101
|
Thomas DC. How the phagocyte NADPH oxidase regulates innate immunity. Free Radic Biol Med 2018; 125:44-52. [PMID: 29953922 DOI: 10.1016/j.freeradbiomed.2018.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/05/2018] [Accepted: 06/11/2018] [Indexed: 11/16/2022]
Abstract
The phagocyte NADPH oxidase is a multi subunit protein complex that generates reactive oxygen species at cell membranes and within phagosomes. It is essential for host defence as evidenced by the severe immunodeficiency syndrome caused by a loss of one of the subunits. This is known as chronic granulomatous disease (CGD). However, the phagocyte NADPH oxidase also has a key role to play in regulating immunity and it is notable that chronic granulomatous disease is also characterised by autoimmune and autoinflammatory manifestations. This is because reactive oxygen species play a role in regulating signalling through their ability to post-translationally modify amino acid residues such as cysteine and methionine. In this review, I will outline the major aspects of innate immunity that are regulated by the phagocyte NADPH oxidase, including control of transcription, autophagy, the inflammasome and type 1 interferon signalling.
Collapse
Affiliation(s)
- David C Thomas
- Department of Medicine, University of Cambridge School of Clinical Medicine, Box 157 Cambridge Biomedical Campus, Cambridge CB2 0QQ, United Kingdom.
| |
Collapse
|
102
|
Comprehensive assessment of the association between genes on JAK-STAT pathway (IFIH1, TYK2, IL-10) and systemic lupus erythematosus: a meta-analysis. Arch Dermatol Res 2018; 310:711-728. [DOI: 10.1007/s00403-018-1858-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/19/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
|
103
|
Lässig C, Lammens K, Gorenflos López JL, Michalski S, Fettscher O, Hopfner KP. Unified mechanisms for self-RNA recognition by RIG-I Singleton-Merten syndrome variants. eLife 2018; 7:e38958. [PMID: 30047865 PMCID: PMC6086658 DOI: 10.7554/elife.38958] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
The innate immune sensor retinoic acid-inducible gene I (RIG-I) detects cytosolic viral RNA and requires a conformational change caused by both ATP and RNA binding to induce an active signaling state and to trigger an immune response. Previously, we showed that ATP hydrolysis removes RIG-I from lower-affinity self-RNAs (Lässig et al., 2015), revealing how ATP turnover helps RIG-I distinguish viral from self-RNA and explaining why a mutation in a motif that slows down ATP hydrolysis causes the autoimmune disease Singleton-Merten syndrome (SMS). Here we show that a different, mechanistically unexplained SMS variant, C268F, which is localized in the ATP-binding P-loop, can signal independently of ATP but is still dependent on RNA. The structure of RIG-I C268F in complex with double-stranded RNA reveals that C268F helps induce a structural conformation in RIG-I that is similar to that induced by ATP. Our results uncover an unexpected mechanism to explain how a mutation in a P-loop ATPase can induce a gain-of-function ATP state in the absence of ATP.
