1
|
Reales G, Amos CI, Benveniste O, Chinoy H, De Bleecker J, De Paepe B, Doria A, Gregersen PK, Lamb JA, Limaye V, Lundberg IE, Machado PM, Maurer B, Miller FW, Molberg Ø, Pachman LM, Padyukov L, Radstake TR, Reed AM, Rider LG, Rothwell S, Selva-O'Callaghan A, Vencovský J, Wedderburn LR, Wallace C. Discovery of new myositis genetic associations through leveraging other immune-mediated diseases. HGG ADVANCES 2024; 5:100336. [PMID: 39044428 PMCID: PMC11350499 DOI: 10.1016/j.xhgg.2024.100336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Genome-wide association studies (GWASs) have been successful at finding associations between genetic variants and human traits, including the immune-mediated diseases (IMDs). However, the requirement of large sample sizes for discovery poses a challenge for learning about less common diseases, where increasing volunteer numbers might not be feasible. An example of this is myositis (or idiopathic inflammatory myopathies [IIM]s), a group of rare, heterogeneous autoimmune diseases affecting skeletal muscle and other organs, severely impairing life quality. Here, we applied a feature engineering method to borrow information from larger IMD GWASs to find new genetic associations with IIM and its subgroups. Combining this approach with two clustering methods, we found 17 IMDs genetically close to IIM, including some common comorbid conditions, such as systemic sclerosis and Sjögren's syndrome, as well as hypo- and hyperthyroidism. All IIM subtypes were genetically similar within this framework. Next, we colocalized IIM signals that overlapped IMD signals, and found seven potentially novel myositis associations mapped to immune-related genes, including BLK, IRF5/TNPO3, and ITK/HAVCR2, implicating a role for both B and T cells in IIM. This work proposes a new paradigm of genetic discovery in rarer diseases by leveraging information from more common IMD, and can be expanded to other conditions and traits beyond IMD.
Collapse
Affiliation(s)
- Guillermo Reales
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| | | | - Olivier Benveniste
- Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière Hospital, Paris, France
| | - Hector Chinoy
- Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK; Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jan De Bleecker
- Department of Neurology, Ghent University, Ghent, Belgium; Neuromuscular Reference Center, Ghent University Hospital, Ghent, Belgium
| | - Boel De Paepe
- Department of Neurology, Ghent University, Ghent, Belgium; Neuromuscular Reference Center, Ghent University Hospital, Ghent, Belgium
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Peter K Gregersen
- The Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute, Manhasset, NY, USA
| | - Janine A Lamb
- Epidemiology and Public Health Group, Division of Population Health, Health Services Research & Primary Care, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Vidya Limaye
- Rheumatology Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Discipline of Medicine, Adelaide University, Adelaide, South Australia, Australia
| | - Ingrid E Lundberg
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Pedro M Machado
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology. London, UK; Centre for Rheumatology, UCL Division of Medicine, University College London, London, UK
| | - Britta Maurer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Frederick W Miller
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, NIH, Bethesda, MD, USA
| | - Øyvind Molberg
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Lauren M Pachman
- Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leonid Padyukov
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Timothy R Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center, Utrecht, the Netherlands
| | - Ann M Reed
- Department of Pediatrics, Duke University, Durham, NC, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, NIH, Bethesda, MD, USA
| | - Simon Rothwell
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Albert Selva-O'Callaghan
- Internal Medicine Department, Vall d'Hebron General Hospital, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Jiri Vencovský
- Institute of Rheumatology and Department of Rheumatology, First Medical Faculty, Charles University, Prague, Czech Republic
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis, UCL Great Ormond Street Institute of Child Health, University College London, London, UK; NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Chris Wallace
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
2
|
Shin JE, Shin N, Park T, Park M. Multipartite network analysis to identify environmental and genetic associations of metabolic syndrome in the Korean population. Sci Rep 2024; 14:20283. [PMID: 39217223 PMCID: PMC11366034 DOI: 10.1038/s41598-024-71217-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Network analysis has become a crucial tool in genetic research, enabling the exploration of associations between genes and diseases. Its utility extends beyond genetics to include the assessment of environmental factors. Unipartite network analysis is commonly used in genomics to visualize initial insights and relationships among variables. Syndromic diseases, such as metabolic syndrome, are characterized by the simultaneous occurrence of various signs, symptoms, and clinicopathological features. Metabolic syndrome encompasses hypertension, diabetes, obesity, and dyslipidemia, and both genetic and environmental factors contribute to its development. Given that relevant data often consist of distinct sets of variables, a more intuitive visualization method is needed. This study applied multipartite network analysis as an effective method to understand the associations among genetic, environmental, and disease components in syndromic diseases. We considered three distinct variable sets: genetic factors, environmental factors, and disease components. The process involved projecting a tripartite network onto a two-mode bipartite network and then simplifying it into a one-mode network. This approach facilitated the visualization of relationships among factors across different sets and within individual sets. To transition from multipartite to unipartite networks, we suggest both sequential and concurrent projection methods. Data from the Korean Association Resource (KARE) project were utilized, including 352,228 SNPs from 8840 individuals, alongside information on environmental factors such as lifestyle, dietary, and socioeconomic factors. The single-SNP analysis step filtered SNPs, supplemented by reference SNPs reported in a genome-wide association study catalog. The resulting network patterns differed significantly by sex: demographic factors and fat intake were crucial for women, while alcohol consumption was central for men. Indirect relationships were identified through projected bipartite networks, revealing that SNPs such as rs4244457, rs2156552, and rs10899345 had lifestyle interactions on metabolic components. Our approach offers several advantages: it simplifies the visualization of complex relationships among different datasets, identifies environmental interactions, and provides insights into SNP clusters sharing common environmental factors and metabolic components. This framework provides a comprehensive approach to elucidate the mechanisms underlying complex diseases like metabolic syndrome.
Collapse
Affiliation(s)
- Ji-Eun Shin
- Department of Biomedical Informatics, Konyang University, Daejeon, Republic of Korea
| | - Nari Shin
- Department of Statistics, Korea University, Seoul, Republic of Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, Republic of Korea
| | - Mira Park
- Department of Preventive Medicine, Eulji University, Daejeon, Republic of Korea.
| |
Collapse
|
3
|
Chiu TY, Lazar DC, Wang WW, Wozniak JM, Jadhav AM, Li W, Gazaniga N, Theofilopoulos AN, Teijaro JR, Parker CG. Chemoproteomic development of SLC15A4 inhibitors with anti-inflammatory activity. Nat Chem Biol 2024; 20:1000-1011. [PMID: 38191941 PMCID: PMC11228132 DOI: 10.1038/s41589-023-01527-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 12/14/2023] [Indexed: 01/10/2024]
Abstract
SLC15A4 is an endolysosome-resident transporter linked with autoinflammation and autoimmunity. Specifically, SLC15A4 is critical for Toll-like receptors (TLRs) 7-9 as well as nucleotide-binding oligomerization domain-containing protein (NOD) signaling in several immune cell subsets. Notably, SLC15A4 is essential for the development of systemic lupus erythematosus in murine models and is associated with autoimmune conditions in humans. Despite its therapeutic potential, the availability of quality chemical probes targeting SLC15A4 functions is limited. In this study, we used an integrated chemical proteomics approach to develop a suite of chemical tools, including first-in-class functional inhibitors, for SLC15A4. We demonstrate that these inhibitors suppress SLC15A4-mediated endolysosomal TLR and NOD functions in a variety of human and mouse immune cells; we provide evidence of their ability to suppress inflammation in vivo and in clinical settings; and we provide insights into their mechanism of action. Our findings establish SLC15A4 as a druggable target for the treatment of autoimmune and autoinflammatory conditions.
