1
|
Hao K, Marshak-Rothstein A. Nucleic acid triggers of autoimmunity and autoinflammation. Curr Opin Immunol 2025; 93:102535. [PMID: 39889356 DOI: 10.1016/j.coi.2025.102535] [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: 12/11/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
The key role of nucleic acid sensing receptors in the development of autoimmune and autoinflammatory diseases is becoming increasingly apparent. Activation of these sensors has been attributed to the failure of professional scavenger cells to adequately clear cell debris, in many cases due to defective scavenger receptors. However, as now summarized in this review, numerous gain-of-function mutations in the nucleic acid sensing receptors, or in molecules that regulate sensor activity, have now been evaluated in gene-targeted murine strains, and critical components of their downstream pathways have been identified as therapeutic targets. In addition, we are beginning to understand how DNases and RNases play crucial roles in both generating and eliminating the distinct ligands that engage the various nucleic acid sensors. Murine models of disease have further provided important insights regarding the function of and synergy between individual endosomal and cytosolic receptors, as well as cell type restricted functions.
Collapse
Affiliation(s)
- Kaiyuan Hao
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01604, USA
| | | |
Collapse
|
2
|
Ma Y, Mayes MD, Guo X, Assassi S, Zhou X. Lack of association of the PLD4 SNP rs2841277 with systemic sclerosis in a European American population. Sci Rep 2024; 14:31068. [PMID: 39730870 DOI: 10.1038/s41598-024-82298-7] [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: 04/09/2024] [Accepted: 12/04/2024] [Indexed: 12/29/2024] Open
Abstract
This study aimed to examine whether a reported SSc-associated SNP rs2841277 in the PLD4 gene identified in an Asian population was also associated with SSc in European American (EA). The EA cohort consisting of 1005 SSc patients and 961 healthy controls was examined in this study. TaqMan genotyping assays were performed to examine the SNP. Exact P-values were obtained from 2 × 2 tables of allele counts and disease status. In contrast to the previous reports in a Japanese population, SSc patients of EA did not show an association of PLD4 rs2841277 with SSc in general (P = 0.231, OR = 0.89, 95% CI = 0.73-1.08), or with clinical subtypes of dcSSc (P = 0.302, OR = 0.86, 95% CI = 0.65-1.13) and lcSSc (P = 0.369, OR = 0.90, 95% CI = 0.72-1.12), or with autoantibody subtypes including ATA (P = 0.126, OR = 0.74, 95% CI = 0.51-1.08), ACA (P = 0.943, OR = 1.01, 95% CI = 0.77-1.34), ARP3 (P = 0.155, OR = 0.77, 95% CI = 0.53-1.1), or Anti-RNP (P = 0.660, OR = 0.73, 95% CI = 0.29-1.84). We found a lack of association of the PLD4 SNP rs2841277 with SSc in an EA population. This is the first study to report a discrepancy in the genetic association between the PLD4 SNP and SSc. This may be explained by genetic heterogeneity between Japanese and EA populations, with genetic ancestry contributing to this variation. Further verification in diverse ancestral populations is warranted.
Collapse
Affiliation(s)
- Yunqing Ma
- Department of Internal Clinical Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Maureen D Mayes
- Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston, Houston, 77030, USA
| | - Xinjian Guo
- Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston, Houston, 77030, USA
| | - Shervin Assassi
- Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston, Houston, 77030, USA
| | - Xiaodong Zhou
- Internal Medicine/Rheumatology, University of Texas Health Science Center at Houston, Houston, 77030, USA.
| |
Collapse
|
3
|
Samulevich ML, Carman LE, Aneskievich BJ. Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders. Biomedicines 2024; 12:2723. [PMID: 39767629 PMCID: PMC11726985 DOI: 10.3390/biomedicines12122723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Successful skin wound healing is dependent on an interplay between epidermal keratinocytes and dermal fibroblasts as they react to local extracellular factors (DAMPs, PAMPs, cytokines, etc.) surveyed from that environment by numerous membrane receptors (e.g., TLRs, cytokine receptors, etc.). In turn, those receptors are the start of a cytoplasmic signaling pathway where balance is key to effective healing and, as needed, cell and matrix regeneration. When directed through NF-κB, these signaling routes lead to transient responses to the benefit of initiating immune cell recruitment, cell replication, local chemokine and cytokine production, and matrix protein synthesis. The converse can also occur, where ongoing canonical NF-κB activation leads to chronic, hyper-responsive states. Here, we assess three key players, TAK1, TNFAIP3, and TNIP1, in cytoplasmic regulation of NF-κB activation, which, because of their distinctive and yet inter-related functions, either promote or limit that activation. Their balanced function is integral to successful wound healing, given their significant control over the expression of inflammation-, fibrosis-, and matrix remodeling-associated genes. Intriguingly, these three proteins have also been emphasized in dysregulated NF-κB signaling central to systemic sclerosis (SSc). Notably, diffuse SSc shares some tissue features similar to an excessive inflammatory/fibrotic wound response without eventual resolution. Taking a cue from certain instances of aberrant wound healing and SSc having some shared aspects, e.g., chronic inflammation and fibrosis, this review looks for the first time, to our knowledge, at what those pathologies might have in common regarding the cytoplasmic progression of NF-κB-mediated signaling. Additionally, while TAK1, TNFAIP3, and TNIP1 are often investigated and reported on individually, we propose them here as three proteins whose consequences of function are very highly interconnected at the signaling focus of NF-κB. We thus highlight the emerging promise for the eventual clinical benefit derived from an improved understanding of these integral signal progression modulators. Depending on the protein, its indirect or direct pharmacological regulation has been reported. Current findings support further intensive studies of these points in NF-κB regulation both for their basic function in healthy cells as well as with the goal of targeting them for translational benefit in multiple cutaneous wound healing situations, whether stemming from acute injury or a dysregulated inflammatory/fibrotic response.
Collapse
Affiliation(s)
- Michael L. Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (M.L.S.); (L.E.C.)
| | - Liam E. Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (M.L.S.); (L.E.C.)
| | - Brian J. Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
4
|
Sun R, Ma T, Zhao Z, Gao Y, Feng J, Yang X. Phospholipase D Family Member 4 Regulates Microglial Phagocytosis and Remyelination via the AKT Pathway in a Cuprizone-Induced Multiple Sclerosis Mouse Model. CNS Neurosci Ther 2024; 30:e70111. [PMID: 39548665 PMCID: PMC11567942 DOI: 10.1111/cns.70111] [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: 07/14/2024] [Revised: 09/30/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024] Open
Abstract
AIMS Remyelination is an endogenous repair process that is often deficient in multiple sclerosis (MS). Stimulation of remyelination is thought to help limit the progression of MS. This study aimed to investigate the expression pattern and function of a microglial phagocytosis-related gene, phospholipase D family member 4 (PLD4), in a cuprizone (CPZ)-induced MS mouse model. METHODS The extent of remyelination was assessed using LFB staining. Myelin phagocytosis assay was used to investigate the effect of Pld4 on microglial phagocytic activity. RESULTS Pld4 was upregulated in the corpus callosum during demyelination and remyelination. AAV9-mediated Pld4 deficiency impaired remyelination and reduced the number of Olig2-positive cells. In the corpus callosum of Pld4-deficient mice, the microglial phagocytosis marker MAC2 was reduced, accompanied by inhibition of TrkA/AKT signaling. Similarly, the phagocytosis assay showed that Pld4 knockdown significantly inhibited myelin debris phagocytosis by BV2 cells. The AKT activator SC79 reversed the Pld4 deficiency-induced inhibition of microglial phagocytic activity and rescued the impaired remyelination in Pld4-deficient mice. CONCLUSION PLD4 is upregulated in CPZ-induced MS and modulates microglial phagocytosis and remyelination via the AKT pathway. Our findings provide experimental evidence for a better understanding of the molecular mechanism of MS.
Collapse
Affiliation(s)
- Ran Sun
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Tengyun Ma
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Zheng Zhao
- Department of Emergency MedicineShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Yan Gao
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Juan Feng
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Xue Yang
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| |
Collapse
|
5
|
Luo Y, Khan A, Liu L, Lee CH, Perreault GJ, Pomenti SF, Gourh P, Kiryluk K, Bernstein EJ. Cross-Phenotype GWAS Supports Shared Genetic Susceptibility to Systemic Sclerosis and Primary Biliary Cholangitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.01.24309721. [PMID: 39006426 PMCID: PMC11245064 DOI: 10.1101/2024.07.01.24309721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Objective An increased risk of primary biliary cholangitis (PBC) has been reported in patients with systemic sclerosis (SSc). Our study aims to investigate the shared genetic susceptibility between the two disorders and to define candidate causal genes using cross-phenotype GWAS meta-analysis. Methods We performed cross-phenotype GWAS meta-analysis and colocalization analysis for SSc and PBC. We performed both genome-wide and locus-based analysis, including tissue and pathway enrichment analyses, fine-mapping, colocalization analyses with expression quantitative trait loci (eQTL) and protein quantitative trait loci (pQTL) datasets, and phenome-wide association studies (PheWAS). Finally, we used an integrative approach to prioritize candidate causal genes from the novel loci. Results We detected a strong genetic correlation between SSc and PBC (rg = 0.84, p = 1.7 × 10-6). In the cross-phenotype GWAS meta-analysis, we identified 44 non-HLA loci that reached genome-wide significance (p < 5 × 10-8). Evidence of shared causal variants between SSc and PBC was found for nine loci, five of which were novel. Integrating multiple sources of evidence, we prioritized CD40, ERAP1, PLD4, SPPL3, and CCDC113 as novel candidate causal genes. The CD40 risk locus colocalized with trans-pQTLs of multiple plasma proteins involved in B cell function. Conclusion Our study supports a strong shared genetic susceptibility between SSc and PBC. Through cross-phenotype analyses, we have prioritized several novel candidate causal genes and pathways for these disorders.
Collapse
Affiliation(s)
- Yiming Luo
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lili Liu
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Cue Hyunkyu Lee
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| | - Gabriel J Perreault
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Sydney F Pomenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Pravitt Gourh
- Scleroderma Genomics and Health Disparities Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Elana J Bernstein
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
6
|
Yuan M, Peng L, Huang D, Gavin A, Luan F, Tran J, Feng Z, Zhu X, Matteson J, Wilson IA, Nemazee D. Structural and mechanistic insights into disease-associated endolysosomal exonucleases PLD3 and PLD4. Structure 2024; 32:766-779.e7. [PMID: 38537643 PMCID: PMC11162324 DOI: 10.1016/j.str.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/12/2024] [Accepted: 02/29/2024] [Indexed: 04/09/2024]
Abstract
Endolysosomal exonucleases PLD3 and PLD4 (phospholipases D3 and D4) are associated with autoinflammatory and autoimmune diseases. We report structures of these enzymes, and the molecular basis of their catalysis. The structures reveal an intra-chain dimer topology forming a basic active site at the interface. Like other PLD superfamily members, PLD3 and PLD4 carry HxKxxxxD/E motifs and participate in phosphodiester-bond cleavage. The enzymes digest ssDNA and ssRNA in a 5'-to-3' manner and are blocked by 5'-phosphorylation. We captured structures in apo, intermediate, and product states and revealed a "link-and-release" two-step catalysis. We also unexpectedly demonstrated phosphatase activity via a covalent 3-phosphohistidine intermediate. PLD4 contains an extra hydrophobic clamp that stabilizes substrate and could affect oligonucleotide substrate preference and product release. Biochemical and structural analysis of disease-associated mutants of PLD3/4 demonstrated reduced enzyme activity or thermostability and the possible basis for disease association. Furthermore, these findings provide insight into therapeutic design.