Collapse
Affiliation(s)
- Charlotte Lässig
- Department of BiochemistryLudwig-Maximilians-Universität MünchenMunichGermany
- Gene Center, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Katja Lammens
- Department of BiochemistryLudwig-Maximilians-Universität MünchenMunichGermany
- Gene Center, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Jacob Lucián Gorenflos López
- Department of BiochemistryLudwig-Maximilians-Universität MünchenMunichGermany
- Gene Center, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Sebastian Michalski
- Department of BiochemistryLudwig-Maximilians-Universität MünchenMunichGermany
- Gene Center, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Olga Fettscher
- Department of BiochemistryLudwig-Maximilians-Universität MünchenMunichGermany
- Gene Center, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Karl-Peter Hopfner
- Department of BiochemistryLudwig-Maximilians-Universität MünchenMunichGermany
- Gene Center, Ludwig-Maximilians-Universität MünchenMunichGermany
- Center for Integrated Protein Science MunichMunichGermany
| |
Collapse
|
104
|
Zhang J, Liu X, Meng Y, Wu H, Wu Y, Yang B, Wang L. Autoimmune disease associated IFIH1 single nucleotide polymorphism related with IL-18 serum levels in Chinese systemic lupus erythematosus patients. Sci Rep 2018; 8:9442. [PMID: 29930297 PMCID: PMC6013496 DOI: 10.1038/s41598-018-27782-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/08/2018] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) has heterogeneous clinical manifestations. IFIH1 (interferon induced with helicase C domain 1) as one of antiviral helicase genes mediating type I interferon production, plays an essential role in the pathogenesis of SLE. The gene variants in IFIH1 could abnormally activate antiviral defenses and increased type I interferon signaling. The present study aimed to validate associations between single nucleotide polymorphisms (SNP) in IFIH1 and the pathogenesis of SLE. In total, rs1990760, rs3747517 and rs10930046 in IFIH1 are genotyped in 400 SLE patients and 659 health controls in Chinese cohort by an improved multiplex ligation detection reaction (iMLDR) technique. Significant associations were observed between alleles of IFIH1 (rs1990760 C > T, P = 0.005, OR = 1.36, 95%CI = 1.10–1.69; rs3747517 T > C, P = 0.004, OR = 1.31, 95%CI = 1.09–1.58, respectively) and SLE susceptibility. IFIH1 rs1990760 TT genotype carriers had lower serum levels of IL-18 (P < 0.001) and granzyme B (P < 0.001) than CC and CT genotype carriers. IFIH1 rs1990760 CT genotype carriers had higher anti-dsDNA–positive than CC and TT genotype carriers. In conclusion, IFIH1 polymorphisms (rs1990760 and rs3747517) were associated with SLE susceptibility and rs1990760 risk T allele related with IL-18 and granzyme B serum levels in SLE patients.
Collapse
Affiliation(s)
- Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xinle Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yanming Meng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hengxu Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yongkang Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Lanlan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
105
|
Krishnan Y, Mukundan S, Akhil S, Gupta S, Viswanad V. Enhanced Lymphatic Uptake of Leflunomide Loaded Nanolipid Carrier via Chylomicron Formation for the Treatment of Rheumatoid Arthritis. Adv Pharm Bull 2018; 8:257-265. [PMID: 30023327 PMCID: PMC6046432 DOI: 10.15171/apb.2018.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/15/2018] [Accepted: 05/19/2018] [Indexed: 12/12/2022] Open
Abstract
