1
|
Yew YW, Loh M, Brown SJ. Understanding Atopic Dermatitis in Asian and European Population Cohorts Using Complementary Omics Techniques. J Invest Dermatol 2024:S0022-202X(24)02171-7. [PMID: 39503693 DOI: 10.1016/j.jid.2024.08.036] [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: 05/16/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 11/08/2024]
Abstract
Atopic dermatitis is highly heterogeneous with respect to pathogenesis, clinical manifestations, and treatment response. There is evidence that ancestry and skin type each contribute to this heterogeneity, indicating the need to improve understanding of disease mechanisms in diverse populations. Methods to integrate multiomics studies have been well-described, but this review focuses on the importance and the strategies needed to integrate data across different ancestral groups, focusing, because of data availability, on Asian and European populations. Skin scientists and clinicians will each benefit from an understanding of how the multiple complimentary layers of omics data may inform future clinical management, from insight into disease pathogenesis and treatment targets.
Collapse
Affiliation(s)
- Yik Weng Yew
- National Skin Centre, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Marie Loh
- National Skin Centre, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom; Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Sara J Brown
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom; Department of Dermatology, NHS Lothian, Edinburgh, United Kingdom
| |
Collapse
|
2
|
da Silva Duarte AJ, Sanabani SS. Deciphering epigenetic regulations in the inflammatory pathways of atopic dermatitis. Life Sci 2024; 348:122713. [PMID: 38735367 DOI: 10.1016/j.lfs.2024.122713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Atopic dermatitis, commonly referred to as atopic eczema, is a persistent inflammatory skin disorder that predominantly manifests in children but may endure into adulthood. Its clinical management poses challenges due to the absence of a definitive cure, and its prevalence varies across ethnicities, genders, and geographic locations. The epigenetic landscape of AD includes changes in DNA methylation, changes in histone acetylation and methylation, and regulation by non-coding RNAs. These changes affect inflammatory and immune mechanisms, and research has identified AD-specific variations in DNA methylation, particularly in the affected epidermis. Histone modifications, including acetylation, have been associated with the disruption of skin barrier function in AD, suggesting the potential therapeutic benefit of histone deacetylase inhibitors such as belinostat. Furthermore, non-coding RNAs, particularly microRNAs and long non-coding RNAs (lncRNAs), have been implicated in modulating various cellular processes central to AD pathogenesis. Therapeutic implications in AD include the potential use of DNA methylation inhibitors and histone deacetylase inhibitors to correct aberrant methylation patterns and modulate gene expression related to immune responses and skin barrier functions. Additionally, the emerging role of lncRNAs suggests the possibility of using small interfering RNAs or antisense oligonucleotides to inhibit lncRNAs and adjust their regulatory impact on gene expression. In conclusion, the importance of epigenetic elements in AD is becoming increasingly clear as studies highlight the contribution of DNA methylation, histone modifications and, control by non-coding RNAs to the onset and progression of the disease. Understanding these epigenetic changes provides valuable insights for developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Alberto José da Silva Duarte
- Laboratory of Medical Investigation LIM-56, Division of Dermatology, Medical School, University of São Paulo, São Paulo 05403-000, Brazil
| | - Sabri Saeed Sanabani
- Laboratory of Medical Investigation LIM-56, Division of Dermatology, Medical School, University of São Paulo, São Paulo 05403-000, Brazil; Laboratory of Medical Investigation Unit 03, Clinics Hospital, Faculty of Medicine, University of Sao Paulo, Sao Paulo 05403-000, Brazil.
| |
Collapse
|
3
|
Hu Y, Sun Y, Li T, Han W, Wang P. Identification of rat Vstm1 with conservative anti-inflammatory effect between rat and human homologs. Genomics 2024; 116:110774. [PMID: 38163574 DOI: 10.1016/j.ygeno.2023.110774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/17/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
Human VSTM1 (also known as SIRL1) is an inhibitory immune checkpoint receptor involved in leukocyte activation. Identification of the homologous genes in other species, such as mice and rats, will undoubtedly contribute to functional studies and clinical applications. Here, we successfully cloned the Vstm1 gene in rats, as supported by high-throughput sequencing data. However, Vstm1 is degenerated to a pseudogene in the mouse genome. Rat Vstm1 mRNA contains a complete open reading frame (ORF) of 630 nucleotides encoding 209 amino acids. Rat Vstm1 is highly expressed in bone marrow, especially in granulocytes. The expression levels of Vstm1 gradually increase with the development of granulocytes in bone marrow but are downregulated in response to inflammatory stimuli. Rat VSTM1 does not have an immunoreceptor tyrosine-based inhibitory motif (ITIM), however, it shows a conservative function of inflammatory inhibition with human VSTM1, and both are anti-correlated with many inflammatory cytokines, such as IL-1α and TNF-α. In bone marrow-derived macrophages (BMDMs), either rat or human VSTM1 suppressed the secretion of inflammatory cytokines in response to LPS stimulation. Further analysis in lung cancer microenvironment revealed that VSTM1 is mainly expressed in myeloid cells, anti-correlated with inflammatory cytokines and associated with tumor development and metastasis.
Collapse
Affiliation(s)
- Yuzhe Hu
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Yingzhe Sun
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Ting Li
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China
| | - Wenling Han
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China.
| | - Pingzhang Wang
- Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Peking University Center for Human Disease Genomics, Beijing, China.
| |
Collapse
|
4
|
Carrascosa-Carrillo JM, Aterido A, Li T, Guillén Y, Martinez S, Marsal S, Julià A. Toward Precision Medicine in Atopic Dermatitis Using Molecular-Based Approaches. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:66-75. [PMID: 37652096 DOI: 10.1016/j.ad.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 09/02/2023] Open
Abstract
Atopic dermatitis is the most common chronic inflammatory skin disorder, affecting up to 20% of children and 10% of adults in developed countries. The pathophysiology of atopic dermatitis is complex and involves a strong genetic predisposition and T-cell driven inflammation. Although our understanding of the pathology and drivers of this disease has improved in recent years, there are still knowledge gaps in the immune pathways involved. Therefore, advances in new omics technologies in atopic dermatitis will play a key role in understanding the pathogenesis of this burden disease and could develop preventive strategies and personalized treatment strategies. In this review, we discuss the latest developments in genetics, transcriptomics, epigenomics, proteomics, and metagenomics and understand how integrating multiple omics datasets will identify potential biomarkers and uncover nets of associations between several molecular levels.