Collapse
Affiliation(s)
- Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Deli Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Amanda Gavin
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Fangkun Luan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jenny Tran
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ziqi Feng
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jeanne Matteson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
7
|
Gumkowska-Sroka O, Kotyla K, Kotyla P. Immunogenetics of Systemic Sclerosis. Genes (Basel) 2024; 15:586. [PMID: 38790215 PMCID: PMC11121022 DOI: 10.3390/genes15050586] [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: 04/07/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Systemic sclerosis (SSc) is a rare autoimmune connective tissue disorder characterized by massive fibrosis, vascular damage, and immune imbalance. Advances in rheumatology and immunology over the past two decades have led to a redefinition of systemic sclerosis, shifting from its initial perception as primarily a "hyperfibrotic" state towards a recognition of systemic sclerosis as an immune-mediated disease. Consequently, the search for genetic markers has transitioned from focusing on fibrotic mechanisms to exploring immune regulatory pathways. Immunogenetics, an emerging field at the intersection of immunology, molecular biology, and genetics has provided valuable insights into inherited factors that influence immunity. Data from genetic studies conducted thus far indicate that alterations in genetic messages can significantly impact disease risk and progression. While certain genetic variations may confer protective effects, others may exacerbate disease susceptibility. This paper presents a comprehensive review of the most relevant genetic changes that influence both the risk and course of systemic sclerosis. Special emphasis is placed on factors regulating the immune response, recognizing their pivotal role in the pathogenesis of the disease.
Collapse
Affiliation(s)
| | | | - Przemysław Kotyla
- Department of Rheumatology and Clinical Immunology, Medical University of Silesia, Voivodeship Hospital No. 5, 41-200 Sosnowiec, Poland; (O.G.-S.); (K.K.)
| |
Collapse
|
8
|
Ishikawa Y, Tanaka N, Asano Y, Kodera M, Shirai Y, Akahoshi M, Hasegawa M, Matsushita T, Saito K, Motegi SI, Yoshifuji H, Yoshizaki A, Kohmoto T, Takagi K, Oka A, Kanda M, Tanaka Y, Ito Y, Nakano K, Kasamatsu H, Utsunomiya A, Sekiguchi A, Niiro H, Jinnin M, Makino K, Makino T, Ihn H, Yamamoto M, Suzuki C, Takahashi H, Nishida E, Morita A, Yamamoto T, Fujimoto M, Kondo Y, Goto D, Sumida T, Ayuzawa N, Yanagida H, Horita T, Atsumi T, Endo H, Shima Y, Kumanogoh A, Hirata J, Otomo N, Suetsugu H, Koike Y, Tomizuka K, Yoshino S, Liu X, Ito S, Hikino K, Suzuki A, Momozawa Y, Ikegawa S, Tanaka Y, Ishikawa O, Takehara K, Torii T, Sato S, Okada Y, Mimori T, Matsuda F, Matsuda K, Amariuta T, Imoto I, Matsuo K, Kuwana M, Kawaguchi Y, Ohmura K, Terao C. GWAS for systemic sclerosis identifies six novel susceptibility loci including one in the Fcγ receptor region. Nat Commun 2024; 15:319. [PMID: 38296975 PMCID: PMC10830486 DOI: 10.1038/s41467-023-44541-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024] Open
Abstract
Here we report the largest Asian genome-wide association study (GWAS) for systemic sclerosis performed to date, based on data from Japanese subjects and comprising of 1428 cases and 112,599 controls. The lead SNP is in the FCGR/FCRL region, which shows a penetrating association in the Asian population, while a complete linkage disequilibrium SNP, rs10917688, is found in a cis-regulatory element for IRF8. IRF8 is also a significant locus in European GWAS for systemic sclerosis, but rs10917688 only shows an association in the presence of the risk allele of IRF8 in the Japanese population. Further analysis shows that rs10917688 is marked with H3K4me1 in primary B cells. A meta-analysis with a European GWAS detects 30 additional significant loci. Polygenic risk scores constructed with the effect sizes of the meta-analysis suggest the potential portability of genetic associations beyond populations. Prioritizing the top 5% of SNPs of IRF8 binding sites in B cells improves the fitting of the polygenic risk scores, underscoring the roles of B cells and IRF8 in the development of systemic sclerosis. The results also suggest that systemic sclerosis shares a common genetic architecture across populations.
Collapse
Affiliation(s)
- Yuki Ishikawa
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Nao Tanaka
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihide Asano
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Dermatology, The University of Tokyo, Tokyo, Japan
| | - Masanari Kodera
- Department of Dermatology, Chukyo Hospital, Japan Community Health Care Organization, Nagoya, Japan
| | - Yuichiro Shirai
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
- Department of Rheumatology, Saga University Hospital, Saga, Japan
| | - Minoru Hasegawa
- Faculty of Medical Sciences, Department of Dermatology, University of Fukui, Fukui, Japan
| | - Takashi Matsushita
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kazuyoshi Saito
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hajime Yoshifuji
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, The University of Tokyo, Tokyo, Japan
| | - Tomohiro Kohmoto
- Aichi Cancer Center Research Institute, Division of Molecular Genetics, Nagoya, Japan
| | - Kae Takagi
- Tokyo Women's Medical University, Adachi Medical Center, Tokyo, Japan
| | - Akira Oka
- Department of Molecular Life Sciences, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Miho Kanda
- Department of Dermatology, Chukyo Hospital, Japan Community Health Care Organization, Nagoya, Japan
| | - Yoshihito Tanaka
- Department of Dermatology, Chukyo Hospital, Japan Community Health Care Organization, Nagoya, Japan
| | - Yumi Ito
- Department of Dermatology, Chukyo Hospital, Japan Community Health Care Organization, Nagoya, Japan
| | - Kazuhisa Nakano
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroshi Kasamatsu
- Faculty of Medical Sciences, Department of Dermatology, University of Fukui, Fukui, Japan
| | - Akira Utsunomiya
- Faculty of Medical Sciences, Department of Dermatology, University of Fukui, Fukui, Japan
| | - Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroaki Niiro
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Masatoshi Jinnin
- Department of Dermatology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Katsunari Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takamitsu Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Motohisa Yamamoto
- Department of Rheumatology and Allergy, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Chisako Suzuki
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Emi Nishida
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Dermatology, Okazaki City Hospital, Okazaki, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiyuki Yamamoto
- Department of Dermatology, Fukushima Medical University, School of Medicine, Fukushima, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuya Kondo
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Daisuke Goto
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takayuki Sumida
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naho Ayuzawa
- Department of Clinical Immunology, National Hospital Organization, Utano National Hospital, Kyoto, Japan
| | - Hidetoshi Yanagida
- Department of Clinical Immunology, National Hospital Organization, Utano National Hospital, Kyoto, Japan
| | - Tetsuya Horita
- Faculty of Medicine and Graduate School of Medicine, Department of Rheumatology, Endocrinology and Nephrology, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Faculty of Medicine and Graduate School of Medicine, Department of Rheumatology, Endocrinology and Nephrology, Hokkaido University, Sapporo, Japan
| | - Hirahito Endo
- Omori Medical Center, Toho University, Rheumatic Disease Center, Tokyo, Japan
| | - Yoshihito Shima
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Hirata
- Immunology Frontier Center, Osaka University, Statistical Immunology, Osaka, Japan
| | - Nao Otomo
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Hiroyuki Suetsugu
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Yoshinao Koike
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Kohei Tomizuka
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Soichiro Yoshino
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Xiaoxi Liu
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Shuji Ito
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan
| | - Keiko Hikino
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Pharmacogenomics, Yokohama, Japan
| | - Akari Suzuki
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Autoimmune Diseases, Yokohama, Japan
| | - Yukihide Momozawa
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Genotyping Development, Yokohama, Japan
| | - Shiro Ikegawa
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Bone and Joint Diseases, Yokohama, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Osamu Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuhiko Takehara
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | | | - Shinichi Sato
- Department of Dermatology, The University of Tokyo, Tokyo, Japan
| | - Yukinori Okada
- Immunology Frontier Center, Osaka University, Statistical Immunology, Osaka, Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Ijinkai Takeada General Hospital, Kyoto, Japan
| | - Fumihiko Matsuda
- Graduate School of Medicine, Kyoto University, Center for Genomic Medicine, Kyoto, Japan
| | - Koichi Matsuda
- Institute of Medical Science, The University of Tokyo, Laboratory of Genome Technology, Human Genome Center, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Tiffany Amariuta
- Center for Data Sciences, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Graduate School of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - Issei Imoto
- Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Keitaro Matsuo
- Aichi Cancer Center Research Institute, Division of Cancer Epidemiology and Prevention, Nagoya, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Yasushi Kawaguchi
- Tokyo Women's Medical University, Division of Rheumatology, Department of Internal Medicine, Tokyo, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, The Laboratory for Statistical and Translational Genetics, Yokohama, Japan.
- Shizuoka General Hospital, The Clinical Research Center, Shizuoka, Japan.
- The Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
9
|
Lee JC, Shirey RJ, Turner LD, Park H, Lairson LL, Janda KD. Discovery of PLD4 modulators by high-throughput screening and kinetic analysis. RESULTS IN CHEMISTRY 2024; 7:101349. [PMID: 38560090 PMCID: PMC10977906 DOI: 10.1016/j.rechem.2024.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
Phospholipase D3 (PLD3) and D4 (PLD4) are endolysosomal exonucleases of ssDNA and ssRNA that regulate innate immunity. Polymorphisms of these enzymes are correlated with numerous human diseases, including Alzheimer's, rheumatoid arthritis, and systemic sclerosis. Pharmacological modulation of these immunoregulatory proteins may yield novel immunotherapies and adjuvants. A previous study reported a high-throughput screen (N = 17,952) that discovered a PLD3-selective activator and inhibitor, as well as a nonselective inhibitor, but failed to identify selective modulators of PLD4. However, modulators selective for PLD4 are therapeutically pertinent, since recent reports have shown that regulating this protein has direct implications in cancer and autoimmune diseases. Furthermore, the high expression of PLD4 in dendritic and myeloid cells, in comparison to the broader expression of PLD3, presents the opportunity for a cell-targeted immunotherapy. Here, we describe screening of an expended diversity library (N = 45,760) with an improved platform and report the discovery of one inhibitor and three activators selective for PLD4. Furthermore, kinetic modeling and structural analysis suggest mechanistic differences in the modulation of these hits. These findings further establish the utility of this screening platform and provide a set of chemical scaffolds to guide future small-molecule development for this novel immunoregulator target.
Collapse
Affiliation(s)
- Jinny Claire Lee
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Ryan J. Shirey
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lewis D. Turner
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Hyeri Park
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Luke L. Lairson
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Kim D. Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
10
|
Miyake K, Shibata T, Fukui R, Murakami Y, Sato R, Hiranuma R. Endosomal Toll-Like Receptors as Therapeutic Targets for Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:97-108. [PMID: 38467975 DOI: 10.1007/978-981-99-9781-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Nucleic acid (NA)-sensing Toll-like receptors (TLRs) reside in the endosomal compartment of innate immune cells, such as macrophages and dendritic cells. NAs transported to the endosomal compartment are degraded by DNases and RNases. Degradation products, including single-stranded DNA, oligoRNA, and nucleosides, are recognized by TLR7, TLR8, and TLR9 to drive the defense responses against pathogens. NA degradation influences endosomal TLR responses by generating and degrading TLR ligands. TLR ligand accumulation because of impaired NA degradation causes constitutive TLR activation, leading to autoinflammatory and autoimmune diseases. Furthermore, some genes associated with these diseases promote endosomal TLR responses. Therefore, endosomal TLRs are promising therapeutic targets for TLR-mediated inflammatory diseases, and novel drugs targeting TLRs are being developed.
Collapse
Affiliation(s)
- Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Hiranuma
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Yuan M, Peng L, Huang D, Gavin A, Luan F, Tran J, Feng Z, Zhu X, Matteson J, Wilson IA, Nemazee D. Structural and mechanistic insights into disease-associated endolysosomal exonucleases PLD3 and PLD4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567917. [PMID: 38045427 PMCID: PMC10690185 DOI: 10.1101/2023.11.20.567917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Endolysosomal exonucleases PLD3 and PLD4 (phospholipases D3 and D4) are associated with autoinflammatory and autoimmune diseases. We report structures of these enzymes, and the molecular basis of their catalysis. The structures reveal an intra-chain dimer topology forming a basic active site at the interface. Like other PLD superfamily members, PLD3 and PLD4 carry HxKxxxxD/E motifs and participate in phosphodiester-bond cleavage. The enzymes digest ssDNA and ssRNA in a 5'-to-3' manner and are blocked by 5'-phosphorylation. We captured structures in apo, intermediate, and product states and revealed a 'link-and-release' two-step catalysis. We also unexpectedly demonstrated phosphatase activity via a covalent 3' phosphistidine intermediate. PLD4 contains an extra hydrophobic clamp that stabilizes substrate and could affect oligonucleotide substrate preference and product release. Biochemical and structural analysis of disease-associated mutants of PLD3/4 demonstrated reduced enzyme activity or thermostability and the possible basis for disease association. Furthermore, these findings provide insight into therapeutic design.
