1
|
Coombes BJ, Ovsyannikova IG, Schaid DJ, Warner ND, Poland GA, Kennedy RB. Polygenic Prediction of Cellular Immune Responses to Mumps Vaccine. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.23.24303277. [PMID: 38464113 PMCID: PMC10925362 DOI: 10.1101/2024.02.23.24303277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
In this report, we provide a follow-up analysis of a previously published genome-wide association study of host genetic variants associated with inter-individual variations in cellular immune responses to mumps vaccine. Here we report the results of a polygenic score (PGS) analysis showing how common variants can predict mumps vaccine response. We found higher PGS for IFNγ, IL-2, and TNFα were predictive of higher post-vaccine IFNγ (p-value = 2e-6), IL-2 (p = 2e-7), and TNFα (p = 0.004) levels, respectively. Control of immune responses after vaccination is complex and polygenic in nature. Our results suggest that the PGS-based approach enables better capture of the combined genetic effects that contribute to mumps vaccine-induced immunity, potentially offering a more comprehensive understanding than traditional single-variant GWAS. This approach will likely have broad utility in studying genetic control of immune responses to other vaccines and to infectious diseases.
Collapse
|
2
|
Betti MJ, Aldrich MC, Gamazon ER. Minimum entropy framework identifies a novel class of genomic functional elements and reveals regulatory mechanisms at human disease loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.11.544507. [PMID: 37398170 PMCID: PMC10312628 DOI: 10.1101/2023.06.11.544507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
We introduce CoRE-BED, a framework trained using 19 epigenomic features in 33 major cell and tissue types to predict cell-type-specific regulatory function. CoRE-BED identifies nine functional classes de-novo, capturing both known and new regulatory categories. Notably, we describe a previously undercharacterized class that we term Development Associated Elements (DAEs), which are highly enriched in cell types with elevated regenerative potential and distinguished by the dual presence of either H3K4me2 and H3K9ac (an epigenetic signature associated with kinetochore assembly) or H3K79me3 and H4K20me1 (a signature associated with transcriptional pause release). Unlike bivalent promoters, which represent a transitory state between active and silenced promoters, DAEs transition directly to or from a non-functional state during stem cell differentiation and are proximal to highly expressed genes. CoRE-BED's interpretability facilitates causal inference and functional prioritization. Across 70 complex traits, distal insulators account for the largest mean proportion of SNP heritability (~49%) captured by the GWAS. Collectively, our results demonstrate the value of exploring non-conventional ways of regulatory classification that enrich for trait heritability, to complement existing approaches for cis-regulatory prediction.
Collapse
Affiliation(s)
| | | | - Eric R Gamazon
- Vanderbilt University Medical Center, Nashville, TN
- Clare Hall, University of Cambridge, Cambridge, England
| |
Collapse
|
3
|
Li D, Farrell JJ, Mez J, Martin ER, Bush WS, Ruiz A, Boada M, de Rojas I, Mayeux R, Haines JL, Vance MAP, Wang LS, Schellenberg GD, Lunetta KL, Farrer LA. Novel loci for Alzheimer's disease identified by a genome-wide association study in Ashkenazi Jews. Alzheimers Dement 2023; 19:5550-5562. [PMID: 37260021 PMCID: PMC10689571 DOI: 10.1002/alz.13117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 06/02/2023]
Abstract
INTRODUCTION Most Alzheimer's disease (AD) loci have been discovered in individuals with European ancestry (EA). METHODS We applied principal component analysis using Gaussian mixture models and an Ashkenazi Jewish (AJ) reference genome-wide association study (GWAS) data set to identify Ashkenazi Jews ascertained in GWAS (n = 42,682), whole genome sequencing (WGS, n = 16,815), and whole exome sequencing (WES, n = 20,504) data sets. The association of AD was tested genome wide (GW) in the GWAS and WGS data sets and exome wide (EW) in all three data sets (EW). Gene-based analyses were performed using aggregated rare variants. RESULTS In addition to apolipoprotein E (APOE), GW analyses (1355 cases and 1661 controls) revealed associations with TREM2 R47H (p = 9.66 × 10-9 ), rs541586606 near RAB3B (p = 5.01 × 10-8 ), and rs760573036 between SPOCK3 and ANXA10 (p = 6.32 × 10-8 ). In EW analyses (1504 cases and 2047 controls), study-wide significant association was observed with rs1003710 near SMAP2 (p = 1.91 × 10-7 ). A significant gene-based association was identified with GIPR (p = 7.34 × 10-7 ). DISCUSSION Our results highlight the efficacy of founder populations for AD genetic studies.
Collapse
Affiliation(s)
- Donghe Li
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - John J Farrell
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Jesse Mez
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Eden R. Martin
- Dr. John T. Macdonald Foundation, University of Miami, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami, Miami, FL 33136, USA
| | - William S. Bush
- Department of Population & Quantitative Health Science and Cleveland Institute for Computational Biology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Agustin Ruiz
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Richard Mayeux
- Taub Institute on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center Department of Neurology, Columbia University, 710 West 168th Street, New York, NY 10032, USA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health Science and Cleveland Institute for Computational Biology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Margaret A. Pericak Vance
- Dr. John T. Macdonald Foundation, University of Miami, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami, Miami, FL 33136, USA
- Department of Neurology, University of Miami, Miami, FL 33136, USA
| | - Li-San Wang
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Gerard D. Schellenberg
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
4
|
Ovsyannikova IG, Haralambieva IH, Schaid DJ, Warner ND, Poland GA, Kennedy RB. Genome-wide determinants of cellular immune responses to mumps vaccine. Vaccine 2023; 41:6579-6588. [PMID: 37778899 DOI: 10.1016/j.vaccine.2023.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/03/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND We have previously described genetic polymorphisms in candidate genes that are associated with inter-individual variations in antibody responses to mumps vaccination. To expand upon our previous work, we performed a genome-wide association study (GWAS) to discover host genetic variants associated with mumps vaccine-induced cellular immune responses. METHODS We performed a GWAS of mumps-specific immune response outcomes (11 secreted cytokines/chemokines) in a cohort of 1,406 subjects. RESULTS Among the 11 cytokine/chemokines we studied, four (IFN-γ, IL-2, IL-1β, and TNFα) demonstrated GWAS signals reaching genome-wide significance (p < 5 × 10-8). A genomic region (encoding Sialic acid-binding immunoglobulin-type lectins/SIGLEC) located on chromosome 19q13 (p < 5 × 10-8) was associated with both IL-1β and TNFα responses. The SIGLEC5/SIGLEC14 region contained 11 statistically significant single nucleotide polymorphisms (SNPs), including the intronic SIGLEC5 rs872629 (p = 1.3E-11) and rs1106476 (p = 1.32E-11) whose alternate alleles were significantly associated with decreased levels of mumps-specific IL-1β (rs872629, p = 1.77E-09; rs1106476, p = 1.78E-09) and TNFα (rs872629, p = 1.3E-11; rs1106476, p = 1.32E-11) production. CONCLUSIONS Our results suggest that SNPs in the SIGLEC5/SIGLEC14 genes play a role in cellular and inflammatory immune responses to mumps vaccination. These findings motivate further research into the functional roles of SIGLEC genes in the regulation of mumps vaccine-induced immunity.
Collapse
Affiliation(s)
| | | | - Daniel J Schaid
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Nathaniel D Warner
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
5
|
Quach HQ, Goergen KM, Grill DE, Haralambieva IH, Ovsyannikova IG, Poland GA, Kennedy RB. Virus-specific and shared gene expression signatures in immune cells after vaccination in response to influenza and vaccinia stimulation. Front Immunol 2023; 14:1168784. [PMID: 37600811 PMCID: PMC10436507 DOI: 10.3389/fimmu.2023.1168784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Background In the vaccine era, individuals receive multiple vaccines in their lifetime. Host gene expression in response to antigenic stimulation is usually virus-specific; however, identifying shared pathways of host response across a wide spectrum of vaccine pathogens can shed light on the molecular mechanisms/components which can be targeted for the development of broad/universal therapeutics and vaccines. Method We isolated PBMCs, monocytes, B cells, and CD8+ T cells from the peripheral blood of healthy donors, who received both seasonal influenza vaccine (within <1 year) and smallpox vaccine (within 1 - 4 years). Each of the purified cell populations was stimulated with either influenza virus or vaccinia virus. Differentially expressed genes (DEGs) relative to unstimulated controls were identified for each in vitro viral infection, as well as for both viral infections (shared DEGs). Pathway enrichment analysis was performed to associate identified DEGs with KEGG/biological pathways. Results We identified 2,906, 3,888, 681, and 446 DEGs in PBMCs, monocytes, B cells, and CD8+ T cells, respectively, in response to influenza stimulation. Meanwhile, 97, 120, 20, and 10 DEGs were identified as gene signatures in PBMCs, monocytes, B cells, and CD8+ T cells, respectively, upon vaccinia stimulation. The majority of DEGs identified in PBMCs were also found in monocytes after either viral stimulation. Of the virus-specific DEGs, 55, 63, and 9 DEGs occurred in common in PBMCs, monocytes, and B cells, respectively, while no DEGs were shared in infected CD8+ T cells after influenza and vaccinia. Gene set enrichment analysis demonstrated that these shared DEGs were over-represented in innate signaling pathways, including cytokine-cytokine receptor interaction, viral protein interaction with cytokine and cytokine receptor, Toll-like receptor signaling, RIG-I-like receptor signaling pathways, cytosolic DNA-sensing pathways, and natural killer cell mediated cytotoxicity. Conclusion Our results provide insights into virus-host interactions in different immune cells, as well as host defense mechanisms against viral stimulation. Our data also highlights the role of monocytes as a major cell population driving gene expression in ex vivo PBMCs in response to viral stimulation. The immune response signaling pathways identified in this study may provide specific targets for the development of novel virus-specific therapeutics and improved vaccines for vaccinia and influenza. Although influenza and vaccinia viruses have been selected in this study as pathogen models, this approach could be applicable to other pathogens.
Collapse
Affiliation(s)
- Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Krista M. Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Diane E. Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Ovsyannikova IG, Haralambieva IH, Schaid DJ, Warner ND, Poland GA, Kennedy RB. Genome-Wide Determinants of Cellular Immune Responses to Mumps Vaccine. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.27.23289213. [PMID: 37205333 PMCID: PMC10187346 DOI: 10.1101/2023.04.27.23289213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background We have previously described genetic polymorphisms in candidate genes that are associated with inter-individual variations in antibody responses to mumps vaccination. To expand upon our previous work, we performed a genome-wide association study (GWAS) to discover host genetic variants associated with mumps vaccine-induced cellular immune responses. Methods We performed a GWAS of mumps-specific immune response outcomes (11 secreted cytokines/chemokines) in a cohort of 1,406 subjects. Results Among the 11 cytokine/chemokines we studied, four (IFN-γ, IL-2, IL-1β, and TNFα) demonstrated GWAS signals reaching genome-wide significance (p<5 x 10 -8 ). A genomic region (encoding Sialic acid-binding immunoglobulin-type lectins/SIGLEC) located on chromosome 19q13 (p<5×10 -8 ) was associated with both IL-1β and TNFα responses. The SIGLEC5/SIGLEC14 region contained 11 statistically significant single nucleotide polymorphisms (SNPs), including the intronic SIGLEC5 rs872629 (p=1.3E-11) and rs1106476 (p=1.32E-11) whose alternate alleles were significantly associated with decreased levels of mumps-specific IL-1β (rs872629, p=1.77E-09; rs1106476, p=1.78E-09) and TNFα (rs872629, p=1.3E-11; rs1106476, p=1.32E-11) production. Conclusions Our results suggest that SNPs in the SIGLEC5/SIGLEC14 genes play a role in cellular and inflammatory immune responses to mumps vaccination. These findings motivate further research into the functional roles of SIGLEC genes in the regulation of mumps vaccine-induced immunity.
Collapse
|
7
|
Couto-Silva CM, Nunes K, Venturini G, Araújo Castro e Silva M, Pereira LV, Comas D, Pereira A, Hünemeier T. Indigenous people from Amazon show genetic signatures of pathogen-driven selection. SCIENCE ADVANCES 2023; 9:eabo0234. [PMID: 36888716 PMCID: PMC9995071 DOI: 10.1126/sciadv.abo0234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Ecological conditions in the Amazon rainforests are historically favorable for the transmission of numerous tropical diseases, especially vector-borne diseases. The high diversity of pathogens likely contributes to the strong selective pressures for human survival and reproduction in this region. However, the genetic basis of human adaptation to this complex ecosystem remains unclear. This study investigates the possible footprints of genetic adaptation to the Amazon rainforest environment by analyzing the genomic data of 19 native populations. The results based on genomic and functional analysis showed an intense signal of natural selection in a set of genes related to Trypanosoma cruzi infection, which is the pathogen responsible for Chagas disease, a neglected tropical parasitic disease native to the Americas that is currently spreading worldwide.
Collapse
Affiliation(s)
- Cainã M. Couto-Silva
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508090, Brazil
| | - Kelly Nunes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508090, Brazil
| | - Gabriela Venturini
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Marcos Araújo Castro e Silva
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508090, Brazil
- Institut de Biologia Evolutiva, Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Lygia V. Pereira
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508090, Brazil
| | - David Comas
- Institut de Biologia Evolutiva, Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Alexandre Pereira
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Tábita Hünemeier
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508090, Brazil
- Institut de Biologia Evolutiva (CSIC/Universitat Pompeu Fabra), Barcelona 08003, Spain
| |
Collapse
|
8
|
Ghabeshi S, Ghasemi S, Mousavizadeh L. The effective factors in human-specific tropism and viral pathogenicity in orthopoxviruses. Cell Biol Int 2023; 47:341-351. [PMID: 36317465 DOI: 10.1002/cbin.11941] [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: 08/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/08/2022]
Abstract
The orthopoxvirus (OPV) genus includes several species that infect humans, including variola, monkeypox, vaccinia, and cowpox. Variola and monkeypox are often life-threatening diseases, while vaccinia and cowpox are usually associated with local lesions. The epidemic potential for OPVs may be lower than respiratory-borne viruses or RNA viruses. However, OPVs are notable for their spread and distribution in different environments and among different hosts. The emergence or re-emergence of OPVs in the human population can also occur in wild or domestic animals as intermediate hosts. More effective and safer vaccines for poxvirus can be developed by understanding how immunity is regulated in poxvirus and vaccines for DNA viruses. Downstream events in cells affected by the virus are regulated functionally by a series of characteristics that are affected by host cell interactions and responses of cells against viral infections, including the interferon pathway and apoptosis. Furthermore, infection outcome is greatly influenced by the distinct selection of host-range and immune-modulatory genes that confer the potential for pathogenesis and host-to-host transmission and the distinct host-range properties of each immune-modulatory gene. The present study reviewed the effective factors in human-restricted tropism and virus pathogenicity in OPVs.