Collapse
Affiliation(s)
- J M Carrascosa-Carrillo
- Dermatology Department, Hospital Germans Trias i Pujol, UAB, IGTP, Badalona, Barcelona, Spain
| | - A Aterido
- IMIDomics, Inc., Barcelona, Spain; Rheumatology Research Group, Vall Hebron Research Institute, Barcelona, Spain
| | - T Li
- IMIDomics, Inc., Barcelona, Spain
| | | | | | - S Marsal
- IMIDomics, Inc., Barcelona, Spain; Rheumatology Research Group, Vall Hebron Research Institute, Barcelona, Spain.
| | - A Julià
- IMIDomics, Inc., Barcelona, Spain; Rheumatology Research Group, Vall Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
5
|
Miao N, Wang J. New insights into the role of the immune inhibitory receptor VSTM1 in autoimmune disease. Cell Mol Immunol 2024; 21:95-96. [PMID: 37935837 PMCID: PMC10757710 DOI: 10.1038/s41423-023-01097-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023] Open
Affiliation(s)
- Naijun Miao
- Center for Immune-related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wang
- Center for Immune-related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Carrascosa-Carrillo JM, Aterido A, Li T, Guillén Y, Martinez S, Marsal S, Julià A. Toward Precision Medicine in Atopic Dermatitis Using Molecular-Based Approaches. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:T66-T75. [PMID: 37923065 DOI: 10.1016/j.ad.2023.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 11/07/2023] Open
Abstract
Atopic dermatitis is the most common chronic inflammatory skin disorder, affecting up to 20% of children and 10% of adults in developed countries. The pathophysiology of atopic dermatitis is complex and involves a strong genetic predisposition and T-cell driven inflammation. Although our understanding of the pathology and drivers of this disease has improved in recent years, there are still knowledge gaps in the immune pathways involved. Therefore, advances in new omics technologies in atopic dermatitis will play a key role in understanding the pathogenesis of this burden disease and could develop preventive strategies and personalized treatment strategies. In this review, we discuss the latest developments in genetics, transcriptomics, epigenomics, proteomics, and metagenomics and understand how integrating multiple omics datasets will identify potential biomarkers and uncover nets of associations between several molecular levels.
Collapse
Affiliation(s)
- J M Carrascosa-Carrillo
- Dermatology Department, Hospital Germans Trias i Pujol, UAB, IGTP, Badalona, Barcelona, España
| | - A Aterido
- IMIDomics, Inc., Barcelona, España; Rheumatology Research Group, Vall Hebron Research Institute, Barcelona, España
| | - T Li
- IMIDomics, Inc., Barcelona, España
| | | | | | - S Marsal
- IMIDomics, Inc., Barcelona, España; Rheumatology Research Group, Vall Hebron Research Institute, Barcelona, España.
| | - A Julià
- IMIDomics, Inc., Barcelona, España; Rheumatology Research Group, Vall Hebron Research Institute, Barcelona, España
| |
Collapse
|
7
|
Dessì A, Di Maria C, Pintus R, Fanos V, Bosco A. Lipidomics and Metabolomics in Infant Atopic Dermatitis: What's the Correlation with Early Nutrition? Curr Pediatr Rev 2024; 20:510-524. [PMID: 37055903 DOI: 10.2174/1573396320666230411093122] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 04/15/2023]
Abstract
To date, the complex picture of atopic dermatitis (AD) has not yet been fully clarified, despite the important prevalence of this disease in the pediatric population (20%) and the possibility of persistence into adulthood, with important implications for the quality of life of those affected, as well as significant social and financial costs. The most recent scientific evidence suggests a new interpretation of AD, highlighting the important role of the environment, particularly that of nutrition in the early stages of development. In fact, the new indications seem to point out the harmful effect of elimination diets, except in rare cases, the uselessness of chrono-insertions during complementary feeding and some benefits, albeit weak, of breastfeeding in those at greater risk. In this context, metabolomics and lipidomics can be necessary for a more in-depth knowledge of the complex metabolic network underlying this pathology. In fact, an alteration of the metabolic contents in children with AD has been highlighted, especially in correlation to the intestinal microbiota. While preliminary lipidomic studies showed the usefulness of a more in-depth knowledge of the alterations of the skin barrier to improve the development of baby skin care products. Therefore, investigating the response of different allergic phenotypes could be useful for better patient management and understanding, thus providing an early intervention on dysbiosis necessary to regulate the immune response from the earliest stages of development.
Collapse
Affiliation(s)
- Angelica Dessì
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, Cagliari, Italy
| | - Camilla Di Maria
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, Cagliari, Italy
| | - Roberta Pintus
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, Cagliari, Italy
| | - Vassilios Fanos
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, Cagliari, Italy
| | - Alice Bosco
- Department of Surgical Sciences, University of Cagliari and Neonatal Intensive Care Unit, AOU Cagliari, Cagliari, Italy
| |
Collapse
|
8
|
Grafanaki K, Antonatos C, Maniatis A, Petropoulou A, Vryzaki E, Vasilopoulos Y, Georgiou S, Gregoriou S. Intrinsic Effects of Exposome in Atopic Dermatitis: Genomics, Epigenomics and Regulatory Layers. J Clin Med 2023; 12:4000. [PMID: 37373692 DOI: 10.3390/jcm12124000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/03/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Atopic dermatitis (AD) or atopic eczema is an increasingly manifested inflammatory skin disorder of complex etiology which is modulated by both extrinsic and intrinsic factors. The exposome includes a person's lifetime exposures and their effects. We recently reviewed the extrinsic exposome's environmental risk factors that contribute to AD. The periods of pregnancy, infancy, and teenage years are recognized as crucial stages in the formation of AD, where the exposome leads to enduring impacts on the immune system. However, research is now focusing on the interactions between intrinsic pathways that are modulated by the extrinsic exposome, including genetic variation, epigenetic modifications, and signals, such as diet, stress, and microbiome interactions. As a result, immune dysregulation, barrier dysfunction, hormonal fluctuations, and skin microbiome dysbiosis are important factors contributing to AD development, and their in-depth understanding is crucial not only for AD treatment but also for similar inflammatory disorders.