Collapse
Affiliation(s)
- Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- These authors contribute equally
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- These authors contribute equally
| | - Deli Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- These authors contribute equally
- Present address: Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Amanda Gavin
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Fangkun Luan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jenny Tran
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ziqi Feng
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jeanne Matteson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
12
|
Ma W, Huang G, Wang Z, Wang L, Gao Q. IRF7: role and regulation in immunity and autoimmunity. Front Immunol 2023; 14:1236923. [PMID: 37638030 PMCID: PMC10449649 DOI: 10.3389/fimmu.2023.1236923] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Interferon regulatory factor (IRF) 7 was originally identified as master transcriptional factor that produced IFN-I and regulated innate immune response, subsequent studies have revealed that IRF7 performs a multifaceted and versatile functions in multiple biological processes. In this review, we provide a comprehensive overview on the current knowledge of the role of IRF7 in immunity and autoimmunity. We focus on the latest regulatory mechanisms of IRF7 in IFN-I, including signaling pathways, transcription, translation, and post-translational levels, the dimerization and nuclear translocation, and the role of IRF7 in IFN-III and COVID-19. In addition to antiviral immunity, we also discuss the role and mechanism of IRF7 in autoimmunity, and the further research will expand our understanding of IRF7.
Collapse
Affiliation(s)
- Wei Ma
- Department of Cell Biology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Gang Huang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhi Wang
- Department of Cell Biology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Wang
- Department of Cell Biology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qiangguo Gao
- Department of Cell Biology, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
13
|
Gavin AL, Blane TR, Thinnes TC, Gerlt E, Marshak-Rothstein A, Huang D, Nemazee D. Disease in the Pld4thss/thss Model of Murine Lupus Requires TLR9. Immunohorizons 2023; 7:577-586. [PMID: 37555846 PMCID: PMC10441812 DOI: 10.4049/immunohorizons.2300058] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/10/2023] Open
Abstract
Phospholipase D4 (PLD4) is an endolysosomal exonuclease of ssRNA and ssDNA, rather than a phospholipase as its name suggests. Human polymorphisms in the PLD4 gene have been linked by genome-wide association studies to systemic sclerosis, rheumatoid arthritis, and systemic lupus erythematosus. However, B6.129 Pld4-/- mice develop features of a distinct disease, macrophage activation syndrome, which is reversed in mice mutated in TLR9. In this article, we compare a Pld4 null mutant identified on the BALB/c background, Pld4thss/thss, which has distinct phenotypes: short stature, thin hair, and features of systemic lupus erythematosus. All phenotypes analyzed were largely normalized in Pld4thss/thssTlr9-/- mice. Thus, Pld4thss/thss represents a rare model in which mouse lupus etiology is TLR9 dependent. Compared with PLD4-deficient B6 mice, Pld4thss/thss mice had elevated levels of serum IgG, IgG anti-dsDNA autoantibodies, BAFF, and IFN-γ and elevated B cell numbers. Overall, the data suggest that PLD4 deficiency can lead to a diverse array of rheumatological abnormalities depending upon background-modifying genes, and that these diseases of PLD4 deficiency are largely driven by TLR9 recognition of ssDNA.
Collapse
Affiliation(s)
- Amanda L. Gavin
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tanya R. Blane
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Theresa C. Thinnes
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | - Deli Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
14
|
Miyake K, Shibata T, Fukui R, Sato R, Saitoh SI, Murakami Y. Nucleic Acid Sensing by Toll-Like Receptors in the Endosomal Compartment. Front Immunol 2022; 13:941931. [PMID: 35812450 PMCID: PMC9259784 DOI: 10.3389/fimmu.2022.941931] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptors (TLRs) respond to pathogen constituents, such as microbial lipids and nucleic acids (NAs). TLRs recognize NAs in endosomal compartments. Structural and functional studies have shown that recognition of NAs by TLRs depends on NA processing by RNases and DNases. DNase II-dependent DNA degradation is required for TLR9 responses to single-stranded DNAs, whereas RNase T2-dependent RNA degradation enables TLR7 and TLR8 to respond to nucleosides and oligoribonucleotides. In contrast, RNases and DNases negatively regulate TLR responses by degrading their ligands. RNase T2 negatively regulates TLR3 responses to degrading the TLR3 ligand double-stranded RNAs. Therefore, NA metabolism in the endosomal compartments affects the endosomal TLR responses. Dysregulation of NA metabolism in the endosomal compartment drives the TLR-dependent pathologies in human diseases.
Collapse
Affiliation(s)
- Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
- *Correspondence: Kensuke Miyake,
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Shin-Ichiroh Saitoh
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| |
Collapse
|
15
|
Huang B, Li J, Zhao J. Screening and identification of potential biomarkers and therapeutic targets for systemic sclerosis-associated interstitial lung disease. Arch Rheumatol 2022; 36:548-559. [PMID: 35382367 PMCID: PMC8957772 DOI: 10.46497/archrheumatol.2021.8625] [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: 12/05/2020] [Accepted: 02/18/2021] [Indexed: 11/03/2022] Open
Abstract
Objectives This study aims to analyze gene expression in lung tissue and lung fibroblasts of patients with systemic sclerosis-associated interstitial lung disease (SSc-ILD) to identify potential biomarkers and therapeutic targets and to examine its possible role in the pathogenesis of SSc-ILD. Patients and methods We obtained datasets from Gene Expression Omnibus (GEO) database, and used Robust Rank Aggregation to calculate the co-expressed differentially-expressed-genes (DEGs) in three chips, then analyzed the function, signaling pathways and the protein-protein interaction network of the DEGs. Finally, we verified the DEGs related to SSc-ILD by three databases of Comparative Toxicogenomics Database (CTD), GENE, and DisGeNET, respectively. Results There were 16 co-expressed DEGs related to SSc-ILD in three GEO series, of which six genes were upregulated, and 10 genes were downregulated. The CTD included 29,936 genes related to SSc, and the GENE and DisGeNET databases had 429 genes related to SSc. Conclusion The results of gene differential expression analysis suggest that interleukin-6, chemokine ligand 2, intercellular adhesion molecule 1, tumor necrosis factor alpha-induced protein 3, pentraxin 3, and cartilage oligomeric matrix protein may be implicated in the pathogenesis of SSc-ILD and are expected to be potential biomarkers and therapeutic targets for SSc-ILD.
Collapse
Affiliation(s)
- Biqing Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
16
|
Langenmayer MC, Jung S, Fux R, Wittlinger C, Tschoner T, Majzoub-Altweck M, Knubben-Schweizer G, Fries R, Hermanns W, Trefz FM. Macrophages in dermal disease progression of phospholipase D4-deficient Fleckvieh calves. Vet Pathol 2022; 59:319-327. [PMID: 34856834 DOI: 10.1177/03009858211062629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A new gene defect in Fleckvieh calves leads to a syndrome with partial phenotype overlap with bovine hereditary zinc deficiency. A mutation in a gene encoding phospholipase D4 (PLD4), an endosomal exonuclease, causes the disorder. In mice, PLD4 activity indirectly regulates the Toll-like receptor 9 (TLR9) pathway via degradation of microbial DNA. PLD4 absence thus results in visceral macrophage activation comparable to human macrophage activation syndrome. In this study, disease progression and the role of macrophages in affected calves were monitored clinically, clinicopathologically, and histologically over time. Breeding data identified 73 risk matings of heterozygous carriers resulting in 54 potentially PLD4-deficient calves born on farms. PLD4 status was examined via 5'-exonuclease assay, detecting 6 calves carrying the defect. These were purchased and monitored daily until final necropsy. The calves developed progressive skin lesions starting with small scaling areas terminating in severe crusting dermatitis, especially in areas with mechanical exposure. Histological and immunohistochemical analyses indicated that macrophages with cytoplasmic vacuolation increased considerably in skin sections obtained weekly during the disease course. Macrophage increase correlated with increased dermal lesion severity. Macrophage activation was confirmed by prominent phagocytic activity in the superficial dermis using electron microscopy. Dermal mRNA abundance of CCL2 and CCL3 measured by quantitative polymerase chain reaction verified macrophage activation. Further increase in mRNA of downstream molecule MyD88 and cytokine IL12b connected bovine PLD4 deficiency to increased TLR9 pathway activation. In contrast to human macrophage activation syndrome, the main feature of bovine PLD4 deficiency was local disease in organs with contact to microbial DNA (skin, intestine, lungs).
Collapse
Affiliation(s)
- Martin C Langenmayer
- Ludwig-Maximilians-Universität München, Munich, Germany
- Specialty Practice for Veterinary Pathology, Munich, Germany
| | - Simone Jung
- Technische Universität München, Freising, Germany
- Bayern-Genetik GmbH, Grub, Germany
| | - Robert Fux
- Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | | | | | - Ruedi Fries
- Technische Universität München, Freising, Germany
| | | | - Florian M Trefz
- Ludwig-Maximilians-Universität München, Munich, Germany
- University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Pu W, Zhang R, Ma Y, Liu Q, Jiang S, Liu J, Zhao Y, Tu W, Guo G, Zuo X, Wang Q, Chen Y, Wu W, Zhou X, Distler JHW, Reveille JD, Zou H, Jin L, Mayes MD, Wang J. Genetic associations of non-MHC susceptibility loci with systemic sclerosis in a Han Chinese population. J Invest Dermatol 2021; 142:2039-2042.e7. [PMID: 34919939 DOI: 10.1016/j.jid.2021.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/16/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Weilin Pu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Research Unit of dissecting the population genetics and developing new technologies for treatment and prevention of skin phenotypes and dermatological diseases (2019RU058), Chinese Academy of Medical Sciences
| | - Rui Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Institute for Six-sector Economy, Fudan University, Shanghai, 200433, China
| | - Yanyun Ma
- Institute for Six-sector Economy, Fudan University, Shanghai, 200433, China; Research Unit of dissecting the population genetics and developing new technologies for treatment and prevention of skin phenotypes and dermatological diseases (2019RU058), Chinese Academy of Medical Sciences
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yinhuan Zhao
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Wenzhen Tu
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Gang Guo
- Department of Rheumatology, Yiling Hospital, Shijiazhuang, China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University
| | - Qingwen Wang
- Rheumatology and Immunology Department, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuanyuan Chen
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Xiaodong Zhou
- Division of Rheumatology and Clinical Immunogenetics, the University of Texas-McGovern Medical School, Houston, TX, USA
| | - Jörg H W Distler
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University of Erlangen, Nuremberg, Germany
| | - John D Reveille
- Division of Rheumatology and Clinical Immunogenetics, the University of Texas-McGovern Medical School, Houston, TX, USA
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Research Unit of dissecting the population genetics and developing new technologies for treatment and prevention of skin phenotypes and dermatological diseases (2019RU058), Chinese Academy of Medical Sciences; Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China
| | - Maureen D Mayes
- Division of Rheumatology and Clinical Immunogenetics, the University of Texas-McGovern Medical School, Houston, TX, USA
| | - Jiucun Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of dissecting the population genetics and developing new technologies for treatment and prevention of skin phenotypes and dermatological diseases (2019RU058), Chinese Academy of Medical Sciences; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Hinchcliff M, Garcia-Milian R, Di Donato S, Dill K, Bundschuh E, Galdo FD. Cellular and Molecular Diversity in Scleroderma. Semin Immunol 2021; 58:101648. [PMID: 35940960 DOI: 10.1016/j.smim.2022.101648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the increasing armamentarium of high-throughput tools available at manageable cost, it is attractive and informative to determine the molecular underpinnings of patient heterogeneity in systemic sclerosis (SSc). Given the highly variable clinical outcomes of patients labelled with the same diagnosis, unravelling the cellular and molecular basis of disease heterogeneity will be crucial to predicting disease risk, stratifying management and ultimately informing a patient-centered precision medicine approach. Herein, we summarise the findings of the past several years in the fields of genomics, transcriptomics, and proteomics that contribute to unraveling the cellular and molecular heterogeneity of SSc. Expansion of these findings and their routine integration with quantitative analysis of histopathology and imaging studies into clinical care promise to inform a scientifically driven patient-centred personalized medicine approach to SSc in the near future.