Collapse
Affiliation(s)
- Soad Ghabeshi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Leila Mousavizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Smatti MK, Alkhatib HA, Al Thani AA, Yassine HM. Will Host Genetics Affect the Response to SARS-CoV-2 Vaccines? Historical Precedents. Front Med (Lausanne) 2022; 9:802312. [PMID: 35360730 PMCID: PMC8962369 DOI: 10.3389/fmed.2022.802312] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
Recent progress in genomics and bioinformatics technologies have allowed for the emergence of immunogenomics field. This intersection of immunology and genetics has broadened our understanding of how the immune system responds to infection and vaccination. While the immunogenetic basis of the huge clinical variability in response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is currently being extensively studied, the host genetic determinants of SARS-CoV-2 vaccines remain largely unknown. Previous reports evidenced that vaccines may not protect all populations or individuals equally, due to multiple host- and vaccine-specific factors. Several studies on vaccine response to measles, rubella, hepatitis B, smallpox, and influenza highlighted the contribution of genetic mutations or polymorphisms in modulating the innate and adaptive immunity following vaccination. Specifically, genetic variants in genes encoding virus receptors, antigen presentation, cytokine production, or related to immune cells activation and differentiation could influence how an individual responds to vaccination. Although such knowledge could be utilized to generate personalized vaccine strategies to optimize the vaccine response, studies in this filed are still scarce. Here, we briefly summarize the scientific literature related to the immunogenetic determinants of vaccine-induced immunity, highlighting the possible role of host genetics in response to SARS-CoV-2 vaccines as well.
Collapse
Affiliation(s)
- Maria K. Smatti
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | | - Hadi M. Yassine
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Peng L, Wang X, Bing D. Identification and Validation of Prognostic Factors of Lipid Metabolism in Obstructive Sleep Apnea. Front Genet 2021; 12:747576. [PMID: 34880901 PMCID: PMC8645574 DOI: 10.3389/fgene.2021.747576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Obstructive sleep apnea (OSA) is considered to be an independent factor affecting lipid metabolism. This study explored the relationship between immune genes and lipid metabolism in OSA. Methods: Immune-related Differentially Expressed Genes (DEGs) were identified by analyzing microarray data sets from the Gene Expression Omnibus (GEO) database. Subsequently, we conducted protein-protein interaction (PPI) network analysis and calculated their Gene Ontology (GO) semantic similarity. The GO, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, Disease Ontology (DO), gene set enrichment analysis (GSEA), and gene set variation analysis (GSVA) were employed for functional enrichment analyses and to determine the most significant functional terms. Combined with the results of boruta and random forest, we selected predictors to build a prognostic model, along with seeking out the potential TFs and target drugs for the predictive genes. Results: Immune-related DEGs included 64 genes upregulated and 98 genes downregulated. The enrichment analysis might closely associate with cell adhesion and T cell-mediated immunity pathways and there were many DEGs involved in lipid and atherosclerosis signaling pathways. The highest-ranking hub gene in PPI network have been reported lowly expressed in OSA. In line with the enrichment analysis, DO analysis reveal that respiratory diseases may be associated with OSA besides immune system disorders. Consistent with the result of the KEGG pathway, the analysis of GSVA revealed that the pro-inflammation pathways are associated with OSA. Monocytes and CD8 T cells were the predominant immune cells in adipose tissue. We built a prognostic model with the top six genes, and the prognostic genes were involved in the polarization of macrophage and differentiation of T lymphocyte subsets. In vivo experimental verification revealed that EPGN, LGR5, NCK1 and VIP were significantly down-regulated while PGRMC2 was significantly up-regulated in mouse model of OSA. Conclusions: Our study demonstrated strong associations between immune genes and the development of dyslipidemia in OSA. This work promoted the molecular mechanisms and potential targets for the regulation of lipid metabolism in OSA.
Collapse
Affiliation(s)
- Lu Peng
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.,Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaodi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Dan Bing
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Teruel M, Barturen G, Martínez-Bueno M, Castellini-Pérez O, Barroso-Gil M, Povedano E, Kerick M, Català-Moll F, Makowska Z, Buttgereit A, Pers JO, Marañón C, Ballestar E, Martin J, Carnero-Montoro E, Alarcón-Riquelme ME. Integrative epigenomics in Sjögren´s syndrome reveals novel pathways and a strong interaction between the HLA, autoantibodies and the interferon signature. Sci Rep 2021; 11:23292. [PMID: 34857786 PMCID: PMC8640069 DOI: 10.1038/s41598-021-01324-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Primary Sjögren's syndrome (SS) is a systemic autoimmune disease characterized by lymphocytic infiltration and damage of exocrine salivary and lacrimal glands. The etiology of SS is complex with environmental triggers and genetic factors involved. By conducting an integrated multi-omics study, we confirmed a vast coordinated hypomethylation and overexpression effects in IFN-related genes, what is known as the IFN signature. Stratified and conditional analyses suggest a strong interaction between SS-associated HLA genetic variation and the presence of Anti-Ro/SSA autoantibodies in driving the IFN epigenetic signature and determining SS. We report a novel epigenetic signature characterized by increased DNA methylation levels in a large number of genes enriched in pathways such as collagen metabolism and extracellular matrix organization. We identified potential new genetic variants associated with SS that might mediate their risk by altering DNA methylation or gene expression patterns, as well as disease-interacting genetic variants that exhibit regulatory function only in the SS population. Our study sheds new light on the interaction between genetics, autoantibody profiles, DNA methylation and gene expression in SS, and contributes to elucidate the genetic architecture of gene regulation in an autoimmune population.
Collapse
Affiliation(s)
- María Teruel
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Guillermo Barturen
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Manuel Martínez-Bueno
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Olivia Castellini-Pérez
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Miguel Barroso-Gil
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Elena Povedano
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Martin Kerick
- IPBLN-CSIC, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, 18016, Granada, Spain
| | - Francesc Català-Moll
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- IDIBELL, Bellvitge Biomedical Research Institute 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Zuzanna Makowska
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | - Anne Buttgereit
- Pharmaceuticals Division, Bayer Pharma Aktiengesellschaft, Berlin, Germany
| | | | - Concepción Marañón
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916, Badalona, Barcelona, Spain
- IDIBELL, Bellvitge Biomedical Research Institute 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Martin
- IPBLN-CSIC, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, 18016, Granada, Spain
| | - Elena Carnero-Montoro
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain.
| | - Marta E Alarcón-Riquelme
- GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, 18016, Granada, Spain.
- Institute for Environmental Medicine, Karolinska Institutet, 171 67, Solna, Sweden.
| |
Collapse
|
12
|
Effect of Serial Passage on the Pathogenicity and Immunogenicity of Vaccinia Virus LC16m8 Strain. BIOLOGY 2021; 10:biology10111158. [PMID: 34827150 PMCID: PMC8614788 DOI: 10.3390/biology10111158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 01/20/2023]
Abstract
The phenotype of an attenuated live vaccine depends on gene mutation achieved by, for example, many passages in cultured cells. Viral clones with preferable phenotypes are selected and the causative genetic mutation(s) are later identified. LC16m8 is an example of a highly attenuated smallpox vaccine that was developed and licensed in Japan in the 1970s. LC16m8 was obtained by the passaging of Lister strain, with indicators of small plaque formation and temperature sensitivity as virus phenotypes. This strain can replicate in mammalian cells and provides robust cellular and humoral immunity, as well as long-term immune memory. Recent studies using proteome-wide antigen arrays have revealed that antibody production against LC16m8 and other VACVs differs largely among individuals. Moreover, associations between SNPs in immune-related genes and immune outcomes have been increasingly found. These results lead to predicting adverse events of a vaccine, which is a purpose of vaccinomics. Studies on VACV will continue to contribute to the understanding of host-pathogen interactions and to development of a vaccine for other infectious and non-infectious diseases. Here, we review studies of VACV, including our recent research on LC16m8, with a focus on the phenotype and genotype, and we discuss future research directions.
Collapse
|
13
|
Hay M, Kumar V, Ricaño-Ponce I. The role of the X chromosome in infectious diseases. Brief Funct Genomics 2021; 21:143-158. [PMID: 34651167 DOI: 10.1093/bfgp/elab039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023] Open
Abstract
Many infectious diseases in humans present with a sex bias. This bias arises from a combination of environmental factors, hormones and genetics. In this study, we review the contribution of the X chromosome to the genetic factor associated with infectious diseases. First, we give an overview of the X-linked genes that have been described in the context of infectious diseases and group them in four main pathways that seem to be dysregulated in infectious diseases: nuclear factor kappa-B, interleukin 2 and interferon γ cascade, toll-like receptors and programmed death ligand 1. Then, we review the infectious disease associations in existing genome-wide association studies (GWAS) from the GWAS Catalog and the Pan-UK Biobank, describing the main associations and their possible implications for the disease. Finally, we highlight the importance of including the X chromosome in GWAS analysis and the importance of sex-specific analysis.
Collapse
|
14
|
Hahn T, Wang J, Preus LM, Karaesmen E, Rizvi A, Clay-Gilmour AI, Zhu Q, Wang Y, Yan L, Liu S, Stram DO, Pooler L, Sheng X, Haiman CA, Berg DVD, Webb A, Brock G, Spellman SR, Onel K, McCarthy PL, Pasquini MC, Sucheston-Campbell LE. Novel genetic variants associated with mortality after unrelated donor allogeneic hematopoietic cell transplantation. EClinicalMedicine 2021; 40:101093. [PMID: 34746714 PMCID: PMC8548922 DOI: 10.1016/j.eclinm.2021.101093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Identification of non-human leukocyte antigen (HLA) genetic risk factors could improve survival after allogeneic blood or marrow transplant (BMT) through matching at additional loci or individualizing risk prediction. We hypothesized that non-HLA loci contributed significantly to 1-year overall survival (OS), disease related mortality (DRM) or transplant related mortality (TRM) after unrelated donor (URD)BMT. METHODS We performed a genome-wide association study (GWAS) in 2,887 acute myeloid leukemia (AML), myelodysplastic syndrome (MDS) and acute lymphoblastic leukemia (ALL) patients and their ≥8/8 HLA-matched URDs comprising two independent cohorts treated from 2000-2011. FINDINGS Using meta-analyses of both cohorts, genome-wide significant associations (p < 5 × 10-8) were identified in: recipient genomes with OS at MBNL1 (rs9990017, HR = 1.4, 95% CI 1.24-1.56, p = 3.3 × 10-8) and donor-recipient genotype mismatch with OS at LINC02774 (rs10927108, HR = 1.34, 95% CI 1.21-1.48, p = 2.0 × 10-8); donor genomes with DRM at PCNX4 (rs79076914, HR = 1.7, 95% CI 1.41-2.05, p = 3.15 × 10-8), LINC01194 (rs79498125, HR = 1.86, 95% CI 1.49-2.31, p = 2.84 × 10-8), ARID5B (rs2167710, HR = 1.5, 95% CI 1.31-1.73, p = 6.9 × 10-9) and CT49 (rs32250, HR = 1.44, 95% CI1.26-1.64, p = 2.6 × 10-8); recipient genomes at PILRB with TRM (rs141591562, HR = 2.33, 95% CI 1.74-3.12, p = 1.26 × 10-8) and donor-recipient genotype mismatch between EPGN and MTHF2DL with TRM (rs75868097, HR = 2.66, 95% CI 1.92-3.58, p = 4.6 × 10-9). Results publicly available at https://fuma.ctglab.nl/browse. INTERPRETATION These data provide the first evidence that non-HLA common genetic variation at novel loci with biochemical function significantly impacts 1-year URD-BMT survival. Our findings have implications for donor selection, could guide treatment strategies and provide individualized risk prediction after future validation and functional studies. FUNDING This project was funded by grants from the National Institutes of Health, USA.
Collapse
Affiliation(s)
- Theresa Hahn
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Junke Wang
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Leah M. Preus
- Department of Epidemiology and Environmental Health, State University of New York at Buffalo, Buffalo, NY, USA
| | - Ezgi Karaesmen
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Abbas Rizvi
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Alyssa I. Clay-Gilmour
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Qianqian Zhu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yiwen Wang
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Daniel O. Stram
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Loreall Pooler
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xin Sheng
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher A. Haiman
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - David Van Den Berg
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy Webb
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Guy Brock
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Stephen R. Spellman
- Center for International Blood and Marrow Transplant Research, Minneapolis, MN, USA
| | - Kenan Onel
- Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip L. McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marcelo C. Pasquini
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lara E. Sucheston-Campbell
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
15
|
McInnes G, Yee SW, Pershad Y, Altman RB. Genomewide Association Studies in Pharmacogenomics. Clin Pharmacol Ther 2021; 110:637-648. [PMID: 34185318 PMCID: PMC8376796 DOI: 10.1002/cpt.2349] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022]
Abstract
The increasing availability of genotype data linked with information about drug-response phenotypes has enabled genomewide association studies (GWAS) that uncover genetic determinants of drug response. GWAS have discovered associations between genetic variants and both drug efficacy and adverse drug reactions. Despite these successes, the design of GWAS in pharmacogenomics (PGx) faces unique challenges. In this review, we analyze the last decade of GWAS in PGx. We review trends in publications over time, including the drugs and drug classes studied and the clinical phenotypes used. Several data sharing consortia have contributed substantially to the PGx GWAS literature. We anticipate increased focus on biobanks and highlight phenotypes that would best enable future PGx discoveries.
Collapse
Affiliation(s)
- Gregory McInnes
- Biomedical Informatics Training Program, Stanford University, Stanford, California, USA
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California, USA
| | - Yash Pershad
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, California, USA.,Departments of Genetics, Medicine, Biomedical Data Science, Stanford, California, USA
| |
Collapse
|
16
|
Okada D, Nakamura N, Setoh K, Kawaguchi T, Higasa K, Tabara Y, Matsuda F, Yamada R. Genome-wide association study of individual differences of human lymphocyte profiles using large-scale cytometry data. J Hum Genet 2021; 66:557-567. [PMID: 33230199 PMCID: PMC8144016 DOI: 10.1038/s10038-020-00874-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/26/2020] [Accepted: 10/31/2020] [Indexed: 01/30/2023]
Abstract
Human immune systems are very complex, and the basis for individual differences in immune phenotypes is largely unclear. One reason is that the phenotype of the immune system is so complex that it is very difficult to describe its features and quantify differences between samples. To identify the genetic factors that cause individual differences in whole lymphocyte profiles and their changes after vaccination without having to rely on biological assumptions, we performed a genome-wide association study (GWAS), using cytometry data. Here, we applied computational analysis to the cytometry data of 301 people before receiving an influenza vaccine, and 1, 7, and 90 days after the vaccination to extract the feature statistics of the lymphocyte profiles in a nonparametric and data-driven manner. We analyzed two types of cytometry data: measurements of six markers for B cell classification and seven markers for T cell classification. The coordinate values calculated by this method can be treated as feature statistics of the lymphocyte profile. Next, we examined the genetic basis of individual differences in human immune phenotypes with a GWAS for the feature statistics, and we newly identified seven significant and 36 suggestive single-nucleotide polymorphisms associated with the individual differences in lymphocyte profiles and their change after vaccination. This study provides a new workflow for performing combined analyses of cytometry data and other types of genomics data.