Collapse
Affiliation(s)
- Tzu-Yuan Chiu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Daniel C Lazar
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Wesley W Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Jacob M Wozniak
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Appaso M Jadhav
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Weichao Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Nathalia Gazaniga
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | | | - John R Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| | | |
Collapse
|
4
|
Pujol-Giménez J, Baumann SP, Ho TM, Augustynek B, Hediger MA. Functional Characterization of the Lysosomal Peptide/Histidine Transporter PHT1 ( SLC15A4) by Solid Supported Membrane Electrophysiology (SSME). Biomolecules 2024; 14:771. [PMID: 39062485 PMCID: PMC11275134 DOI: 10.3390/biom14070771] [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: 06/07/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The peptide/histidine transporter PHT1 (SLC15A4) is expressed in the lysosomal membranes of immune cells where it plays an important role in metabolic and inflammatory signaling. PHT1 is an H+-coupled/histidine symporter that can transport a wide range of oligopeptides, including a variety of bacterial-derived peptides. Moreover, it enables the scaffolding of various metabolic signaling molecules and interacts with key regulatory elements of the immune response. Not surprisingly, PHT1 has been implicated in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE). Unfortunately, the pharmacological development of PHT1 modulators has been hampered by the lack of suitable transport assays. To address this shortcoming, a novel transport assay based on solid-supported membrane-based electrophysiology (SSME) is presented. Key findings of the present SSME studies include the first recordings of electrophysiological properties, a pH dependence analysis, an assessment of PHT1 substrate selectivity, as well as the transport kinetics of the identified substrates. In contrast to previous work, PHT1 is studied in its native lysosomal environment. Moreover, observed substrate selectivity is validated by molecular docking. Overall, this new SSME-based assay is expected to contribute to unlocking the pharmacological potential of PHT1 and to deepen the understanding of its functional properties.
Collapse
Affiliation(s)
- Jonai Pujol-Giménez
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Sven P. Baumann
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Tin Manh Ho
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Bartlomiej Augustynek
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Matthias A. Hediger
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| |
Collapse
|
5
|
Dong M, Li P, Luo J, Chen B, Jiang H. Oligopeptide/Histidine Transporter PHT1 and PHT2 - Function, Regulation, and Pathophysiological Implications Specifically in Immunoregulation. Pharm Res 2023; 40:2585-2596. [PMID: 37610621 DOI: 10.1007/s11095-023-03589-8] [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: 05/28/2023] [Accepted: 08/15/2023] [Indexed: 08/24/2023]
Abstract
The oligopeptide/histidine transporters PHT1 and PHT2, two mammalian solute carrier family 15A proteins, mediate the transmembrane transport of histidine and some di/tripeptides via proton gradient. PHT1 and PHT2 are distributed in a variety of tissues but are preferentially expressed in immune cells and localize to the lysosome-related organelles. Studies have reported the relationships between PHT1/PHT2 and immune diseases. PHT1 and PHT2 participate in the regulation of lysosomal homeostasis and lysosome-associated signaling pathways through their transport and nontransport functions, playing important roles in inflammatory diseases. In this review, we summarize recent research on PHT1 and PHT2, aiming to provide reference for their further biological research and as targets for drug design.
Collapse
Affiliation(s)
- Minlei Dong
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Li
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Luo
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Binxin Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Huidi Jiang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Xiang J, Keep RF. Proton-Coupled Oligopeptide Transport (Slc15) in the Brain: Past and Future Research. Pharm Res 2023; 40:2533-2540. [PMID: 37308743 DOI: 10.1007/s11095-023-03550-9] [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: 05/10/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
This mini-review describes the role of the solute carrier (SLC)15 family of proton-coupled oligopeptide transporters (POTs) and particularly Pept2 (Slc15A2) and PhT1 (Slc15A4) in the brain. That family transports endogenous di- and tripeptides and peptidomimetics but also a number of drugs. The review focuses on the pioneering work of David E. Smith in the field in identifying the impact of PepT2 at the choroid plexus (the blood-CSF barrier) as well as PepT2 and PhT1 in brain parenchymal cells. It also discusses recent findings and future directions in relation to brain POTs including cellular and subcellular localization, regulatory pathways, transporter structure, species differences and disease states.
Collapse
Affiliation(s)
- Jianming Xiang
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
7
|
Kobayashi T, Toyama-Sorimachi N. Metabolic control from the endolysosome: lysosome-resident amino acid transporters open novel therapeutic possibilities. Front Immunol 2023; 14:1243104. [PMID: 37781390 PMCID: PMC10540624 DOI: 10.3389/fimmu.2023.1243104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Amino acid transporters are generally recognized as machinery that transport amino acids from the extracellular environment into the cytoplasm. Although their primary function is the uptake of amino acids to supply the cell with nutrients and energy, endolysosome-resident amino acid (EL-aa) transporters possess several unique functions in accordance with their localization in intracellular vesicular membranes. They play pivotal roles in the maintenance of metabolic homeostasis via direct involvement in the amino acid sensing pathway, which regulates the activity of mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of cellular metabolism. Additionally, some EL-aa transporters contribute to the maintenance of dynamic homeostasis of endolysosomes, including the regulation of endolysosomal acidity, by carrying amino acids out of endolysosomes. In addition, EL-aa transporters act as a scaffold to gather signaling molecules and multiple enzymes to control cellular metabolism on the endolysosomal membrane. Among EL-aa transporters, solute carrier family 15 member 4 (SLC15A4) is preferentially expressed in immune cells, including macrophages, dendritic cells, and B cells, and plays a key role in the integration of metabolic and inflammatory signals. In this review, we summarize our recent findings on EL-aa transporter contributions to inflammatory and metabolic signaling in the endolysosomes of immune cells by focusing on the SLC15 family, including SLC15A4 and SLC15A3, and discuss their uniqueness and universality. We also discuss the potential of targeting these EL-aa transporters in immune cells for the development of novel therapeutic strategies for inflammatory diseases. Because these transporters are highly expressed in immune cells and significantly alter the functions of immune cells, targeting them would provide a great advantage in ensuring a wide safety margin.
