswered questions with a focus on these challenges. Additionally, we will review the investigation of biology and the development of drugs related to the IL-23/IL-17 pathway, which has been partially driven by the AS genetics, and discuss what can be learned from these studies for the future functional and translational study of AS-associated genes.
Collapse
Affiliation(s)
- Zaarour Nancy
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Li Yan
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Shi Hui
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Bowness Paul
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Chen Liye
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Palmer RHC, Benca-Bachman CE, Huggett SB, Bubier JA, McGeary JE, Ramgiri N, Srijeyanthan J, Yang J, Visscher PM, Yang J, Knopik VS, Chesler EJ. Multi-omic and multi-species meta-analyses of nicotine consumption. Transl Psychiatry 2021; 11:98. [PMID: 33542196 PMCID: PMC7862377 DOI: 10.1038/s41398-021-01231-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/23/2022] Open
Abstract
Cross-species translational approaches to human genomic analyses are lacking. The present study uses an integrative framework to investigate how genes associated with nicotine use in model organisms contribute to the genetic architecture of human tobacco consumption. First, we created a model organism geneset by collecting results from five animal models of nicotine exposure (RNA expression changes in brain) and then tested the relevance of these genes and flanking genetic variation using genetic data from human cigarettes per day (UK BioBank N = 123,844; all European Ancestry). We tested three hypotheses: (1) DNA variation in, or around, the 'model organism geneset' will contribute to the heritability to human tobacco consumption, (2) that the model organism genes will be enriched for genes associated with human tobacco consumption, and (3) that a polygenic score based off our model organism geneset will predict tobacco consumption in the AddHealth sample (N = 1667; all European Ancestry). Our results suggested that: (1) model organism genes accounted for ~5-36% of the observed SNP-heritability in human tobacco consumption (enrichment: 1.60-31.45), (2) model organism genes, but not negative control genes, were enriched for the gene-based associations (MAGMA, H-MAGMA, SMultiXcan) for human cigarettes per day, and (3) polygenic scores based on our model organism geneset predicted cigarettes per day in an independent sample. Altogether, these findings highlight the advantages of using multiple species evidence to isolate genetic factors to better understand the etiological complexity of tobacco and other nicotine consumption.
Collapse
Affiliation(s)
- Rohan H. C. Palmer
- grid.189967.80000 0001 0941 6502Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA USA
| | - Chelsie E. Benca-Bachman
- grid.189967.80000 0001 0941 6502Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA USA
| | - Spencer B. Huggett
- grid.189967.80000 0001 0941 6502Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA USA
| | - Jason A. Bubier
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory, Bar Harbor, ME USA
| | - John E. McGeary
- grid.40263.330000 0004 1936 9094Department of Psychiatry and Human Behavior, Brown University, Providence, RI USA ,grid.413904.b0000 0004 0420 4094Providence Veterans Affairs Medical Center, Providence, RI USA
| | - Nikhil Ramgiri
- grid.189967.80000 0001 0941 6502Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA USA
| | - Jenani Srijeyanthan
- grid.189967.80000 0001 0941 6502Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA USA
| | - Jingjing Yang
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University School of Medicine, Atlanta, GA USA
| | - Peter M. Visscher
- grid.1003.20000 0000 9320 7537Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD Australia
| | - Jian Yang
- grid.1003.20000 0000 9320 7537Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD Australia
| | - Valerie S. Knopik
- grid.169077.e0000 0004 1937 2197Department of Human Development and Family Studies, Purdue University, West Lafayette, IN USA
| | - Elissa J. Chesler
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory, Bar Harbor, ME USA
| |
Collapse
|
22
|