Purpose: The current study aims the lymphatic delivery of leflunomide loaded nanostructured lipid carriers (LNLC) for the treatment of rheumatoid arthritis, mainly focussed to enhance the lymphatic delivery via chylomicron formation, improved bioavailability and reduced systemic toxicity. Methods: Melt emulsification ultra-sonication method was used to formulate the nanostructured lipid carrier (NLC) containing leflunomide. Four batches were prepared by using various concentration of surfactants (tween 80 and poloxmer 188) and lipid mixtures (stearic acid and oleic acid). All the formulations were studied for various physiochemical properties Results: The formulation with increased concentration of lipid and surfactants showed highest entrapment efficiency (93.96 ± 0.47%) and better drug release (90.35%) at the end of 48 hrs. In vivo tests were carried out to determine the antiarthritic potential of the formulation in Sprague-dawley rats for a duration of 30d. The effect was evaluated by measuring the reduction in knee thickness. LNLC showed a marked reduction in inflammation compared to standard drug. Intestinal lymphatic uptake studies of LNLC were performed by intraduodenal administration and compared with leflunomide drug solution. The mesenteric lymph node was analysed by HPLC method and the concentration of drug was estimated. It showed that LNLC having highest uptake (40.34μg/ml) when compared with leflunomide drug solution (10.04μg/ml). Radiographic analysis and histopathological studies showed the formation of healthy cartilage after treatment period. Conclusion: The results suggested that LNLC has the potential to reduce the systemic toxicities associated with conventional therapy along with improved efficacy in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Yadhu Krishnan
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi - 682041, India
| | - Shilpa Mukundan
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi - 682041, India
| | - Suresh Akhil
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi - 682041, India
| | - Swati Gupta
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi - 682041, India
| | - Vidya Viswanad
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi - 682041, India
| |
Collapse
|
106
|
Enerbäck C, Sandin C, Lambert S, Zawistowski M, Stuart PE, Verma D, Tsoi LC, Nair RP, Johnston A, Elder JT. The psoriasis-protective TYK2 I684S variant impairs IL-12 stimulated pSTAT4 response in skin-homing CD4+ and CD8+ memory T-cells. Sci Rep 2018; 8:7043. [PMID: 29728633 PMCID: PMC5935702 DOI: 10.1038/s41598-018-25282-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/16/2018] [Indexed: 01/11/2023] Open
Abstract
Tyrosine kinase 2 (TYK2) belongs to the Janus kinase (JAK) family of tyrosine kinases, which transmit signals from activated cytokine receptors. GWAS have consistently implicated TYK2 in psoriasis susceptibility. We performed an in-depth association analysis of TYK2 using GWAS and resequencing data. Strong genetic association of three nonsynonymous variants in the exonic regions of the TYK2 gene (rs34536443, rs12720356, and rs2304256) were found. rs12720356 encoding I684S is predicted to be deleterious based on its location in the pseudokinase domain. We analyzed PBMCs from 29 individuals representing the haplotypes containing each of the significantly associated signals. STAT4 phosphorylation was evaluated by phospho-flow cytometry after CD3/CD28 activation of cells followed by IL-12 stimulation. Individuals carrying the protective I684S variant manifested significantly reduced p-STAT4 levels in CD4 + CD25 + CD45RO+ (mean Stimulation Index (S.I.) 48.08, n = 10) and CD8 + CD25 + CD45RO + cells (S.I. 55.71, n = 10), compared to controls homozygous for the ancestral haplotype (S.I. 68.19, n = 10 (p = 0.002) and 76.76 n = 10 (p = 0.0008) respectively). Reduced p-STAT4 levels were also observed in skin-homing, cutaneous lymphocyte associated antigen (CLA)-positive CD4 and CD8 cells from I684S carriers. No significant changes in p-STAT4 for the psoriasis-associated variant rs34536443 was found. These data establish the functional significance of the TYK2 I684S variant in psoriasis susceptibility.
Collapse
Affiliation(s)
- C Enerbäck
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA. .,Ingrid Asp Psoriasis Research Center, Department of Dermatology, Linköping University, Linköping, Sweden.