Collapse
Affiliation(s)
- Monique Hinchcliff
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA.
| | | | - Stefano Di Donato
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK
| | | | - Elizabeth Bundschuh
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA
| | - Francesco Del Galdo
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK.
| |
Collapse
|
19
|
Zhou D, Wu Y, Jiang K, Xu F, Hong R, Wang S. Identification of a risk prediction model for clinical prognosis in HER2 positive breast cancer patients. Genomics 2021; 113:4088-4097. [PMID: 34666190 DOI: 10.1016/j.ygeno.2021.10.010] [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] [Received: 07/21/2020] [Revised: 10/03/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
Background New biomarkers are needed to identify different clinical outcomes for HER2+ breast cancer (BC). Methods Differential genes of HER2+ BC were screened based on TCGA database. We used WGCNA to identify the genes related to the survival. Genetic Algorithm was used to structure risk prediction model. The prognostic model was validated in GSE data. Results We constructed a risk prediction model of 6 genes to identify prognosis of HER2+ BC, including CLEC9A, PLD4, PIM1, PTK2B, AKNAD1 and C15orf27. Kaplan-Meier curve showed that the model effectively distinguished the survival of HER2+ BC patients. The multivariate Cox regression suggested that the risk model was an independent predictor for HER2+ BC. Analysis related to immune showed that significant differences in immune infiltration between high- and low-risk groups classified by the prognostic model. Conclusions Our study identified a risk prediction model of 6 genes that could distinguish the prognosis of HER2+ BC.
Collapse
Affiliation(s)
- Danyang Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Ying Wu
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Kuikui Jiang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Fei Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Ruoxi Hong
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Shusen Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| |
Collapse
|
20
|
Cleavage of DNA and RNA by PLD3 and PLD4 limits autoinflammatory triggering by multiple sensors. Nat Commun 2021; 12:5874. [PMID: 34620855 PMCID: PMC8497607 DOI: 10.1038/s41467-021-26150-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/15/2021] [Indexed: 11/26/2022] Open
Abstract
Phospholipase D3 (PLD3) and PLD4 polymorphisms have been associated with several important inflammatory diseases. Here, we show that PLD3 and PLD4 digest ssRNA in addition to ssDNA as reported previously. Moreover, Pld3−/−Pld4−/− mice accumulate small ssRNAs and develop spontaneous fatal hemophagocytic lymphohistiocytosis (HLH) characterized by inflammatory liver damage and overproduction of Interferon (IFN)-γ. Pathology is rescued in Unc93b13d/3dPld3−/−Pld4−/− mice, which lack all endosomal TLR signaling; genetic codeficiency or antibody blockade of TLR9 or TLR7 ameliorates disease less effectively, suggesting that both RNA and DNA sensing by TLRs contributes to inflammation. IFN-γ made a minor contribution to pathology. Elevated type I IFN and some other remaining perturbations in Unc93b13d/3dPld3−/−Pld4−/− mice requires STING (Tmem173). Our results show that PLD3 and PLD4 regulate both endosomal TLR and cytoplasmic/STING nucleic acid sensing pathways and have implications for the treatment of nucleic acid-driven inflammatory disease. Loss of function polymorphisms of phospholipase D3 and D4 are associated with inflammatory diseases and their function is unclear. Here the authors show that PLD3/4 function as RNAses and deletion of these proteins in mice leads to accumulation of ssRNA which exacerbates inflammation through TLR signalling.
Collapse
|
21
|
Ototake Y, Yamaguchi Y, Asami M, Komitsu N, Akita A, Watanabe T, Kanaoka M, Kurotaki D, Tamura T, Aihara M. Downregulated IRF8 in Monocytes and Macrophages of Patients with Systemic Sclerosis May Aggravate the Fibrotic Phenotype. J Invest Dermatol 2021; 141:1954-1963. [PMID: 33705797 DOI: 10.1016/j.jid.2021.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
Monocytes and macrophages may be involved in the pathogenesis of systemic sclerosis (SSc); however, the etiology and regulation of monocyte and macrophage function in SSc remain unknown. IRF8 is a transcriptional regulator that is essential for the differentiation and function of monocytes and macrophages and thus may be involved in the regulation of macrophage phenotypes in SSc fibrosis. In this study, we measured IRF8 levels in circulating monocytes of 26 patients with SSc (diffuse cutaneous SSc, n = 11; limited cutaneous SSc, n = 15) and 14 healthy controls. IRF8 levels were significantly suppressed in monocytes of patients with diffuse cutaneous SSc and correlated negatively with the modified Rodnan total skin thickness score. Next, we assessed expression levels of cell surface markers, cytokine profiles, and components of extracellular matrix in IRF8-silenced monocyte-derived macrophages. IRF8-silenced monocyte-derived macrophages displayed an M2 phenotype and significantly upregulated mRNA and protein levels of profibrotic factors and extracellular matrix components. Finally, we assessed skin fibrosis in myeloid cell-specific IRF8 conditional knockout (Irf8flox/flox; Lyz2Cre/+) mice and found upregulated mRNA levels of extracellular matrix components and increased bleomycin-induced skin fibrosis. In conclusion, altered IRF8 regulation in monocytes and macrophages may be involved in SSc pathogenesis.
Collapse
Affiliation(s)
- Yasushi Ototake
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Miho Asami
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Komitsu
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Asami Akita
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoya Watanabe
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miwa Kanaoka
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Michiko Aihara
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
22
|
Ota Y, Kuwana M. Updates on genetics in systemic sclerosis. Inflamm Regen 2021; 41:17. [PMID: 34130729 PMCID: PMC8204536 DOI: 10.1186/s41232-021-00167-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is a complex disease, in which an interaction of genetic and environmental factors plays an important role in its development and pathogenesis. A number of genetic studies, including candidate gene analysis and genome-wide association study, have found that the associated genetic variants are mainly localized in noncoding regions in the expression quantitative trait locus and influence corresponding gene expression. The gene variants identified as a risk for SSc susceptibility include those associated with innate immunity, adaptive immune response, and cell death, while there are only few SSc-associated genes involved in the fibrotic process or vascular homeostasis. Human leukocyte antigen class II genes are associated with SSc-related autoantibodies rather than SSc itself. Since the pathways between the associated genotype and phenotype are still poorly understood, further investigations using multi-omics technologies are necessary to characterize the complex molecular architecture of SSc, identify biomarkers useful to predict future outcomes and treatment responses, and discover effective drug targets.
Collapse
Affiliation(s)
- Yuko Ota
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603-8582, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603-8582, Japan.
| |
Collapse
|
23
|
Leylek R, Alcántara-Hernández M, Granja JM, Chavez M, Perez K, Diaz OR, Li R, Satpathy AT, Chang HY, Idoyaga J. Chromatin Landscape Underpinning Human Dendritic Cell Heterogeneity. Cell Rep 2021; 32:108180. [PMID: 32966789 PMCID: PMC7546547 DOI: 10.1016/j.celrep.2020.108180] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/18/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Human dendritic cells (DCs) comprise subsets with distinct phenotypic and functional characteristics, but the transcriptional programs that dictate their identity remain elusive. Here, we analyze global chromatin accessibility profiles across resting and stimulated human DC subsets by means of the assay for transposase-accessible chromatin using sequencing (ATAC-seq). We uncover specific regions of chromatin accessibility for each subset and transcriptional regulators of DC function. By comparing plasmacytoid DC responses to IFN-I-producing and non-IFN-I-producing conditions, we identify genetic programs related to their function. Finally, by intersecting chromatin accessibility with genome-wide association studies, we recognize DC subset-specific enrichment of heritability in autoimmune diseases. Our results unravel the basis of human DC subset heterogeneity and provide a framework for their analysis in disease pathogenesis. Human dendritic cells (DCs) orchestrate immune responses by a division of labor between functionally specialized subsets; however, the transcriptional basis of this heterogeneity is poorly understood. Using ATAC-seq, Leylek et al. profile the chromatin landscape of human DC subsets, providing insight into the underlying regulatory mechanisms that modulate their function.
Collapse
Affiliation(s)
- Rebecca Leylek
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marcela Alcántara-Hernández
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey M Granja
- Biophysics Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Chavez
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kimberly Perez
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Oscar R Diaz
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Juliana Idoyaga
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
24
|
Santa P, Garreau A, Serpas L, Ferriere A, Blanco P, Soni C, Sisirak V. The Role of Nucleases and Nucleic Acid Editing Enzymes in the Regulation of Self-Nucleic Acid Sensing. Front Immunol 2021; 12:629922. [PMID: 33717156 PMCID: PMC7952454 DOI: 10.3389/fimmu.2021.629922] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/21/2021] [Indexed: 12/24/2022] Open
Abstract
Detection of microbial nucleic acids by the innate immune system is mediated by numerous intracellular nucleic acids sensors. Upon the detection of nucleic acids these sensors induce the production of inflammatory cytokines, and thus play a crucial role in the activation of anti-microbial immunity. In addition to microbial genetic material, nucleic acid sensors can also recognize self-nucleic acids exposed extracellularly during turn-over of cells, inefficient efferocytosis, or intracellularly upon mislocalization. Safeguard mechanisms have evolved to dispose of such self-nucleic acids to impede the development of autoinflammatory and autoimmune responses. These safeguard mechanisms involve nucleases that are either specific to DNA (DNases) or RNA (RNases) as well as nucleic acid editing enzymes, whose biochemical properties, expression profiles, functions and mechanisms of action will be detailed in this review. Fully elucidating the role of these enzymes in degrading and/or processing of self-nucleic acids to thwart their immunostimulatory potential is of utmost importance to develop novel therapeutic strategies for patients affected by inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Pauline Santa
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Anne Garreau
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Lee Serpas
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | | | - Patrick Blanco
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
- Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| | - Chetna Soni
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Vanja Sisirak
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| |
Collapse
|
25
|
Ishikawa Y, Terao C. Genetics of systemic sclerosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:192-201. [PMID: 35382527 PMCID: PMC8922623 DOI: 10.1177/2397198320913695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/23/2020] [Indexed: 01/05/2024]
Abstract
Systemic sclerosis is an autoimmune disease characterized by generalized fibrosis in connective tissues and internal organs as consequences of microvascular dysfunction and immune dysfunctions, which leads to premature death in affected individuals. The etiology of systemic sclerosis is complex and poorly understood, but as with most autoimmune diseases, it is widely accepted that both environmental and genetic factors contribute to disease risk. During the last decade, the number of genetic markers convincingly associated with systemic sclerosis has exponentially increased. In this article, we briefly mention the genetic components of systemic sclerosis. Then, we review the classical and novel genetic associations with systemic sclerosis, analyzing the firmest and replicated signals within non-human leukocyte antigen genes, identified by both candidate gene approach and genome-wide association studies. We also provide an insight into the future perspectives that will shed more light into the complex genetic background of the disease. Despite the remarkable advance of systemic sclerosis genetics during the last decade, the use of the new genetic technologies such as next-generation sequencing, as well as the deep phenotyping of the study cohorts, to fully characterize the genetic component of this disease is imperative to identify causal variants, which leads to more targeted and effective treatment of systemic sclerosis.
Collapse
Affiliation(s)
- Yuki Ishikawa
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
26
|
Yoo HJ, Hwang WC, Min DS. Targeting of Phospholipase D1 Ameliorates Collagen-Induced Arthritis via Modulation of Treg and Th17 Cell Imbalance and Suppression of Osteoclastogenesis. Int J Mol Sci 2020; 21:ijms21093230. [PMID: 32370217 PMCID: PMC7247592 DOI: 10.3390/ijms21093230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Phospholipase D1 (PLD1) plays a crucial role in various inflammatory and autoimmune diseases. Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease. However, the role of PLD1 in the pathogenesis of RA remains unknown. Here, we first investigated the role and effects of PLD1 in collagen-induced arthritis (CIA) and found that genetic and pharmacological inhibition of PLD1 in DBA1/J mice with CIA reduced the incidence of CIA, decreased the clinical score, and abrogated disease symptoms including infiltration of leukocytes, synovial inflammation, bone erosion, and cartilage destruction. Moreover, ablation and inhibition of PLD1 suppressed the production of type II collagen-specific IgG2a autoantibody and proinflammatory cytokines, accompanied by an increase in the regulatory T (Treg) cell population and a decrease in the Th17 cell population in CIA mice. The PLD1 inhibitor also promoted differentiation of Treg cells and suppressed differentiation of Th17 cells in vitro. Furthermore, the PLD1 inhibitor attenuated pathologic bone destruction in CIA mice by suppressing osteoclastogenesis and bone resorption. Thus, our findings indicate that the targeting of PLD1 can ameliorate CIA by modulating the imbalance of Treg and Th17 cells and suppressing osteoclastogenesis, which might be a novel strategy to treat autoimmune diseases, such as RA.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/prevention & control
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/prevention & control
- Benzimidazoles/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Cytokines/blood
- Disease Models, Animal
- Knee Joint/drug effects
- Knee Joint/metabolism
- Knee Joint/pathology
- Male
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Phospholipase D/antagonists & inhibitors
- Phospholipase D/genetics
- Phospholipase D/metabolism
- Piperidines/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- X-Ray Microtomography
Collapse
Affiliation(s)
- Hyun Jung Yoo
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46241, Korea; (H.J.Y.); (W.C.H.)