Collapse
Affiliation(s)
- Daigo Okada
- Department of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naotoshi Nakamura
- Department of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuya Setoh
- Department of Human Disease Genomics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahisa Kawaguchi
- Department of Human Disease Genomics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichiro Higasa
- Department of Human Disease Genomics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yasuharu Tabara
- Department of Human Disease Genomics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Department of Human Disease Genomics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Yamada
- Department of Statistical Genetics, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
17
|
Kottyan LC, Trimarchi MP, Lu X, Caldwell JM, Maddox A, Parameswaran S, Lape M, D'Mello RJ, Bonfield M, Ballaban A, Mukkada V, Putnam PE, Abonia P, Ben-Baruch Morgenstern N, Eapen AA, Wen T, Weirauch MT, Rothenberg ME. Replication and meta-analyses nominate numerous eosinophilic esophagitis risk genes. J Allergy Clin Immunol 2021; 147:255-266. [PMID: 33446330 PMCID: PMC8082436 DOI: 10.1016/j.jaci.2020.10.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is an emerging, chronic, rare allergic disease associated with marked eosinophil accumulation in the esophagus. Previous genome-wide association studies have provided strong evidence for 3 genome-wide susceptibility loci. OBJECTIVE We sought to replicate known and suggestive EoE genetic risk loci and conduct a meta-analysis of previously reported data sets. METHODS An EoE-Custom single-nucleotide polymophism (SNP) Chip containing 956 candidate EoE risk single-nucleotide polymorphisms was used to genotype 627 cases and 365 controls. Statistical power was enhanced by adding 1959 external controls and performing meta-analyses with 2 independent EoE genome-wide association studies. RESULTS Meta-analysis identified replicated association and genome-wide significance at 6 loci: 2p23 (2 independent genetic effects) and 5q22, 10p14, 11q13, and 16p13. Seven additional loci were identified at suggestive significance (P < 10-6): 1q31, 5q23, 6q15, 6q21, 8p21, 17q12, and 22q13. From these risk loci, 13 protein-coding EoE candidate risk genes were expressed in a genotype-dependent manner. EoE risk genes were expressed in disease-relevant cell types, including esophageal epithelia, fibroblasts, and immune cells, with some expressed as a function of disease activity. The genetic risk burden of EoE-associated genetic variants was markedly larger in cases relative to controls (P < 10-38); individuals with the highest decile of genetic burden had greater than 12-fold risk of EoE compared with those within the lowest decile. CONCLUSIONS This study extends the genetic underpinnings of EoE, highlighting 13 genes whose genotype-dependent expression expands our etiologic understanding of EoE and provides a framework for a polygenic risk score to be validated in future studies.
Collapse
Affiliation(s)
- Leah C Kottyan
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Michael P Trimarchi
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Xiaoming Lu
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Julie M Caldwell
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Avery Maddox
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael Lape
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Graduate Program in Biomedical Informatics, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Rahul J D'Mello
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Immunology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Madeline Bonfield
- Immunology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Adina Ballaban
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Vincent Mukkada
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio; Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Philip E Putnam
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio; Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Pablo Abonia
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Amy A Eapen
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ting Wen
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
18
|
Pierce CF, Brown VR, Olsen SC, Boggiatto P, Pedersen K, Miller RS, Speidel SE, Smyser TJ. Loci Associated With Antibody Response in Feral Swine ( Sus scrofa) Infected With Brucella suis. Front Vet Sci 2020; 7:554674. [PMID: 33324693 PMCID: PMC7724110 DOI: 10.3389/fvets.2020.554674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Feral swine (Sus scrofa) are a destructive invasive species widespread throughout the United States that disrupt ecosystems, damage crops, and carry pathogens of concern for the health of domestic stock and humans including Brucella suis-the causative organism for swine brucellosis. In domestic swine, brucellosis results in reproductive failure due to abortions and infertility. Contact with infected feral swine poses spillover risks to domestic pigs as well as humans, companion animals, wildlife, and other livestock. Genetic factors influence the outcome of infectious diseases; therefore, genome wide association studies (GWAS) of differential immune responses among feral swine can provide an understanding of disease dynamics and inform management to prevent the spillover of brucellosis from feral swine to domestic pigs. We sought to identify loci associated with differential antibody responses among feral swine naturally infected with B. suis using a case-control GWAS. Tissue, serum, and genotype data (68,516 bi-allelic single nucleotide polymorphisms) collected from 47 feral swine were analyzed in this study. The 47 feral swine were culture positive for Brucella spp. Of these 47, 16 were antibody positive (cases) whereas 31 were antibody negative (controls). Single-locus GWAS were performed using efficient mixed-model association eXpedited (EMMAX) methodology with three genetic models: additive, dominant, and recessive. Eight loci associated with seroconversion were identified on chromosome 4, 8, 9, 10, 12, and 18. Subsequent bioinformatic analyses revealed nine putative candidate genes related to immune function, most notably phagocytosis and induction of an inflammatory response. Identified loci and putative candidate genes may play an important role in host immune responses to B. suis infection, characterized by a detectable bacterial presence yet a differential antibody response. Given that antibody tests are used to evaluate brucellosis infection in domestic pigs and for disease surveillance in invasive feral swine, additional studies are needed to fully understand the genetic component of the response to B. suis infection and to more effectively translate estimates of Brucella spp. antibody prevalence among feral swine to disease control management action.
Collapse
Affiliation(s)
- Courtney F. Pierce
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Vienna R. Brown
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Feral Swine Damage Management Program, Fort Collins, CO, United States
| | - Steven C. Olsen
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Paola Boggiatto
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Kerri Pedersen
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Raleigh, NC, United States
| | - Ryan S. Miller
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, Center for Epidemiology and Animal Health, Fort Collins, CO, United States
| | - Scott E. Speidel
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Timothy J. Smyser
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
| |
Collapse
|
19
|
Kennedy RB, Haralambieva IH, Ovsyannikova IG, Voigt EA, Larrabee BR, Schaid DJ, Zimmermann MT, Oberg AL, Poland GA. Polymorphisms in STING Affect Human Innate Immune Responses to Poxviruses. Front Immunol 2020; 11:567348. [PMID: 33154747 PMCID: PMC7591719 DOI: 10.3389/fimmu.2020.567348] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/24/2020] [Indexed: 01/22/2023] Open
Abstract
We conducted a large genome-wide association study (GWAS) of the immune responses to primary smallpox vaccination in a combined cohort of 1,653 subjects. We did not observe any polymorphisms associated with standard vaccine response outcomes (e.g., neutralizing antibody, T cell ELISPOT response, or T cell cytokine production); however, we did identify a cluster of SNPs on chromosome 5 (5q31.2) that were significantly associated (p-value: 1.3 x 10-12 - 1.5x10-36) with IFNα response to in vitro poxvirus stimulation. Examination of these SNPs led to the functional testing of rs1131769, a non-synonymous SNP in TMEM173 causing an Arg-to-His change at position 232 in the STING protein-a major regulator of innate immune responses to viral infections. Our findings demonstrate differences in the ability of the two STING variants to phosphorylate the downstream intermediates TBK1 and IRF3 in response to multiple STING ligands. Further downstream in the STING pathway, we observed significantly reduced expression of type I IFNs (including IFNα) and IFN-response genes in cells carrying the H232 variant. Subsequent molecular modeling of both alleles predicted altered ligand binding characteristics between the two variants, providing a potential mechanism underlying differences in inter-individual responses to poxvirus infection. Our data indicate that possession of the H232 variant may impair STING-mediated innate immunity to poxviruses. These results clarify prior studies evaluating functional effects of genetic variants in TMEM173 and provide novel data regarding genetic control of poxvirus immunity.
Collapse
Affiliation(s)
- Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| | | | | | - Emily A. Voigt
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| | - Beth R. Larrabee
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN, United States
| | - Daniel J. Schaid
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN, United States
| | - Michael T. Zimmermann
- Bioinformatics Research and Development Laboratory, Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ann L. Oberg
- Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic, Rochester, MN, United States
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
20
|
Pick JL, Hatakeyama M, Ihle KE, Gasparini J, Haussy C, Ishishita S, Matsuda Y, Yoshimura T, Kanaoka MM, Shimizu‐Inatsugi R, Shimizu KK, Tschirren B. Artificial selection reveals the role of transcriptional constraints in the maintenance of life history variation. Evol Lett 2020; 4:200-211. [PMID: 32547781 PMCID: PMC7293072 DOI: 10.1002/evl3.166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/23/2020] [Accepted: 02/18/2020] [Indexed: 11/06/2022] Open
Abstract
The trade-off between reproduction and self-maintenance is a cornerstone of life history theory, yet its proximate underpinnings are elusive. Here, we used an artificial selection approach to create replicated lines of Japanese quail (Coturnix japonica) that differ genetically in their reproductive investment. Whole transcriptome sequencing revealed that females from lines selected for high reproductive output show a consistent upregulation of genes associated with reproduction but a simultaneous downregulation of immune genes. Concordant phenotypic differences in immune function (i.e., specific antibody response against keyhole limpet hemocyanin) were observed between the selection lines, even in males who do not provide parental care. Our findings demonstrate the key role of obligate transcriptional constraints in the maintenance of life history variation. These constraints set fundamental limits to productivity and health in natural and domestic animal populations.
Collapse
Affiliation(s)
- Joel L. Pick
- Department of Evolutionary Biology and Environmental StudiesUniversity of ZurichZurich8057Switzerland
- School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyAustralia
- Current Address: Institute of Evolutionary BiologyUniversity of EdinburghEdinburghUnited Kingdom
| | - Masaomi Hatakeyama
- Department of Evolutionary Biology and Environmental StudiesUniversity of ZurichZurich8057Switzerland
- Functional Genomics Center ZurichZurich8057Switzerland
| | - Kate E. Ihle
- Department of Evolutionary Biology and Environmental StudiesUniversity of ZurichZurich8057Switzerland
| | - Julien Gasparini
- Laboratoire Ecologie and Evolution UMR 7625Université Pierre et Marie Curie CNRS ENSParisFrance
| | - Claudy Haussy
- Laboratoire Ecologie and Evolution UMR 7625Université Pierre et Marie Curie CNRS ENSParisFrance
| | - Satoshi Ishishita
- Graduate School of Bioagricultural SciencesNagoya UniversityNagoya464–8602Japan
| | - Yoichi Matsuda
- Graduate School of Bioagricultural SciencesNagoya UniversityNagoya464–8602Japan
| | - Takashi Yoshimura
- Graduate School of Bioagricultural SciencesNagoya UniversityNagoya464–8602Japan
| | | | - Rie Shimizu‐Inatsugi
- Department of Evolutionary Biology and Environmental StudiesUniversity of ZurichZurich8057Switzerland
| | - Kentaro K. Shimizu
- Department of Evolutionary Biology and Environmental StudiesUniversity of ZurichZurich8057Switzerland
- Kihara Institute for Biological ResearchYokohama City UniversityYokohama244–0813Japan
| | - Barbara Tschirren
- Centre for Ecology and ConservationUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| |
Collapse
|
21
|
Mitchell KA, Shah E, Bowman ED, Zingone A, Nichols N, Pine SR, Kittles RA, Ryan BM. Relationship between West African ancestry with lung cancer risk and survival in African Americans. Cancer Causes Control 2019; 30:1259-1268. [PMID: 31468279 PMCID: PMC8274945 DOI: 10.1007/s10552-019-01212-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/30/2019] [Indexed: 01/12/2023]
Abstract
PURPOSE African Americans, especially men, have a higher incidence of lung cancer compared with all other racial and ethnic groups in the US. Self-reported race is frequently used in genomic research studies to capture an individual's race or ethnicity. However, it is clear from studies of genetic admixture that human genetic variation does not segregate into the same biologically discrete categories as socially defined categories of race. Previous studies have suggested that the degree of West African ancestry among African Americans can contribute to cancer risk in this population, though few studies have addressed this question in lung cancer. METHODS Using a genetic ancestry panel of 100 SNPs, we estimated West African, European, and Native American ancestry in 1,407 self-described African Americans and 2,413 European Americans. RESULTS We found that increasing West African ancestry was associated with increased risk of lung cancer among African American men (ORQ5 vs Q1 = 2.55 (1.45-4.48), p = 0.001), while no association was observed in African American women (ORQ5 vs Q1 = 0.90 (0.51-1.59), p = 0.56). This relationship diminished following adjustment for income and education. CONCLUSIONS Genetic ancestry is not a major contributor to lung cancer risk or survival disparities.
Collapse
Affiliation(s)
- Khadijah A Mitchell
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Ebony Shah
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Elise D Bowman
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Adriana Zingone
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Noah Nichols
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Sharon R Pine
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08854, USA
| | - Rick A Kittles
- Division of Health Equities, Department of Populations Sciences, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.
- Laboratory of Human Carcinogenesis, Center for Cancer Research, NCI, Building 37, Room 3060C, Bethesda, MD, 20892, USA.
| |
Collapse
|
22
|
Voigt EA, Haralambieva IH, Larrabee BL, Kennedy RB, Ovsyannikova IG, Schaid DJ, Poland GA. Polymorphisms in the Wilms Tumor Gene Are Associated With Interindividual Variations in Rubella Virus-Specific Cellular Immunity After Measles-Mumps-Rubella II Vaccination. J Infect Dis 2019; 217:560-566. [PMID: 29253144 DOI: 10.1093/infdis/jix538] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/03/2017] [Indexed: 02/07/2023] Open
Abstract
Rubella vaccination induces widely variable immune responses in vaccine recipients. While rubella vaccination is effective at inducing immunity to rubella infection in most subjects, up to 5% of individuals do not achieve or maintain long-term protective immunity. To expand upon our previous work identifying genetic polymorphisms that are associated with these interindividual differences in humoral immunity to rubella virus, we performed a genome-wide association study in a large cohort of 1843 subjects to discover single-nucleotide polymorphisms (SNPs) associated with rubella virus-specific cellular immune responses. We identified SNPs in the Wilms tumor protein gene (WT1) that were significantly associated (P < 5 × 10-8) with interindividual variations in rubella-specific interleukin 6 secretion from subjects' peripheral blood mononuclear cells postvaccination. No SNPs were found to be significantly associated with variations in rubella-specific interferon-γ secretion. Our findings demonstrate that genetic polymorphisms in the WT1 gene in subjects of European ancestry are associated with interindividual differences in rubella virus-specific cellular immunity after measles-mumps-rubella II vaccination.