| | - C Sandin
- Ingrid Asp Psoriasis Research Center, Department of Dermatology, Linköping University, Linköping, Sweden
| | - S Lambert
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - M Zawistowski
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - P E Stuart
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - D Verma
- Ingrid Asp Psoriasis Research Center, Department of Dermatology, Linköping University, Linköping, Sweden
| | - L C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA.,Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - R P Nair
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - A Johnston
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - J T Elder
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA.,Ann Arbor Veterans Affairs Health System, Ann Arbor, MI, USA
| |
Collapse
|
107
|
NADPH oxidase activation regulates apoptotic neutrophil clearance by murine macrophages. Blood 2018; 131:2367-2378. [PMID: 29618478 DOI: 10.1182/blood-2017-09-809004] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/29/2018] [Indexed: 02/07/2023] Open
Abstract
The phagocyte reduced NAD phosphate (NADPH) oxidase generates superoxide, the precursor to reactive oxygen species (ROS) that has both antimicrobial and immunoregulatory functions. Inactivating mutations in NADPH oxidase alleles cause chronic granulomatous disease (CGD), characterized by enhanced susceptibility to life-threatening microbial infections and inflammatory disorders; hypomorphic NADPH oxidase alleles are associated with autoimmunity. Impaired apoptotic cell (AC) clearance is implicated as an important contributing factor in chronic inflammation and autoimmunity, but the role of NADPH oxidase-derived ROS in this process is incompletely understood. Here, we demonstrate that phagocytosis of AC (efferocytosis) potently activated NADPH oxidase in mouse peritoneal exudate macrophages (PEMs). ROS generation was dependent on macrophage CD11b, Toll-like receptor 2 (TLR2), TLR4, and myeloid differentiation primary response 88 (MyD88), and was also regulated by phosphatidylinositol 3-phosphate binding to the p40 phox oxidase subunit. Maturation of efferosomes containing apoptotic neutrophils was significantly delayed in CGD PEMs, including acidification and acquisition of proteolytic activity, and was associated with slower digestion of apoptotic neutrophil proteins. Treatment of wild-type macrophages with the vacuolar-type H+ ATPase inhibitor bafilomycin also delayed proteolysis within efferosomes, showing that luminal acidification was essential for efficient digestion of efferosome proteins. Finally, cross-presentation of AC-associated antigens by CGD PEMs to CD8 T cells was increased. These studies unravel a key role for the NADPH oxidase in the disposal of ACs by inflammatory macrophages. The oxidants generated promote efferosome maturation and acidification that facilitate the degradation of ingested ACs.
Collapse
|
108
|
Malkiel S, Barlev AN, Atisha-Fregoso Y, Suurmond J, Diamond B. Plasma Cell Differentiation Pathways in Systemic Lupus Erythematosus. Front Immunol 2018; 9:427. [PMID: 29556239 PMCID: PMC5845388 DOI: 10.3389/fimmu.2018.00427] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/16/2018] [Indexed: 01/20/2023] Open
Abstract
Plasma cells (PCs) are responsible for the production of protective antibodies against infectious agents but they also produce pathogenic antibodies in autoimmune diseases, such as systemic lupus erythematosus (SLE). Traditionally, high affinity IgG autoantibodies are thought to arise through germinal center (GC) responses. However, class switching and somatic hypermutation can occur in extrafollicular (EF) locations, and this pathway has also been implicated in SLE. The pathway from which PCs originate may determine several characteristics, such as PC lifespan and sensitivity to therapeutics. Although both GC and EF responses have been implicated in SLE, we hypothesize that one of these pathways dominates in each individual patient and genetic risk factors may drive this predominance. While it will be important to distinguish polymorphisms that contribute to a GC-driven or EF B cell response to develop targeted treatments, the challenge will be not only to identify the differentiation pathway but the molecular mechanisms involved. In B cells, this task is complicated by the cross-talk between the B cell receptor, toll-like receptors (TLR), and cytokine signaling molecules, which contribute to both GC and EF responses. While risk variants that affect the function of dendritic cells and T follicular helper cells are likely to primarily influence GC responses, it will be important to discover whether some risk variants in the interferon and TLR pathways preferentially influence EF responses. Identifying the pathways of autoreactive PC differentiation in SLE may help us to understand patient heterogeneity and thereby guide precision therapy.