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46241, Korea; (H.J.Y.); (W.C.H.)
- College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Correspondence: ; Tel.: +82-32-749-4522
| |
Collapse
|
27
|
Carvalheiro T, Zimmermann M, Radstake TRDJ, Marut W. Novel insights into dendritic cells in the pathogenesis of systemic sclerosis. Clin Exp Immunol 2020; 201:25-33. [PMID: 31970748 PMCID: PMC7290079 DOI: 10.1111/cei.13417] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (SSc) is a severe autoimmune fibrotic disease characterized by fibrosis, vasculopathy, and immune dysregulation. Dendritic cells (DCs) are the most potent antigen-presenting cells, specialized in pathogen sensing, with high capacity to shape the immune responses. The most recent technological advances have allowed the discovery of new DC subsets with potential implications in inflammatory conditions. Alterations of DC distribution in circulation and affected tissue as well as impaired DC function have been described in SSc patients, pointing towards a crucial role of these cells in SSc pathogenesis. In particular, recent studies have shown the importance of plasmacytoid DCs either by their high capacity to produce type I interferon or other inflammatory mediators implicated in SSc pathology, such as chemokine C-X-C motif ligand 4 (CXCL4). In-vivo models of SSc have been vital to clarify the implications of DCs in this disease, especially DCs depletion and specific gene knock-down studies. This review provides these new insights into the contribution of the different DCs subsets in the pathogenesis of SSc, as well as to the novel developments on DCs in in-vivo models of SSc and the potential use of DCs and their mediators as therapeutic targets.
Collapse
Affiliation(s)
- T Carvalheiro
- Center for 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
| | - M Zimmermann
- Center for 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
| | - T R D J Radstake
- Center for 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
| | - W Marut
- Center for 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
| |
Collapse
|
28
|
Soni C, Reizis B. Self-DNA at the Epicenter of SLE: Immunogenic Forms, Regulation, and Effects. Front Immunol 2019; 10:1601. [PMID: 31354738 PMCID: PMC6637313 DOI: 10.3389/fimmu.2019.01601] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Self-reactive B cells generated through V(D)J recombination in the bone marrow or through accrual of random mutations in secondary lymphoid tissues are mostly purged or edited to prevent autoimmunity. Yet, 10–20% of all mature naïve B cells in healthy individuals have self-reactive B cell receptors (BCRs). In patients with serologically active systemic lupus erythematosus (SLE) the percentage increases up to 50%, with significant self-DNA reactivity that correlates with disease severity. Endogenous or self-DNA has emerged as a potent antigen in several autoimmune disorders, particularly in SLE. However, the mechanism(s) regulating or preventing anti-DNA antibody production remain elusive. It is likely that in healthy subjects, DNA-reactive B cells avoid activation due to the unavailability of endogenous DNA, which is efficiently degraded through efferocytosis and various DNA-processing proteins. Genetic defects, physiological, and/or pathological conditions can override these protective checkpoints, leading to autoimmunity. Plausibly, increased availability of immunogenic self-DNA may be the key initiating event in the loss of tolerance of otherwise quiescent DNA-reactive B cells. Indeed, mutations impairing apoptotic cell clearance pathways and nucleic acid metabolism-associated genes like DNases, RNases, and their sensors are known to cause autoimmune disorders including SLE. Here we review the literature supporting the idea that increased availability of DNA as an immunogen or adjuvant, or both, may cause the production of pathogenic anti-DNA antibodies and subsequent manifestations of clinical disease such as SLE. We discuss the main cellular players involved in anti-DNA responses; the physical forms and sources of immunogenic DNA in autoimmunity; the DNA-protein complexes that render DNA immunogenic; the regulation of DNA availability by intracellular and extracellular DNases and the autoimmune pathologies associated with their dysfunction; the cytosolic and endosomal sensors of immunogenic DNA; and the cytokines such as interferons that drive auto-inflammatory and autoimmune pathways leading to clinical disease. We propose that prevention of DNA availability by aiding extracellular DNase activity could be a viable therapeutic modality in controlling SLE.
Collapse
Affiliation(s)
- Chetna Soni
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
29
|
Akizuki S, Ishigaki K, Kochi Y, Law SM, Matsuo K, Ohmura K, Suzuki A, Nakayama M, Iizuka Y, Koseki H, Ohara O, Hirata J, Kamatani Y, Matsuda F, Sumida T, Yamamoto K, Okada Y, Mimori T, Terao C. PLD4 is a genetic determinant to systemic lupus erythematosus and involved in murine autoimmune phenotypes. Ann Rheum Dis 2019; 78:509-518. [PMID: 30679154 DOI: 10.1136/annrheumdis-2018-214116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/22/2018] [Accepted: 12/17/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is an autoimmune disease that is characterised by autoantibody production and widespread inflammation damaging many organs. Previous genome-wide association studies (GWASs) have revealed over 80 genetic determinants of SLE, but they collectively explain a fraction of the heritability, and only a few were proven in vivo for the involvement in SLE. We conducted a meta-analysis of SLE GWAS in the Japanese population, followed by functional analyses of a susceptibility gene with use of mutant mice. METHODS We conducted a meta-analysis of two GWASs comprising a total of 1363 cases and 5536 controls using the 1000 Genome Project data as an imputation reference. Enrichment analyses for functional annotations were conducted. We examined Phospholipase D4 (Pld4) mutant mice to assess functional involvement of a genetic determinant. RESULTS We found a total of 14 significant loci, which included rs2582511 in AHNAK2/PLD4 recently reported in a Chinese study and a novel locus of rs143181706 in MAMLD1 (p=7.9×10-11 and 3.7×10-8, respectively). PLD4 risk allele was associated with anti-dsDNA antibody production. Enrichment analysis of genetic signals revealed involvement of a wide range of immune-related cells and pathways. Pld4 mutant mice revealed remarkably low body weight. The mice demonstrated autoimmune phenotypes compatible with SLE, including splenomegaly and lymphadenopathy, expansion of B cells and hypersecretion of BAFF and production of autoantibodies especially anti-nuclear antibody and anti-dsDNA antibody. CONCLUSIONS We found a novel susceptibility gene to SLE. Pld4 mutant mice revealed autoimmune phenotypes suggesting functional involvement of PLD4 with the basics of SLE.
Collapse
Affiliation(s)
- Shuji Akizuki
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Sze-Ming Law
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Manabu Nakayama
- Department of Frontier Research and Development, Kazusa DNA Research Institute, Chiba, Japan
| | - Yusuke Iizuka
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Jun Hirata
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Pharmaceutical Discovery Research Laboratories, TEIJIN PHARMA LIMITED, Hino, Japan
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chikashi Terao
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
30
|
Skaug B, Assassi S. Type I interferon dysregulation in Systemic Sclerosis. Cytokine 2019; 132:154635. [PMID: 30685202 DOI: 10.1016/j.cyto.2018.12.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/28/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022]
Abstract
Systemic Sclerosis (Scleroderma, SSc) is a multifaceted disease characterized by autoimmunity, vasculopathy, and fibrosis affecting the skin and internal organs. Despite advances in the understanding and treatment of SSc in recent years, SSc continues to cause reduced quality of life and premature mortality. Type I interferons (IFNs), a family of cytokines with essential roles in the immune response to microbial infection, play a pathogenic role in certain autoimmune diseases (reviewed elsewhere in this edition). Polymorphisms in interferon-regulatory factors confer an increased risk of SSc, and IFN excess is evident in the blood and skin of a large percentage of SSc patients. Here we describe the evidence of Type I IFN dysregulation in SSc, revealed predominately by genetics and gene expression profiling. We also discuss evidence regarding mechanisms by which Type I IFN might contribute to SSc pathogenesis, mechanisms driving excess Type I IFN production in SSc, and the potential roles of Type I IFNs as biomarkers and therapeutic targets in SSc.
Collapse
Affiliation(s)
- Brian Skaug
- The University of Texas Health Science Center in Houston, Division of Rheumatology, 6431 Fannin, MSB 5.262, Houston, TX 77030, United States
| | - Shervin Assassi
- The University of Texas Health Science Center in Houston, Division of Rheumatology, 6431 Fannin, MSB 5.262, Houston, TX 77030, United States.
| |
Collapse
|
31
|
Phospholipase D and the Mitogen Phosphatidic Acid in Human Disease: Inhibitors of PLD at the Crossroads of Phospholipid Biology and Cancer. Handb Exp Pharmacol 2019; 259:89-113. [PMID: 31541319 DOI: 10.1007/164_2019_216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lipids are key building blocks of biological membranes and are involved in complex signaling processes such as metabolism, proliferation, migration, and apoptosis. Extracellular signaling by growth factors, stress, and nutrients is transmitted through receptors that activate lipid-modifying enzymes such as the phospholipases, sphingosine kinase, or phosphoinositide 3-kinase, which then modify phospholipids, sphingolipids, and phosphoinositides. One such important enzyme is phospholipase D (PLD), which cleaves phosphatidylcholine to yield phosphatidic acid and choline. PLD isoforms have dual role in cells. The first involves maintaining cell membrane integrity and cell signaling, including cell proliferation, migration, cytoskeletal alterations, and invasion through the PLD product PA, and the second involves protein-protein interactions with a variety of binding partners. Increased evidence of elevated PLD expression and activity linked to many pathological conditions, including cancer, neurological and inflammatory diseases, and infection, has motivated the development of dual- and isoform-specific PLD inhibitors. Many of these inhibitors are reported to be efficacious and safe in cells and mouse disease models, suggesting the potential for PLD inhibitors as therapeutics for cancer and other diseases. Current knowledge and ongoing research of PLD signaling networks will help to evolve inhibitors with increased efficacy and safety for clinical studies.
Collapse
|
32
|
Gavin AL, Huang D, Huber C, Mårtensson A, Tardif V, Skog PD, Blane TR, Thinnes TC, Osborn K, Chong HS, Kargaran F, Kimm P, Zeitjian A, Sielski RL, Briggs M, Schulz SR, Zarpellon A, Cravatt B, Pang ES, Teijaro J, de la Torre JC, O'Keeffe M, Hochrein H, Damme M, Teyton L, Lawson BR, Nemazee D. PLD3 and PLD4 are single-stranded acid exonucleases that regulate endosomal nucleic-acid sensing. Nat Immunol 2018; 19:942-953. [PMID: 30111894 PMCID: PMC6105523 DOI: 10.1038/s41590-018-0179-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/28/2018] [Indexed: 01/06/2023]
Abstract
The sensing of microbial genetic material by leukocytes often elicits beneficial pro-inflammatory cytokines, but dysregulated responses can cause severe pathogenesis. Genome-wide association studies have linked the gene encoding phospholipase D3 (PLD3) to Alzheimer's disease and have linked PLD4 to rheumatoid arthritis and systemic sclerosis. PLD3 and PLD4 are endolysosomal proteins whose functions are obscure. Here, PLD4-deficient mice were found to have an inflammatory disease, marked by elevated levels of interferon-γ (IFN-γ) and splenomegaly. These phenotypes were traced to altered responsiveness of PLD4-deficient dendritic cells to ligands of the single-stranded DNA sensor TLR9. Macrophages from PLD3-deficient mice also had exaggerated TLR9 responses. Although PLD4 and PLD3 were presumed to be phospholipases, we found that they are 5' exonucleases, probably identical to spleen phosphodiesterase, that break down TLR9 ligands. Mice deficient in both PLD3 and PLD4 developed lethal liver inflammation in early life, which indicates that both enzymes are needed to regulate inflammatory cytokine responses via the degradation of nucleic acids.