Collapse
Affiliation(s)
- Emily A Voigt
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester
| | | | - Beth L Larrabee
- Mayo Clinic Division of Biostatistics, Mayo Clinic, Rochester, Minnesota
| | | | | | - Daniel J Schaid
- Mayo Clinic Division of Biostatistics, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
23
|
Mekli K, Stevens A, Marshall AD, Arpawong TE, Phillips DF, Tampubolon G, Lee J, Prescott CA, Nazroo JY, Pendleton N. Frailty Index associates with GRIN2B in two representative samples from the United States and the United Kingdom. PLoS One 2018; 13:e0207824. [PMID: 30475886 PMCID: PMC6258126 DOI: 10.1371/journal.pone.0207824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023] Open
Abstract
The concept of frailty has been used in the clinical and research field for more than two decades. It is usually described as a clinical state of heightened vulnerability to poor resolution of homeostasis after a stressor event, which thereby increases the risk of adverse outcomes, including falls, delirium, disability and mortality. Here we report the results of the first genome-wide association scan and comparative gene ontology analyses where we aimed to identify genes and pathways associated with the deficit model of frailty. We used a discovery-replication design with two independent, nationally representative samples of older adults. The square-root transformed Frailty Index (FI) was the outcome variable, and age and sex were included as covariates. We report one hit exceeding genome-wide significance: the rs6765037 A allele was significantly associated with a decrease in the square-root transformed FI score in the Discovery sample (beta = -0.01958, p = 2.14E-08), without confirmation in the Replication sample. We also report a nominal replication: the rs7134291 A allele was significantly associated with a decrease in the square-root transformed FI score (Discovery sample: beta = -0.01021, p = 1.85E-06, Replication sample: beta = -0.005013, p = 0.03433). These hits represent the KBTBD12 and the GRIN2B genes, respectively. Comparative gene ontology analysis identified the pathways ‘Neuropathic pain signalling in dorsal horn neurons’ and the ‘GPCR-Mediated Nutrient Sensing in Enteroendocrine Cells’, exceeding the p = 0.01 significance in both samples, although this result does not survive correction for multiple testing. Considering the crucial role of GRIN2B in brain development, synaptic plasticity and cognition, this gene appears to be a potential candidate to play a role in frailty. In conclusion, we conducted genome-wide association scan and pathway analyses and have identified genes and pathways with potential roles in frailty. However, frailty is a complex condition. Therefore, further research is required to confirm our results and more thoroughly identify relevant biological mechanisms.
Collapse
Affiliation(s)
- Krisztina Mekli
- Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
- * E-mail:
| | - Adam Stevens
- Division of Developmental Biology and Medicine, The University of Manchester, Manchester, United Kingdom
| | - Alan D. Marshall
- School of Social and Political Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Thalida E. Arpawong
- Department of Psychology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, United States of America
| | - Drystan F. Phillips
- Dornsife Center for Economic and Social Research, University of Southern California, Los Angeles, CA, United States of America
- RAND Corporation, Santa Monica, CA, United States of America
| | - Gindo Tampubolon
- Institute for Social Change, The University of Manchester, Manchester, United Kingdom
| | - Jinkook Lee
- Dornsife Center for Economic and Social Research, University of Southern California, Los Angeles, CA, United States of America
- RAND Corporation, Santa Monica, CA, United States of America
| | - Carol A. Prescott
- Department of Psychology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, United States of America
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States of America
| | - James Y. Nazroo
- Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
| | - Neil Pendleton
- Division of Neuroscience and Experimental Psychology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
24
|
Sjöstedt E, Sivertsson Å, Hikmet Noraddin F, Katona B, Näsström Å, Vuu J, Kesti D, Oksvold P, Edqvist PH, Olsson I, Uhlén M, Lindskog C. Integration of Transcriptomics and Antibody-Based Proteomics for Exploration of Proteins Expressed in Specialized Tissues. J Proteome Res 2018; 17:4127-4137. [DOI: 10.1021/acs.jproteome.8b00406] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Evelina Sjöstedt
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm SE 171 21, Sweden
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Åsa Sivertsson
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm SE 171 21, Sweden
| | - Feria Hikmet Noraddin
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Borbala Katona
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Åsa Näsström
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Jimmy Vuu
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Dennis Kesti
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Per Oksvold
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm SE 171 21, Sweden
| | - Per-Henrik Edqvist
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Ingmarie Olsson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Stockholm SE 171 21, Sweden
| | - Cecilia Lindskog
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala SE 752 37, Sweden
| |
Collapse
|
25
|
Patel S, Jin L. TMEM173 variants and potential importance to human biology and disease. Genes Immun 2018; 20:82-89. [PMID: 29728611 PMCID: PMC6212339 DOI: 10.1038/s41435-018-0029-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 12/23/2022]
Abstract
TMEM173 gene encodes the protein STING (stimulator of interferon genes), a key player in host defense against pathogens. Mutations in the human TMEM173 gene cause a life-threatening auto-inflammatory disease called SAVI (STING-associated vasculopathy with onset in infancy). Human STING is also a promising therapeutic target for cancers and infectious diseases. Recently, Aduro Biotech and Novartis announced a $250M-plus initiative to develop STING-targeting cancer immunotherapies. Thus, understanding the genetics of the human TMEM173 gene is important for both basic and translational research. The human TMEM173 gene has great heterogeneity and population stratification. R232 of STING is the most common human TMEM173 allele. However, >50% of Americans are not R232/R232. HAQ (R71H-G230A-R293Q) is the second most common human TMEM173 allele. While R232/R232 is the dominant TMEM173 genotype in Europeans, R232/HAQ is the most common TMEM173 genotype in East Asians. Importantly, recent studies suggested that HAQ and H232 are likely loss-of-function TMEM173 alleles. In all, ~30% of East Asians and ~10% of Europeans are HAQ/HAQ, HAQ/H232, or H232/H232. Here, we reviewed human TMEM173 alleles, mutations and their potential impact on human health and medicine.
Collapse
Affiliation(s)
- Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
26
|
Mozzi A, Pontremoli C, Sironi M. Genetic susceptibility to infectious diseases: Current status and future perspectives from genome-wide approaches. INFECTION GENETICS AND EVOLUTION 2017; 66:286-307. [PMID: 28951201 PMCID: PMC7106304 DOI: 10.1016/j.meegid.2017.09.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
Abstract
Genome-wide association studies (GWASs) have been widely applied to identify genetic factors that affect complex diseases or traits. Presently, the GWAS Catalog includes > 2800 human studies. Of these, only a minority have investigated the susceptibility to infectious diseases or the response to therapies for the treatment or prevention of infections. Despite their limited application in the field, GWASs have provided valuable insights by pinpointing associations to both innate and adaptive immune response loci, as well as novel unexpected risk factors for infection susceptibility. Herein, we discuss some issues and caveats of GWASs for infectious diseases, we review the most recent findings ensuing from these studies, and we provide a brief summary of selected GWASs for infections in non-human mammals. We conclude that, although the general trend in the field of complex traits is to shift from GWAS to next-generation sequencing, important knowledge on infectious disease-related traits can be still gained by GWASs, especially for those conditions that have never been investigated using this approach. We suggest that future studies will benefit from the leveraging of information from the host's and pathogen's genomes, as well as from the exploration of models that incorporate heterogeneity across populations and phenotypes. Interactions within HLA genes or among HLA variants and polymorphisms located outside the major histocompatibility complex may also play an important role in shaping the susceptibility and response to invading pathogens. Relatively few GWASs for infectious diseases were performed. Phenotype heterogeneity and case/control misclassification can affect GWAS power. Adaptive and innate immunity loci were identified in several infectious disease GWASs. Unexpected loci (e.g., lncRNAs) were also associated with infection susceptibility. GWASs should integrate host and pathogen diversity and use complex association models.
Collapse
Affiliation(s)
- Alessandra Mozzi
- Bioinformatics, Scientific Institute IRCCS E.MEDEA, 23842 Bosisio Parini, Italy
| | - Chiara Pontremoli
- Bioinformatics, Scientific Institute IRCCS E.MEDEA, 23842 Bosisio Parini, Italy
| | - Manuela Sironi
- Bioinformatics, Scientific Institute IRCCS E.MEDEA, 23842 Bosisio Parini, Italy.
| |
Collapse
|
27
|
Ding L, McDonald DJ. Predicting phenotypes from microarrays using amplified, initially marginal, eigenvector regression. Bioinformatics 2017; 33:i350-i358. [PMID: 28881997 PMCID: PMC5870707 DOI: 10.1093/bioinformatics/btx265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Motivation The discovery of relationships between gene expression measurements and phenotypic responses is hampered by both computational and statistical impediments. Conventional statistical methods are less than ideal because they either fail to select relevant genes, predict poorly, ignore the unknown interaction structure between genes, or are computationally intractable. Thus, the creation of new methods which can handle many expression measurements on relatively small numbers of patients while also uncovering gene-gene relationships and predicting well is desirable. Results We develop a new technique for using the marginal relationship between gene expression measurements and patient survival outcomes to identify a small subset of genes which appear highly relevant for predicting survival, produce a low-dimensional embedding based on this small subset, and amplify this embedding with information from the remaining genes. We motivate our methodology by using gene expression measurements to predict survival time for patients with diffuse large B-cell lymphoma, illustrate the behavior of our methodology on carefully constructed synthetic examples, and test it on a number of other gene expression datasets. Our technique is computationally tractable, generally outperforms other methods, is extensible to other phenotypes, and also identifies different genes (relative to existing methods) for possible future study. Availability and Implementation All of the code and data are available at http://mypage.iu.edu/∼dajmcdon/research/ . Contact dajmcdon@indiana.edu. Supplementary information Supplementary material is available at Bioinformatics online.
Collapse
Affiliation(s)
- Lei Ding
- Department of Statistics, Indiana University, Bloomington, IN, USA
| | | |
Collapse
|
28
|
Patel S, Blaauboer SM, Tucker HR, Mansouri S, Ruiz-Moreno JS, Hamann L, Schumann RR, Opitz B, Jin L. Response to Comment on "The Common R71H-G230A-R293Q Human TMEM173 Is a Null Allele". JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:4185-4188. [PMID: 28533277 PMCID: PMC8344094 DOI: 10.4049/jimmunol.1700322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610
| | - Steven M Blaauboer
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208; and
| | - Heidi R Tucker
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208; and
| | - Samira Mansouri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208; and
| | - Juan Sebastian Ruiz-Moreno
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - University Medicine Berlin, 10117 Berlin, Germany
| | - Lutz Hamann
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - University Medicine Berlin, 10117 Berlin, Germany
| | - Ralf R Schumann
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - University Medicine Berlin, 10117 Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - University Medicine Berlin, 10117 Berlin, Germany
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610;
| |
Collapse
|
29
|
A systematic review on the association between inflammatory genes and cognitive decline in non-demented elderly individuals. Eur Neuropsychopharmacol 2017; 27:568-588. [PMID: 26718789 DOI: 10.1016/j.euroneuro.2015.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/27/2015] [Accepted: 12/01/2015] [Indexed: 01/15/2023]
Abstract
Cognitive impairment, or decline, is not only a feature of Alzheimer׳s disease and other forms of dementia but also normal ageing. Abundant evidence from epidemiological studies points towards perturbed inflammatory mechanisms in aged individuals, though the cause-effect nature of this apparent relationship is difficult to establish. Genetic association studies focusing on polymorphism in and around inflammatory genes represent a viable approach to establish whether inflammatory mechanisms might play a causal role in cognitive decline, whilst also enabling the identification of specific genes potentially influencing specific cognitive facets. Thus, here we provide a review of published genetic association studies investigating inflammatory genes in the context of cognitive decline in elderly, non-demented, samples. Numerous candidate gene association studies have been performed to date, focusing almost exclusively on genes encoding major cytokines. Some of these studies report significant cognitive domain-specific associations implicating Interleukin 1β (IL1β) (rs16944), Tumour Necrosis Factor α (TNFα) (rs1800629) and C-reactive protein (CRP) in various domains of cognitive function. However, the majority of these studies are lacking in statistical power and have other methodological limitations, suggesting some of them may have yielded false positive results. Genome-wide association studies have implicated less direct and less obvious regulators of inflammatory processes (i.e., PDE7A, HS3ST4, SPOCK3), indicating that a shift away from the major cytokine-encoding genes in future studies will be important. Furthermore, better cohesion across studies with regards to the cognitive test batteries administered to participants along with the continued application of longitudinal designs will be vital.
Collapse
|
30
|
Langlais D, Fodil N, Gros P. Genetics of Infectious and Inflammatory Diseases: Overlapping Discoveries from Association and Exome-Sequencing Studies. Annu Rev Immunol 2017; 35:1-30. [DOI: 10.1146/annurev-immunol-051116-052442] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- David Langlais
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec H3G 0B1, Canada;, ,
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Nassima Fodil
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec H3G 0B1, Canada;, ,
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Philippe Gros
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec H3G 0B1, Canada;, ,
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 0B1, Canada
| |
Collapse
|
31
|
Zimmermann MT, Kennedy RB, Grill DE, Oberg AL, Goergen KM, Ovsyannikova IG, Haralambieva IH, Poland GA. Integration of Immune Cell Populations, mRNA-Seq, and CpG Methylation to Better Predict Humoral Immunity to Influenza Vaccination: Dependence of mRNA-Seq/CpG Methylation on Immune Cell Populations. Front Immunol 2017; 8:445. [PMID: 28484452 PMCID: PMC5399034 DOI: 10.3389/fimmu.2017.00445] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/31/2017] [Indexed: 12/21/2022] Open
Abstract
The development of a humoral immune response to influenza vaccines occurs on a multisystems level. Due to the orchestration required for robust immune responses when multiple genes and their regulatory components across multiple cell types are involved, we examined an influenza vaccination cohort using multiple high-throughput technologies. In this study, we sought a more thorough understanding of how immune cell composition and gene expression relate to each other and contribute to interindividual variation in response to influenza vaccination. We first hypothesized that many of the differentially expressed (DE) genes observed after influenza vaccination result from changes in the composition of participants' peripheral blood mononuclear cells (PBMCs), which were assessed using flow cytometry. We demonstrated that DE genes in our study are correlated with changes in PBMC composition. We gathered DE genes from 128 other publically available PBMC-based vaccine studies and identified that an average of 57% correlated with specific cell subset levels in our study (permutation used to control false discovery), suggesting that the associations we have identified are likely general features of PBMC-based transcriptomics. Second, we hypothesized that more robust models of vaccine response could be generated by accounting for the interplay between PBMC composition, gene expression, and gene regulation. We employed machine learning to generate predictive models of B-cell ELISPOT response outcomes and hemagglutination inhibition (HAI) antibody titers. The top HAI and B-cell ELISPOT model achieved an area under the receiver operating curve (AUC) of 0.64 and 0.79, respectively, with linear model coefficients of determination of 0.08 and 0.28. For the B-cell ELISPOT outcomes, CpG methylation had the greatest predictive ability, highlighting potentially novel regulatory features important for immune response. B-cell ELISOT models using only PBMC composition had lower performance (AUC = 0.67), but highlighted well-known mechanisms. Our analysis demonstrated that each of the three data sets (cell composition, mRNA-Seq, and DNA methylation) may provide distinct information for the prediction of humoral immune response outcomes. We believe that these findings are important for the interpretation of current omics-based studies and set the stage for a more thorough understanding of interindividual immune responses to influenza vaccination.