Collapse
Affiliation(s)
- Susan Malkiel
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Ashley N Barlev
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Yemil Atisha-Fregoso
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States.,Tecnologico de Monterrey, Monterrey, Mexico
| | - Jolien Suurmond
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Betty Diamond
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| |
Collapse
|
109
|
Lindén M, Ramírez Sepúlveda JI, James T, Thorlacius GE, Brauner S, Gómez-Cabrero D, Olsson T, Kockum I, Wahren-Herlenius M. Sex influences eQTL effects of SLE and Sjögren's syndrome-associated genetic polymorphisms. Biol Sex Differ 2017; 8:34. [PMID: 29070082 PMCID: PMC5657123 DOI: 10.1186/s13293-017-0153-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/09/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) and primary Sjögren's syndrome (pSS) are autoimmune disorders characterized by autoantibodies, dysregulated B cells, and notably high female-to-male incidence ratios. Genome-wide association studies have identified several susceptibility SNPs for both diseases. Many SNPs in the genome are expression quantitative trait loci (eQTLs), with context-dependent effects. Assuming that sex is a biological context, we investigated whether SLE/pSS SNPs act as eQTLs in B cells and used a disease-targeted approach to understand if they display sex-specific effects. METHODS We used genome-wide genotype and gene expression data from primary B cells from 125 males and 162 females. The MatrixEQTL R package was used to identify eQTLs within a genomic window of 2 Mb centered on each of 22 established SLE and/or pSS susceptibility SNPs. To find sex-specific eQTLs, we used a linear model with a SNP * sex interaction term. RESULTS We found ten SNPs affecting the expression of 16 different genes (FDR < 0.05). rs7574865-INPP1, rs7574865-MYO1B, rs4938573-CD3D, rs11755393-SNRPC, and rs4963128-PHRF1 were novel observations for the immune compartment and B cells. By analyzing the SNP * sex interaction terms, we identified six genes with differentially regulated expression in females compared to males, depending on the genotype of SLE/pSS-associated SNPs: SLC39A8 (BANK1 locus), CD74 (TNIP1 locus), PXK, CTSB (BLK/FAM167A locus), ARCN1 (CXCR5 locus), and DHX9 (NCF2 locus). CONCLUSIONS We identified several unknown sex-specific eQTL effects of SLE/pSS-associated genetic polymorphisms and provide novel insight into how gene-sex interactions may contribute to the sex bias in systemic autoimmune diseases.
Collapse
Affiliation(s)
- Magdalena Lindén
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jorge I Ramírez Sepúlveda
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tojo James
- Unit of Neuroimmunology, Department of Clinical Neuroscience, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Gudny Ella Thorlacius
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Brauner
- Unit of Neuroimmunology, Department of Clinical Neuroscience, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - David Gómez-Cabrero
- Unit of Computational Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.,Unit of Clinical Epidemiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, 17121, Solna, Sweden.,Mucosal and Salivary Biology Division, King's College London Dental Institute, London, SE1 9RT, UK
| | - Tomas Olsson
- Unit of Neuroimmunology, Department of Clinical Neuroscience, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Unit of Neuroimmunology, Department of Clinical Neuroscience, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
110
|
Luo W, Zhou B, Luo Q, Fang H, Zuo X, Zou Y. Polymorphism of keratin 1 associates with systemic lupus erythematosus and systemic sclerosis in a south Chinese population. PLoS One 2017; 12:e0186409. [PMID: 29028840 PMCID: PMC5640249 DOI: 10.1371/journal.pone.0186409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/29/2017] [Indexed: 12/15/2022] Open
Abstract
Both systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) diseases are related to the genetic and environmental factors, causing damage to the skin. The mutations of keratin 1 gene (KRT1) were reported to associate with skin diseases. The single-nucleotide polymorphism (SNP, rs14024) and the indel polymorphism (cds-indel, rs267607656), consisting mostly of the common haplotypes and could be used for genotyping of KRT1. We used the PCR with sequence specific primers (PCR-SSP) to determine the genotype of KRT1 in 164 SLE, 99 SSc patients, and 418 healthy controls. The results showed that the mutant with G at SNP rs14024 was associated with the high risk to SLE (p = 6.48×10-5) and SSc (p = 8.75×10-5), while the deletion allele at rs267607656 was associated with the low risk to SSc (p = 4.89×10-4) comparing to the normal controls. Haplogenotype, Del-/MU+ was associated with high susceptibility to SLE (OR = 1.87, p = 0.001) and SSc (OR = 2.29, p = 2.34×10-4). In contrast, the Haplogenotype Del+/MU- was associated with resistance to SLE (OR = 0.35, p = 6.24×10-5) and SSc (OR = 0.34, p = 0.001). This study demonstrates that the variations in KRT1 and the specific polymorphism of KRT1 in this Chinese Han population are associated with autoimmune diseases SLE and SSc. Typing KRT1 might be helpful to identify SLE and SSc patients.