Collapse
Affiliation(s)
- Amanda L Gavin
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Deli Huang
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Christoph Huber
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- , Bottmingen, Switzerland
| | - Annica Mårtensson
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Sophiris Bio, La Jolla, CA, USA
| | - Virginie Tardif
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University, Philadelphia, PA, USA
| | - Patrick D Skog
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tanya R Blane
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Therese C Thinnes
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Kent Osborn
- The University of California, San Diego, La Jolla, CA, USA
| | - Hayley S Chong
- The University of California, San Diego, La Jolla, CA, USA
| | | | - Phoebe Kimm
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Armen Zeitjian
- The University of California, San Diego, La Jolla, CA, USA
| | | | - Megan Briggs
- The University of California, San Diego, La Jolla, CA, USA
| | - Sebastian R Schulz
- Division of Molecular Immunology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Alessandro Zarpellon
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin Cravatt
- The Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ee Shan Pang
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - John Teijaro
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Juan Carlos de la Torre
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Meredith O'Keeffe
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | | | - Markus Damme
- Biochemisches Institut, Christian-Albrechts-Universität, Kiel, Germany
| | - Luc Teyton
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Brian R Lawson
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - David Nemazee
- The Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
33
|
Rezaei R, Aslani S, Dashti N, Jamshidi A, Gharibdoost F, Mahmoudi M. Genetic implications in the pathogenesis of systemic sclerosis. Int J Rheum Dis 2018; 21:1478-1486. [DOI: 10.1111/1756-185x.13344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ramazan Rezaei
- Rheumatology Research Center Tehran University of Medical Sciences Tehran Iran
- Department of Immunology School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Saeed Aslani
- Rheumatology Research Center Tehran University of Medical Sciences Tehran Iran
| | - Navid Dashti
- Rheumatology Research Center Tehran University of Medical Sciences Tehran Iran
- Department of Immunology School of Medicine Tehran University of Medical Sciences Tehran Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center Tehran University of Medical Sciences Tehran Iran
| | - Farhad Gharibdoost
- Rheumatology Research Center Tehran University of Medical Sciences Tehran Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
34
|
Wen L, Zhu C, Zhu Z, Yang C, Zheng X, Liu L, Zuo X, Sheng Y, Tang H, Liang B, Zhou Y, Li P, Zhu J, Ding Y, Chen G, Gao J, Tang L, Cheng Y, Sun J, Elango T, Kafle A, Yu R, Xue K, Zhang Y, Li F, Li Z, Guo J, Zhang X, Zhou C, Tang Y, Shen N, Wang M, Yu X, Liu S, Fan X, Gao M, Xiao F, Wang P, Wang Z, Zhang A, Zhou F, Sun L, Yang S, Xu J, Yin X, Cui Y, Zhang X. Exome-wide association study identifies four novel loci for systemic lupus erythematosus in Han Chinese population. Ann Rheum Dis 2018; 77:417. [PMID: 29233832 DOI: 10.1136/annrheumdis-2017-211823] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 11/11/2017] [Accepted: 11/19/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is a chronic autoimmune disease of considerable genetic predisposition. Genome-wide association studies have identified tens of common variants for SLE. However, the majority of them reside in non-coding sequences. The contributions of coding variants have not yet been systematically evaluated. METHODS We performed a large-scale exome-wide study in 5004 SLE cases and 8179 healthy controls in a Han Chinese population using a custom exome array, and then genotyped 32 variants with suggestive evidence in an independent cohort of 13 246 samples. We further explored the regulatory effect of one novel non-coding single nucleotide polymorphism (SNP) in ex vivo experiments. RESULTS We discovered four novel SLE gene regions (LCT, TPCN2, AHNAK2 and TNFRSF13B) encompassing three novel missense variants (XP_016859577.1:p.Asn1639Ser, XP_016859577.1:p.Val219Phe and XP_005267356.1:p.Thr4664Ala) and two non-coding variants (rs10750836 and rs4792801) with genome-wide significance (pmeta <5.00×10-8). These variants are enriched in several chromatin states of primary B cells. The novel intergenic variant rs10750836 exhibited an expression quantitative trait locus effect on the TPCN2 gene in immune cells. Clones containing this novel SNP exhibited gene promoter activity for TPCN2 (P=1.38×10-3) whose expression level was reduced significantly in patients with SLE (P<2.53×10-2) and was suggested to be further modulated by rs10750836 in CD19+ B cells (P=7.57×10-5) in ex vivo experiments. CONCLUSIONS This study identified three novel coding variants and four new susceptibility gene regions for SLE. The results provide insights into the biological mechanism of SLE.
Collapse
Affiliation(s)
- Leilei Wen
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Caihong Zhu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Zhengwei Zhu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Chao Yang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaodong Zheng
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lu Liu
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xianbo Zuo
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yujun Sheng
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Huayang Tang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Bo Liang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yi Zhou
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Pan Li
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jun Zhu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yantao Ding
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Gang Chen
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinping Gao
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lili Tang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yuyan Cheng
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jingying Sun
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Tamilselvi Elango
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Anjana Kafle
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Ruixing Yu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Ke Xue
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Yaohua Zhang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Feng Li
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangdong, China
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangdong, China
| | - Shengxiu Liu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xing Fan
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Min Gao
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Fengli Xiao
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Peiguang Wang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Zaixing Wang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Anping Zhang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Fusheng Zhou
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Liangdan Sun
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Sen Yang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhua Xu
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Xianyong Yin
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Xuejun Zhang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
35
|
Langenmayer MC, Jung S, Majzoub-Altweck M, Trefz FM, Seifert C, Knubben-Schweizer G, Fries R, Hermanns W, Gollnick NS. Zinc Deficiency-Like Syndrome in Fleckvieh Calves: Clinical and Pathological Findings and Differentiation from Bovine Hereditary Zinc Deficiency. J Vet Intern Med 2018; 32:853-859. [PMID: 29424482 PMCID: PMC5866964 DOI: 10.1111/jvim.15040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/17/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022] Open
Abstract
Background Zinc deficiency‐like (ZDL) syndrome is an inherited defect of Fleckvieh calves, with striking similarity to bovine hereditary zinc deficiency (BHZD). However, the causative mutation in a phospholipase D4 encoding gene (PLD4) shows no connection to zinc metabolism. Objectives To describe clinical signs, laboratory variables, and pathological findings of ZDL syndrome and their utility to differentiate ZDL from BHZD and infectious diseases with similar phenotype. Animals Nine hospitalized calves with crusting dermatitis and confirmed mutation in PLD4 and medical records from 25 calves with crusting dermatitis or suspected zinc deficiency. Methods Prospective and retrospective case series. Results The 9 calves (age: 5–53 weeks) displayed a moderate to severe crusting dermatitis mainly on the head, ventrum, and joints. Respiratory and digestive tract inflammations were frequently observed. Zinc supplementation did not lead to remission of clinical signs in 4 calves. Laboratory variables revealed slight anemia in 8 calves, hypoalbuminemia in 6 calves, but reduced serum zinc concentrations in only 3 calves. Mucosal erosions/ulcerations were present in 7 calves and thymus atrophy or reduced thymic weights in 8 calves. Histologically, skin lesions were indistinguishable from BHZD. Retrospective analysis of medical records revealed the presence of this phenotype since 1988 and pedigree analysis revealed a common ancestor of several affected calves. Conclusions and Clinical Importance ZDL syndrome should be suspected in Fleckvieh calves with crusting dermatitis together with diarrhea or respiratory tract inflammations without response to oral zinc supplementation. Definite diagnosis requires molecular genetic confirmation of the PLD4 mutation.
Collapse
Affiliation(s)
- M C Langenmayer
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany.,Institute for Infectious Diseases and Zoonoses, LMU Munich, Munich, Germany
| | - S Jung
- Chair of Animal Breeding, Technische Universität München, Freising-Weihenstephan, Germany
| | - M Majzoub-Altweck
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - F M Trefz
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| | - C Seifert
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| | - G Knubben-Schweizer
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| | - R Fries
- Chair of Animal Breeding, Technische Universität München, Freising-Weihenstephan, Germany
| | - W Hermanns
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - N S Gollnick
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| |
Collapse
|
36
|
Ciechomska M, Skalska U. Targeting interferons as a strategy for systemic sclerosis treatment. Immunol Lett 2017; 195:45-54. [PMID: 29106987 DOI: 10.1016/j.imlet.2017.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022]
Abstract
Systemic Sclerosis (SSc) is an autoimmune disease characterised by vasculopathy, uncontrolled inflammation and enhanced fibrosis which can subsequently lead to the loss of organ function or even premature death. Interferons (IFNs) are pleiotropic cytokines that are critical not only in mounting an effective immune response against viral and bacterial infections but also strongly contribute to the pathogenesis of SSc. Furthermore, elevated levels of IFNs are found in SSc patients and correlate with skin thickness and disease activity suggesting potential role of IFNs as biomarkers. In this review, we summarise existing knowledge regarding all types of IFNs and IFN-inducible genes in the pathogenesis of SSc. We then argue why IFN-blocking strategies are promising therapeutic targets in SSc and other autoimmune diseases.
Collapse
Affiliation(s)
- Marzena Ciechomska
- National Institute of Geriatrics Rheumatology and Rehabilitation, Warsaw, Poland
| | - Urszula Skalska
- National Institute of Geriatrics Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
37
|
Li L, Chen S, Wang Q, Wu C, Wen X, Yang F, Liu C, Zhang F, Li Y. GLIS3 and TYK2 Single Nucleotide Polymorphisms Are Not Associated with Dermatomyositis/Polymyositis in Chinese Han Population. Genet Test Mol Biomarkers 2017; 21:565-570. [PMID: 28846454 DOI: 10.1089/gtmb.2017.0059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIM Racial differences and genetic overlap have been shown to be responsible for the difference in susceptibility to dermatomyositis (DM)/polymyositis (PM) in a variety of populations. Single nucleotide polymorphisms (SNPs) in the GLI-similar 3 (GLIS3) and tyrosine kinase 2 (TYK2) genes have been associated with various autoimmune diseases. The aim of this study was to investigate whether SNPs in GLIS3 (rs7020673, rs10758593, and rs10814916) and TYK2 (rs280519, rs2304256, rs17000730, and rs280501) were associated with an increase in susceptibility to DM/PM in a Chinese Han population. MATERIALS AND METHODS SNPs in GLIS3 and TYK2 were analyzed in a cohort of 1017 patients with DM/PM and 1280 healthy controls using a Sequenom MassArray system. Association analyses were performed using the PLINK v1.07 software. RESULTS In our study, the GLIS3 polymorphisms rs7020673 and rs10758593 were initially found to be predisposing risk factors for PM and PM with interstitial lung disease (p < 0.05). Both rs7020673 and rs10758593 were associated with PM in both additive and dominant models (p < 0.05); however, these observed associations were not apparent after Bonferroni correction. Other SNPs examined in our study were not associated with susceptibility to DM/PM. CONCLUSION GLIS3 and TYK2 polymorphisms are not associated with DM/PM in the Chinese Han population. As associations of SNPs in these genes in patients with DM/PM have been previously reported in other populations, we may conclude that the lack of association in our study may be the result of differences in genetic background. Further studies in other populations are needed to confirm our findings.