Collapse
Affiliation(s)
- Michael T Zimmermann
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | | | - Diane E Grill
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Ann L Oberg
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Krista M Goergen
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Genome-wide associations of CD46 and IFI44L genetic variants with neutralizing antibody response to measles vaccine. Hum Genet 2017; 136:421-435. [PMID: 28289848 DOI: 10.1007/s00439-017-1768-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/14/2017] [Indexed: 12/27/2022]
Abstract
Population-based studies have revealed 2-10% measles vaccine failure rate even after two vaccine doses. While the mechanisms behind this remain unknown, we hypothesized that host genetic factors are likely to be involved. We performed a genome-wide association study of measles specific neutralizing antibody and IFNγ ELISPOT response in a combined sample of 2872 subjects. We identified two distinct chromosome 1 regions (previously associated with MMR-related febrile seizures), associated with vaccine-induced measles neutralizing antibody titers. The 1q32 region contained 20 significant SNPs in/around the measles virus receptor-encoding CD46 gene, including the intronic rs2724384 (p value = 2.64 × 10-09) and rs2724374 (p value = 3.16 × 10-09) SNPs. The 1q31.1 region contained nine significant SNPs in/around IFI44L, including the intronic rs1333973 (p value = 1.41 × 10-10) and the missense rs273259 (His73Arg, p value = 2.87 × 10-10) SNPs. Analysis of differential exon usage with mRNA-Seq data and RT-PCR suggests the involvement of rs2724374 minor G allele in the CD46 STP region exon B skipping, resulting in shorter CD46 isoforms. Our study reveals common CD46 and IFI44L SNPs associated with measles-specific humoral immunity, and highlights the importance of alternative splicing/virus cellular receptor isoform usage as a mechanism explaining inter-individual variation in immune response after live measles vaccine.
Collapse
|
33
|
Ovsyannikova IG, Schaid DJ, Larrabee BR, Haralambieva IH, Kennedy RB, Poland GA. A large population-based association study between HLA and KIR genotypes and measles vaccine antibody responses. PLoS One 2017; 12:e0171261. [PMID: 28158231 PMCID: PMC5291460 DOI: 10.1371/journal.pone.0171261] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/17/2017] [Indexed: 12/15/2022] Open
Abstract
Human antibody response to measles vaccine is highly variable in the population. Host genes contribute to inter-individual antibody response variation. The killer cell immunoglobulin-like receptors (KIR) are recognized to interact with HLA molecules and possibly influence humoral immune response to viral antigens. To expand on and improve our previous work with HLA genes, and to explore the genetic contribution of KIR genes to the inter-individual variability in measles vaccine-induced antibody responses, we performed a large population-based study in 2,506 healthy immunized subjects (ages 11 to 41 years) to identify HLA and KIR associations with measles vaccine-induced neutralizing antibodies. After correcting for the large number of statistical tests of allele effects on measles-specific neutralizing antibody titers, no statistically significant associations were found for either HLA or KIR loci. However, suggestive associations worthy of follow-up in other cohorts include B*57:01, DQB1*06:02, and DRB1*15:05 alleles. Specifically, the B*57:01 allele (1,040 mIU/mL; p = 0.0002) was suggestive of an association with lower measles antibody titer. In contrast, the DQB1*06:02 (1,349 mIU/mL; p = 0.0004) and DRB1*15:05 (2,547 mIU/mL; p = 0.0004) alleles were suggestive of an association with higher measles antibodies. Notably, the associations with KIR genotypes were strongly nonsignificant, suggesting that KIR loci in terms of copy number and haplotypes are not likely to play a major role in antibody response to measles vaccination. These findings refine our knowledge of the role of HLA and KIR alleles in measles vaccine-induced immunity.
Collapse
Affiliation(s)
- Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States of America
| | - Daniel J. Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Beth R. Larrabee
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States of America
| | - Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States of America
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
34
|
Pasanen A, Karjalainen MK, Bont L, Piippo-Savolainen E, Ruotsalainen M, Goksör E, Kumawat K, Hodemaekers H, Nuolivirta K, Jartti T, Wennergren G, Hallman M, Rämet M, Korppi M. Genome-Wide Association Study of Polymorphisms Predisposing to Bronchiolitis. Sci Rep 2017; 7:41653. [PMID: 28139761 PMCID: PMC5282585 DOI: 10.1038/srep41653] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/20/2016] [Indexed: 11/10/2022] Open
Abstract
Bronchiolitis is a major cause of hospitalization among infants. Severe bronchiolitis is associated with later asthma, suggesting a common genetic predisposition. Genetic background of bronchiolitis is not well characterized. To identify polymorphisms associated with bronchiolitis, we conducted a genome-wide association study (GWAS) in which 5,300,000 single nucleotide polymorphisms (SNPs) were tested for association in a Finnish–Swedish population of 217 children hospitalized for bronchiolitis and 778 controls. The most promising SNPs (n = 77) were genotyped in a Dutch replication population of 416 cases and 432 controls. Finally, we used a set of 202 Finnish bronchiolitis cases to further investigate candidate SNPs. We did not detect genome-wide significant associations, but several suggestive association signals (p < 10−5) were observed in the GWAS. In the replication population, three SNPs were nominally associated (p < 0.05). Of them, rs269094 was an expression quantitative trait locus (eQTL) for KCND3, previously shown to be associated with occupational asthma. In the additional set of Finnish cases, the association for another SNP (rs9591920) within a noncoding RNA locus was further strengthened. Our results provide a first genome-wide examination of the genetics underlying bronchiolitis. These preliminary findings require further validation in a larger sample size.
Collapse
Affiliation(s)
- Anu Pasanen
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Minna K Karjalainen
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Louis Bont
- Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Marja Ruotsalainen
- Kuopio University Hospital, Pediatrics, University of Eastern Finland, Kuopio, Finland
| | - Emma Goksör
- Department of Pediatrics, University of Gothenburg, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Kuldeep Kumawat
- Department of Immunology, Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hennie Hodemaekers
- RIVM, National Institute for Public Health and the Environment, GZB, Center for Health Protection, Bilthoven, The Netherlands
| | - Kirsi Nuolivirta
- Department of Pediatrics, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Tuomas Jartti
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Göran Wennergren
- Department of Pediatrics, University of Gothenburg, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Mikko Hallman
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Mika Rämet
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.,BioMediTech, University of Tampere, Tampere, Finland
| | - Matti Korppi
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
35
|
Bustamante M, Standl M, Bassat Q, Vilor-Tejedor N, Medina-Gomez C, Bonilla C, Ahluwalia TS, Bacelis J, Bradfield JP, Tiesler CMT, Rivadeneira F, Ring S, Vissing NH, Fink NR, Jugessur A, Mentch FD, Ballester F, Kriebel J, Kiefte-de Jong JC, Wolsk HM, Llop S, Thiering E, Beth SA, Timpson NJ, Andersen J, Schulz H, Jaddoe VWV, Evans DM, Waage J, Hakonarson H, Grant SFA, Jacobsson B, Bønnelykke K, Bisgaard H, Davey Smith G, Moll HA, Heinrich J, Estivill X, Sunyer J. A genome-wide association meta-analysis of diarrhoeal disease in young children identifies FUT2 locus and provides plausible biological pathways. Hum Mol Genet 2016; 25:4127-4142. [PMID: 27559109 PMCID: PMC5291237 DOI: 10.1093/hmg/ddw264] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/21/2016] [Accepted: 07/28/2016] [Indexed: 02/02/2023] Open
Abstract
More than a million childhood diarrhoeal episodes occur worldwide each year, and in developed countries a considerable part of them are caused by viral infections. In this study, we aimed to search for genetic variants associated with diarrhoeal disease in young children by meta-analyzing genome-wide association studies, and to elucidate plausible biological mechanisms. The study was conducted in the context of the Early Genetics and Lifecourse Epidemiology (EAGLE) consortium. Data about diarrhoeal disease in two time windows (around 1 year of age and around 2 years of age) was obtained via parental questionnaires, doctor interviews or medical records. Standard quality control and statistical tests were applied to the 1000 Genomes imputed genotypic data. The meta-analysis (N = 5758) followed by replication (N = 3784) identified a genome-wide significant association between rs8111874 and diarrhoea at age 1 year. Conditional analysis suggested that the causal variant could be rs601338 (W154X) in the FUT2 gene. Children with the A allele, which results in a truncated FUT2 protein, had lower risk of diarrhoea. FUT2 participates in the production of histo-blood group antigens and has previously been implicated in the susceptibility to infections, including Rotavirus and Norovirus Gene-set enrichment analysis suggested pathways related to the histo-blood group antigen production, and the regulation of ion transport and blood pressure. Among others, the gastrointestinal tract, and the immune and neuro-secretory systems were detected as relevant organs. In summary, this genome-wide association meta-analysis suggests the implication of the FUT2 gene in diarrhoeal disease in young children from the general population.
Collapse
Affiliation(s)
- Mariona Bustamante
- ISGlobal, Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Quique Bassat
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Natalia Vilor-Tejedor
- ISGlobal, Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Carolina Medina-Gomez
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Carolina Bonilla
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Tarunveer S Ahluwalia
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Bacelis
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonathan P Bradfield
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Carla M T Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Division of Metabolic Diseases and Nutritional Medicine, Munich, Germany
| | - Fernando Rivadeneira
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Susan Ring
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Nadja H Vissing
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nadia R Fink
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Astanand Jugessur
- Department of Genetics and Bioinformatics, Area of Health Data and Digitalisation, Institute of Public Health, Oslo, Norway
| | - Frank D Mentch
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ferran Ballester
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, València, Spain
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Jessica C Kiefte-de Jong
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
- Leiden University College, The Hague, The Netherlands
| | - Helene M Wolsk
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Sabrina Llop
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, València, Spain
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Division of Metabolic Diseases and Nutritional Medicine, Munich, Germany
| | - Systke A Beth
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | - Nicholas J Timpson
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Josefine Andersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | - David M Evans
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Johannes Waage
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F A Grant
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Area of Health Data and Digitalisation, Institute of Public Health, Oslo, Norway
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - George Davey Smith
- MRC/University of Bristol Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Henriette A Moll
- Department of Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Inner City Clinic, University Hospital of Munich (LMU), Munich, Germany
| | - Xavier Estivill
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Experimental Genetics, Sidra Medical and Research Centre, Doha, Qatar
| | - Jordi Sunyer
- ISGlobal, Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Pompeu Fabra University (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
36
|
Naviaux RK, Naviaux JC, Li K, Bright AT, Alaynick WA, Wang L, Baxter A, Nathan N, Anderson W, Gordon E. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci U S A 2016; 113:E5472-80. [PMID: 27573827 PMCID: PMC5027464 DOI: 10.1073/pnas.1607571113] [Citation(s) in RCA: 247] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
More than 2 million people in the United States have myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). We performed targeted, broad-spectrum metabolomics to gain insights into the biology of CFS. We studied a total of 84 subjects using these methods. Forty-five subjects (n = 22 men and 23 women) met diagnostic criteria for ME/CFS by Institute of Medicine, Canadian, and Fukuda criteria. Thirty-nine subjects (n = 18 men and 21 women) were age- and sex-matched normal controls. Males with CFS were 53 (±2.8) y old (mean ± SEM; range, 21-67 y). Females were 52 (±2.5) y old (range, 20-67 y). The Karnofsky performance scores were 62 (±3.2) for males and 54 (±3.3) for females. We targeted 612 metabolites in plasma from 63 biochemical pathways by hydrophilic interaction liquid chromatography, electrospray ionization, and tandem mass spectrometry in a single-injection method. Patients with CFS showed abnormalities in 20 metabolic pathways. Eighty percent of the diagnostic metabolites were decreased, consistent with a hypometabolic syndrome. Pathway abnormalities included sphingolipid, phospholipid, purine, cholesterol, microbiome, pyrroline-5-carboxylate, riboflavin, branch chain amino acid, peroxisomal, and mitochondrial metabolism. Area under the receiver operator characteristic curve analysis showed diagnostic accuracies of 94% [95% confidence interval (CI), 84-100%] in males using eight metabolites and 96% (95% CI, 86-100%) in females using 13 metabolites. Our data show that despite the heterogeneity of factors leading to CFS, the cellular metabolic response in patients was homogeneous, statistically robust, and chemically similar to the evolutionarily conserved persistence response to environmental stress known as dauer.
Collapse
Affiliation(s)
- Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA 92103-8467;
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA 92103-8467
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA 92103-8467
| | - A Taylor Bright
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA 92103-8467
| | - William A Alaynick
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA 92103-8467
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA 92103-8467; Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA 92103-8467
| | - Asha Baxter
- Gordon Medical Associates, Santa Rosa, CA 95403
| | - Neil Nathan
- Gordon Medical Associates, Santa Rosa, CA 95403
| | | | - Eric Gordon
- Gordon Medical Associates, Santa Rosa, CA 95403
| |
Collapse
|
37
|
Voigt EA, Ovsyannikova IG, Haralambieva IH, Kennedy RB, Larrabee BR, Schaid DJ, Poland GA. Genetically defined race, but not sex, is associated with higher humoral and cellular immune responses to measles vaccination. Vaccine 2016; 34:4913-4919. [PMID: 27591105 DOI: 10.1016/j.vaccine.2016.08.060] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022]
Abstract
In addition to host genetic and environmental factors, variations in immune responses to vaccination are influenced by demographic variables, such as race and sex. The influence of genetic race and sex on measles vaccine responses is not well understood, yet important for the development of much-needed improved measles vaccines with lower failure rates. We assessed associations between genetically defined race and sex with measles humoral and cellular immunity after measles vaccination in three independent and geographically distinct cohorts totaling 2872 healthy racially diverse children, older adolescents, and young adults. We found no associations between biological sex and either humoral or cellular immunity to measles vaccine, and no correlation between humoral and cellular immunity in these study subjects. Genetically defined race was, however, significantly associated with both measles vaccine-induced humoral and cellular immune responses, with subjects genetically classified as having African-American ancestry demonstrating significantly higher antibody and cell-mediated immune responses relative to subjects of Caucasian ancestry. This information may be useful in designing novel measles vaccines that are optimally effective across human genetic backgrounds.