Tian J, Cai Y, Li Y, Lu Z, Huang J, Deng Y, Yang N, Wang X, Ying P, Zhang S, Zhu Y, Zhang H, Zhong R, Chang J, Miao X. CancerImmunityQTL: a database to systematically evaluate the impact of genetic variants on immune infiltration in human cancer. Nucleic Acids Res 2021; 49:D1065-D1073. [PMID: 33010176 PMCID: PMC7778991 DOI: 10.1093/nar/gkaa805] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/10/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor-infiltrating immune cells as integral component of the tumor microenvironment are associated with tumor progress, prognosis and responses to immunotherapy. Genetic variants have been demonstrated to impact tumor-infiltrating, underscoring the heritable character of immune landscape. Therefore, identification of immunity quantitative trait loci (immunQTLs), which evaluate the effect of genetic variants on immune cells infiltration, might present a critical step toward fully understanding the contribution of genetic variants in tumor development. Although emerging studies have demonstrated the determinants of germline variants on immune infiltration, no database has yet been developed to systematically analyze immunQTLs across multiple cancer types. Using genotype data from TCGA database and immune cell fractions estimated by CIBERSORT, we developed a computational pipeline to identify immunQTLs in 33 cancer types. A total of 913 immunQTLs across different cancer types were identified. Among them, 5 immunQTLs are associated with patient overall survival. Furthermore, by integrating immunQTLs with GWAS data, we identified 527 immunQTLs overlapping with known GWAS linkage disequilibrium regions. Finally, we constructed a user-friendly database, CancerImmunityQTL (http://www.cancerimmunityqtl-hust.com/) for users to browse, search and download data of interest. This database provides an informative resource to understand the germline determinants of immune infiltration in human cancer and benefit from personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Jianbo Tian
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yimin Cai
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinyu Huang
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yao Deng
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Nan Yang
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoyang Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pingting Ying
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shanshan Zhang
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huilan Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Zhong
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiang Chang
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, Key Laboratory of Environmental Health of Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
23
|
Liu Y, Liu X, Zheng Z, Ma T, Liu Y, Long H, Cheng H, Fang M, Gong J, Li X, Zhao S, Xu X. Genome-wide analysis of expression QTL (eQTL) and allele-specific expression (ASE) in pig muscle identifies candidate genes for meat quality traits. Genet Sel Evol 2020; 52:59. [PMID: 33036552 PMCID: PMC7547458 DOI: 10.1186/s12711-020-00579-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 09/28/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Genetic analysis of gene expression level is a promising approach for characterizing candidate genes that are involved in complex economic traits such as meat quality. In the present study, we conducted expression quantitative trait loci (eQTL) and allele-specific expression (ASE) analyses based on RNA-sequencing (RNAseq) data from the longissimus muscle of 189 Duroc × Luchuan crossed pigs in order to identify some candidate genes for meat quality traits. RESULTS Using a genome-wide association study based on a mixed linear model, we identified 7192 cis-eQTL corresponding to 2098 cis-genes (p ≤ 1.33e-3, FDR ≤ 0.05) and 6400 trans-eQTL corresponding to 863 trans-genes (p ≤ 1.13e-6, FDR ≤ 0.05). ASE analysis using RNAseq SNPs identified 9815 significant ASE-SNPs in 2253 unique genes. Integrative analysis between the cis-eQTL and ASE target genes identified 540 common genes, including 33 genes with expression levels that were correlated with at least one meat quality trait. Among these 540 common genes, 63 have been reported previously as candidate genes for meat quality traits, such as PHKG1 (q-value = 1.67e-6 for the leading SNP in the cis-eQTL analysis), NUDT7 (q-value = 5.67e-13), FADS2 (q-value = 8.44e-5), and DGAT2 (q-value = 1.24e-3). CONCLUSIONS The present study confirmed several previously published candidate genes and identified some novel candidate genes for meat quality traits via eQTL and ASE analyses, which will be useful to prioritize candidate genes in further studies.