Collapse
Affiliation(s)
- Katerina Grafanaki
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Alexandros Maniatis
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Antonia Petropoulou
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Eleftheria Vryzaki
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Sophia Georgiou
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Stamatis Gregoriou
- Department of Dermatology-Venereology, Faculty of Medicine, Andreas Sygros Hospital, National and Kapodistrian University of Athens, 16121 Athens, Greece
| |
Collapse
|
9
|
Aygün N, Liang D, Crouse WL, Keele GR, Love MI, Stein JL. Inferring cell-type-specific causal gene regulatory networks during human neurogenesis. Genome Biol 2023; 24:130. [PMID: 37254169 PMCID: PMC10230710 DOI: 10.1186/s13059-023-02959-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Genetic variation influences both chromatin accessibility, assessed in chromatin accessibility quantitative trait loci (caQTL) studies, and gene expression, assessed in expression QTL (eQTL) studies. Genetic variants can impact either nearby genes (cis-eQTLs) or distal genes (trans-eQTLs). Colocalization between caQTL and eQTL, or cis- and trans-eQTLs suggests that they share causal variants. However, pairwise colocalization between these molecular QTLs does not guarantee a causal relationship. Mediation analysis can be applied to assess the evidence supporting causality versus independence between molecular QTLs. Given that the function of QTLs can be cell-type-specific, we performed mediation analyses to find epigenetic and distal regulatory causal pathways for genes within two major cell types of the developing human cortex, progenitors and neurons. RESULTS We find that the expression of 168 and 38 genes is mediated by chromatin accessibility in progenitors and neurons, respectively. We also find that the expression of 11 and 12 downstream genes is mediated by upstream genes in progenitors and neurons. Moreover, we discover that a genetic locus associated with inter-individual differences in brain structure shows evidence for mediation of SLC26A7 through chromatin accessibility, identifying molecular mechanisms of a common variant association to a brain trait. CONCLUSIONS In this study, we identify cell-type-specific causal gene regulatory networks whereby the impacts of variants on gene expression were mediated by chromatin accessibility or distal gene expression. Identification of these causal paths will enable identifying and prioritizing actionable regulatory targets perturbing these key processes during neurodevelopment.
Collapse
Affiliation(s)
- Nil Aygün
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dan Liang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wesley L Crouse
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Gregory R Keele
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
10
|
von Richthofen HJ, Westerlaken GH, Gollnast D, Besteman S, Delemarre EM, Rodenburg K, Moerer P, Stapels DA, Andiappan AK, Rötzschke O, Nierkens S, Leavis HL, Bont LJ, Rooijakkers SH, Meyaard L. Soluble Signal Inhibitory Receptor on Leukocytes-1 Is Released from Activated Neutrophils by Proteinase 3 Cleavage. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:389-397. [PMID: 36637221 PMCID: PMC9915861 DOI: 10.4049/jimmunol.2200169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/05/2022] [Indexed: 01/14/2023]
Abstract
Signal inhibitory receptor on leukocytes-1 (SIRL-1) is an immune inhibitory receptor expressed on human granulocytes and monocytes that dampens antimicrobial functions. We previously showed that sputum neutrophils from infants with severe respiratory syncytial virus (RSV) bronchiolitis have decreased SIRL-1 surface expression compared with blood neutrophils and that SIRL-1 surface expression is rapidly lost from in vitro activated neutrophils. This led us to hypothesize that activated neutrophils lose SIRL-1 by ectodomain shedding. Here, we developed an ELISA and measured the concentration of soluble SIRL-1 (sSIRL-1) in patients with RSV bronchiolitis and hospitalized patients with COVID-19, which are both characterized by neutrophilic inflammation. In line with our hypothesis, sSIRL-1 concentration was increased in sputum compared with plasma of patients with RSV bronchiolitis and in serum of hospitalized patients with COVID-19 compared with control serum. In addition, we show that in vitro activated neutrophils release sSIRL-1 by proteolytic cleavage and that this diminishes the ability to inhibit neutrophilic reactive oxygen species production via SIRL-1. Finally, we found that SIRL-1 shedding is prevented by proteinase 3 inhibition and by extracellular adherence protein from Staphylococcus aureus. Notably, we recently showed that SIRL-1 is activated by PSMα3 from S. aureus, suggesting that S. aureus may counteract SIRL-1 shedding to benefit from preserved inhibitory function of SIRL-1. In conclusion, we report that SIRL-1 is released from activated neutrophils by proteinase 3 cleavage and that endogenous sSIRL-1 protein is present in vivo.
Collapse
Affiliation(s)
- Helen J. von Richthofen
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| | - Geertje H.A. Westerlaken
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| | - Doron Gollnast
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| | - Sjanna Besteman
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eveline M. Delemarre
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Karlijn Rodenburg
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Petra Moerer
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Daphne A.C. Stapels
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Anand K. Andiappan
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; and
| | - Olaf Rötzschke
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; and
| | - Stefan Nierkens
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Helen L. Leavis
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Louis J. Bont
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Suzan H.M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Linde Meyaard
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| |
Collapse
|
11
|
von Richthofen HJ, Hafkamp FMJ, van Haperen A, de Jong EC, Meyaard L. VSTM1-v2 does not drive human Th17 cell differentiation: A replication study. PLoS One 2023; 18:e0284404. [PMID: 37053248 PMCID: PMC10101491 DOI: 10.1371/journal.pone.0284404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/28/2023] [Indexed: 04/14/2023] Open
Abstract
Signal inhibitory receptor on leukocytes-1 (SIRL-1) is an immune inhibitory receptor expressed on human myeloid cells. We previously showed that dendritic cell (DC)-driven Th17 cell differentiation of human naive CD4+ T cells requires presence of neutrophils, which is inhibited by SIRL-1 ligation. VSTM1-v2 is a soluble isoform of SIRL-1, which was previously proposed to function as a Th17 polarizing cytokine. Here, we investigated the effect of VSTM1-v2 on DC-driven Th17 cell development. Neutrophils induced DC-driven Th17 cell differentiation, which was not enhanced by VSTM1-v2. Similarly, we found no effect of VSTM1-v2 on cytokine-driven Th17 cell development. Thus, our results do not support a role for VSTM1-v2 in Th17 cell differentiation.