Collapse
Affiliation(s)
- Emily A Voigt
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | - Beth R Larrabee
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
38
|
Statistical Methods for Testing Genetic Pleiotropy. Genetics 2016; 204:483-497. [PMID: 27527515 DOI: 10.1534/genetics.116.189308] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/11/2016] [Indexed: 12/28/2022] Open
Abstract
Genetic pleiotropy is when a single gene influences more than one trait. Detecting pleiotropy and understanding its causes can improve the biological understanding of a gene in multiple ways, yet current multivariate methods to evaluate pleiotropy test the null hypothesis that none of the traits are associated with a variant; departures from the null could be driven by just one associated trait. A formal test of pleiotropy should assume a null hypothesis that one or no traits are associated with a genetic variant. For the special case of two traits, one can construct this null hypothesis based on the intersection-union (IU) test, which rejects the null hypothesis only if the null hypotheses of no association for both traits are rejected. To allow for more than two traits, we developed a new likelihood-ratio test for pleiotropy. We then extended the testing framework to a sequential approach to test the null hypothesis that [Formula: see text] traits are associated, given that the null of k traits are associated was rejected. This provides a formal testing framework to determine the number of traits associated with a genetic variant, while accounting for correlations among the traits. By simulations, we illustrate the type I error rate and power of our new methods; describe how they are influenced by sample size, the number of traits, and the trait correlations; and apply the new methods to multivariate immune phenotypes in response to smallpox vaccination. Our new approach provides a quantitative assessment of pleiotropy, enhancing current analytic practice.
Collapse
|
39
|
Kennedy RB, Poland GA, Ovsyannikova IG, Oberg AL, Asmann YW, Grill DE, Vierkant RA, Jacobson RM. Impaired innate, humoral, and cellular immunity despite a take in smallpox vaccine recipients. Vaccine 2016; 34:3283-90. [PMID: 27177944 PMCID: PMC5528000 DOI: 10.1016/j.vaccine.2016.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 11/29/2022]
Abstract
Smallpox vaccine is highly effective, inducing protective immunity to smallpox and diseases caused by related orthopoxviruses. Smallpox vaccine efficacy was historically defined by the appearance of a lesion or "take" at the vaccine site, which leaves behind a characteristic scar. Both the take and scar are readily recognizable and were used during the eradication effort to indicate successful vaccination and to categorize individuals as "protected." However, the development of a typical vaccine take may not equate to the successful development of a robust, protective immune response. In this report, we examined two large (>1000) cohorts of recipients of either Dryvax(®) or ACAM2000 using a testing and replication study design and identified subgroups of individuals who had documented vaccine takes, but who failed to develop robust neutralizing antibody titers. Examination of these individuals revealed that they had suboptimal cellular immune responses as well. Further testing indicated these low responders had a diminished innate antiviral gene expression pattern (IFNA1, CXCL10, CXCL11, OASL) upon in vitro stimulation with vaccinia virus, perhaps indicative of a dysregulated innate response. Our results suggest that poor activation of innate antiviral pathways may result in suboptimal immune responses to the smallpox vaccine. These genes and pathways may serve as suitable targets for adjuvants in new attenuated smallpox vaccines and/or effective antiviral therapy targets against poxvirus infections.
Collapse
Affiliation(s)
- Richard B Kennedy
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gregory A Poland
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Inna G Ovsyannikova
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Diane E Grill
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Robert A Vierkant
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Robert M Jacobson
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
40
|
Do C, Lang C, Lin J, Darbary H, Krupska I, Gaba A, Petukhova L, Vonsattel JP, Gallagher M, Goland R, Clynes R, Dwork A, Kral J, Monk C, Christiano A, Tycko B. Mechanisms and Disease Associations of Haplotype-Dependent Allele-Specific DNA Methylation. Am J Hum Genet 2016; 98:934-955. [PMID: 27153397 DOI: 10.1016/j.ajhg.2016.03.027] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/25/2016] [Indexed: 10/21/2022] Open
Abstract
Haplotype-dependent allele-specific methylation (hap-ASM) can impact disease susceptibility, but maps of this phenomenon using stringent criteria in disease-relevant tissues remain sparse. Here we apply array-based and Methyl-Seq approaches to multiple human tissues and cell types, including brain, purified neurons and glia, T lymphocytes, and placenta, and identify 795 hap-ASM differentially methylated regions (DMRs) and 3,082 strong methylation quantitative trait loci (mQTLs), most not previously reported. More than half of these DMRs have cell type-restricted ASM, and among them are 188 hap-ASM DMRs and 933 mQTLs located near GWAS signals for immune and neurological disorders. Targeted bis-seq confirmed hap-ASM in 12/13 loci tested, including CCDC155, CD69, FRMD1, IRF1, KBTBD11, and S100A(∗)-ILF2, associated with immune phenotypes, MYT1L, PTPRN2, CMTM8 and CELF2, associated with neurological disorders, NGFR and HLA-DRB6, associated with both immunological and brain disorders, and ZFP57, a trans-acting regulator of genomic imprinting. Polymorphic CTCF and transcription factor (TF) binding sites were over-represented among hap-ASM DMRs and mQTLs, and analysis of the human data, supplemented by cross-species comparisons to macaques, indicated that CTCF and TF binding likelihood predicts the strength and direction of the allelic methylation asymmetry. These results show that hap-ASM is highly tissue specific; an important trans-acting regulator of genomic imprinting is regulated by this phenomenon; and variation in CTCF and TF binding sites is an underlying mechanism, and maps of hap-ASM and mQTLs reveal regulatory sequences underlying supra- and sub-threshold GWAS peaks in immunological and neurological disorders.
Collapse
|
41
|
Parolo S, Lisa A, Gentilini D, Di Blasio AM, Barlera S, Nicolis EB, Boncoraglio GB, Parati EA, Bione S. Characterization of the biological processes shaping the genetic structure of the Italian population. BMC Genet 2015; 16:132. [PMID: 26553317 PMCID: PMC4640365 DOI: 10.1186/s12863-015-0293-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022] Open
Abstract
Background The genetic structure of human populations is the outcome of the combined action of different processes such as demographic dynamics and natural selection. Several efforts toward the characterization of population genetic architectures and the identification of adaptation signatures were recently made. In this study, we provide a genome-wide depiction of the Italian population structure and the analysis of the major determinants of the current existing genetic variation. Results We defined and characterized 210 genomic loci associated with the first Principal Component calculated on the Italian genotypic data and correlated to the North–south genetic gradient. Using a gene-enrichment approach we identified the immune function as primarily involved in the Italian population differentiation and we described a locus on chromosome 13 showing combined evidence of North–south diversification in allele frequencies and signs of recent positive selection. In this region our bioinformatics analysis pinpointed an uncharacterized long intergenic non-coding (lincRNA), whose expression appeared specific for immune-related tissues suggesting its relevance for the immune function. Conclusions Our study, combining population genetic analyses with biological insights provides a description of the Italian genetic structure that in future could contribute to the evaluation of complex diseases risk in the population context. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0293-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silvia Parolo
- Computational Biology Unit, Institute of Molecular Genetics-National Research Council, Pavia, Italy.
| | - Antonella Lisa
- Computational Biology Unit, Institute of Molecular Genetics-National Research Council, Pavia, Italy.
| | - Davide Gentilini
- Molecular Biology Laboratory, Istituto Auxologico Italiano, Milan, Italy.
| | | | - Simona Barlera
- Department of Cardiovascular Research, IRCCS Mario Negri Institute for Pharmacological Research, Milan, Italy.
| | - Enrico B Nicolis
- Department of Cardiovascular Research, IRCCS Mario Negri Institute for Pharmacological Research, Milan, Italy.
| | - Giorgio B Boncoraglio
- Department of Cerebrovascular Diseases, IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Eugenio A Parati
- Department of Cerebrovascular Diseases, IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Silvia Bione
- Computational Biology Unit, Institute of Molecular Genetics-National Research Council, Pavia, Italy.
| |
Collapse
|
42
|
Simon WL, Salk HM, Ovsyannikova IG, Kennedy RB, Poland GA. Cytokine production associated with smallpox vaccine responses. Immunotherapy 2015; 6:1097-112. [PMID: 25428648 DOI: 10.2217/imt.14.72] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Smallpox was eradicated 34 years ago due to the success of the smallpox vaccine; yet, the vaccine continues to be studied because of its importance in responding to potential biological warfare and the adverse events associated with current smallpox vaccines. Interindividual variations in vaccine response are observed and are, in part, due to genetic variation. In some cases, these varying responses lead to adverse events, which occur at a relatively high rate for the smallpox vaccine compared with other vaccines. Here, we aim to summarize the cytokine responses associated with smallpox vaccine response to date. Along with a description of each of these cytokines, we describe the genetic and adverse event data associated with cytokine responses to smallpox vaccination.
Collapse
Affiliation(s)
- Whitney L Simon
- Mayo Vaccine Research Group, Mayo Clinic, Guggenheim 611C, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
43
|
Fine Mapping Causal Variants with an Approximate Bayesian Method Using Marginal Test Statistics. Genetics 2015; 200:719-36. [PMID: 25948564 DOI: 10.1534/genetics.115.176107] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/04/2015] [Indexed: 01/08/2023] Open
Abstract
Two recently developed fine-mapping methods, CAVIAR and PAINTOR, demonstrate better performance over other fine-mapping methods. They also have the advantage of using only the marginal test statistics and the correlation among SNPs. Both methods leverage the fact that the marginal test statistics asymptotically follow a multivariate normal distribution and are likelihood based. However, their relationship with Bayesian fine mapping, such as BIMBAM, is not clear. In this study, we first show that CAVIAR and BIMBAM are actually approximately equivalent to each other. This leads to a fine-mapping method using marginal test statistics in the Bayesian framework, which we call CAVIAR Bayes factor (CAVIARBF). Another advantage of the Bayesian framework is that it can answer both association and fine-mapping questions. We also used simulations to compare CAVIARBF with other methods under different numbers of causal variants. The results showed that both CAVIARBF and BIMBAM have better performance than PAINTOR and other methods. Compared to BIMBAM, CAVIARBF has the advantage of using only marginal test statistics and takes about one-quarter to one-fifth of the running time. We applied different methods on two independent cohorts of the same phenotype. Results showed that CAVIARBF, BIMBAM, and PAINTOR selected the same top 3 SNPs; however, CAVIARBF and BIMBAM had better consistency in selecting the top 10 ranked SNPs between the two cohorts. Software is available at https://bitbucket.org/Wenan/caviarbf.
Collapse
|
44
|
Debette S, Ibrahim Verbaas CA, Bressler J, Schuur M, Smith A, Bis JC, Davies G, Wolf C, Gudnason V, Chibnik LB, Yang Q, deStefano AL, de Quervain DJF, Srikanth V, Lahti J, Grabe HJ, Smith JA, Priebe L, Yu L, Karbalai N, Hayward C, Wilson JF, Campbell H, Petrovic K, Fornage M, Chauhan G, Yeo R, Boxall R, Becker J, Stegle O, Mather KA, Chouraki V, Sun Q, Rose LM, Resnick S, Oldmeadow C, Kirin M, Wright AF, Jonsdottir MK, Au R, Becker A, Amin N, Nalls MA, Turner ST, Kardia SLR, Oostra B, Windham G, Coker LH, Zhao W, Knopman DS, Heiss G, Griswold ME, Gottesman RF, Vitart V, Hastie ND, Zgaga L, Rudan I, Polasek O, Holliday EG, Schofield P, Choi SH, Tanaka T, An Y, Perry RT, Kennedy RE, Sale MM, Wang J, Wadley VG, Liewald DC, Ridker PM, Gow AJ, Pattie A, Starr JM, Porteous D, Liu X, Thomson R, Armstrong NJ, Eiriksdottir G, Assareh AA, Kochan NA, Widen E, Palotie A, Hsieh YC, Eriksson JG, Vogler C, van Swieten JC, Shulman JM, Beiser A, Rotter J, Schmidt CO, Hoffmann W, Nöthen MM, Ferrucci L, Attia J, Uitterlinden AG, Amouyel P, Dartigues JF, Amieva H, Räikkönen K, Garcia M, et alDebette S, Ibrahim Verbaas CA, Bressler J, Schuur M, Smith A, Bis JC, Davies G, Wolf C, Gudnason V, Chibnik LB, Yang Q, deStefano AL, de Quervain DJF, Srikanth V, Lahti J, Grabe HJ, Smith JA, Priebe L, Yu L, Karbalai N, Hayward C, Wilson JF, Campbell H, Petrovic K, Fornage M, Chauhan G, Yeo R, Boxall R, Becker J, Stegle O, Mather KA, Chouraki V, Sun Q, Rose LM, Resnick S, Oldmeadow C, Kirin M, Wright AF, Jonsdottir MK, Au R, Becker A, Amin N, Nalls MA, Turner ST, Kardia SLR, Oostra B, Windham G, Coker LH, Zhao W, Knopman DS, Heiss G, Griswold ME, Gottesman RF, Vitart V, Hastie ND, Zgaga L, Rudan I, Polasek O, Holliday EG, Schofield P, Choi SH, Tanaka T, An Y, Perry RT, Kennedy RE, Sale MM, Wang J, Wadley VG, Liewald DC, Ridker PM, Gow AJ, Pattie A, Starr JM, Porteous D, Liu X, Thomson R, Armstrong NJ, Eiriksdottir G, Assareh AA, Kochan NA, Widen E, Palotie A, Hsieh YC, Eriksson JG, Vogler C, van Swieten JC, Shulman JM, Beiser A, Rotter J, Schmidt CO, Hoffmann W, Nöthen MM, Ferrucci L, Attia J, Uitterlinden AG, Amouyel P, Dartigues JF, Amieva H, Räikkönen K, Garcia M, Wolf PA, Hofman A, Longstreth WT, Psaty BM, Boerwinkle E, DeJager PL, Sachdev PS, Schmidt R, Breteler MMB, Teumer A, Lopez OL, Cichon S, Chasman DI, Grodstein F, Müller-Myhsok B, Tzourio C, Papassotiropoulos A, Bennett DA, Ikram MA, Deary IJ, van Duijn CM, Launer L, Fitzpatrick AL, Seshadri S, Mosley TH. Genome-wide studies of verbal declarative memory in nondemented older people: the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium. Biol Psychiatry 2015; 77:749-63. [PMID: 25648963 PMCID: PMC4513651 DOI: 10.1016/j.biopsych.2014.08.027] [Show More Authors] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 08/23/2014] [Accepted: 08/25/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Memory performance in older persons can reflect genetic influences on cognitive function and dementing processes. We aimed to identify genetic contributions to verbal declarative memory in a community setting. METHODS We conducted genome-wide association studies for paragraph or word list delayed recall in 19 cohorts from the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium, comprising 29,076 dementia- and stroke-free individuals of European descent, aged ≥45 years. Replication of suggestive associations (p < 5 × 10(-6)) was sought in 10,617 participants of European descent, 3811 African-Americans, and 1561 young adults. RESULTS rs4420638, near APOE, was associated with poorer delayed recall performance in discovery (p = 5.57 × 10(-10)) and replication cohorts (p = 5.65 × 10(-8)). This association was stronger for paragraph than word list delayed recall and in the oldest persons. Two associations with specific tests, in subsets of the total sample, reached genome-wide significance in combined analyses of discovery and replication (rs11074779 [HS3ST4], p = 3.11 × 10(-8), and rs6813517 [SPOCK3], p = 2.58 × 10(-8)) near genes involved in immune response. A genetic score combining 58 independent suggestive memory risk variants was associated with increasing Alzheimer disease pathology in 725 autopsy samples. Association of memory risk loci with gene expression in 138 human hippocampus samples showed cis-associations with WDR48 and CLDN5, both related to ubiquitin metabolism. CONCLUSIONS This largest study to date exploring the genetics of memory function in ~40,000 older individuals revealed genome-wide associations and suggested an involvement of immune and ubiquitin pathways.