Collapse
Affiliation(s)
- Yan Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Xiaolei Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Zhiwei Zheng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Tingting Ma
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Ying Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Huan Long
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Huijun Cheng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Ming Fang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021 China
| | - Jing Gong
- Colleges of Informatics, Huazhong Agricultural University, Wuhan, 430070 China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| | - Xuewen Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education & College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070 China
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, 430070 China
| |
Collapse
|
24
|
Spurr LF, Alomran N, Bousounis P, Reece-Stremtan D, Prashant NM, Liu H, Słowiński P, Li M, Zhang Q, Sein J, Asher G, Crandall KA, Tsaneva-Atanasova K, Horvath A. ReQTL: identifying correlations between expressed SNVs and gene expression using RNA-sequencing data. Bioinformatics 2020; 36:1351-1359. [PMID: 31589315 PMCID: PMC7058180 DOI: 10.1093/bioinformatics/btz750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
MOTIVATION By testing for associations between DNA genotypes and gene expression levels, expression quantitative trait locus (eQTL) analyses have been instrumental in understanding how thousands of single nucleotide variants (SNVs) may affect gene expression. As compared to DNA genotypes, RNA genetic variation represents a phenotypic trait that reflects the actual allele content of the studied system. RNA genetic variation at expressed SNV loci can be estimated using the proportion of alleles bearing the variant nucleotide (variant allele fraction, VAFRNA). VAFRNA is a continuous measure which allows for precise allele quantitation in loci where the RNA alleles do not scale with the genotype count. We describe a method to correlate VAFRNA with gene expression and assess its ability to identify genetically regulated expression solely from RNA-sequencing (RNA-seq) datasets. RESULTS We introduce ReQTL, an eQTL modification which substitutes the DNA allele count for the variant allele fraction at expressed SNV loci in the transcriptome (VAFRNA). We exemplify the method on sets of RNA-seq data from human tissues obtained though the Genotype-Tissue Expression (GTEx) project and demonstrate that ReQTL analyses are computationally feasible and can identify a subset of expressed eQTL loci. AVAILABILITY AND IMPLEMENTATION A toolkit to perform ReQTL analyses is available at https://github.com/HorvathLab/ReQTL. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Liam F Spurr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Cancer Program, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Nawaf Alomran
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Pavlos Bousounis
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Dacian Reece-Stremtan
- Computer Applications Support Services, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - N M Prashant
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Hongyu Liu
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Piotr Słowiński
- Department of Mathematics & Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Muzi Li
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Qianqian Zhang
- Department of Biochemistry and Molecular Medicine.,Department of Biostatistics and Bioinformatics, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Justin Sein
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Gabriel Asher
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center
| | - Keith A Crandall
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics & Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.,EPSRC Centre for Predictive Modelling in Healthcare, University of Exeter, Exeter EX4 4QJ, UK
| | - Anelia Horvath
- Biochemistry and Molecular Medicine, McCormick Genomics and Proteomics Center.,Department of Biochemistry and Molecular Medicine.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
25
|
Gleason KJ, Yang F, Pierce BL, He X, Chen LS. Primo: integration of multiple GWAS and omics QTL summary statistics for elucidation of molecular mechanisms of trait-associated SNPs and detection of pleiotropy in complex traits. Genome Biol 2020; 21:236. [PMID: 32912334 PMCID: PMC7488447 DOI: 10.1186/s13059-020-02125-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/29/2020] [Indexed: 01/10/2023] Open
Abstract
To provide a comprehensive mechanistic interpretation of how known trait-associated SNPs affect complex traits, we propose a method, Primo, for integrative analysis of GWAS summary statistics with multiple sets of omics QTL summary statistics from different cellular conditions or studies. Primo examines association patterns of SNPs to complex and omics traits. In gene regions harboring known susceptibility loci, Primo performs conditional association analysis to account for linkage disequilibrium. Primo allows for unknown study heterogeneity and sample correlations. We show two applications using Primo to examine the molecular mechanisms of known susceptibility loci and to detect and interpret pleiotropic effects.
Collapse
Affiliation(s)
- Kevin J. Gleason
- Department of Public Health Sciences, University of Chicago, 5841 South Maryland Ave MC2000, Chicago, 60637 IL USA
| | - Fan Yang
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Aurora, 80045 CO USA
| | - Brandon L. Pierce
- Department of Public Health Sciences, University of Chicago, 5841 South Maryland Ave MC2000, Chicago, 60637 IL USA
- Department of Human Genetics, University of Chicago, 920 E 58th St, Chicago, 60637 IL USA
| | - Xin He
- Department of Human Genetics, University of Chicago, 920 E 58th St, Chicago, 60637 IL USA
| | - Lin S. Chen
- Department of Public Health Sciences, University of Chicago, 5841 South Maryland Ave MC2000, Chicago, 60637 IL USA
| |
Collapse
|
26
|