Collapse
Affiliation(s)
- Helen J von Richthofen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Florianne M J Hafkamp
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anouk van Haperen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| |
Collapse
|
12
|
Wang L, Yuan J, Cheng Y, Xu Z, Ding M, Li J, Si Y, Zong M, Fan L. Signal inhibitory receptor on leukocytes-1 regulates the formation of the neutrophil extracellular trap in rheumatoid arthritis. Mol Immunol 2022; 151:242-251. [PMID: 36182788 DOI: 10.1016/j.molimm.2022.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/16/2022] [Accepted: 09/14/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Neutrophil extracellular trap (NET) has been demonstrated to play important roles in the pathogenesis and progression of rheumatoid arthritis (RA). Emerging evidence indicates that ligation of signal inhibitory receptor on leukocytes-1 (SIRL-1) can dampen Fc receptor-induced reactive oxygen species (ROS) production in primary human neutrophils by reducing extracellular signal-regulated kinase (ERK) activation. The current study aimed to determine the regulatory effects of SIRL-1 on the NET formation and ROS production by comparing RA patients and healthy controls (HC). METHODS Multiple assays were employed to detect the expression level of SIRL-1, including immunohistochemical staining, quantitative reverse transcription polymerase chain reaction (RT-PCR) and flow cytometry. Peripheral blood neutrophils from both HC and RA patients were freshly isolated. The NET formation was assessed spontaneously before and after exposure to serum samples from HC and RA patients, respectively. The quantification of NET formation was determined by fluorescence microscopy and Spectra Max M5 fluorescent plate reader. The ROS production was examined by flow cytometry. RESULTS The expression level of SIRL-1 in peripheral blood neutrophils was decreased in RA, comparing to HC. The RA-originated neutrophils showed higher levels of ROS production and NET formation. Ligation of SIRL-1 to neutrophils suppressed ROS production and NET formation. Stimulation of neutrophils with severe anti-cyclic citrullinated peptides (CCP) induced NET formation, which could be inhibited by application of SIRL-1 ligation. CONCLUSION The current study identified SIRL-1 differentially expressed in neutrophils between RA and HC. Ligation of SIRL-1 inhibited ROS production and NET formation. Downregulation of SIRL-1 showed correlation with upregulation of NET formation in RA. These findings showed the regulation of SIRL-1 on NET formation and provided a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Lan Wang
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Jiayi Yuan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Yu Cheng
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Zhen Xu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Menglei Ding
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Jing Li
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Yuying Si
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Ming Zong
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| | - Lieying Fan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China.
| |
Collapse
|
13
|
Lin S, Luo B, Ma J. Multiple datasets to explore the molecular mechanism of sepsis. BMC Genom Data 2022; 23:66. [PMID: 35971090 PMCID: PMC9380322 DOI: 10.1186/s12863-022-01078-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022] Open
Abstract
Background This study aimed to identify potential biomarkers, by means of bioinformatics, affecting the occurrence and development of septic shock. Methods Download GSE131761 septic shock data set from NCBI geo database, including 33 control samples and 81 septic shock samples. GSE131761 and sequencing data were used to identify and analyze differentially expressed genes in septic shock patients and normal subjects. In addition, with sequencing data as training set and GSE131761 as validation set, a diagnostic model was established by lasso regression to identify key genes. ROC curve verified the stability of the model. Finally, immune infiltration analysis, enrichment analysis, transcriptional regulation analysis and correlation analysis of key genes were carried out to understand the potential molecular mechanism of key genes affecting septic shock. Results A total of 292 differential genes were screened out from the self-test data, 294 differential genes were screened out by GSE131761, Lasso regression was performed on the intersection genes of the two, a diagnostic model was constructed, and 5 genes were identified as biomarkers of septic shock. These 5 genes were SIGLEC10, VSTM1, GYPB, OPTN, and GIMAP7. The five key genes were strongly correlated with immune cells, and the ROC results showed that the five genes had good predictive performance on the occurrence and development of diseases. In addition, the key genes were strongly correlated with immune regulatory genes. Conclusion In this study, a series of algorithms were used to identify five key genes that are associated with septic shock, which may become potential candidate targets for septic shock diagnosis and treatment. Trial registration Approval number:2019XE0149-1.
Collapse
|
14
|
Ignatieva EV, Matrosova EA. Disease-associated genetic variants in the regulatory regions of human genes: mechanisms of action on transcription and genomic resources for dissecting these mechanisms. Vavilovskii Zhurnal Genet Selektsii 2021; 25:18-29. [PMID: 34541447 PMCID: PMC8408020 DOI: 10.18699/vj21.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 11/21/2022] Open
Abstract
Whole genome and whole exome sequencing technologies play a very important role in the studies of the genetic aspects of the pathogenesis of various diseases. The ample use of genome-wide and exome-wide association study
methodology (GWAS and EWAS) made it possible to identify a large number of genetic variants associated with diseases.
This information is accumulated in the databases like GWAS central, GWAS catalog, OMIM, ClinVar, etc. Most of the variants identified by the GWAS technique are located in the noncoding regions of the human genome. According to the
ENCODE project, the fraction of regions in the human genome potentially involved in transcriptional control is many times
greater than the fraction of coding regions. Thus, genetic variation in noncoding regions of the genome can increase the
susceptibility to diseases by disrupting various regulatory elements (promoters, enhancers, silencers, insulator regions,
etc.). However, identification of the mechanisms of influence of pathogenic genetic variants on the diseases risk is difficult
due to a wide variety of regulatory elements. The present review focuses on the molecular genetic mechanisms by which
pathogenic genetic variants affect gene expression. At the same time, attention is concentrated on the transcriptional level
of regulation as an initial step in the expression of any gene. A triggering event mediating the effect of a pathogenic genetic
variant on the level of gene expression can be, for example, a change in the functional activity of transcription factor binding sites (TFBSs) or DNA methylation change, which, in turn, affects the functional activity of promoters or enhancers. Dissecting the regulatory roles of polymorphic loci have been impossible without close integration of modern experimental
approaches with computer analysis of a growing wealth of genetic and biological data obtained using omics technologies.
The review provides a brief description of a number of the most well-known public genomic information resources containing data obtained using omics technologies, including (1) resources that accumulate data on the chromatin states and the
regions of transcription factor binding derived from ChIP-seq experiments; (2) resources containing data on genomic loci,
for which allele-specific transcription factor binding was revealed based on ChIP-seq technology; (3) resources containing
in silico predicted data on the potential impact of genetic variants on the transcription factor binding sites
Collapse
Affiliation(s)
- E V Ignatieva
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - E A Matrosova
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
15
|
Rumpret M, von Richthofen HJ, van der Linden M, Westerlaken GHA, Talavera Ormeño C, van Strijp JAG, Landau M, Ovaa H, van Sorge NM, Meyaard L. Signal inhibitory receptor on leukocytes-1 recognizes bacterial and endogenous amphipathic α-helical peptides. FASEB J 2021; 35:e21875. [PMID: 34533845 DOI: 10.1096/fj.202100812r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 01/31/2023]
Abstract
Signal inhibitory receptor on leukocytes-1 (SIRL-1) is a negative regulator of myeloid cell function and dampens antimicrobial responses. We here show that different species of the genus Staphylococcus secrete SIRL-1-engaging factors. By screening a library of single-gene transposon mutants in Staphylococcus aureus, we identified these factors as phenol-soluble modulins (PSMs). PSMs are amphipathic α-helical peptides involved in multiple aspects of staphylococcal virulence and physiology. They are cytotoxic and activate the chemotactic formyl peptide receptor 2 (FPR2) on immune cells. Human cathelicidin LL-37 is also an amphipathic α-helical peptide with antimicrobial and chemotactic activities, structurally and functionally similar to α-type PSMs. We demonstrate that α-type PSMs from multiple staphylococcal species as well as human cathelicidin LL-37 activate SIRL-1, suggesting that SIRL-1 recognizes α-helical peptides with an amphipathic arrangement of hydrophobicity, although we were not able to show direct binding to SIRL-1. Upon rational peptide design, we identified artificial peptides in which the capacity to ligate SIRL-1 is segregated from cytotoxic and FPR2-activating properties, allowing specific engagement of SIRL-1. In conclusion, we propose staphylococcal PSMs and human LL-37 as a potential new class of natural ligands for SIRL-1.