Collapse
Affiliation(s)
| | - Noriko Toyama-Sorimachi
- Division of Human Immunology, International Research and Development Center for Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
8
|
Crow MK. Pathogenesis of systemic lupus erythematosus: risks, mechanisms and therapeutic targets. Ann Rheum Dis 2023; 82:999-1014. [PMID: 36792346 DOI: 10.1136/ard-2022-223741] [Citation(s) in RCA: 89] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023]
Abstract
Research elucidating the pathogenesis of systemic lupus erythematosus (SLE) has defined two critical families of mediators, type I interferon (IFN-I) and autoantibodies targeting nucleic acids and nucleic acid-binding proteins, as fundamental contributors to the disease. On the fertile background of significant genetic risk, a triggering stimulus, perhaps microbial, induces IFN-I, autoantibody production or most likely both. When innate and adaptive immune system cells are engaged and collaborate in the autoimmune response, clinical SLE can develop. This review describes recent data from genetic analyses of patients with SLE, along with current studies of innate and adaptive immune function that contribute to sustained IFN-I pathway activation, immune activation and autoantibody production, generation of inflammatory mediators and tissue damage. The goal of these studies is to understand disease mechanisms, identify therapeutic targets and stimulate development of therapeutics that can achieve improved outcomes for patients.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
9
|
Shu Y, Ma X, Chen C, Wang Y, Sun X, Zhang L, Lu Z, Petersen F, Qiu W, Yu X. Myelin oligodendrocyte glycoprotein-associated disease is associated with BANK1, RNASET2 and TNIP1 polymorphisms. J Neuroimmunol 2022; 372:577937. [PMID: 36054934 DOI: 10.1016/j.jneuroim.2022.577937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/01/2022] [Accepted: 07/25/2022] [Indexed: 12/31/2022]
Abstract
AIM Here we aimed to compare association of common immune-related genetic variants with three autoimmune central nervous system (CNS) demyelinating diseases, namely myelin oligodendrocyte glycoprotein-associated disease (MOGAD), multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). METHODS In this retrospective cross-sectional study, 26 common immune-related single nucleotide polymorphisms were genotyped in 102 patients with MOGAD, 100 patients with MS, 198 patients with NMOSD and 541 healthy control subjects recruited from Guangzhou, China. RESULTS Among all tested genetic variations, one polymorphism, B cell scaffold protein with ankyrin repeats 1 (BANK1) rs4522865 was associated with multiple disorders, namely MOGAD (OR = 1.94, 95% CI:1.19-3.17, P = 0.0059) and NMOSD (OR = 1.69, 95% CI:1.17-2.45). Besides BANK1 rs4522865, two other non-HLA loci, ribonuclease T2 (RNASET2) rs9355610 (OR = 0.47, 95% CI: 0.26-0.85) and TNFAIP3 interacting protein 1 (TNIP1) rs10036748 (OR = 1.76, 95% CI: 1.16-2.71), were associated with MOGAD. In addition, NMOSD was associated with signal transducer and activator of transcription 4 (STAT4) rs7574865 (OR = 1.58, 95% CI: 1.12-2.24) and general transcription factor Iii (GTF2I) rs73366469 (OR = 1.60, 95% CI:1.12-2.29), while MS was associated with a killer cell lectin like receptor G1 (KLRG1) rs1805673 (OR = 0.61, 95% CI: 0.40-0.94) and T-box transcription factor 21 (TBX21) rs17244587 (OR = 2.25, 95% CI: 1.25-4.06). CONCLUSION The current study suggests for the first time three non-HLA susceptibility loci for MOGAD. In addition, comparison of association of 26 immune-related polymorphisms with three autoimmune CNS demyelinating diseases demonstrates substantial difference in genetic basis of those disorders.
Collapse
Affiliation(s)
- Yaqing Shu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyu Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chen Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaobo Sun
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Liang Zhang
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Frank Petersen
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Xinhua Yu
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.
| |
Collapse
|
10
|
Skopelitou D, Srivastava A, Miao B, Kumar A, Dymerska D, Paramasivam N, Schlesner M, Lubinski J, Hemminki K, Försti A, Reddy Bandapalli O. Whole exome sequencing identifies novel germline variants of SLC15A4 gene as potentially cancer predisposing in familial colorectal cancer. Mol Genet Genomics 2022; 297:965-979. [PMID: 35562597 PMCID: PMC9250485 DOI: 10.1007/s00438-022-01896-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 04/02/2022] [Indexed: 11/25/2022]
Abstract
About 15% of colorectal cancer (CRC) patients have first-degree relatives affected by the same malignancy. However, for most families the cause of familial aggregation of CRC is unknown. To identify novel high-to-moderate-penetrance germline variants underlying CRC susceptibility, we performed whole exome sequencing (WES) on four CRC cases and two unaffected members of a Polish family without any mutation in known CRC predisposition genes. After WES, we used our in-house developed Familial Cancer Variant Prioritization Pipeline and identified two novel variants in the solute carrier family 15 member 4 (SLC15A4) gene. The heterozygous missense variant, p. Y444C, was predicted to affect the phylogenetically conserved PTR2/POT domain and to have a deleterious effect on the function of the encoded peptide/histidine transporter. The other variant was located in the upstream region of the same gene (GRCh37.p13, 12_129308531_C_T; 43 bp upstream of transcription start site, ENST00000266771.5) and it was annotated to affect the promoter region of SLC15A4 as well as binding sites of 17 different transcription factors. Our findings of two distinct variants in the same gene may indicate a synergistic up-regulation of SLC15A4 as the underlying genetic cause and implicate this gene for the first time in genetic inheritance of familial CRC.
Collapse
Affiliation(s)
- Diamanto Skopelitou
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Aayushi Srivastava
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Beiping Miao
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Abhishek Kumar
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104 India
| | - Dagmara Dymerska
- Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Nagarajan Paramasivam
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Kari Hemminki
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
| | - Asta Försti
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Obul Reddy Bandapalli
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
11
|
Fu Y, Lin Q, Zhang ZR. Association of TNFSF4 polymorphisms with systemic lupus erythematosus: a meta-analysis. Adv Rheumatol 2021; 61:59. [PMID: 34538280 DOI: 10.1186/s42358-021-00215-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/02/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To more precisely estimate the association between the tumor necrosis factor ligand superfamily member 4 (TNFSF4) gene polymorphisms and systemic lupus erythematosus (SLE) susceptibility, we performed a meta-analysis on the association of the following single nucleotide polymorphisms (SNPs) of TNFSF4 with SLE: rs1234315, rs844648, rs2205960, rs704840, rs844644, rs10489265. METHODS A literature-based search was conducted using PubMed, MEDLINE, Embase, Web of Science databases, and Cochrane Library databases to identify all relevant studies. And the association of TNFSF4 gene polymorphisms and SLE susceptibility was evaluated by pooled odds ratio (OR) with 95% confidence interval (CI). RESULTS The meta-analysis produced overall OR of 1.42 (95% CI 1.36-1.49, P < 0.00001), 1.41 (95% CI 1.36-1.46, P < 0.00001) and 1.34 (95% CI 1.26-1.42, P < 0.00001) for the rs2205960, rs1234315 and rs704840 polymorphisms respectively, confirming these three SNPs confer a significant risk for the development of SLE. On the other hand, the meta-analysis produced overall OR of 0.92 (95% CI 0.70-1.21, P = 0.54) for the rs844644 polymorphism, suggesting no significant association. And no association was also found between either rs844648 1.11 (OR 1.11, 95% CI 0.86-1.43, P = 0.41) or rs10489265 (OR 1.17, 95% CI 0.94-1.47, P = 0.17) polymorphism with SLE susceptibility, respectively. CONCLUSIONS Our meta-analysis demonstrated that the TNFSF4 rs2205960, rs1234315 and rs844840 SNPs was significantly associated with an increased risk of SLE.