Collapse
Affiliation(s)
- Stéphanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux; Department of Neurology, University Hospital of Bordeaux, Bordeaux, France.
| | - Carla A Ibrahim Verbaas
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Neurology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Maaike Schuur
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Neurology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Albert Smith
- Icelandic Heart Association, Kopavogur; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Gail Davies
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh; Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Lori B Chibnik
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Anita L deStefano
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Dominique J F de Quervain
- Psychiatric University Clinics and Department of Psychology, Division of Cognitive Neuroscience, University of Basel, Basel, Switzerland
| | - Velandai Srikanth
- Stroke and Ageing Research Centre, Southern Clinical School, Department of Medicine, Monash University, Melbourne; Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki; Folkhälsan Research Centre, Helsinki, Finland
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, HELIOS-Hospital Stralsund, Stralsund; German Center for Neurodegenerative Diseases, Site Rostock/ Greifswald, Rostock, Germany
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Lutz Priebe
- Institute of Human Genetics, Universitätsklinikum Bonn, Bonn, Germany
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | | | | | - James F Wilson
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | - Harry Campbell
- Division of General Neurology, Department of Neurology, Medical University and General Hospital of Graz, Austria
| | - Katja Petrovic
- Division of General Neurology, Department of Neurology, Medical University and General Hospital of Graz, Austria
| | - Myriam Fornage
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas; Institute of Molecular Medicine, University of Texas-Houston Health Science Center, Houston, Texas
| | - Ganesh Chauhan
- Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux
| | - Robin Yeo
- Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux
| | - Ruth Boxall
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh
| | - James Becker
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Psychology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Oliver Stegle
- Max Planck Institute for Intelligent Systems, Tübingen, Germany; Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia
| | - Vincent Chouraki
- Institut National de la Santé et de la Recherche Médicale Unit 744, Institut Pasteur de Lille, and Université Lille Nord de France, Lille, France
| | - Qi Sun
- Department of Nutrition, Harvard School of Public Health; Channing Division of Network Medicine, Department of Medicine, Brigham and Women׳s Hospital and Harvard Medical School; Harvard Medical School, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Lynda M Rose
- Division of Preventive Medicine, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Susan Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Christopher Oldmeadow
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia; Public Health, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Mirna Kirin
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | - Alan F Wright
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | | | - Rhoda Au
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Albert Becker
- Institute of Neuropathology, Universitätsklinikum Bonn, Bonn, Germany
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Mike A Nalls
- Molecular Genetics Section , Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Stephen T Turner
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Ben Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Clinical Genetics, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Gwen Windham
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Laura H Coker
- Division of Public Health Sciences and Neurology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | | | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael E Griswold
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rebecca F Gottesman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | - Lina Zgaga
- Medical Research Council Human Genetics Unit
| | - Igor Rudan
- Institute of Genetics and Molecular Medicine, University of Edinburgh, United Kingdom, Centre for Population Health Sciences, University of Edinburgh, United Kingdom
| | - Ozren Polasek
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia
| | - Elizabeth G Holliday
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia
| | - Peter Schofield
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia; Department of Medicine, John Hunter Hospital, Newcastle, Australia
| | - Seung Hoan Choi
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Yang An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Rodney T Perry
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard E Kennedy
- Division of Gerontology, Geriatrics, and Palliative Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Michèle M Sale
- Center for Public Health Genomics, Department of Medicine, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Jing Wang
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Virginia G Wadley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David C Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh
| | - Paul M Ridker
- Harvard Medical School, Brigham and Women׳s Hospital, Boston, Massachusetts; Division of Preventive Medicine, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Alan J Gow
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh
| | - Alison Pattie
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - David Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Medical Genetics Section, Molecular Medicine Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Xuan Liu
- Department of Biostatistics, Boston University School of Public Health, Boston
| | - Russell Thomson
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Australia
| | - Nicola J Armstrong
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst; School of Mathematics & Statistics and Prince of Wales Clinical School, University of New South Wales, Sydney
| | | | - Arezoo A Assareh
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Neuroscience Research Australia and Primary Dementia Collaborative Research Centre-Assessment and Better Care, University of New South Wales, Sydney
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick New South Wales, Australia
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Finland; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom; Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Yi-Chen Hsieh
- Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Johan G Eriksson
- Folkhälsan Research Centre, Helsinki, Finland; National Institute for Health and Welfare, Helsinki, Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Helsinki University Central Hospital, Unit of General Practice, Helsinki, Vasa Central Hospital, Vasa, Finland
| | - Christian Vogler
- Psychiatric University Clinics and Department of Psychology, Division of Molecular Neuroscience, University of Basel, Basel, Switzerland
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Joshua M Shulman
- Departments of Neurology and Molecular and Human Genetics, Baylor College of Medicine and The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
| | - Alexa Beiser
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Jerome Rotter
- Institute for Translational Genomics and Populaton Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California
| | | | - Wolfgang Hoffmann
- German Center for Neurodegenerative Diseases, Site Rostock/ Greifswald, Rostock, Germany; Section Epidemiology of Health Care and Community Health, Greifswald
| | - Markus M Nöthen
- Institute of Human Genetics, Department of Genomics, Life & Brain Research Center, University of Bonn, Bonn; German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - John Attia
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine, Newcastle, Australia; Public Health, Faculty of Health, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia; Department of Medicine, John Hunter Hospital, Newcastle, Australia
| | - Andre G Uitterlinden
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Internal Medicine, Erasmus Medical Center University Medical Center, Rotterdam, the Netherlands
| | - Philippe Amouyel
- Institut National de la Santé et de la Recherche Médicale Unit 744, Institut Pasteur de Lille, and Université Lille Nord de France, Lille, France; Centre Hospitalier Régional Universitaire de Lille, Lille
| | - Jean-François Dartigues
- Institut National de la Santé et de la Recherche Médicale, Bordeaux University, Talence, France
| | - Hélène Amieva
- Institut National de la Santé et de la Recherche Médicale, Bordeaux University, Talence, France
| | | | - Melissa Garcia
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland
| | - Philip A Wolf
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Albert Hofman
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - W T Longstreth
- Departments of Neurology, University of Washington; Epidemiology, University of Washington
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington; Epidemiology, University of Washington; Health Services, University of Washington; Group Health Research Institute, Group Health Cooperative, Seattle, Washington
| | - Eric Boerwinkle
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas
| | - Philip L DeJager
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales Medicine, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick New South Wales, Australia
| | - Reinhold Schmidt
- Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Monique M B Breteler
- German Center for Neurodegenerative Diseases, Site Rostock/ Greifswald, Rostock, Germany; Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Population Health Sciences, University of Bonn, Bonn, Germany; Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, Massachusetts
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; The Alzheimer׳s Disease Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sven Cichon
- Institute of Human Genetics, Universitätsklinikum Bonn, Bonn, Germany; Institute of Neuroscience and Medicine, Research Center Julich, Julich, Germany; Division of Medical Genetics, Department of Biomedicine, University of Basel, Switzerland
| | - Daniel I Chasman
- Harvard Medical School, Brigham and Women׳s Hospital, Boston, Massachusetts; Division of Preventive Medicine, Brigham and Women׳s Hospital, Boston, Massachusetts
| | - Francine Grodstein
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women׳s Hospital and Harvard Medical School; Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, Massachusetts
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christophe Tzourio
- Institut National de la Santé et de la Recherche Médicale, Epidemiology, University of Bordeaux; University Bordeaux Segalen, Bordeaux, France
| | - Andreas Papassotiropoulos
- Psychiatric University Clinics and Department of Psychology, Division of Molecular Neuroscience, University of Basel, Basel, Switzerland; Department Biozentrum, Life Sciences Training Facility, Basel, Switzerland
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - M Arfan Ikram
- Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh; Department of Psychology, The University of Edinburgh
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center University Medical Center, Rotterdam, The Netherlands; Netherlands Consortium for Healthy Ageing, Leiden, the Netherlands; Center for Medical Systems Biology, Netherlands Genomics Initiative, Leiden University Medical Center, Leiden, The Netherlands
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, Maryland
| | | | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston; The National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, Massachusetts
| | - Thomas H Mosley
- Department of Medicine and Neurology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
45
|
Li D, Zhao H, Kranzler HR, Li MD, Jensen KP, Zayats T, Farrer LA, Gelernter J. Genome-wide association study of copy number variations (CNVs) with opioid dependence. Neuropsychopharmacology 2015; 40:1016-26. [PMID: 25345593 PMCID: PMC4330517 DOI: 10.1038/npp.2014.290] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 08/18/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022]
Abstract
Single-nucleotide polymorphisms that have been associated with opioid dependence (OD) altogether account for only a small proportion of the known heritability. Most of the genetic risk factors are unknown. Some of the 'missing heritability' might be explained by copy number variations (CNVs) in the human genome. We used Illumina HumanOmni1 arrays to genotype 5152 African-American and European-American OD cases and screened controls and implemented combined CNV calling methods. After quality control measures were applied, a genome-wide association study (GWAS) of CNVs with OD was performed. For common CNVs, two deletions and one duplication were significantly associated with OD genome-wide (eg, P=2 × 10(-8) and OR (95% CI)=0.64 (0.54-0.74) for a chromosome 18q12.3 deletion). Several rare or unique CNVs showed suggestive or marginal significance with large effect sizes. This study is the first GWAS of OD using CNVs. Some identified CNVs harbor genes newly identified here to be of biological importance in addiction, whereas others affect genes previously known to contribute to substance dependence risk. Our findings augment our specific knowledge of the importance of genomic variation in addictive disorders, and provide an addiction CNV pool for further research. These findings require replication.
Collapse
Affiliation(s)
- Dawei Li
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, USA
- Department of Computer Science, University of Vermont, Burlington, VT, USA
- Neuroscience, Behavior, and Health Initiative, University of Vermont, Burlington, VT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania School of Medicine and VISN 4 MIRECC, Philadelphia VAMC, Philadelphia, PA, USA
| | - Ming D Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA
| | - Kevin P Jensen
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Tetyana Zayats
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
| | - Lindsay A Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Genetics and Genomics, Biostatistics, and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Joel Gelernter
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
- VA Connecticut Healthcare Center, Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
46
|
Newport MJ. The genetic regulation of infant immune responses to vaccination. Front Immunol 2015; 6:18. [PMID: 25699041 PMCID: PMC4313718 DOI: 10.3389/fimmu.2015.00018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/09/2015] [Indexed: 12/16/2022] Open
Abstract
A number of factors are recognized to influence immune responses to vaccinations including age, gender, the dose, and quality of the antigen used, the number of doses given, the route of administration, and the nutritional status of the recipient. Additionally, several immunogenetic studies have identified associations between polymorphisms in genes encoding immune response proteins, both innate and adaptive, and variation in responses to vaccines. Variants in the genes encoding Toll-like receptors, HLA molecules, cytokines, and cytokine receptors have associated with heterogeneity of responses to a wide range of vaccines including measles, hepatitis B, influenza A, BCG, Haemophilus influenzae type b, and certain Neisseria meningitidis serotypes, amongst others. However, the vast majority of these studies have been conducted in older children and adults and there are very few data available from studies conducted in infants. This paper reviews the evidence to date that host genes influencing vaccines responses in these older population and identifies a large gap in our understanding of the genetic regulation of responses in early life. Given the high mortality from infection in early life and the challenges of developing vaccines that generate effective immune responses in the context of the developing immune system further research on infant populations is required.
Collapse
Affiliation(s)
- Melanie J. Newport
- Division of Clinical Medicine, Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
47
|
Peprah E, Xu H, Tekola-Ayele F, Royal CD. Genome-wide association studies in Africans and African Americans: expanding the framework of the genomics of human traits and disease. Public Health Genomics 2014; 18:40-51. [PMID: 25427668 DOI: 10.1159/000367962] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/29/2014] [Indexed: 01/11/2023] Open
Abstract
Genomic research is one of the tools for elucidating the pathogenesis of diseases of global health relevance and paving the research dimension to clinical and public health translation. Recent advances in genomic research and technologies have increased our understanding of human diseases, genes associated with these disorders, and the relevant mechanisms. Genome-wide association studies (GWAS) have proliferated since the first studies were published several years ago and have become an important tool in helping researchers comprehend human variation and the role genetic variants play in disease. However, the need to expand the diversity of populations in GWAS has become increasingly apparent as new knowledge is gained about genetic variation. Inclusion of diverse populations in genomic studies is critical to a more complete understanding of human variation and elucidation of the underpinnings of complex diseases. In this review, we summarize the available data on GWAS in recent African ancestry populations within the western hemisphere (i.e. African Americans and peoples of the Caribbean) and continental African populations. Furthermore, we highlight ways in which genomic studies in populations of recent African ancestry have led to advances in the areas of malaria, HIV, prostate cancer, and other diseases. Finally, we discuss the advantages of conducting GWAS in recent African ancestry populations in the context of addressing existing and emerging global health conditions.