Collapse
Affiliation(s)
- Matevž Rumpret
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Helen J von Richthofen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Maarten van der Linden
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geertje H A Westerlaken
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Cami Talavera Ormeño
- Oncode Institute, Utrecht, The Netherlands.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Meytal Landau
- Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
| | - Huib Ovaa
- Oncode Institute, Utrecht, The Netherlands.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nina M van Sorge
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| |
Collapse
|
16
|
Schmidt AD, de Guzman Strong C. Current understanding of epigenetics in atopic dermatitis. Exp Dermatol 2021; 30:1150-1155. [PMID: 34008901 PMCID: PMC8361700 DOI: 10.1111/exd.14392] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/19/2021] [Accepted: 05/09/2021] [Indexed: 02/06/2023]
Abstract
Atopic dermatitis (AD) is an inflammatory skin disorder affecting up to 20% of the paediatric population worldwide. AD patients commonly exhibit dry skin and pruritus and are at a higher risk for developing asthma as well as allergic rhinitis. Filaggrin loss‐of‐function variants are the most widely replicated genetic risk factor among >40 genes associated with AD susceptibility. The prevalence of AD has tripled in the past 30 years in industrial countries around the world. This urgent public health issue has prompted the field to more thoroughly investigate the mechanisms that underlie AD pathogenesis amidst environmental exposures. Epigenetics is the study of heritable, yet reversible, modifications to the genome that affect gene expression. The past decade has seen an emergence of exciting studies identifying a role for epigenetic regulation associated with AD and at the interface of environmental factors. Such epigenetic studies have been empowered by sequencing technologies and human genome variation and epigenome maps. miRNAs that post‐transcriptionally modify gene expression and circRNAs have also been discovered to be associated with AD. Here, we review our current understanding of epigenetics associated with atopic dermatitis. We discuss studies identifying distinct DNA methylation changes in keratinocytes and T cells, eQTLs as DNA methylation switches that impact gene expression, and histone modification changes associated with AD‐related microbial dysbiosis. We further highlight the need for integrative and collaborative analyses to elucidate the impact of these epigenetic findings as potential drivers for AD pathogenesis and the translation of this new knowledge to develop newer targeted treatments.
Collapse
Affiliation(s)
- Alina D Schmidt
- Division of Dermatology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Center for Pharmacogenomics, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Center for the Study of Itch & Sensory Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Cristina de Guzman Strong
- Division of Dermatology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Center for Pharmacogenomics, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Center for the Study of Itch & Sensory Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
17
|
Tseng CC, Wong MC, Liao WT, Chen CJ, Lee SC, Yen JH, Chang SJ. Genetic Variants in Transcription Factor Binding Sites in Humans: Triggered by Natural Selection and Triggers of Diseases. Int J Mol Sci 2021; 22:ijms22084187. [PMID: 33919522 PMCID: PMC8073710 DOI: 10.3390/ijms22084187] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Variants of transcription factor binding sites (TFBSs) constitute an important part of the human genome. Current evidence demonstrates close links between nucleotides within TFBSs and gene expression. There are multiple pathways through which genomic sequences located in TFBSs regulate gene expression, and recent genome-wide association studies have shown the biological significance of TFBS variation in human phenotypes. However, numerous challenges remain in the study of TFBS polymorphisms. This article aims to cover the current state of understanding as regards the genomic features of TFBSs and TFBS variants; the mechanisms through which TFBS variants regulate gene expression; the approaches to studying the effects of nucleotide changes that create or disrupt TFBSs; the challenges faced in studies of TFBS sequence variations; the effects of natural selection on collections of TFBSs; in addition to the insights gained from the study of TFBS alleles related to gout, its associated comorbidities (increased body mass index, chronic kidney disease, diabetes, dyslipidemia, coronary artery disease, ischemic heart disease, hypertension, hyperuricemia, osteoporosis, and prostate cancer), and the treatment responses of patients.
Collapse
Affiliation(s)
- Chia-Chun Tseng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (J.-H.Y.)
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Man-Chun Wong
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Wei-Ting Liao
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Correspondence: (W.-T.L.); (S.-J.C.); Tel.: +886-7-3121101 (W.-T.L.); +886-7-5916679 (S.-J.C.); Fax:+886-7-3125339 (W.-T.L.); +886-7-5919264 (S.-J.C.)
| | - Chung-Jen Chen
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan;
| | - Su-Chen Lee
- Laboratory Diagnosis of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Jeng-Hsien Yen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (J.-H.Y.)
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Biological Science and Technology, National Chiao-Tung University, Hsinchu 30010, Taiwan
| | - Shun-Jen Chang
- Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung 81148, Taiwan
- Correspondence: (W.-T.L.); (S.-J.C.); Tel.: +886-7-3121101 (W.-T.L.); +886-7-5916679 (S.-J.C.); Fax:+886-7-3125339 (W.-T.L.); +886-7-5919264 (S.-J.C.)
| |
Collapse
|
18
|
Liu W, Li M, Zhang W, Zhou G, Wu X, Wang J, Lu Q, Zhao H. Leveraging functional annotation to identify genes associated with complex diseases. PLoS Comput Biol 2020; 16:e1008315. [PMID: 33137096 PMCID: PMC7660930 DOI: 10.1371/journal.pcbi.1008315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 11/12/2020] [Accepted: 09/05/2020] [Indexed: 02/06/2023] Open
Abstract
To increase statistical power to identify genes associated with complex traits, a number of transcriptome-wide association study (TWAS) methods have been proposed using gene expression as a mediating trait linking genetic variations and diseases. These methods first predict expression levels based on inferred expression quantitative trait loci (eQTLs) and then identify expression-mediated genetic effects on diseases by associating phenotypes with predicted expression levels. The success of these methods critically depends on the identification of eQTLs, which may not be functional in the corresponding tissue, due to linkage disequilibrium (LD) and the correlation of gene expression between tissues. Here, we introduce a new method called T-GEN (Transcriptome-mediated identification of disease-associated Genes with Epigenetic aNnotation) to identify disease-associated genes leveraging epigenetic information. Through prioritizing SNPs with tissue-specific epigenetic annotation, T-GEN can better identify SNPs that are both statistically predictive and biologically functional. We found that a significantly higher percentage (an increase of 18.7% to 47.2%) of eQTLs identified by T-GEN are inferred to be functional by ChromHMM and more are deleterious based on their Combined Annotation Dependent Depletion (CADD) scores. Applying T-GEN to 207 complex traits, we were able to identify more trait-associated genes (ranging from 7.7% to 102%) than those from existing methods. Among the identified genes associated with these traits, T-GEN can better identify genes with high (>0.99) pLI scores compared to other methods. When T-GEN was applied to late-onset Alzheimer's disease, we identified 96 genes located at 15 loci, including two novel loci not implicated in previous GWAS. We further replicated 50 genes in an independent GWAS, including one of the two novel loci.