Diedisheim M, Carcarino E, Vandiedonck C, Roussel R, Gautier JF, Venteclef N. Regulation of inflammation in diabetes: From genetics to epigenomics evidence. Mol Metab 2020; 41:101041. [PMID: 32603690 PMCID: PMC7394913 DOI: 10.1016/j.molmet.2020.101041] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Background Diabetes is one of the greatest public health challenges worldwide, and we still lack complementary approaches to significantly enhance the efficacy of preventive and therapeutic approaches. Genetic and environmental factors are the culprits involved in diabetes risk. Evidence from the last decade has highlighted that deregulation in the immune and inflammatory responses increase susceptibility to type 1 and type 2 diabetes. Spatiotemporal patterns of gene expression involved in immune cell polarisation depend on genomic enhancer elements in response to inflammatory and metabolic cues. Several studies have reported that most regulatory genetic variants are located in the non-protein coding regions of the genome and particularly in enhancer regions. The progress of high-throughput technologies has permitted the characterisation of enhancer chromatin properties. These advances support the concept that genetic alteration of enhancers may influence the immune and inflammatory responses in relation to diabetes. Scope of review Results from genome-wide association studies (GWAS) combined with functional and integrative analyses have elucidated the impacts of some diabetes risk-associated variants that are involved in the regulation of the immune system. Additionally, genetic variant mapping to enhancer regions may alter enhancer status, which in turn leads to aberrant expression of inflammatory genes associated with diabetes susceptibility. The focus of this review was to provide an overview of the current indications that inflammatory processes are regulated at the genetic and epigenomic levels in diabetes, along with perspectives on future research avenues that may improve understanding of the disease. Major conclusions In this review, we provide genetic evidence in support of a deregulated immune response as a risk factor in diabetes. We also argue about the importance of enhancer regions in the regulation of immune cell polarisation and how the recent advances using genome-wide methods for enhancer identification have enabled the determination of the impact of enhancer genetic variation on diabetes onset and phenotype. This could eventually lead to better management plans and improved treatment responses in human diabetes.
Collapse
Affiliation(s)
- Marc Diedisheim
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, IMMEDIAB Laboratory, F-75006, Paris, France
| | - Elena Carcarino
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, IMMEDIAB Laboratory, F-75006, Paris, France
| | - Claire Vandiedonck
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, IMMEDIAB Laboratory, F-75006, Paris, France
| | - Ronan Roussel
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, IMMEDIAB Laboratory, F-75006, Paris, France; Bichat-Claude Bernard, Hospital, AP-HP, Diabetology Department, Université de Paris, Paris, France
| | - Jean-François Gautier
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, IMMEDIAB Laboratory, F-75006, Paris, France; Lariboisière Hospital, AP-HP, Diabetology Department, Université de Paris, Paris, France
| | - Nicolas Venteclef
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, IMMEDIAB Laboratory, F-75006, Paris, France.
| |
Collapse
|
27
|
Neumeyer S, Hemani G, Zeggini E. Strengthening Causal Inference for Complex Disease Using Molecular Quantitative Trait Loci. Trends Mol Med 2020; 26:232-241. [PMID: 31718940 DOI: 10.1016/j.molmed.2019.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
Abstract
Large genome-wide association studies (GWAS) have identified loci that are associated with complex traits and diseases, but index variants are often not causal and reside in non-coding regions of the genome. To gain a better understanding of the relevant biological mechanisms, intermediate traits such as gene expression and protein levels are increasingly being investigated because these are likely mediators between genetic variants and disease outcome. Genetic variants associated with intermediate traits, termed molecular quantitative trait loci (molQTLs), can then be used as instrumental variables in a Mendelian randomization (MR) approach to identify the causal features and mechanisms of complex traits. Challenges such as pleiotropy and the non-specificity of molQTLs remain, and further approaches and methods need to be developed.
Collapse
Affiliation(s)
- Sonja Neumeyer
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Gibran Hemani
- Medical Research Council (MRC) Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
28
|
Zheng Z, Huang D, Wang J, Zhao K, Zhou Y, Guo Z, Zhai S, Xu H, Cui H, Yao H, Wang Z, Yi X, Zhang S, Sham PC, Li MJ. QTLbase: an integrative resource for quantitative trait loci across multiple human molecular phenotypes. Nucleic Acids Res 2020; 48:D983-D991. [PMID: 31598699 PMCID: PMC6943073 DOI: 10.1093/nar/gkz888] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/24/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Recent advances in genome sequencing and functional genomic profiling have promoted many large-scale quantitative trait locus (QTL) studies, which connect genotypes with tissue/cell type-specific cellular functions from transcriptional to post-translational level. However, no comprehensive resource can perform QTL lookup across multiple molecular phenotypes and investigate the potential cascade effect of functional variants. We developed a versatile resource, named QTLbase, for interpreting the possible molecular functions of genetic variants, as well as their tissue/cell-type specificity. Overall, QTLbase has five key functions: (i) curating and compiling genome-wide QTL summary statistics for 13 human molecular traits from 233 independent studies; (ii) mapping QTL-relevant tissue/cell types to 78 unified terms according to a standard anatomogram; (iii) normalizing variant and trait information uniformly, yielding >170 million significant QTLs; (iv) providing a rich web client that enables phenome- and tissue-wise visualization; and (v) integrating the most comprehensive genomic features and functional predictions to annotate the potential QTL mechanisms. QTLbase provides a one-stop shop for QTL retrieval and comparison across multiple tissues and multiple layers of molecular complexity, and will greatly help researchers interrogate the biological mechanism of causal variants and guide the direction of functional validation. QTLbase is freely available at http://mulinlab.org/qtlbase.