Collapse
Affiliation(s)
- Yu Fu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.,Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Zhi-Rong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
12
|
Zeng Z, Sun QQ, Zhang W, Wen QW, Wang TH, Qin W, Xiao DM, Zhang Z, Huang H, Mo YJ, Wu XD, Cen H. Assessment of genetic polymorphisms within nuclear factor-κB signaling pathway genes in rheumatoid arthritis: Evidence for replication and genetic interaction. Int Immunopharmacol 2021; 100:108089. [PMID: 34464884 DOI: 10.1016/j.intimp.2021.108089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/07/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE This study was performed to replicate the associations of genetic polymorphisms within nuclear factor-κB (NF-κB) signaling pathway genes with rheumatoid arthritis (RA), and to further examine genetic interactions in a Chinese population. METHODS A total of eleven single-nucleotide polymorphisms (SNPs) were genotyped in 594 RA patients and 604 healthy controls. RESULTS Genetic association analysis revealed that NFKBIE rs2233434, TNIP1 rs10036748 and BLK rs13277113 were significantly associated with RA, cyclic citrullinated peptide (CCP)-positive RA and rheumatoid factor (RF)-positive RA, and TNFAIP3 rs2230926 was significantly associated with CCP-positive RA. Significant additive interaction was observed between NFKB1 rs28362491 and IKBKE rs12142086 (RERI = 0.76, 95% CI 0.13-1.38; AP = 0.57, 95% CI 0.11-1.03), NFKBIE rs2233434 and BLK rs13277113 (RERI = 1.41, 95% CI 0.88-1.94; AP = 0.85, 95% CI 0.50-1.20), NFKBIL rs2071592 and TNIP1 rs10036748 (RERI = 0.59, 95% CI 0.17-1.02; AP = 0.46, 95% CI 0.05-0.87), UBE2L3 rs5754217 and TNFSF4 rs2205960 (RERI = 0.50, 95% CI 0.16-0.84; AP = 0.57, 95% CI 0.09-1.05). Significant multiplicative interaction was detected between BLK rs13277113 and UBE2L3 rs5754217 (p = 0.02), BLK rs13277113 and TNFSF4 rs2205960 (p = 0.03). CONCLUSIONS Our results lent further support to the role of NF-κB signaling pathway in the pathogenesis of RA from a genetic perspective.
Collapse
Affiliation(s)
- Zhen Zeng
- Department of Preventive Medicine, Medical School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China; Zhejiang Provincial Key Laboratory of Pathophysiology, Medical School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Qing-Qing Sun
- Department of Health Education and Chronic Disease Prevention, Yinzhou District Center for Disease Control and Prevention, 1221 Xueshi Road, Ningbo, Zhejiang, 315100, PR China
| | - Wei Zhang
- Department of Preventive Medicine, Medical School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China; Zhejiang Provincial Key Laboratory of Pathophysiology, Medical School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Qin-Wen Wen
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Ting-Hui Wang
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Wen Qin
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Dong-Mei Xiao
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Zhen Zhang
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Hua Huang
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Yi-Jun Mo
- Department of Clinical Laboratory, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Xiu-Di Wu
- Department of Rheumatology, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, 59 Liuting Road, Ningbo, Zhejiang, 315010, PR China
| | - Han Cen
- Department of Preventive Medicine, Medical School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China; Zhejiang Provincial Key Laboratory of Pathophysiology, Medical School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China.
| |
Collapse
|
13
|
Katewa A, Suto E, Hui J, Heredia J, Liang J, Hackney J, Anderson K, Alcantar TM, Bacarro N, Dunlap D, Eastham J, Paler-Martinez A, Rairdan XY, Modrusan Z, Lee WP, Austin CD, Lafkas D, Ghilardi N. The peptide symporter SLC15a4 is essential for the development of systemic lupus erythematosus in murine models. PLoS One 2021; 16:e0244439. [PMID: 33444326 PMCID: PMC7808665 DOI: 10.1371/journal.pone.0244439] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/09/2020] [Indexed: 11/18/2022] Open
Abstract
Systemic Lupus Erythematosus (SLE) is a chronic autoimmune disease representing a serious unmet medical need. The disease is associated with the loss of self-tolerance and exaggerated B cell activation, resulting in autoantibody production and the formation of immune complexes that accumulate in the kidney, causing glomerulonephritis. TLR7, an important mediator of the innate immune response, drives the expression of type-1 interferon (IFN), which leads to expression of type-1 IFN induced genes and aggravates lupus pathology. Because the lysosomal peptide symporter slc15a4 is critically required for type-1 interferon production by pDC, and for certain B cell functions in response to TLR7 and TLR9 signals, we considered it as a potential target for pharmacological intervention in SLE. We deleted the slc15a4 gene in C57BL/6, NZB, and NZW mice and found that pristane-challenged slc15a4-/- mice in the C57BL/6 background and lupus prone slc15a4-/- NZB/W F1 mice were both completely protected from lupus like disease. In the NZB/W F1 model, protection persisted even when disease development was accelerated with an adenovirus encoding IFNα, emphasizing a broad role of slc15a4 in disease initiation. Our results establish a non-redundant function of slc15a4 in regulating both innate and adaptive components of the immune response in SLE pathobiology and suggest that it may be an attractive drug target.
Collapse
Affiliation(s)
- Arna Katewa
- Dept. Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, United States of America
| | - Eric Suto
- Dept. Translational Immunology, Genentech, South San Francisco, CA, United States of America
| | - Jessica Hui
- Evercore ISI, New York, NY, United States of America
| | - Jose Heredia
- Dept. Immunology, Genentech, South San Francisco, CA, United States of America
| | - Jie Liang
- Dept. Molecular Oncology, Genentech, South San Francisco, CA, United States of America
| | - Jason Hackney
- Dept. Bioinformatics, Genentech, South San Francisco, CA, United States of America
| | - Keith Anderson
- Dept. Molecular Biology, Genentech, South San Francisco, CA, United States of America
| | - Tuija M. Alcantar
- Dept. Molecular Biology, Genentech, South San Francisco, CA, United States of America
| | - Natasha Bacarro
- Dept. Molecular Biology, Genentech, South San Francisco, CA, United States of America
| | - Debra Dunlap
- Dept. Pathology, Genentech, South San Francisco, CA, United States of America
| | - Jeffrey Eastham
- Dept. Pathology, Genentech, South San Francisco, CA, United States of America
| | - Andres Paler-Martinez
- Dept. Translational Immunology, Genentech, South San Francisco, CA, United States of America
| | - Xin Y. Rairdan
- gRED Animal Resources, South San Francisco, CA, United States of America
| | - Zora Modrusan
- Dept. Microchemistry, Proteomics, & Lipidomics, Genentech, South San Francisco, CA, United States of America
| | - Wyne P. Lee
- Dept. Translational Immunology, Genentech, South San Francisco, CA, United States of America
| | - Cary D. Austin
- Dept. Pathology, Genentech, South San Francisco, CA, United States of America
| | - Daniel Lafkas
- Dept. Immunology, Genentech, South San Francisco, CA, United States of America
| | - Nico Ghilardi
- DiCE Molecules, South San Francisco, CA, United States of America
| |
Collapse
|
14
|
Moreno-Eutimio MA, Martínez-Alemán CE, Aranda-Uribe IS, Aquino-Jarquin G, Cabello-Gutierrez C, Fragoso JM, Barbosa-Cobos RE, Saavedra MA, Ramírez-Bello J. TNFSF4 is a risk factor to systemic lupus erythematosus in a Latin American population. Clin Rheumatol 2020; 40:929-939. [PMID: 32809147 DOI: 10.1007/s10067-020-05332-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/03/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The aim of this study was to examine the association of three TNFSF4 single nucleotide variants (SNVs) with systemic lupus erythematosus (SLE) susceptibility in Mexican patients. METHODS Genotypes of the TNFSF4 rs1234315T/C, rs2205960G/T, and rs704840T/G SNVs were determined using a TaqMan assay. In our study, we included 395 patients with SLE and 500 controls. RESULTS Our information shows a significant difference in the allelic and genotypic frequency of the three TNFSF4 SNVs between cases and controls. Thus, our data showed an association between TNFSF4 rs1234315T/C (T vs. C, OR 1.40, p = 0.00087), rs2205960G/T (G vs. T, OR 1.32, p = 0.0037), and rs704840T/G (T vs. G, OR 1.41, p = 0.0003) and SLE susceptibility in Mexican subjects. Besides, we conducted a meta-analysis to determine the role of TNFSF4 rs2205960G/T and SLE susceptibility; our results showed that this variant is a risk factor for SLE in Latin Americans and Asians. CONCLUSION Our results show that TNFSF4 rs1234315T/C, rs2205960G/T, and rs704840T/G are risk factors to SLE in Mexicans. This is the first study to document an association between TNFSF4 rs704840T/G and SLE in a Latin American population. In addition, our meta-analysis showed that TNFSF4 rs2205960G/T is a risk factor for Asians and Latin Americans. Key Point • The TNFSF4 rs1234315T/C, rs2205960G/T, and rs704849T/G SNVs are risk factors to SLE in patients from Mexico.