Collapse
Affiliation(s)
- Wei Liu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, United States of America
| | - Mo Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States of America
| | - Wenfeng Zhang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States of America
| | - Geyu Zhou
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, United States of America
| | - Xing Wu
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, United States of America
| | - Jiawei Wang
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, United States of America
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, WI, United States of America
- Department of Statistics, University of Wisconsin-Madison, WI, United States of America
- Center for Demography of Health and Aging, University of Wisconsin-Madison, WI, United States of America
| | - Hongyu Zhao
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, United States of America
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States of America
- Department of Genetics, Yale School of Medicine, New Haven, CT, United States of America
| |
Collapse
|
19
|
Epigenetic alterations in skin homing CD4 +CLA + T cells of atopic dermatitis patients. Sci Rep 2020; 10:18020. [PMID: 33093567 PMCID: PMC7582180 DOI: 10.1038/s41598-020-74798-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
T cells expressing the cutaneous lymphocyte antigen (CLA) mediate pathogenic inflammation in atopic dermatitis (AD). The molecular alterations contributing to their dysregulation remain unclear. With the aim to elucidate putative altered pathways in AD we profiled DNA methylation levels and miRNA expression in sorted T cell populations (CD4+, CD4+CD45RA+ naïve, CD4+CLA+, and CD8+) from adult AD patients and healthy controls (HC). Skin homing CD4+CLA+ T cells from AD patients showed significant differences in DNA methylation in 40 genes compared to HC (p < 0.05). Reduced DNA methylation levels in the upstream region of the interleukin-13 gene (IL13) in CD4+CLA+ T cells from AD patients correlated with increased IL13 mRNA expression in these cells. Sixteen miRNAs showed differential expression in CD4+CLA+ T cells from AD patients targeting genes in 202 biological processes (p < 0.05). An integrated network analysis of miRNAs and CpG sites identified two communities of strongly interconnected regulatory elements with strong antagonistic behaviours that recapitulated the differences between AD patients and HC. Functional analysis of the genes linked to these communities revealed their association with key cytokine signaling pathways, MAP kinase signaling and protein ubiquitination. Our findings support that epigenetic mechanisms play a role in the pathogenesis of AD by affecting inflammatory signaling molecules in skin homing CD4+CLA+ T cells and uncover putative molecules participating in AD pathways.
Collapse
|
20
|
Vohra M, Sharma AR, Prabhu B N, Rai PS. SNPs in Sites for DNA Methylation, Transcription Factor Binding, and miRNA Targets Leading to Allele-Specific Gene Expression and Contributing to Complex Disease Risk: A Systematic Review. Public Health Genomics 2020; 23:155-170. [PMID: 32966991 DOI: 10.1159/000510253] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 07/16/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The complex genetic diversity among human populations results from an assortment of factors acting at various sequential levels, including mutations, population migrations, genetic drift, and selection. Although there are a plethora of DNA sequence variations identified through genome-wide association studies (GWAS), the challenge remains to explain the mechanisms underlying interindividual phenotypic disparity accounting for disease susceptibility. Single nucleotide polymorphisms (SNPs) present in the sites for DNA methylation, transcription factor (TF) binding, or miRNA targets can alter the gene expression. The systematic review aimed to evaluate the complex crosstalk among SNPs, miRNAs, DNA methylation, and TFs for complex multifactorial disease risk. METHODS PubMed and Scopus databases were used from inception until May 15, 2019. Initially, screening of articles involved studies assessing the interaction of SNPs with TFs, DNA methylation, or miRNAs resulting in allele-specific gene expression in complex multifactorial diseases. We also included the studies which provided experimental validation of the interaction of SNPs with each of these factors. The results from various studies on multifactorial diseases were assessed. RESULTS A total of 11 articles for SNPs interacting with DNA methylation, 30 articles for SNPs interacting with TFs, and 11 articles for SNPs in miRNA binding sites were selected. The interactions of SNPs with epigenetic factors were found to be implicated in different types of cancers, autoimmune diseases, cardiovascular diseases, diabetes, and asthma. CONCLUSION The systematic review provides evidence for the interplay between genetic and epigenetic risk factors through allele-specific gene expression in various complex multifactorial diseases.
Collapse
Affiliation(s)
- Manik Vohra
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Anu Radha Sharma
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Navya Prabhu B
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India,
| |
Collapse
|
21
|
von Richthofen HJ, Gollnast D, van Capel TMM, Giovannone B, Westerlaken GHA, Lutter L, Oldenburg B, Hijnen D, van der Vlist M, de Jong EC, Meyaard L. Signal Inhibitory Receptor on Leukocytes-1 is highly expressed on lung monocytes, but absent on mononuclear phagocytes in skin and colon. Cell Immunol 2020; 357:104199. [PMID: 32942189 DOI: 10.1016/j.cellimm.2020.104199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/07/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023]
Abstract
Signal Inhibitory Receptor on Leukocytes-1 (SIRL-1) is expressed on human blood monocytes and granulocytes and inhibits myeloid effector functions. On monocytes, but not granulocytes, SIRL-1 expression is low or absent in individuals with the single nucleotide polymorphism (SNP) rs612529C. The expression of SIRL-1 in tissue and the influence of rs612529 hereon is currently unknown. Here, we used flow cytometry to determine SIRL-1 expression on immune cells in human blood and three barrier tissues; skin, colon and lung. SIRL-1 was expressed by virtually all neutrophils and eosinophils in these tissues. In contrast, SIRL-1 was not expressed by monocyte-derived cells in skin and colon, whereas it was highly expressed by lung classical monocytes. Lung monocytes from individuals with a rs612529C allele had decreased SIRL-1 expression, consistent with the genotype association in blood. Within the different monocyte subsets in blood and lung, SIRL-1 expression was highest in classical monocytes and lowest in nonclassical monocytes. SIRL-1 was not expressed by dendritic cells in blood and barrier tissues. Together, these results indicate that SIRL-1 is differentially expressed on phagocyte subsets in blood and barrier tissues, and that its expression on monocytes is genotype- and tissue-specific. Immune regulation of monocytes by SIRL-1 may be of particular importance in the lung.