Collapse
Affiliation(s)
- Zhanye Zheng
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Dandan Huang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Jianhua Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Ke Zhao
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Yao Zhou
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Zhenyang Guo
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Sinan Zhai
- School of Biomedical Engineering, Tianjin Medical University, Tianjin 300070, China
| | - Hang Xu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Hui Cui
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Hongcheng Yao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Zhao Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Xianfu Yi
- School of Biomedical Engineering, Tianjin Medical University, Tianjin 300070, China
| | - Shijie Zhang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Pak Chung Sham
- Centre of Genomics Sciences, State Key Laboratory of Brain and Cognitive Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Mulin Jun Li
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
29
|
Fischer D. Performing QTL and eQTL Analyses with the R-Package GenomicTools. Methods Mol Biol 2020; 2082:15-38. [PMID: 31849005 DOI: 10.1007/978-1-0716-0026-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We present the R-package GenomicTools that can be used to perform QTL and eQTL analyses in a user-friendly way. First, the theoretical backgrounds of both implemented methods are explained. These are (a) the linear model approach that is commonly used in the standard QTL/eQTL testing as well as (b) a non-parametrical directional testing method implemented in GenomicTools. The directional test overcomes some of the drawbacks of the standard way and is robust against outliers in the data. The main focus, however, is on a detailed explanation, how the R-package is used in practice. Starting from the installation of the package, followed by the data import and also the required steps to perform the analyses, all necessary steps are explained in detail with examples. Also, the commands to create publication-ready figures are presented. The last chapter concludes and discusses general topics related to the analysis of QTL and eQTL data in particular and genomic data in general.
Collapse
Affiliation(s)
- Daniel Fischer
- Applied Statistical Methods, Natural Resources Institute Finland (Luke), Jokioinen, Finland.
| |
Collapse
|
30
|
Saul MC, Philip VM, Reinholdt LG, Chesler EJ. High-Diversity Mouse Populations for Complex Traits. Trends Genet 2019; 35:501-514. [PMID: 31133439 DOI: 10.1016/j.tig.2019.04.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/21/2022]
Abstract
Contemporary mouse genetic reference populations are a powerful platform to discover complex disease mechanisms. Advanced high-diversity mouse populations include the Collaborative Cross (CC) strains, Diversity Outbred (DO) stock, and their isogenic founder strains. When used in systems genetics and integrative genomics analyses, these populations efficiently harnesses known genetic variation for precise and contextualized identification of complex disease mechanisms. Extensive genetic, genomic, and phenotypic data are already available for these high-diversity mouse populations and a growing suite of data analysis tools have been developed to support research on diverse mice. This integrated resource can be used to discover and evaluate disease mechanisms relevant across species.
Collapse
Affiliation(s)
- Michael C Saul
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - Vivek M Philip
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | -
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA; UNC Chapel Hill, Chapel Hill, NC, USA; SUNY Binghamton, Binghamton, NY, USA; Pittsburgh University, Pittsburgh, PA, USA
| | - Elissa J Chesler
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA.
| |
Collapse
|
31
|
Maynard RD, Ackert-Bicknell CL. Mouse Models and Online Resources for Functional Analysis of Osteoporosis Genome-Wide Association Studies. Front Endocrinol (Lausanne) 2019; 10:277. [PMID: 31133984 PMCID: PMC6515928 DOI: 10.3389/fendo.2019.00277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is a complex genetic disease in which the number of loci associated with the bone mineral density, a clinical risk factor for fracture, has increased at an exponential rate in the last decade. The identification of the causative variants and candidate genes underlying these loci has not been able to keep pace with the rate of locus discovery. A large number of tools and data resources have been built around the use of the mouse as model of human genetic disease. Herein, we describe resources available for functional validation of human Genome Wide Association Study (GWAS) loci using mouse models. We specifically focus on large-scale phenotyping efforts focused on bone relevant phenotypes and repositories of genotype-phenotype data that exist for transgenic and mutant mice, which can be readily mined as a first step toward more targeted efforts designed to deeply characterize the role of a gene in bone biology.