Collapse
Affiliation(s)
| | | | | | - Guillermo Aquino-Jarquin
- Laboratorio de Investigación en Genómica, Genética y Bioinformática, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Carlos Cabello-Gutierrez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - José Manuel Fragoso
- Departamento de Biología Molecular, Instituto Nacional de Cardiología, Mexico City, Mexico
| | | | | | | |
Collapse
|
15
|
Kim T, Bae SC, Kang C. Synergistic activation of NF-κB by TNFAIP3 (A20) reduction and UBE2L3 (UBCH7) augment that synergistically elevate lupus risk. Arthritis Res Ther 2020; 22:93. [PMID: 32334614 PMCID: PMC7183688 DOI: 10.1186/s13075-020-02181-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/02/2020] [Indexed: 02/03/2023] Open
Abstract
Background Systemic lupus erythematosus (SLE) is an autoimmune inflammatory rheumatic disease. SLE susceptibility is affected by multiple genetic elements, environmental factors, and their interactions. We aimed in this study to statistically and functionally characterize a gene-gene interaction (epistasis) recently documented to affect SLE risk. Methods Two single-nucleotide polymorphisms, rs2230926 in TNFAIP3 (A20) gene and rs131654 in UBE2L3 (UBCH7) gene, were genotyped in all 3525 Korean participants, and their SLE risk association and epistasis were statistically analyzed by calculating odds ratio (OR), 95% confidence interval (CI), and P values in genotype comparisons between 1318 SLE patients and 2207 healthy controls. Furthermore, their effects on gene functions were assessed by comparatively examining separate and combined effects of TNFAIP3 and UBE2L3 knockdowns on NF-κB transcription factor activity in human cells. Results SLE susceptibility is associated with TNFAIP3 rs2230926 (OR = 1.9, 95% CI 1.6–2.4, P = 8.6 × 10−11) and UBE2L3 rs131654 (OR = 1.2, 95% CI 1.1–1.4, P = 1.1 × 10−4) in a Korean population of this study. Their risk-associated alleles synergistically elevate SLE susceptibility in both multivariate logistic regression analysis (ORinteraction = 1.6, P = 0.0028) and genotype-stratified analysis (ORinteraction = 2.4), confirming the synergistic TNFAIP3-UBE2L3 interaction in SLE risk. Additionally, the SLE-susceptible alleles confer decreased TNFAIP3 expression (P = 1.1 × 10−6, n = 610) and increased UBE2L3 expression (P = 9.5 × 10−11, n = 475), respectively, in B cell analysis of the International HapMap Project individuals with adjustment for ethnicity. Furthermore, when compared with TNFAIP3 non-knockdown and UBE2L3 knockdown in human HeLa cells, TNFAIP3 knockdown and UBE2L3 non-knockdown synergistically increase three cytokines, CCL2, CXCL8 (IL8), and IL6, all regulated by NF-κB in the human TNFR signaling pathway. Conclusions A synergistic interaction between TNFAIP3 and UBE2L3 genes is observed in SLE risk, as being evident in comparison of genotype distributions between SLE patients and controls. Additionally, the synergistic gene-gene interaction is functionally validated, as TNFAIP3 reduction and UBE2L3 augment exert synergism in activation of NF-κB and subsequent induction of inflammatory cytokines. Accordingly, SLE inflammation and risk could be synergistically alleviated by TNFAIP3 upregulation and UBE2L3 downregulation.
Collapse
Affiliation(s)
- Taehyeung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| | - Changwon Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
16
|
Yang Y, Li X, Li B, Mu L, Wang J, Cheng Y, Gu Y, Wu H. Associations between TNFSF4 gene polymorphisms (rs2205960 G > A, rs704840 T > G and rs844648 G > A) and susceptibility to autoimmune diseases in Asians: a meta-analysis. Immunol Invest 2020; 50:184-200. [PMID: 32208776 DOI: 10.1080/08820139.2020.1718693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Tumor necrosis factor superfamily member 4 (TNFSF4) has significant role in modulating autoimmune diseases (ADs) and single nucleotide polymorphism (SNP) is also related with the susceptibility to some diseases. So a meta-analysis aimed at systematically assessing the associations between TNFSF4 polymorphisms (rs2205960 G > A, rs704840 T > G and rs844648 G > A) and ADs risk was performed in Asians. METHODS Total 14 eligible articles published before March 2019 involving 35 studies, of which 21 studies (16,109 cases and 26,378 controls) for rs2205960 G > A, 8 studies (2,424 cases and 3,692 controls) for rs704840 T > G, and 6 studies (3,839 cases and 5,867 controls) for rs844648 G > A were included. Effects of the three respective polymorphisms on the susceptibility to ADs were estimated by pooling the odds ratios (ORs) with their corresponding 95% confidence interval (95% CI) in allelic, dominant, recessive, heterozygous and homozygous models. RESULTS The overall analysis revealed that all the rs2205960 G > A, rs704840 T > G and rs844648 G > A polymorphisms could increase the risk of ADs in allelic, dominant, recessive, heterozygous and homozygous models. Furthermore, subgroup analysis showed that both rs2205960 G > A and rs704840 T > G were significantly associated with the susceptibility to systemic lupus erythematosus (SLE). What's more, statistically significant association between rs2205960 G > A polymorphism and primary Sjögren's syndrome (pSS) susceptibility was also observed in allelic, dominant and heterozygous models. CONCLUSIONS This current meta-analysis suggested that all of the three TNFSF4 polymorphisms may be associated with ADs susceptibility in Asians.