Collapse
Affiliation(s)
- Helen J von Richthofen
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Oncode Institute, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Doron Gollnast
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Oncode Institute, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Toni M M van Capel
- Department of Experimental Immunology, Amsterdam University Medical Center, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands
| | - Barbara Giovannone
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Department of Dermatology and Allergology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Geertje H A Westerlaken
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Oncode Institute, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Lisanne Lutter
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - DirkJan Hijnen
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Department of Dermatology and Allergology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Department of Dermatology, Diakonessenhuis Utrecht, Bosboomstraat 1, 3582 KE Utrecht, the Netherlands
| | - Michiel van der Vlist
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Oncode Institute, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, Meibergdreef 15, 1105 AZ Amsterdam, the Netherlands
| | - Linde Meyaard
- Center of Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Oncode Institute, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands.
| |
Collapse
|
22
|
Coassin S, Hermann-Kleiter N, Haun M, Wahl S, Wilson R, Paulweber B, Kunze S, Meitinger T, Strauch K, Peters A, Waldenberger M, Kronenberg F, Lamina C. A genome-wide analysis of DNA methylation identifies a novel association signal for Lp(a) concentrations in the LPA promoter. PLoS One 2020; 15:e0232073. [PMID: 32343731 PMCID: PMC7188291 DOI: 10.1371/journal.pone.0232073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/19/2020] [Indexed: 12/24/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is a major cardiovascular risk factor, which is largely genetically determined by one major gene locus, the LPA gene. Many aspects of the transcriptional regulation of LPA are poorly understood and the role of epigenetics has not been addressed yet. Therefore, we conducted an epigenome-wide analysis of DNA methylation on Lp(a) levels in two population-based studies (total n = 2208). We identified a CpG site in the LPA promoter which was significantly associated with Lp(a) concentrations. Surprisingly, the identified CpG site was found to overlap the SNP rs76735376. We genotyped this SNP de-novo in three studies (total n = 7512). The minor allele of rs76735376 (1.1% minor allele frequency) was associated with increased Lp(a) values (p = 1.01e-59) and explained 3.5% of the variation of Lp(a). Statistical mediation analysis showed that the effect on Lp(a) is rather originating from the base change itself and is not mediated by DNA methylation levels. This finding is supported by eQTL data from 208 liver tissue samples from the GTEx project, which shows a significant association of the rs76735376 minor allele with increased LPA expression. To evaluate, whether the association signal at rs76735376 may actually be derived from a stronger eQTL signal in LD with this SNP, eQTL association results of all correlated SNPs (r2≥0.1) were integrated with genetic association results. This analysis pinpointed to rs10455872 as the potential trigger of the effect of rs76735376. Furthermore, both SNPs coincide with short apo(a) isoforms. Adjusting for both, rs10455872 and the apo(a) isoforms diminished the effect size of rs76735376 to 5.38 mg/dL (p = 0.0463). This indicates that the effect of rs76735376 can be explained by both an independent effect of the SNP and a strong correlation with rs10455872 and apo(a) isoforms.
Collapse
Affiliation(s)
- Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Department of Genetics and Pharmacology, Institute of Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Margot Haun
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Simone Wahl
- 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
| | - Rory Wilson
- 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
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Private Medical University, Salzburg, Austria
| | - Sonja Kunze
- 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
| | - Thomas Meitinger
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- German Research Center for Environmental Health, Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Konstantin Strauch
- German Research Center for Environmental Health, Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- 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
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
23
|
Martin MJ, Estravís M, García-Sánchez A, Dávila I, Isidoro-García M, Sanz C. Genetics and Epigenetics of Atopic Dermatitis: An Updated Systematic Review. Genes (Basel) 2020; 11:E442. [PMID: 32325630 PMCID: PMC7231115 DOI: 10.3390/genes11040442] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Atopic dermatitis is a common inflammatory skin disorder that affects up to 15-20% of the population and is characterized by recurrent eczematous lesions with intense itching. As a heterogeneous disease, multiple factors have been suggested to explain the nature of atopic dermatitis (AD), and its high prevalence makes it necessary to periodically compile and update the new information available. In this systematic review, the focus is set at the genetic and epigenetic studies carried out in the last years. METHODS A systematic literature review was conducted in three scientific publication databases (PubMed, Cochrane Library, and Scopus). The search was restricted to publications indexed from July 2016 to December 2019, and keywords related to atopic dermatitis genetics and epigenetics were used. RESULTS A total of 73 original papers met the inclusion criteria established, including 9 epigenetic studies. A total of 62 genes and 5 intergenic regions were described as associated with AD. CONCLUSION Filaggrin (FLG) polymorphisms are confirmed as key genetic determinants for AD development, but also epigenetic regulation and other genes with functions mainly related to the immune system and extracellular matrix, reinforcing the notion of skin homeostasis breakage in AD.
Collapse
Affiliation(s)
- Maria J Martin
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
| | - Miguel Estravís
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Asunción García-Sánchez
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Ignacio Dávila
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Immunoallergy, Salamanca University Hospital, 37007 Salamanca, Spain
| | - María Isidoro-García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Catalina Sanz
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
24
|
Kumar D, Lee B, Puan KJ, Lee W, Luis BS, Yusof N, Andiappan AK, Del Rosario R, Poschmann J, Kumar P, DeLibero G, Singhal A, Prabhakar S, De Yun W, Poidinger M, Rötzschke O. Resistin expression in human monocytes is controlled by two linked promoter SNPs mediating NFKB p50/p50 binding and C-methylation. Sci Rep 2019; 9:15245. [PMID: 31645609 PMCID: PMC6811637 DOI: 10.1038/s41598-019-51592-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Resistin is a key cytokine associated with metabolic and inflammatory diseases. Especially in East Asian populations, the expression levels are strongly influenced by genetic polymorphisms. Mechanisms and functional implications of this genetic control are still unknown. By employing reporter assays, EMSA, inhibition studies, bisulphite sequencing, ChIP-Seq and gene-editing we show that the p50/p50 homodimer known to act as repressor for a number of pro-inflammatory genes plays a central role in the genetic regulation of resistin in monocytes along with promoter methylation. In the common RETN haplotype p50/p50 constitutively dampens the expression by binding to the promoter. In an Asian haplotype variant however this interaction is disrupted by the A allele of rs3219175. The SNP is in very close linkage to rs34861192, a CpG SNP, located 280 bp upstream which provides an allele-specific C-methylation site. rs34861192 is located in a 100 bp region found to be methylated in the common but not in the Asian haplotype, resulting in the latter having a higher basal expression, which also associates with elevated histone acetylation (H3K27ac). Genotype associations within cohort data of 200 East Asian individuals revealed significant associations between this haplotype and the plasma levels of factors such as TGF-b, S100B, sRAGE and IL-8 as well as with myeloid DC counts. Thus, the common RETN haplotype is tightly regulated by the epigenetic mechanism linked to p50/p50-binding. This control is lost in the Asian haplotype, which may have evolved to balance the antagonistic RETN effects on pathogen protection vs. metabolic and inflammatory disease induction.