Collapse
Affiliation(s)
- Robert D. Maynard
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
| | - Cheryl L. Ackert-Bicknell
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester, Rochester, NY, United States
- *Correspondence: Cheryl L. Ackert-Bicknell
| |
Collapse
|
32
|
Gene expression variation and parental allele inheritance in a Xiphophorus interspecies hybridization model. PLoS Genet 2018; 14:e1007875. [PMID: 30586357 PMCID: PMC6324826 DOI: 10.1371/journal.pgen.1007875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/08/2019] [Accepted: 12/04/2018] [Indexed: 01/06/2023] Open
Abstract
Understanding the genetic mechanisms underlying segregation of phenotypic variation through successive generations is important for understanding physiological changes and disease risk. Tracing the etiology of variation in gene expression enables identification of genetic interactions, and may uncover molecular mechanisms leading to the phenotypic expression of a trait, especially when utilizing model organisms that have well-defined genetic lineages. There are a plethora of studies that describe relationships between gene expression and genotype, however, the idea that global variations in gene expression are also controlled by genotype remains novel. Despite the identification of loci that control gene expression variation, the global understanding of how genome constitution affects trait variability is unknown. To study this question, we utilized Xiphophorus fish of different, but tractable genetic backgrounds (inbred, F1 interspecies hybrids, and backcross hybrid progeny), and measured each individual’s gene expression concurrent with the degrees of inter-individual expression variation. We found, (a) F1 interspecies hybrids exhibited less variability than inbred animals, indicting gene expression variation is not affected by the fraction of heterozygous loci within an individual genome, and (b), that mixing genotypes in backcross populations led to higher levels of gene expression variability, supporting the idea that expression variability is caused by heterogeneity of genotypes of cis or trans loci. In conclusion, heterogeneity of genotype, introduced by inheritance of different alleles, accounts for the largest effects on global phenotypical variability. Phenotypical variability is a multi-factorial phenomenon. Although it has been shown that inheriting certain gene is associated with lower phenotypical variability, how genome complexity affect phenotypical variability is still unclear. To study this question, we used inbred Xiphophorus fish, backcross interspecies hybrids, and F1 interspecies hybrids between select Xiphophorus species to model genetic composition with minimum, medium, and maximum heterozygosity respectively, and measured their global gene expression variability. We found gene expression variation is not affected by the percentage of heterozygous loci in individual genome, but instead related to heterogeneity of genotype at local or remote loci.
Collapse
|
33
|
Costantino F, Breban M, Garchon HJ. Genetics and Functional Genomics of Spondyloarthritis. Front Immunol 2018; 9:2933. [PMID: 30619293 PMCID: PMC6305624 DOI: 10.3389/fimmu.2018.02933] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Spondyloarthritis (SpA) is a chronic inflammatory disorder with high heritability but with complex genetics. It encompasses several entities that share common clinical features. Most of the genetic studies in SpA have been restricted to ankylosing spondylitis (AS), the prototypical form of SpA. However, there is growing evidence of shared genetic background between all the SpA subtypes and also with some other immune-mediated diseases. The most important part of SpA heritability comes from the HLA-B27 allele in the major histocompatibility complex (MHC) that explains around 25% of the attributable heredity. Several other loci outside of the MHC have been shown to be involved in the disease. However, all these non-MHC loci explain only a small additional fraction of disease predisposition. Thus, a substantial fraction of SpA genetic basis remains poorly understood. Gene expression profiling is a complementary approach to elucidate the underlying mechanisms and pathways that drive the disease. Several expression profiling studies have been undertaken in SpA. However, results have been quite disappointing with little overlap between the studies largely due to the small sample sizes, resulting in limited power to discover small effects. In this review, we summarize current knowledge on genetic findings concerning SpA and we describe strategic approaches for identification of additional variants, with a focus on rare variants in familial forms. We also provide an overview of gene expression studies in SpA and discuss the possibilities offered by high-throughput RNA sequencing technologies, in particular in sorted cells. Finally, issues in establishing molecular mechanisms underlying genetic association hits and potential translational applications will be addressed.