Collapse
Affiliation(s)
- Yangyang Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| | - Xiahui Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| | - Bowen Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| | - Liying Mu
- School of Life Science and Medicine, Dalian University of Technology , Panjin, China
| | - Jin Wang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| | - Yunmeng Cheng
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| | - Yao Gu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology , Dalian, China
| |
Collapse
|
17
|
Griffith AD, Zaidi AK, Pietro A, Hadiono M, Yang JS, Davis R, Popkin DL. A requirement for slc15a4 in imiquimod-induced systemic inflammation and psoriasiform inflammation in mice. Sci Rep 2018; 8:14451. [PMID: 30262916 PMCID: PMC6160456 DOI: 10.1038/s41598-018-32668-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/04/2018] [Indexed: 01/08/2023] Open
Abstract
There is competing evidence that plasmacytoid dendritic cells (pDC), the most potent source of IFN-I, may initiate psoriasis. We targeted pDC function using the slc15a4feeble loss-of-function mouse whose pDC are unresponsive to TLR agonists. slc15a4feeble treated with the topical TLR7-agonist imiquimod (IMQ) demonstrated decreased epidermal thickening 24 hours post-treatment which was more pronounced by day 5 as compared to wildtype mice. These findings were specific to the acute IMQ model and not the protracted IL23 model that drives inflammation downstream of TLR activation. Systemically, slc15a4 was required for IMQ-induced weight loss and cutaneous accumulation of CD4+ and Siglec H+, but not CD11b+ cells. Consistent with this phenotype and the function of slc15a4, induction of IFN-I was virtually absent systemically and via cutaneous gene expression. Induction of other inflammatory cytokines (cytokine storm) was modestly blunted in slc15a4feeble except for inflammasome-associated genes consistent with slc15a4 being required for TLR7-mediated (but not inflammasome-mediated) inflammation downstream of IMQ. Surprisingly, only IFN-I gene expression was suppressed within IMQ-treated skin. Other genes including conserved psoriasiform trademark gene expression were augmented in slc15a4feeble versus littermate controls. Taken together, we have identified a role for slc15a4 but not canonical psoriasiform genes in the imiquimod model of psoriasiform dermatitis.
Collapse
Affiliation(s)
- Alexis D Griffith
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA
| | - Asifa K Zaidi
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA
| | - Ashley Pietro
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA
| | - Matthew Hadiono
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA
| | - Jessica S Yang
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA
| | - Rachel Davis
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA
| | - Daniel L Popkin
- Department of Dermatology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA. .,Departments of Dermatology, Pathology, Molecular Biology and Microbiology, Case Western Reserve University Hospitals, Cleveland, OH, 44106, USA.
| |
Collapse
|
18
|
Wang Y, Hu Y, Li P, Weng Y, Kamada N, Jiang H, Smith DE. Expression and regulation of proton-coupled oligopeptide transporters in colonic tissue and immune cells of mice. Biochem Pharmacol 2018; 148:163-173. [PMID: 29305856 PMCID: PMC5801143 DOI: 10.1016/j.bcp.2017.12.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/29/2017] [Indexed: 02/06/2023]
Abstract
A number of studies have implicated proton-coupled oligopeptide transporters (POTs) in the initiation and/or progression of inflammatory bowel disease and immune cell signaling. With this in mind, the aim of this study was to delineate the expression of POTs in mouse colonic tissues and immune cells, and characterize the potential role of these transporters in nucleotide-binding oligomerization domain (NOD) signaling. Using a dextran sodium sulfate (DSS)-induced colitis mouse model, we found that DSS down regulated Pht1 gene expression and up regulated Pht2 gene expression in colonic tissue and immune cells. In contrast, PEPT1 protein was absent from the colonic tissue and immune cells of normal and DSS-treated mice. NOD ligands, muramyl dipeptide (MDP) and l-Ala-γ-d-Glu-meso-diaminopimelic acid (tri-DAP), were shown to be substrates of PHT2 in MDCK-hPHT219,20AA cells. Subsequent studies revealed that the immune response of lamina propia mononuclear cells may be regulated by PHT1 and PHT2, and that PHT2 facilitated the NOD-dependent immune response in RAW264.7 macrophages. These results clarified the expression of POTs in mouse colonic segments, cells and subtypes, and the role of increased Pht2 expression during chemically-induced colitis in facilitating NOD-dependent immune response. The findings further suggest that intestinal PHT2 may serve as a therapeutic target for IBD therapy.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA; Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Yongjun Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Ping Li
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Yayun Weng
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Nobuhiko Kamada
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - David E Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
19
|
Song F, Hu Y, Wang Y, Smith DE, Jiang H. Functional Characterization of Human Peptide/Histidine Transporter 1 in Stably Transfected MDCK Cells. Mol Pharm 2018; 15:385-393. [PMID: 29224352 DOI: 10.1021/acs.molpharmaceut.7b00728] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The proton-coupled oligopeptide transporter PHT1 (SLC15A4), which facilitates cross-membrane transport of histidine and small peptides from inside the endosomes or lysosomes to cytosol, plays an important role in intracellular peptides homeostasis and innate immune responses. However, it remains a challenge to elucidate functional properties of the PHT1 transporter because of its subcellular localization. The purpose of this study was to resort hPHT1 protein from the subcellular to outer cell membrane of MDCK cells stably transfected with human PHT1 mutants, and to characterize its functional activity in these cells. Using this model, the functional activity of hPHT1 was evaluated by cellular uptake studies with d3-l-histidine, GlySar, and the bacterial peptidoglycan products MDP and Tri-DAP. We found that the disruption of two dileucine motifs was indispensable for hPHT1 transporter being preferentially targeting to plasma membranes. hPHT1 showed high affinity for d3-l-histidine and low affinity for GlySar, with Km values of 16.3 ± 1.9 μM and 1.60 ± 0.30 mM, respectively. Moreover, the bacterial peptidoglycan components MDP and Tri-DAP were shown conclusively to be hPHT1 substrates. The uptake of MDP by hPHT1 was inhibited by di/tripeptides and peptide-like drugs, but not by glycine and acyclovir. The functional activity of hPHT1 was also pH-dependent, with an optimal cellular uptake in buffer pH 6.5. Taken together, we established a novel cell model to evaluate the function of hPHT1 in vitro, and confirmed that MDP and Tri-DAP were substrates of hPHT1. Our findings suggest that PHT1 may serve as a potential target for reducing the immune responses and for drug treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Feifeng Song
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University , Zhejiang 310058, China
| | - Yongjun Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Yuqing Wang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University , Zhejiang 310058, China
| | - David E Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University , Zhejiang 310058, China
| |
Collapse
|
20
|
Shen Y, Liu Y, Wang XQ, Ke X, Kang HY, Hong SL. Association between TNFSF4 and BLK gene polymorphisms and susceptibility to allergic rhinitis. Mol Med Rep 2017; 16:3224-3232. [PMID: 28713926 PMCID: PMC5547929 DOI: 10.3892/mmr.2017.6954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 04/12/2017] [Indexed: 02/06/2023] Open
Abstract
Allergic rhinitis (AR) is a common inflammatory disease of the upper airway. Recent evidence suggests that gene‑gene interactions between tumor necrosis factor receptor superfamily 4 (TNFSF4) and B cell lymphocyte kinase (BLK) may have a synergistic effect on T and B cells in determining immunologic aberration, via the nuclear factor‑κB pathway. The present study was performed to evaluate the potential association between specific single nucleotide polymorphisms (SNPs) in the TNFSF4 and BKL genes with susceptibility to AR in Chinese subjects. A population‑based case‑control study was performed in 600 Chinese AR patients and 700 controls. Blood was drawn for DNA extraction, and 9 SNPs (6 in TNFSF4 and 3 in BKL genes) were selected and genotyped. The TNFSF4 SNPs rs1234314 and rs1234315, and the BLK SNPs rs13277113 and rs1600249 were observed to occur in different frequencies between the AR patients and the controls. The CC (rs1234314, rs1234315) and AA (rs1600249, rs13277113) genotypes provided protective effects against AR, whereas the AG (rs13277113) genotype presented a risk factor for AR. The haplotypes ACC in the rs1234313‑rs1234314‑rs1234315 block and GA in the rs2254546‑rs13277113 block significantly decreased the risk of AR, whereas the GGT and AG haplotypes served protective roles. SNP interaction analysis further indicated that there may be synergistic effects among the selected sets of polymorphisms. The present study suggests a novel association between specific TNFSF4 and BLK gene polymorphisms and AR risk, highlighting their potential utility as genetic biomarkers for AR susceptibility in a Chinese Han population.