Collapse
Affiliation(s)
- Dilip Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| | - Bernett Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Wendy Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Boris San Luis
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Nurhashikin Yusof
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Ricardo Del Rosario
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA, 02142, USA
| | - Jeremie Poschmann
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Centre de Recherche en Transplantation et Immunologie, Université de Nantes, Nantes, France
| | - Pavanish Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Gennaro DeLibero
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Amit Singhal
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore
| | - Wang De Yun
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Olaf Rötzschke
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| |
Collapse
|
25
|
Wang H, Lou D, Wang Z. Crosstalk of Genetic Variants, Allele-Specific DNA Methylation, and Environmental Factors for Complex Disease Risk. Front Genet 2019; 9:695. [PMID: 30687383 PMCID: PMC6334214 DOI: 10.3389/fgene.2018.00695] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 12/12/2018] [Indexed: 01/04/2023] Open
Abstract
Over the past decades, genome-wide association studies (GWAS) have identified thousands of phenotype-associated DNA sequence variants for potential explanations of inter-individual phenotypic differences and disease susceptibility. However, it remains a challenge for translating the associations into causative mechanisms for complex diseases, partially due to the involved variants in the noncoding regions and the inconvenience of functional studies in human population samples. So far, accumulating evidence has suggested a complex crosstalk among genetic variants, allele-specific binding of transcription factors (ABTF), and allele-specific DNA methylation patterns (ASM), as well as environmental factors for disease risk. This review aims to summarize the current studies regarding the interactions of the aforementioned factors with a focus on epigenetic insights. We present two scenarios of single nucleotide polymorphisms (SNPs) in coding regions and non-coding regions for disease risk, via potentially impacting epigenetic patterns. While a SNP in a coding region may confer disease risk via altering protein functions, a SNP in non-coding region may cause diseases, via SNP-altering ABTF, ASM, and allele-specific gene expression (ASE). The allelic increases or decreases of gene expression are key for disease risk during development. Such ASE can be achieved via either a "SNP-introduced ABTF to ASM" or a "SNP-introduced ASM to ABTF." Together with our additional in-depth review on insulator CTCF, we are convinced to propose a working model that the small effect of a SNP acts through altered ABTF and/or ASM, for ASE and eventual disease outcome (named as a "SNP intensifier" model). In summary, the significance of complex crosstalk among genetic factors, epigenetic patterns, and environmental factors requires further investigations for disease susceptibility.
Collapse
Affiliation(s)
- Huishan Wang
- Laboratory of Human Environmental Epigenome, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Dan Lou
- Laboratory of Human Environmental Epigenome, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Zhibin Wang
- Laboratory of Human Environmental Epigenome, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
26
|
Aberg KA, Shabalin AA, Chan RF, Zhao M, Kumar G, van Grootheest G, Clark SL, Xie LY, Milaneschi Y, Penninx BWJH, van den Oord EJCG. Convergence of evidence from a methylome-wide CpG-SNP association study and GWAS of major depressive disorder. Transl Psychiatry 2018; 8:162. [PMID: 30135428 PMCID: PMC6105579 DOI: 10.1038/s41398-018-0205-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 06/04/2018] [Accepted: 06/10/2018] [Indexed: 01/19/2023] Open
Abstract
DNA methylation is an epigenetic modification that provides stability and diversity to the cellular phenotype. It is influenced by both genetic sequence variation and environmental factors, and can therefore potentially account for variation of heritable phenotypes and disorders. Therefore, methylome-wide association studies (MWAS) are promising complements to genome-wide association studies (GWAS) of genetic variants. Of particular interest are methylation sites (CpGs) that are created or destroyed by the alleles of single-nucleotide polymorphisms (SNPs), as these so-called CpG-SNPs may show variation in methylation levels on top of what can be explained by the sequence variation. Using sequencing-based data from 1132 major depressive disorder (MDD) cases and controls, we performed a MWAS of 970,414 common CpG-SNPs. The analysis identified 27 suggestively significant (P < 1.00 × 10-5) CpG-SNPs associations. Furthermore, the MWAS results were over-represented (odds ratios ranging 1.36-5.00; P ranging 4.9 × 10-3-8.1 × 10-2) among findings from three recent GWAS for MDD-related phenotypes. Overlapping loci included, e.g., ROBO2, ASIC2, and DCC. As the CpG-SNP analysis accounts for the number of alleles that creates CpGs, the methylation differences could not be explained by differences in allele frequencies. Thus, the results show that the MWAS and GWASs provide independent lines of evidence for the involvement of these loci in MDD. In conclusion, our methylation study of MDD contributes novel information about loci of relevance that complements previous findings and generates new hypothesis about MDD etiology, such as that the functional effects of genetic association may be partly mediated and/or enhanced by the methylation status in these loci.
Collapse
Affiliation(s)
- Karolina A. Aberg
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Andrey A. Shabalin
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Robin F. Chan
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Min Zhao
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Gaurav Kumar
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Gerard van Grootheest
- 0000 0004 0435 165Xgrid.16872.3aDepartment of Psychiatry, Amsterdam Neuroscience, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Shaunna L. Clark
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Lin Y. Xie
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| | - Yuri Milaneschi
- 0000 0004 0435 165Xgrid.16872.3aDepartment of Psychiatry, Amsterdam Neuroscience, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Brenda W. J. H. Penninx
- 0000 0004 0435 165Xgrid.16872.3aDepartment of Psychiatry, Amsterdam Neuroscience, VU University Medical Center/GGZ inGeest, Amsterdam, The Netherlands
| | - Edwin J. C. G. van den Oord
- 0000 0004 0458 8737grid.224260.0Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA USA
| |
Collapse
|
27
|
Yazd NKK, Patel RR, Dellavalle RP, Dunnick CA. Genetic Risk Factors for Development of Atopic Dermatitis: a Systematic Review. CURRENT DERMATOLOGY REPORTS 2017. [DOI: 10.1007/s13671-017-0199-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|