Collapse
Affiliation(s)
- Félicie Costantino
- UMR 1173 INSERM/Versailles Saint-Quentin-en-Yvelines University, Montigny le Bretonneux, France.,Rheumatology Division Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Maxime Breban
- UMR 1173 INSERM/Versailles Saint-Quentin-en-Yvelines University, Montigny le Bretonneux, France.,Rheumatology Division Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Henri-Jean Garchon
- UMR 1173 INSERM/Versailles Saint-Quentin-en-Yvelines University, Montigny le Bretonneux, France.,Genetics Division Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| |
Collapse
|
34
|
Mobasheri A, Bay-Jensen AC, Gualillo O, Larkin J, Levesque MC, Henrotin Y. Soluble biochemical markers of osteoarthritis: Are we close to using them in clinical practice? Best Pract Res Clin Rheumatol 2018; 31:705-720. [PMID: 30509415 DOI: 10.1016/j.berh.2018.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/17/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis and a major cause of pain and disability. Recent work suggests that the global burden of OA is increasing, and costs associated with treatment are expected to increase dramatically as the aging human population expands. OA is currently diagnosed using radiography, but this technique is an indirect and insensitive measure of alterations in articular cartilage and fails to measure dynamic inflammatory processes in the joint. Radiographic changes detected overtime are small and occur in only a subset (progressors) of patients with OA. Therefore, we diagnose patients with OA on the basis of a diagnostic classification that is outdated. We also use the same tools and approaches for assessing the efficacy of new pharmacological and nonpharmacological interventions. In this review, we discuss the utility of soluble biochemical markers as biomarkers of OA and discuss whether we are close to using them in clinical practice. Combining patient information, functional imaging and carefully selected panels of biomarkers can help in achieving enhanced patient stratification and lead to better designed clinical trials. Biomarkers can be used for molecular endotyping and for developing more effective and more personalized treatments that will enhance clinical care for patients with OA.
Collapse
Affiliation(s)
- Ali Mobasheri
- The D-BOARD FP7 Consortium(1), European Union; The APPROACH IMI Consortium(2), European Union; Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom; Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham, United Kingdom; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| | - Anne-Christine Bay-Jensen
- The D-BOARD FP7 Consortium(1), European Union; The APPROACH IMI Consortium(2), European Union; Rheumatology, Biomarkers and Research, Nordic Bioscience A/S, Herlev, Denmark
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Jonanthan Larkin
- The APPROACH IMI Consortium(2), European Union; C3 DPU, Immunoinflammation Therapeutic Area, GlaxoSmithKline, King of Prussia, PA, 19406, United States
| | - Marc C Levesque
- The APPROACH IMI Consortium(2), European Union; AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Yves Henrotin
- The D-BOARD FP7 Consortium(1), European Union; The APPROACH IMI Consortium(2), European Union; Bone and Cartilage Research Unit, Arthropôle Liege, University of Liège, Liège, Belgium; Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
| |
Collapse
|
35
|
Heinig M. Using Gene Expression to Annotate Cardiovascular GWAS Loci. Front Cardiovasc Med 2018; 5:59. [PMID: 29922679 PMCID: PMC5996083 DOI: 10.3389/fcvm.2018.00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/15/2018] [Indexed: 01/27/2023] Open
Abstract
Genetic variants at hundreds of loci associated with cardiovascular phenotypes have been identified by genome wide association studies. Most of these variants are located in intronic or intergenic regions rendering the functional and mechanistic follow up difficult. These non-protein-coding regions harbor regulatory sequences. Thus the study of genetic variants associated with transcription—so called expression quantitative trait loci—has emerged as a promising approach to identify regulatory sequence variants. The genes and pathways they control constitute candidate causal drivers at cardiovascular risk loci. This review provides an overview of the expression quantitative trait loci resources available for cardiovascular genetics research and the most commonly used approaches for candidate gene identification.
Collapse
Affiliation(s)
- Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany.,Department of Informatics, Technical University of Munich, Munich, Germany
| |
Collapse
|