Collapse
Affiliation(s)
- Yang Shen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yun Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiao-Qiang Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xia Ke
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hou-Yong Kang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Su-Ling Hong
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
21
|
Teruel M, Chamberlain C, Alarcón-Riquelme ME. Omics studies: their use in diagnosis and reclassification of SLE and other systemic autoimmune diseases. Rheumatology (Oxford) 2017; 56:i78-i87. [PMID: 28339517 DOI: 10.1093/rheumatology/kew339] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Indexed: 12/18/2022] Open
Abstract
Omics studies of systemic autoimmune diseases (SADs) in general, and SLE in particular, have delivered isolated information from transcriptome, epigenome, genome, cytokine and metabolome analyses. Such analyses have resulted in the identification of disease susceptibility genes and the description of IFN expression signatures, allowing extensive insight into the mechanisms of disease and the development of new therapies. Access to such technologies allows the recognition of patterns of disease at a pathway level, thereby, to reclassify SLE and other SADs and to develop new therapeutics from a personalized perspective. The use of omic information allows the discovery of correlative patterns involving drugs not currently suspected to be of value in SADs. In this review, we summarize the omics findings for SLE and propose ways of using the data for the identification of new biomarkers, finding new drugs and reclassifying patients not only with SLE, but also with other SADs.
Collapse
Affiliation(s)
- Maria Teruel
- Parque Tecnológico de la Salud, Medical Genomics, Centre Pfizer, University of Granada, Andalusian Regional Government for Genomics and Oncological Research, Granada, Spain
| | | | - Marta E Alarcón-Riquelme
- Parque Tecnológico de la Salud, Medical Genomics, Centre Pfizer, University of Granada, Andalusian Regional Government for Genomics and Oncological Research, Granada, Spain.,Chronic Inflammatory Diseases Unit, Institute for Environmental Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
22
|
Dang J, Li J, Xin Q, Shan S, Bian X, Yuan Q, Liu N, Ma X, Li Y, Liu Q. Gene-gene interaction of ATG5, ATG7, BLK and BANK1 in systemic lupus erythematosus. Int J Rheum Dis 2016; 19:1284-1293. [PMID: 26420661 DOI: 10.1111/1756-185x.12768] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIM Autophagy-related gene 5 (ATG5), ATG7, B-lymphoid tyrosine kinase (BLK) and B-cell scaffold protein with ankyrin repeats 1 (BANK1) are involved in B-cell signaling; several genome-wide association studies detected these genes as candidates involved in systemic lupus erythematosus (SLE). We aimed to replicate the association of these genes with SLE in Chinese Han and to search for possible gene-gene interactions. METHODS TaqMan single-nucleotide polymorphism (SNP) genotyping was used to detect rs548234, rs665791 in ATG5, rs11706903 in ATG7, rs2736340 in BLK and rs10516487 in BANK1 in 382 SLE patients and 660 healthy controls. The epistasis effect was analyzed by logistic regression, multifactor dimensionality reduction (MDR) and linear regression analysis. RESULTS SLE was associated with frequency of rs548234 (P = 0.010; odds ratio [OR] = 1.298), rs2736340 (P = 2.47 × 10-5 ; OR = 1.574) and rs10516487 (P = 0.002; OR = 0.642). Although no epistasis effects were found among three autophagy-related gene loci or with rs2736340 and rs10516487, BLK and BANK1 had the closest interaction effect on logistic regression analysis (P = 0.013; OR = 1.205), MDR (P < 0.0001), and linear regression analysis (P = 0.0017; R2 = 0.1806). The risk genotype TT of rs2736340 was associated with decreased messenger RNA level of BLK; BLK transcript level was lower in SLE patients than healthy controls. CONCLUSION We confirmed the association of rs548234, rs2736340 and rs10516487 with SLE in Chinese Han and reinforced our hypothesis of their epistasis effect in regulating B-cell signaling in SLE.
Collapse
Affiliation(s)
- Jie Dang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
- Department of Medical Genetics and Cell Biology, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jiangxia Li
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qian Xin
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Shan Shan
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xianli Bian
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qianqian Yuan
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Na Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xiaochun Ma
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Yan Li
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qiji Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, Shandong, China
| |
Collapse
|
23
|
Zhang MY, Yang XK, Pan HF, Ye DQ. Associations between TNFAIP3 gene polymorphisms and systemic lupus erythematosus risk: an updated meta-analysis. HLA 2016; 88:245-252. [PMID: 27726311 DOI: 10.1111/tan.12908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/07/2016] [Accepted: 09/15/2016] [Indexed: 11/29/2022]
Abstract
In order to determine whether tumor necrosis factor alpha inducible protein 3 (TNFAIP3) gene polymorphisms confers susceptibility to systemic lupus erythematosus (SLE) in ethnically different populations. A meta-analysis was conducted to examine the association between TNFAIP3 polymorphisms and susceptibility to SLE. A systematic literature search was conducted to identify all relevant studies. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were used to estimate the strength of the association. A total of 23 studies from 20 articles, involving 18,501 patients with SLE and 30,435 healthy controls were included in this meta-analysis. Overall, we found significant association between SLE and the TNFAIP3 rs2230926, rs5029937, rs5029939, and rs3757173 polymorphisms (all P < 0.001). Stratification by ethnicity indicated that rs5029939 polymorphism was associated with SLE in Europeans, while rs2230926, rs5029937, and rs3757173 polymorphisms were associated with SLE both in Europeans and Asians (all P < 0.001). The results of our meta-analysis suggest that TNFAIP3 (rs2230926, rs5029937, rs5029939, and rs3757173) polymorphisms are associated with susceptibility to SLE.
Collapse
Affiliation(s)
- M-Y Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, PR China
| | - X-K Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, PR China
| | - H-F Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, PR China
| | - D-Q Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
24
|
Chua KH, Ooh YY, Chai HC. TNFSF4 polymorphisms are associated with systemic lupus erythematosus in the Malaysian population. Int J Immunogenet 2016; 43:303-9. [PMID: 27519474 DOI: 10.1111/iji.12287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 06/01/2016] [Accepted: 07/21/2016] [Indexed: 02/05/2023]
Abstract
Tumour necrosis factor superfamily 4 (TNFSF4) gene has been reported to be associated with systemic lupus erythematosus (SLE) susceptibility due to its encoding for OX40L protein that can increase autoantibody production and cause imbalance of T-cell proliferation. The purpose of this study was to investigate the association of TNFSF4 rs2205960, rs1234315, rs8446748 and rs704840 with SLE in the Malaysian population. A total of 476 patients with SLE and 509 healthy controls were recruited. Real-time polymerase chain reaction (PCR) was applied to genotype the selected single nucleotide polymorphisms (SNPs). Allelic and genotypic frequencies of each SNP were calculated for each ethnic group, and association test was performed using logistic regression. The overall association of each SNP in Malaysian patients with SLE was determined with meta-analysis. The frequency of minor T allele of TNFSF4 rs2205960 was significant in Chinese and Indian patients with SLE, with P values of 0.05 (OR = 1.27, 95% CI: 1.00-1.61) and 0.004 (OR = 3.16, 95% CI: 1.41-7.05), respectively. Significant association of minor G allele of rs704840 with SLE was also observed in Chinese (P = 0.03, OR = 1.26, 95% CI: 1.02-1.56). However, after Bonferroni correction, only T allele of rs2205960 remained significantly associated with Indian cohort. Overall, minor G allele of rs704840 showed significant association with SLE in the Malaysian population with P values of 0.05 (OR = 1.20, 95% CI: 1.00-1.43). We suggested TNFSF4 rs704840 could be the potential SLE risk factors in the Malaysian population.