Collapse
Affiliation(s)
- Liubing Li
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Si Chen
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China .,2 Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University , Beijing, China
| | - Qian Wang
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Chanyuan Wu
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaoting Wen
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Funing Yang
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China .,3 Department of Medical Laboratory, The First Hospital of Jilin University , Changchun, China
| | - Chenxi Liu
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Fengchun Zhang
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yongzhe Li
- 1 Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
38
|
Rossato M, Affandi AJ, Thordardottir S, Wichers CGK, Cossu M, Broen JCA, Moret FM, Bossini-Castillo L, Chouri E, van Bon L, Wolters F, Marut W, van der Kroef M, Silva-Cardoso S, Bekker CPJ, Dolstra H, van Laar JM, Martin J, van Roon JAG, Reedquist KA, Beretta L, Radstake TRDJ. Association of MicroRNA-618 Expression With Altered Frequency and Activation of Plasmacytoid Dendritic Cells in Patients With Systemic Sclerosis. Arthritis Rheumatol 2017; 69:1891-1902. [PMID: 28556560 DOI: 10.1002/art.40163] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 05/23/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Plasmacytoid dendritic cells (PDCs) are a critical source of type I interferons (IFNs) that can contribute to the onset and maintenance of autoimmunity. Molecular mechanisms leading to PDC dysregulation and a persistent type I IFN signature are largely unexplored, especially in patients with systemic sclerosis (SSc), a disease in which PDCs infiltrate fibrotic skin lesions and produce higher levels of IFNα than those in healthy controls. This study was undertaken to investigate potential microRNA (miRNA)-mediated epigenetic mechanisms underlying PDC dysregulation and type I IFN production in SSc. METHODS We performed miRNA expression profiling and validation in highly purified PDCs obtained from the peripheral blood of 3 independent cohorts of healthy controls and SSc patients. Possible functions of miRNA-618 (miR-618) on PDC biology were identified by overexpression in healthy PDCs. RESULTS Expression of miR-618 was up-regulated in PDCs from SSc patients, including those with early disease who did not present with skin fibrosis. IFN regulatory factor 8, a crucial transcription factor for PDC development and activation, was identified as a target of miR-618. Overexpression of miR-618 reduced the development of PDCs from CD34+ cells in vitro and enhanced their ability to secrete IFNα, mimicking the PDC phenotype observed in SSc patients. CONCLUSION Up-regulation of miR-618 suppresses the development of PDCs and increases their ability to secrete IFNα, potentially contributing to the type I IFN signature observed in SSc patients. Considering the importance of PDCs in the pathogenesis of SSc and other diseases characterized by a type I IFN signature, miR-618 potentially represents an important epigenetic target to regulate immune system homeostasis in these conditions.
Collapse
Affiliation(s)
- Marzia Rossato
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | - Marta Cossu
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Lara Bossini-Castillo
- Consejo Superior de Investigaciones Científicas, Granada, Spain, and Wellcome Trust Sanger Institute, Cambridge, UK
| | - Eleni Chouri
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lenny van Bon
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Femke Wolters
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wioleta Marut
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | - Harry Dolstra
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Javier Martin
- Consejo Superior de Investigaciones Científicas, Granada, Spain
| | | | | | - Lorenzo Beretta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | | |
Collapse
|
39
|
Chen WC, Wang WC, Okada Y, Chang WP, Chou YH, Chang HH, Huang JD, Chen DY, Chang WC. rs2841277 ( PLD4) is associated with susceptibility and rs4672495 is associated with disease activity in rheumatoid arthritis. Oncotarget 2017; 8:64180-64190. [PMID: 28969061 PMCID: PMC5609993 DOI: 10.18632/oncotarget.19419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases, can lead to long-term joint damage, chronic pain, and loss of motor function in the hands, and may share some common genetic factors with other autoimmune disorders, such as ankylosing spondylitis (AS). Many single-nucleotide polymorphisms (SNPs) were reported by genome-wide association studies (GWASs) of RA, but some of them have not been examined in the Taiwanese population. In this study, for 15 SNPs reported in previous RA and AS GWASs, we investigated their association with RA in a Taiwanese population. Based on 334 RA patients recruited from the Taichung Veterans General Hospital and 16,036 healthy subjects from the Taiwan Biobank (TWB) project, we observed that subjects having minor allele C at rs2841277 (phospholipase D family, member 4 (PLD4)) have lower susceptibility of RA, compare to those having genotype TT (Odds ratio (OR) = 0.6, p = 3.0 × 10−6). Among the RA patients, we observed that subjects having GG at rs4672495 have a lower proportion of severe RA, compare to other subjects (OR = 0.09, p = 5.6 × 10−3). Results of a bioinformatics approach showed that rs2841277 is able to influence expression of LINC00638 and AHNAK2 and rs4672495 is able to influence the expression of B3GNT2. In summary, this study replicated an association of rs2841277 with RA susceptibility and showed an AS-associated SNP, rs4672495, is associated with RA activity in the Taiwanese population.
Collapse
Affiliation(s)
- Wei-Chiao Chen
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Chang Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yukinori Okada
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Wei-Pin Chang
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei, Taiwan
| | - Yii-Her Chou
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jin-Ding Huang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Der-Yuan Chen
- Department of Internal Medicine and Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, National Yang Ming University, Taipei, Taiwan.,Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Chiao Chang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacy, Taipei Medical University-Wanfang Hospital, Taipei, Taiwan
| |
Collapse
|
40
|
Terao C, Kawaguchi T, Dieude P, Varga J, Kuwana M, Hudson M, Kawaguchi Y, Matucci-Cerinic M, Ohmura K, Riemekasten G, Kawasaki A, Airo P, Horita T, Oka A, Hachulla E, Yoshifuji H, Caramaschi P, Hunzelmann N, Baron M, Atsumi T, Hassoun P, Torii T, Takahashi M, Tabara Y, Shimizu M, Tochimoto A, Ayuzawa N, Yanagida H, Furukawa H, Tohma S, Hasegawa M, Fujimoto M, Ishikawa O, Yamamoto T, Goto D, Asano Y, Jinnin M, Endo H, Takahashi H, Takehara K, Sato S, Ihn H, Raychaudhuri S, Liao K, Gregersen P, Tsuchiya N, Riccieri V, Melchers I, Valentini G, Cauvet A, Martinez M, Mimori T, Matsuda F, Allanore Y. Transethnic meta-analysis identifies GSDMA and PRDM1 as susceptibility genes to systemic sclerosis. Ann Rheum Dis 2017; 76:1150-1158. [PMID: 28314753 PMCID: PMC6733404 DOI: 10.1136/annrheumdis-2016-210645] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/08/2017] [Accepted: 02/21/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is an autoimmune disease characterised by skin and systemic fibrosis culminating in organ damage. Previous genetic studies including genome-wide association studies (GWAS) have identified 12 susceptibility loci satisfying genome-wide significance. Transethnic meta-analyses have successfully expanded the list of susceptibility genes and deepened biological insights for other autoimmune diseases. METHODS We performed transethnic meta-analysis of GWAS in the Japanese and European populations, followed by a two-staged replication study comprising a total of 4436 cases and 14 751 controls. Associations between significant single nuclear polymorphisms (SNPs) and neighbouring genes were evaluated. Enrichment analysis of H3K4Me3, a representative histone mark for active promoter was conducted with an expanded list of SSc susceptibility genes. RESULTS We identified two significant SNP in two loci, GSDMA and PRDM1, both of which are related to immune functions and associated with other autoimmune diseases (p=1.4×10-10 and 6.6×10-10, respectively). GSDMA also showed a significant association with limited cutaneous SSc. We also replicated the associations of previously reported loci including a non-GWAS locus, TNFAIP3. PRDM1 encodes BLIMP1, a transcription factor regulating T-cell proliferation and plasma cell differentiation. The top SNP in GSDMA was a missense variant and correlated with gene expression of neighbouring genes, and this could explain the association in this locus. We found different human leukocyte antigen (HLA) association patterns between the two populations. Enrichment analysis suggested the importance of CD4-naïve primary T cell. CONCLUSIONS GSDMA and PRDM1 are associated with SSc. These findings provide enhanced insight into the genetic and biological basis of SSc.
Collapse
Affiliation(s)
- Chikashi Terao
- Department of Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University Graduate School of
Medicine, Kyoto, Japan
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital, Boston,
Massachusetts, USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts,
USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Takahisa Kawaguchi
- Department of Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Philippe Dieude
- Rheumatology Bichat Hospital, Paris, University, Paris, France
| | - John Varga
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Masataka Kuwana
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo,
Japan
| | - Marie Hudson
- Jewish General Hospital and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Yasushi Kawaguchi
- Institute of Rheumatology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Marco Matucci-Cerinic
- Division of Rheumatology AOUC, Department of Experimental and Clinical Medicine, Department of Medical
& Geriatrics Medicine, University of Florence, Firenze, Italy
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto,
Japan
| | - Gabriela Riemekasten
- Clinic for Rheumatology, University of Lübeck, Lübeck, Germany
- German Lung Center Borstel, Leibniz Institute, Germany
| | - Aya Kawasaki
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba,
Japan
| | - Paolo Airo
- Rheumatology Unit, Spedali Civili, Brescia, Italy
| | - Tetsuya Horita
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University, Graduate School of Medicine,
Sapporo, Japan
| | - Akira Oka
- The Institute of Medical Science, Tokai University, Isehara, Japan
| | - Eric Hachulla
- Internal Medicine Department, FHU Immune-Mediated Inflammatory Diseases and Targeted Therapies, Lille
University, Lille, France
| | - Hajime Yoshifuji
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto,
Japan
| | - Paola Caramaschi
- Rheumatology Department, University of Verona, Azienda Ospedaliera Universitaria Integrata, Italy
| | | | - Murray Baron
- Jewish General Hospital and Lady Davis Research Institute, Montreal, Quebec, Canada
| | - Tatsuya Atsumi
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University, Graduate School of Medicine,
Sapporo, Japan
| | - Paul Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University,
Baltimore, Maryland, USA
| | | | - Meiko Takahashi
- Department of Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuharu Tabara
- Department of Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masakazu Shimizu
- Department of Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akiko Tochimoto
- Institute of Rheumatology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Naho Ayuzawa
- Department of Rheumatology, National Hospital Organization, Utano National Hospital, Kyoto, Japan
| | - Hidetoshi Yanagida
- Department of Rheumatology, National Hospital Organization, Utano National Hospital, Kyoto, Japan
| | - Hiroshi Furukawa
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba,
Japan
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization,
Sagamihara, Japan
| | - Shigeto Tohma
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization,
Sagamihara, Japan
| | - Minoru Hasegawa
- Division of Medicine, Faculty of Medical Sciences, Department of Dermatology, University of Fukui, Fukui,
Japan
| | - Manabu Fujimoto
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Osamu Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Toshiyuki Yamamoto
- Department of Dermatology, Fukushima Medical University, Fukushima, Japan
| | - Daisuke Goto
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masatoshi Jinnin
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto,
Japan
| | - Hirahito Endo
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Toho University, Tokyo,
Japan
| | - Hiroki Takahashi
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo,
Hokkaido, Japan
| | - Kazuhiko Takehara
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences,
Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto,
Japan
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital, Boston,
Massachusetts, USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts,
USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Faculty of
Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Katherine Liao
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital, Boston,
Massachusetts, USA
| | - Peter Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research,
Manhasset, New York, USA
| | - Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba,
Japan
| | | | | | - Gabriele Valentini
- Department of Clinical and Experimental Medicine, Rheumatology Section, Second University of Naples,
Naples, Italy
| | - Anne Cauvet
- INSERM U1016/UMR 8104, Cochin Institute, Paris Descartes University, Paris, France
| | - Maria Martinez
- INSERM U1220—IRSD—Batiment B Purpan Hospital Toulouse, Paris, France
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto,
Japan
| | - Fumihiko Matsuda
- Department of Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yannick Allanore
- Rheumatology A Department, INSERM U1016/uMr 8104, Cochin Institute, Paris Descartes University,
Paris, France
| |
Collapse
|
41
|
Tsou PS, Sawalha AH. Unfolding the pathogenesis of scleroderma through genomics and epigenomics. J Autoimmun 2017; 83:73-94. [PMID: 28526340 DOI: 10.1016/j.jaut.2017.05.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 12/12/2022]
Abstract
With unknown etiology, scleroderma (SSc) is a multifaceted disease characterized by immune activation, vascular complications, and excessive fibrosis in internal organs. Genetic studies, including candidate gene association studies, genome-wide association studies, and whole-exome sequencing have supported the notion that while genetic susceptibility to SSc appears to be modest, SSc patients are genetically predisposed to this disease. The strongest genetic association for SSc lies within the MHC region, with loci in HLA-DRB1, HLA-DQB1, HLA-DPB1, and HLA-DOA1 being the most replicated. The non-HLA genes associated with SSc are involved in various functions, with the most robust associations including genes for B and T cell activation and innate immunity. Other pathways include genes involved in extracellular matrix deposition, cytokines, and autophagy. Among these genes, IRF5, STAT4, and CD247 were replicated most frequently while SNPs rs35677470 in DNASE1L3, rs5029939 in TNFAIP3, and rs7574685 in STAT4 have the strongest associations with SSc. In addition to genetic predisposition, it became clear that environmental factors and epigenetic influences also contribute to the development of SSc. Epigenetics, which refers to studies that focus on heritable phenotypes resulting from changes in chromatin structure without affecting the DNA sequence, is one of the most rapidly expanding fields in biomedical research. Indeed extensive epigenetic changes have been described in SSc. Alteration in enzymes and mediators involved in DNA methylation and histone modification, as well as dysregulated non-coding RNA levels all contribute to fibrosis, immune dysregulation, and impaired angiogenesis in this disease. Genes that are affected by epigenetic dysregulation include ones involved in autoimmunity, T cell function and regulation, TGFβ pathway, Wnt pathway, extracellular matrix, and transcription factors governing fibrosis and angiogenesis. In this review, we provide a comprehensive overview of the current findings of SSc genetic susceptibility, followed by an extensive description and a systematic review of epigenetic research that has been carried out to date in SSc. We also summarize the therapeutic potential of drugs that affect epigenetic mechanisms, and outline the future prospective of genomics and epigenomics research in SSc.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
42
|
Positive Association between ANKRD55 Polymorphism 7731626 and Dermatomyositis/Polymyositis with Interstitial Lung Disease in Chinese Han Population. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2905987. [PMID: 28470010 PMCID: PMC5392395 DOI: 10.1155/2017/2905987] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 12/22/2022]
Abstract
Single nucleotide polymorphisms (SNPs) in TNFSF4 and ANKRD55 genes have been shown to be associated with several autoimmune diseases, although whether these genes are susceptibility genes for dermatomyositis/polymyositis (DM/PM) has, to date, not been reported. This study aimed to investigate the potential associations of these SNPs with DM/PM in a Chinese Han population. Five SNPs in TNFSF4 (rs2205960, rs844644, and rs844648) and ANKRD55 (rs6859219, rs7731626) genes were genotyped using the SequenomMassArray system in 2297 Chinese individuals. In total, 1017 DM/PM patients and 1280 gender-matched healthy controls were genotyped. No significant associations were observed in DM/PM patients for the five SNPs analyzed. The association between SNPs and interstitial lung disease (ILD) was also investigated. Both DM-ILD (Pc = 0.030, OR = 0.65, 95% CI: 0.47–0.88) and DM/PM-ILD (Pc = 0.015, OR = 0.67, 95% CI: 0.51–0.87) exhibited a significant association with the rs7731626-A allele. Rs7731626-A was less frequently found in DM-ILD and DM/PM-ILD patients compared with healthy controls. This is the first study to demonstrate a positive association between ANKRD55 polymorphism and DM-ILD and DM/PM-ILD. A decreased frequency of rs7731626-A in DM-ILD and DM/PM-ILD patients suggests that the A variant may be protective against DM/PM-ILD.