Collapse
Affiliation(s)
- Weiguang Luo
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bin Zhou
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qizhi Luo
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Huilong Fang
- Department of pathogenic Biology and Immunology, Xiangnan University, Chenzhou, Hunan, China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhou Zou
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Cooperative Innovation Center of Engineering and new Products for Developmental Biology of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
111
|
Thomas DC. The phagocyte respiratory burst: Historical perspectives and recent advances. Immunol Lett 2017; 192:88-96. [PMID: 28864335 DOI: 10.1016/j.imlet.2017.08.016] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 11/18/2022]
Abstract
When exposed to certain stimuli, phagocytes (including neutrophils, macrophages and eosinophils) undergo marked changes in the way they handle oxygen. Firstly, their rate of oxygen uptake increases greatly. This is accompanied by (i) the production of large amounts of superoxide and hydrogen peroxide and (ii) the metabolism of large quantities of glucose through the hexose monophosphate shunt. We now know that the oxygen used is not for respiration but for the production of powerful microbiocidal agents downstream of the initial production of superoxide. Concomitantly, glucose is oxidised through the hexose monophosphate shunt to re-generate the NADPH that has been consumed through the reduction of molecular oxygen to generate superoxide. This phagocyte respiratory burst is generated by an NADPH oxidase multi-protein complex that has a catalytic core consisting of membrane-bound gp91phox (CYBB) and p22phox (CYBA) sub-units and cytosolic components p47phox (NCF1), p67phox (NCF2) and p40phox (NCF4). Finally, another cytosolic component, the small G-protein Rac (Rac2 in neutrophils and Rac1 in macrophages) is also required for full activation. The importance of the complex in host defence is underlined by chronic granulomatous disease, a severe life-limiting immunodeficiency caused by mutations in the genes encoding the individual subunits. In this review, I will discuss the experimental evidence that underlies our knowledge of the respiratory burst, outlining how elegant biochemical analysis, coupled with study of patients deficient in the various subunits has helped elucidate the function of this essential part of innate immunity. I will also discuss some exciting recent studies that shed new light on how the abundance of the various components is controlled. Finally, I will explore the emerging role of reactive oxygen species such as superoxide and hydrogen peroxide in the pathogenesis of major human diseases including auto-inflammatory diseases.
Collapse
Affiliation(s)
- David C Thomas
- Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Box 157, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, United Kingdom.
| |
Collapse
|
112
|
Nakayama Y, Kosek J, Capone L, Hur EM, Schafer PH, Ringheim GE. Aiolos Overexpression in Systemic Lupus Erythematosus B Cell Subtypes and BAFF-Induced Memory B Cell Differentiation Are Reduced by CC-220 Modulation of Cereblon Activity. THE JOURNAL OF IMMUNOLOGY 2017; 199:2388-2407. [PMID: 28848067 DOI: 10.4049/jimmunol.1601725] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 08/03/2017] [Indexed: 12/26/2022]
Abstract
BAFF is a B cell survival and maturation factor implicated in the pathogenesis of systemic lupus erythematosus (SLE). In this in vitro study, we describe that soluble BAFF in combination with IL-2 and IL-21 is a T cell contact-independent inducer of human B cell proliferation, plasmablast differentiation, and IgG secretion from circulating CD27+ memory and memory-like CD27-IgD- double-negative (DN) B cells, but not CD27-IgD+ naive B cells. In contrast, soluble CD40L in combination with IL-2 and IL-21 induces these activities in both memory and naive B cells. Blood from healthy donors and SLE patients have similar circulating levels of IL-2, whereas SLE patients exhibit elevated BAFF and DN B cells and reduced IL-21. B cell differentiation transcription factors in memory, DN, and naive B cells in SLE show elevated levels of Aiolos, whereas Ikaros levels are unchanged. Treatment with CC-220, a modulator of the cullin ring ligase 4-cereblon E3 ubiquitin ligase complex, reduces Aiolos and Ikaros protein levels and BAFF- and CD40L-induced proliferation, plasmablast differentiation, and IgG secretion. The observation that the soluble factors BAFF, IL-2, and IL-21 induce memory and DN B cell activation and differentiation has implications for extrafollicular plasmablast development within inflamed tissue. Inhibition of B cell plasmablast differentiation by reduction of Aiolos and Ikaros may have utility in the treatment of SLE, where elevated levels of BAFF and Aiolos may prime CD27+ memory and DN memory-like B cells to become Ab-producing plasmablasts in the presence of BAFF and proinflammatory cytokines.