Collapse
|
48
|
Boon ACM, Williams RW, Sinasac DS, Webby RJ. A novel genetic locus linked to pro-inflammatory cytokines after virulent H5N1 virus infection in mice. BMC Genomics 2014; 15:1017. [PMID: 25418976 PMCID: PMC4256927 DOI: 10.1186/1471-2164-15-1017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 10/22/2014] [Indexed: 12/15/2022] Open
Abstract
Background Genetic variation in the human population is a key determinant of influenza disease severity. A single nucleotide polymorphism in the antiviral gene IFITM3 was linked to outcomes during the 2009 H1N1 pandemic. To identify variant host genes associated with increased virus replication and severe disease, we performed a quantitative trait locus analysis on pro-inflammatory cytokine production 48 hours after intranasal infection with highly pathogenic H5N1 influenza virus. Results Pro-inflammatory cytokines CCL2, TNFα and IFN-α, were measured by ELISA in lung homogenates of DBA/2J (D2), C57BL/6J (B6) and 44 different BXD recombinant inbred mouse strains. Virus titer was also assessed in a subset of these animals. CCL2 (8-fold), TNFα (24-fold) and IFN-α (8-fold) concentrations varied significantly among the different BXD RI strains. Importantly, cytokine concentration correlated very well (r =0.86-0.96, P <0.0001) with virus titer suggesting that early cytokine production is due to increased virus infection and replication. Linkage analysis of cytokine concentration revealed a significant locus on chromosome 6 associated with differences in TNFα, IFN-α and CCL2 cytokine concentration (LRS =26). This locus accounted for nearly 20% of the observed phenotypic variation in the BXD population studied. Sequence and RNA expression analysis identified several candidate host genes containing missense mutations or deletions; Samd9l, Ica1, and Slc25a13. To study the role of Slc25a13, we obtained Slc25a13 knockout line, but upon challenge with H5N1 influenza virus observed no effect on CCL2 production, or morbidity and mortality. Conclusion A novel genetic locus on chromosome 6 modulates early pro-inflammatory cytokine production and virus replication after highly pathogenic influenza virus infection. Candidate genes, Samd9l and Ica1, may be important for the control of influenza virus infection and pathogenesis. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1017) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adrianus C M Boon
- Departments of Internal Medicine, Division of Infectious Diseases, Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
49
|
Lambert ND, Haralambieva IH, Kennedy RB, Ovsyannikova IG, Pankratz VS, Poland GA. Polymorphisms in HLA-DPB1 are associated with differences in rubella virus-specific humoral immunity after vaccination. J Infect Dis 2014; 211:898-905. [PMID: 25293367 DOI: 10.1093/infdis/jiu553] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Vaccination with live attenuated rubella virus induces a strong immune response in most individuals. However, small numbers of subjects never reach or maintain protective antibody levels, and there is a high degree of variability in immune response. We have previously described genetic polymorphisms in HLA and other candidate genes that are associated with interindividual differences in humoral immunity to rubella virus. To expand our previous work, we performed a genome-wide association study (GWAS) to discover single-nucleotide polymorphisms (SNPs) associated with rubella virus-specific neutralizing antibodies. We identified rs2064479 in the HLA-DPB1 genetic region as being significantly associated with humoral immune response variations after rubella vaccination (P = 8.62 × 10(-8)). All other significant SNPs in this GWAS were located near the HLA-DPB1 gene (P ≤ 1 × 10(-7)). These findings demonstrate that polymorphisms in HLA-DPB1 are strongly associated with interindividual differences in neutralizing antibody levels to rubella vaccination and represent a validation of our previous HLA work.
Collapse
Affiliation(s)
| | | | | | | | | | - Gregory A Poland
- Mayo Vaccine Research Group Program in Translational Immunovirology and Biodefense
| |
Collapse
|
50
|
Wu C, Wang Z, Song X, Feng XS, Abnet CC, He J, Hu N, Zuo XB, Tan W, Zhan Q, Hu Z, He Z, Jia W, Zhou Y, Yu K, Shu XO, Yuan JM, Zheng W, Zhao XK, Gao SG, Yuan ZQ, Zhou FY, Fan ZM, Cui JL, Lin HL, Han XN, Li B, Chen X, Dawsey SM, Liao L, Lee MP, Ding T, Qiao YL, Liu Z, Liu Y, Yu D, Chang J, Wei L, Gao YT, Koh WP, Xiang YB, Tang ZZ, Fan JH, Han JJ, Zhou SL, Zhang P, Zhang DY, Yuan Y, Huang Y, Liu C, Zhai K, Qiao Y, Jin G, Guo C, Fu J, Miao X, Lu C, Yang H, Wang C, Wheeler WA, Gail M, Yeager M, Yuenger J, Guo ET, Li AL, Zhang W, Li XM, Sun LD, Ma BG, Li Y, Tang S, Peng XQ, Liu J, Hutchinson A, Jacobs K, Giffen C, Burdette L, Fraumeni JF, Shen H, Ke Y, Zeng Y, Wu T, Kraft P, Chung CC, Tucker MA, Hou ZC, Liu YL, Hu YL, Liu Y, Wang L, Yuan G, Chen LS, Liu X, Ma T, Meng H, Sun L, Li XM, Li XM, Ku JW, Zhou YF, et alWu C, Wang Z, Song X, Feng XS, Abnet CC, He J, Hu N, Zuo XB, Tan W, Zhan Q, Hu Z, He Z, Jia W, Zhou Y, Yu K, Shu XO, Yuan JM, Zheng W, Zhao XK, Gao SG, Yuan ZQ, Zhou FY, Fan ZM, Cui JL, Lin HL, Han XN, Li B, Chen X, Dawsey SM, Liao L, Lee MP, Ding T, Qiao YL, Liu Z, Liu Y, Yu D, Chang J, Wei L, Gao YT, Koh WP, Xiang YB, Tang ZZ, Fan JH, Han JJ, Zhou SL, Zhang P, Zhang DY, Yuan Y, Huang Y, Liu C, Zhai K, Qiao Y, Jin G, Guo C, Fu J, Miao X, Lu C, Yang H, Wang C, Wheeler WA, Gail M, Yeager M, Yuenger J, Guo ET, Li AL, Zhang W, Li XM, Sun LD, Ma BG, Li Y, Tang S, Peng XQ, Liu J, Hutchinson A, Jacobs K, Giffen C, Burdette L, Fraumeni JF, Shen H, Ke Y, Zeng Y, Wu T, Kraft P, Chung CC, Tucker MA, Hou ZC, Liu YL, Hu YL, Liu Y, Wang L, Yuan G, Chen LS, Liu X, Ma T, Meng H, Sun L, Li XM, Li XM, Ku JW, Zhou YF, Yang LQ, Wang Z, Li Y, Qige Q, Yang WJ, Lei GY, Chen LQ, Li EM, Yuan L, Yue WB, Wang R, Wang LW, Fan XP, Zhu FH, Zhao WX, Mao YM, Zhang M, Xing GL, Li JL, Han M, Ren JL, Liu B, Ren SW, Kong QP, Li F, Sheyhidin I, Wei W, Zhang YR, Feng CW, Wang J, Yang YH, Hao HZ, Bao QD, Liu BC, Wu AQ, Xie D, Yang WC, Wang L, Zhao XH, Chen SQ, Hong JY, Zhang XJ, Freedman ND, Goldstein AM, Lin D, Taylor PR, Wang LD, Chanock SJ. Joint analysis of three genome-wide association studies of esophageal squamous cell carcinoma in Chinese populations. Nat Genet 2014; 46:1001-1006. [PMID: 25129146 PMCID: PMC4212832 DOI: 10.1038/ng.3064] [Show More Authors] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 07/23/2014] [Indexed: 02/05/2023]
Abstract
We conducted a joint (pooled) analysis of three genome-wide association studies (GWAS) of esophageal squamous cell carcinoma (ESCC) in individuals of Chinese ancestry (5,337 ESCC cases and 5,787 controls) with 9,654 ESCC cases and 10,058 controls for follow-up. In a logistic regression model adjusted for age, sex, study and two eigenvectors, two new loci achieved genome-wide significance, marked by rs7447927 at 5q31.2 (per-allele odds ratio (OR) = 0.85, 95% confidence interval (CI) = 0.82-0.88; P = 7.72 × 10(-20)) and rs1642764 at 17p13.1 (per-allele OR = 0.88, 95% CI = 0.85-0.91; P = 3.10 × 10(-13)). rs7447927 is a synonymous SNP in TMEM173, and rs1642764 is an intronic SNP in ATP1B2, near TP53. Furthermore, a locus in the HLA class II region at 6p21.32 (rs35597309) achieved genome-wide significance in the two populations at highest risk for ESSC (OR = 1.33, 95% CI = 1.22-1.46; P = 1.99 × 10(-10)). Our joint analysis identifies new ESCC susceptibility loci overall as well as a new locus unique to the population in the Taihang Mountain region at high risk of ESCC.
Collapse
Affiliation(s)
- Chen Wu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Xin Song
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Shan Feng
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Christian C. Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Jie He
- Department of Thoracic Surgery, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Xian-Bo Zuo
- Key Laboratory of Dermatology, Anhui Medical University, Anhui, Hefei, China
| | - Wen Tan
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Zhonghu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Weihua Jia
- State Key Laboratory of Oncology in Southern China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yifeng Zhou
- Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, China
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Xiao-Ou Shu
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Jian-Min Yuan
- University of Pittsburgh Cancer Institute, Pittsburg, PA, USA
| | - Wei Zheng
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Xue-Ke Zhao
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - She-Gan Gao
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Zhi-Qing Yuan
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | | | - Zong-Min Fan
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ji-Li Cui
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Hong-Li Lin
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue-Na Han
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bei Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xi Chen
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Basic Oncology and Pathology at College of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Sanford M. Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Linda Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Maxwell P. Lee
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ti Ding
- Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - You-Lin Qiao
- Department of Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dianke Yu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Chang
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lixuan Wei
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Tang Gao
- Shanghai Cancer Institute, Shanghai, People's Republic of China
| | - Woon-Puay Koh
- Duke-National University of Singapore, Graduate Medical School, Singapore
| | - Yong-Bing Xiang
- Shanghai Cancer Institute, Shanghai, People's Republic of China
| | | | - Jin-Hu Fan
- Department of Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Jing Han
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Basic Oncology and Pathology at College of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Sheng-Li Zhou
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Dong-Yun Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Yuan
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Huang
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunling Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kan Zhai
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Qiao
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Chuanhai Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in Southern China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiaoping Miao
- Key Laboratory for Environment and Health (Ministry of Education), School of Public Health, Huazhong University of Sciences and Technology, Wuhan, China
| | | | | | - Chaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | | | - Mitchell Gail
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Jeff Yuenger
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Er-Tao Guo
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ai-Li Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Medical Oncology, Tumor Hospital of Linzhou, Linzhou, Henan, China
| | - Wei Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue-Min Li
- Department of Pathology and Thoracic Surgery, Centre for Health Screening and Endoscopy, Cixian Hospital, Cixian, Hebei, China
| | - Liang-Dan Sun
- Key Laboratory of Dermatology, Anhui Medical University, Anhui, Hefei, China
| | - Bao-Gen Ma
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Central Laboratory, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yan Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Sa Tang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiu-Qing Peng
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Jing Liu
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Kevin Jacobs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Carol Giffen
- Information Management Services Inc., Silver Spring, Maryland, USA
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Joseph F. Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, China
| | - Yixin Zeng
- State Key Laboratory of Oncology in Southern China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tangchun Wu
- Key Laboratory for Environment and Health (Ministry of Education), School of Public Health, Huazhong University of Sciences and Technology, Wuhan, China
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Charles C. Chung
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
- Cancer Genome Research Laboratory, Division of Cancer Epidemiology and Genetics, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Margaret A. Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Zhi-Chao Hou
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ya-Li Liu
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan-Long Hu
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Liu
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Li Wang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Department of Basic Oncology, Pathology and Gastroenterology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Guo Yuan
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Sha Chen
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao Liu
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Teng Ma
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Meng
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Li Sun
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xin-Min Li
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiu-Min Li
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jian-Wei Ku
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The Second Affiliated Hospital of Nanyang Medical College, Nanyang, Henan, China
| | - Ying-Fa Zhou
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Liu-Qin Yang
- Department of Radiotherapy, Pathology and Pediatrics, Central Hospital of Xinxiang, Xinxiang, Henan, China
| | - Zhou Wang
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yin Li
- Department of Thoracic Surgery and Radiotherapy, Cancer Hospital of Henan Province, Zhengzhou, Henan, China
| | - Qirenwang Qige
- Department of Internal Mongolia Medicine, The Affiliated Hospital, Inner Mongolia Medical College, Huhhot, Inner Mongolia, China
| | - Wen-Jun Yang
- Department of Biotechnology, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Guang-Yan Lei
- Department of Thoracic Surgery, Tumor Hospital of Shaanxi Province, Xi'an, Shaanxi, China
| | - Long-Qi Chen
- Department of Thoracic and Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - En-Min Li
- Department of Oncologic Pathology, Medical College of Shantou University, Shantou, Guangdong, China
- Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou, Guangdong, China
| | - Ling Yuan
- Department of Thoracic Surgery and Radiotherapy, Cancer Hospital of Henan Province, Zhengzhou, Henan, China
| | - Wen-Bin Yue
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, Puyang City Oil Field General Hospital, Puyang, Henan, China
| | - Ran Wang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Lu-Wen Wang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue-Ping Fan
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Fang-Heng Zhu
- Department of Radiotherapy, Pathology and Pediatrics, Central Hospital of Xinxiang, Xinxiang, Henan, China
| | - Wei-Xing Zhao
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yi-Min Mao
- Department of Oncology, Pathology, Clinical Laboratory and Respiratory Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Mei Zhang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Guo-Lan Xing
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ji-Lin Li
- Department of Pathology and Thoracic Surgery, Linzhou Esophageal Cancer Hospital, Linzhou, Henan, China
| | - Min Han
- Department of Gastroenterology and Thoracic Surgery, The First People's Hospital of Shangqiu, Shangqiu, Henan, China
| | - Jing-Li Ren
- Department of Basic Oncology, Pathology and Gastroenterology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Liu
- Department of Gastroenterology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China
| | - Shu-Wei Ren
- Department of Oncology, Xinyang Central Hospital, Xinyang, Henan, China
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resource and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Feng Li
- Department of Pathology, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Ilyar Sheyhidin
- Department of Thoracic Surgery, Xinjiang Medical University, Urumqi, Xinjiang, China
- Medical Research Center, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wu Wei
- Institute of Hematologic Disease, Changzhi Medical University, Changzhi, Shanxi, China
- Department of Pathology, Changzhi Medical University, Changzhi, Shanxi, China
| | - Yan-Rui Zhang
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Central Laboratory, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Chang-Wei Feng
- Department of Basic Oncology, Pathology and Gastroenterology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jin Wang
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu-Hua Yang
- Department of Surgery, Hebi Dahejian Hospital, Hebi, Henan, China
| | | | - Qi-De Bao
- Anyang District Hospital, Anyang, Henan, China
| | - Bao-Chi Liu
- Surgical Department of Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ai-Qun Wu
- Department of Anatomy, the Second Military Medical University of Chinese PLA, Shanghai, China
| | - Dong Xie
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wan-Cai Yang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Liang Wang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Medical College of Wisconsin, Cancer Research Center, Milwaukee, WI, USA
| | - Xiao-Hang Zhao
- National Laboratory of Molecular Oncology, Cancer Institute & Hospital, CAMS, Beijing, China
| | - Shu-Qing Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun-Yan Hong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - Xue-Jun Zhang
- Key Laboratory of Dermatology, Anhui Medical University, Anhui, Hefei, China
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Alisa M. Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Philip R. Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Li-Dong Wang
- Department of Pathology, Cancer Research Center, Basic Medical College, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|