Collapse
|
43
|
Pardo J, Abba MC, Lacunza E, Francelle L, Morel GR, Outeiro TF, Goya RG. Identification of a conserved gene signature associated with an exacerbated inflammatory environment in the hippocampus of aging rats. Hippocampus 2017; 27:435-449. [DOI: 10.1002/hipo.22703] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Joaquín Pardo
- INIBIOLP, Histology B–Pathology B, School of Medicine, UNLPLa Plata Argentina
| | | | | | - Laetitia Francelle
- Department of Neurodegeneration and Restorative ResearchUniversity Medical Center GöttingenGöttingen Germany
| | - Gustavo R. Morel
- INIBIOLP, Histology B–Pathology B, School of Medicine, UNLPLa Plata Argentina
| | - Tiago F. Outeiro
- Department of Neurodegeneration and Restorative ResearchUniversity Medical Center GöttingenGöttingen Germany
| | - Rodolfo G. Goya
- INIBIOLP, Histology B–Pathology B, School of Medicine, UNLPLa Plata Argentina
| |
Collapse
|
44
|
Gao L, Zhou Y, Zhou SX, Yu XJ, Xu JM, Zuo L, Luo YH, Li XA. PLD4 promotes M1 macrophages to perform antitumor effects in colon cancer cells. Oncol Rep 2016; 37:408-416. [PMID: 27840999 DOI: 10.3892/or.2016.5216] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/17/2016] [Indexed: 11/06/2022] Open
Abstract
Phospholipase D4 (PLD4) is a newly identified protein expressed in microglia. However, the function of PLD4 in tumor-associated macrophages (TAMs) is unknown. In the present study, we revealed that the expression of PLD4 was located in macrophages in the colon cancer mesenchymal and lymph nodes as shown by immunohistochemical analysis. furthermore, its expression was associated with clinical staging of colon cancer. Then, THP-1 as a cell model induced into TAMs. Western blot and RT-PCR analysis showed that PLD4 was mainly presented in M1 phenotype TAMs. The secretion of pro-inflammatory cytokines in M1 macrophages was significantly reduced after the expression of PLD4 inhibited by PLD4-siRNA. Furthermore, co-cultured with condition-medium from control or PLD4-siRNA M1 macrophages for 24 h, cell apoptosis, cycle and proliferation of cancer cells improved compared to control. These results indicated that PLD4 could be involved in the activation process of M1 phenotype macrophages.
Collapse
Affiliation(s)
- Long Gao
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Yan Zhou
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Shu-Xian Zhou
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Xian-Jing Yu
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Jin-Mei Xu
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Luo Zuo
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Yong-Hui Luo
- Burn and Plastic Surgery Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiao-An Li
- The Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
45
|
Multiple genes, especially immune-regulating genes, contribute to disease susceptibility in systemic sclerosis. Curr Opin Rheumatol 2016; 28:595-605. [DOI: 10.1097/bor.0000000000000334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Chairta P, Nicolaou P, Christodoulou K. Genomic and genetic studies of systemic sclerosis: A systematic review. Hum Immunol 2016; 78:153-165. [PMID: 27984087 DOI: 10.1016/j.humimm.2016.10.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
Systemic sclerosis is an autoimmune rheumatic disease characterised by fibrosis, vasculopathy and inflammation. The exact aetiology of SSc remains unknown but evidences show that various genetic factors may be involved. This review aimed to assess HLA alleles/non-HLA polymorphisms, microsatellites and chromosomal abnormalities that have thus far been associated with SSc. PubMed, Embase and Scopus databases were searched up to July 29, 2015 using a combination of search-terms. Articles retrieved were evaluated based on set exclusion and inclusion criteria. A total of 150 publications passed the filters. HLA and non-HLA studies showed that particular alleles in the HLA-DRB1, HLA-DQB1, HLA-DQA1, HLA-DPB1 genes and variants in STAT4, IRF5 and CD247 are frequently associated with SSc. Non-HLA genes analysis was performed using the PANTHER and STRING10 databases. PANTHER classification revealed that inflammation mediated by chemokine and cytokine, interleukin and integrin signalling pathways are among the common extracted pathways associated with SSc. STRING10 analysis showed that NFKB1, CSF3R, STAT4, IFNG, PRL and ILs are the main "hubs" of interaction network of the non-HLA genes associated with SSc. This study gathers data of valid genetic factors associated with SSc and discusses the possible interactions of implicated molecules.
Collapse
Affiliation(s)
- Paraskevi Chairta
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus
| | - Paschalis Nicolaou
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus
| | - Kyproula Christodoulou
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus.
| |
Collapse
|
47
|
Philip F, Ha EE, Seeliger MA, Frohman MA. Measuring Phospholipase D Enzymatic Activity Through Biochemical and Imaging Methods. Methods Enzymol 2016; 583:309-325. [PMID: 28063496 DOI: 10.1016/bs.mie.2016.09.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The phospholipase D (PLD) enzymatic superfamily regulates a wide range of cell biological and physiological pathways, including platelet activation, immune responses, cancer, and spermatogenesis. The three main enzymatic actions of the superfamily entail (i) hydrolyzing membrane phospholipids (phosphatidylcholine (PC) and cardiolipin) to generate choline and the second messenger signaling lipid phosphatidic acid (PA), (ii) using ethanol to transphosphatidylate PC to generate the long-lived metabolite phosphatidylethanol, and (iii) hydrolyzing RNA transcripts to generate piRNAs, the third form of endogenous RNAi. We discuss briefly previously published methods for in vitro and in vivo detection and imaging of PA, and focus on production, purification, and in vitro endonuclease activity analysis for human PLD6, a mitochondrial-tethered isoform with roles in fertility, cancer, and neuronal homeostasis.
Collapse
Affiliation(s)
- F Philip
- Center for Developmental Genetics, Stony Brook University School of Medicine, Stony Brook, NY, United States
| | - E E Ha
- Center for Developmental Genetics, Stony Brook University School of Medicine, Stony Brook, NY, United States
| | - M A Seeliger
- Center for Developmental Genetics, Stony Brook University School of Medicine, Stony Brook, NY, United States
| | - M A Frohman
- Center for Developmental Genetics, Stony Brook University School of Medicine, Stony Brook, NY, United States.
| |
Collapse
|
48
|
Abstract
Purpose of review Large-scale and follow-up genetic association studies in systemic sclerosis (SSc) have implicated over 40 regions in disease risk, 15 of which with robust associations. Nevertheless, the causal variants and the functional mechanisms underlying the genetic associations remain elusive, and the reasons for the higher disease burden in African Americans unknown. Incorporating tools from diverse fields is beginning to unveil the role of genetic diversity and regulatory variation in SSc susceptibility. This review will summarize recent advances in SSc genetics, including autoimmune disease overlap, evidence of natural selection, and current progress towards the dissection of the functional role of associated risk variants. Recent findings In the past year, multiple large-scale studies reported novel strong and suggestive SSc associations. These results, coupled with the regions shared with other autoimmune diseases, emphasize the role of dysregulation of immune pathways as a key causative factor in SSc pathogenesis. Strong evidence implicates natural selection as a mechanism contributing to the maintenance of some of these SSc alleles in the population. Studies integrating genomic, transcriptomic, and epigenomic datasets in specific cell types to identify causal autoimmune disease variants are emerging. Summary The identification and comprehensive understanding of the factors and mechanisms contributing to SSc will contribute to improved diagnosis and disease management.
Collapse
|
49
|
Fuschiotti P. Current perspectives on the immunopathogenesis of systemic sclerosis. Immunotargets Ther 2016; 5:21-35. [PMID: 27529059 PMCID: PMC4970639 DOI: 10.2147/itt.s82037] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic sclerosis (SSc or scleroderma) is a progressive and highly debilitating autoimmune disorder characterized by inflammation, vasculopathy, and extensive fibrosis. SSc is highly heterogeneous in its clinical presentation, extent and severity of skin and internal organ involvement, and clinical course and has the highest fatality rate among connective tissue diseases. While clinical outcomes have improved in recent years, no current therapy is able to reverse or slow the natural progression of SSc, a reflection of its complex pathogenesis. Although activation of the immune system has long been recognized, the mechanisms responsible for the initiation of autoimmunity and the role of immune effector pathways in the pathogenesis of SSc remain incompletely understood. This review summarizes recent progress in disease pathogenesis with particular focus on the immunopathogenetic mechanisms of SSc.
Collapse
Affiliation(s)
- Patrizia Fuschiotti
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
50
|
Abstract
Phospholipases are lipid-metabolizing enzymes that hydrolyze phospholipids. In some cases, their activity results in remodeling of lipids and/or allows the synthesis of other lipids. In other cases, however, and of interest to the topic of adrenal steroidogenesis, phospholipases produce second messengers that modify the function of a cell. In this review, the enzymatic reactions, products, and effectors of three phospholipases, phospholipase C, phospholipase D, and phospholipase A2, are discussed. Although much data have been obtained concerning the role of phospholipases C and D in regulating adrenal steroid hormone production, there are still many gaps in our knowledge. Furthermore, little is known about the involvement of phospholipase A2, perhaps, in part, because this enzyme comprises a large family of related enzymes that are differentially regulated and with different functions. This review presents the evidence supporting the role of each of these phospholipases in steroidogenesis in the adrenal cortex.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical CenterOne Freedom Way, Augusta, GA, USA Department of PhysiologyMedical College of Georgia, Augusta University (formerly Georgia Regents University), Augusta, GA, USA
| |
Collapse
|