Collapse
Affiliation(s)
- K H Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Y Y Ooh
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - H C Chai
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Teruel M, Alarcón-Riquelme ME. The genetic basis of systemic lupus erythematosus: What are the risk factors and what have we learned. J Autoimmun 2016; 74:161-175. [PMID: 27522116 DOI: 10.1016/j.jaut.2016.08.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 08/02/2016] [Indexed: 12/19/2022]
Abstract
The genome-wide association study is a free-hypothesis approach based on screening of thousands or even millions of genetic variants distributed throughout the whole human genome in relation to a phenotype. The relevant role of the genome-wide association studies in the last decade is undisputed because it has permitted to elucidate multiple risk genetic factors associated with the susceptibility to several human complex diseases. Regarding systemic lupus erythematosus (SLE) this approach has allowed to identify more than 60 risk loci for SLE susceptibility across populations to date, increasing our understanding on the pathogenesis of this disease. We present the latest findings in the genetic of SLE across populations using genome-wide approaches. These studies revealed that most of the genetic risk is shared across borders and ethnicities. Finally, we focus on describing the most important risk loci for SLE attempting to cover the genetic findings in relation to functional polymorphisms, such as missense single nucleotide polymorphisms (SNPs) or regulatory variants involved in the development of the disease. The functional studies try to identify the causality of some GWAS-associated variants, many of which fall in non-coding regions of the genome, suggesting a regulatory role. Many loci show an environmental interaction, another aspect revealed by the studies of epigenetic modifications and those associated with genetic variants. Finally, new-generation sequencing technologies can open other paths in the research on SLE genetics, the role of rare variants and the detailed identification of causal regulatory variation. The clinical relevance of the genetic factors will be shown when we are able to use them or in combination with other molecular measurements to re-classify a heterogeneous disease such as SLE.
Collapse
Affiliation(s)
- Maria Teruel
- Center for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Government, PTS, Granada, 18016, Spain.
| | - Marta E Alarcón-Riquelme
- Center for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Government, PTS, Granada, 18016, Spain; Institute of Environmental Medicine, Karolinska Institute, Stockholm, 171 67, Sweden.
| |
Collapse
|
26
|
Zhang M, Chen F, Zhang D, Zhai Z, Hao F. Association Study Between SLC15A4 Polymorphisms and Haplotypes and Systemic Lupus Erythematosus in a Han Chinese Population. Genet Test Mol Biomarkers 2016; 20:451-8. [PMID: 27362648 PMCID: PMC4991581 DOI: 10.1089/gtmb.2015.0289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Objective: The gene SLC15A4 (solute carrier family 15 [oligopeptide transporter], member 4) has been reported as contributing to the pathogenesis of systemic lupus erythematosus (SLE). We performed a case–control replication study to investigate further the association between single-nucleotide polymorphisms (SNPs) in the SLC15A4 gene and systemic SLE in a Han Chinese population. Methods: In Han Chinese SLE patients and healthy individuals (n = 355, 375, respectively), 18 SNPs in the SLC15A4 gene were genotyped using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and TaqMan SNP genotyping assays. Analyses of allele frequencies and genotypes using codominant, dominant, and recessive models were conducted, as well as a linkage disequilibrium analysis. P values < 0.05 were considered significant. Results: Allele frequencies of five of the analyzed SNPs were significantly associated with SLE. Under a codominant model the genotype frequencies of rs3765108 AG and rs7308691 AT were significantly higher in the SLE group than the control group (p = 0.019, 0.049, respectively). Under a dominant model the rs1385374 (TT+CT) SNP carried a higher risk of SLE than (CC) (p = 0.042). One SLC15A4 haplotype (TA), which consists of 2 SNPs (rs959989 and rs983492), was associated with SLE (p = 0.024). Conclusion: Our study determined that five SNPs (rs959989, rs1385374, rs983492, rs12298615, and rs10847697) are associated with SLE. Thus, SLC15A4 may be important in the pathogenesis of SLE in Han Chinese patients.
Collapse
Affiliation(s)
- Mingwang Zhang
- 1 Department of Dermatology, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Fangru Chen
- 2 Department of Dermatology, Affiliated Hospital of Guilin Medical College , Guilin, China
| | - Dongmei Zhang
- 1 Department of Dermatology, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Zhifang Zhai
- 1 Department of Dermatology, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Fei Hao
- 1 Department of Dermatology, Southwest Hospital, Third Military Medical University , Chongqing, China
| |
Collapse
|
27
|
G'Sell RT, Gaffney PM, Powell DW. A20-Binding Inhibitor of NF-κB Activation 1 is a Physiologic Inhibitor of NF-κB: A Molecular Switch for Inflammation and Autoimmunity. Arthritis Rheumatol 2015; 67:2292-302. [PMID: 26097105 DOI: 10.1002/art.39245] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/09/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Rachel T G'Sell
- University of Louisville School of Medicine, Louisville, Kentucky
| | | | - David W Powell
- University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
28
|
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder that has a broad spectrum of effects on the majority of organs, including the kidneys. Approximately 40-70% of patients with SLE will develop lupus nephritis. Renal assault during SLE is initiated by genes that breach immune tolerance and promote autoantibody production. These genes might act in concert with other genetic factors that augment innate immune signalling and IFN-I production, which in turn can generate an influx of effector leucocytes, inflammatory mediators and autoantibodies into end organs, such as the kidneys. The presence of cognate antigens in the glomerular matrix, together with intrinsic molecular abnormalities in resident renal cells, might further accentuate disease progression. This Review discusses the genetic insights and molecular mechanisms for key pathogenic contributors in SLE and lupus nephritis. We have categorized the genes identified in human studies of SLE into one of four pathogenic events that lead to lupus nephritis. We selected these categories on the basis of the cell types in which these genes are expressed, and the emerging paradigms of SLE pathogenesis arising from murine models. Deciphering the molecular basis of SLE and/or lupus nephritis in each patient will help physicians to tailor specific therapies.
Collapse
|
29
|
Shen C, Liu L, Jiang Z, Zheng X, Meng L, Yin X, Gao J, Sheng Y, Gao J, Li Y, Zhou F, Xiao F, Sun L, Cui Y, Yang S, Zuo X, Zhang X. Four genetic variants interact to confer susceptibility to atopic dermatitis in Chinese Han population. Mol Genet Genomics 2015; 290:1493-8. [DOI: 10.1007/s00438-015-1014-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 01/27/2015] [Indexed: 12/14/2022]
|
30
|
Lee HS, Bae SC. Recent advances in systemic lupus erythematosus genetics in an Asian population. Int J Rheum Dis 2014; 18:192-9. [DOI: 10.1111/1756-185x.12498] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hye-Soon Lee
- Hanyang University Hospital for Rheumatic Diseases; Seoul Korea
| | - Sang Cheol Bae
- Hanyang University Hospital for Rheumatic Diseases; Seoul Korea
| |
Collapse
|