Collapse
Affiliation(s)
- Yumi Nakayama
- Inflammation and Immunology Translational Development, Celgene Corporation, Summit, NJ 07901
| | - Jolanta Kosek
- Inflammation and Immunology Translational Development, Celgene Corporation, Summit, NJ 07901
| | - Lori Capone
- Inflammation and Immunology Translational Development, Celgene Corporation, Summit, NJ 07901
| | - Eun Mi Hur
- Inflammation and Immunology Translational Development, Celgene Corporation, Summit, NJ 07901
| | - Peter H Schafer
- Inflammation and Immunology Translational Development, Celgene Corporation, Summit, NJ 07901
| | - Garth E Ringheim
- Inflammation and Immunology Translational Development, Celgene Corporation, Summit, NJ 07901
| |
Collapse
|
113
|
Abstract
PURPOSE OF REVIEW To describe the recent studies on the genetics of systemic lupus erythematosus (SLE) and Sjögren's syndrome. RECENT FINDINGS We overview the most recent findings on the genetic susceptibility of the diseases and provide information on their genetic similarities and differences. SUMMARY SLE and Sjögren's syndrome are two closely related systemic autoimmune diseases that share multiple clinical and molecular aspects, including a significant number of susceptibility genes. Several genome-wide association studies were recently published in different populations that provide a better picture of their molecular mechanisms. It is becoming clear that their genetic architecture is quite well established, but more information is required on expression quantitative trait loci, epigenetic genome-wide analyses, gene × gene interactions and the role of rare variants.
Collapse
|
114
|
Lood C, Arve S, Ledbetter J, Elkon KB. TLR7/8 activation in neutrophils impairs immune complex phagocytosis through shedding of FcgRIIA. J Exp Med 2017; 214:2103-2119. [PMID: 28606989 PMCID: PMC5502427 DOI: 10.1084/jem.20161512] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/21/2016] [Accepted: 04/19/2017] [Indexed: 12/24/2022] Open
Abstract
Lood et al. find that neutrophil TLR7/8 activation shifts neutrophils from phagocytosis of immune complexes to NETosis. Reduced phagocytosis of immune complexes is associated with partial proteolytic cleavage of FcgRIIA. Cleaved FcgRIIA is found in SLE neutrophils ex vivo. Neutrophils play a crucial role in host defense. However, neutrophil activation is also linked to autoimmune diseases such as systemic lupus erythematosus (SLE), where nucleic acid–containing immune complexes (IC) drive inflammation. The role of Toll-like receptor (TLR) signaling in processing of SLE ICs and downstream inflammatory neutrophil effector functions is not known. We observed that TLR7/8 activation leads to a furin-dependent proteolytic cleavage of the N-terminal part of FcgRIIA, shifting neutrophils away from phagocytosis of ICs toward the programmed form of necrosis, NETosis. TLR7/8-activated neutrophils promoted cleavage of FcgRIIA on plasmacytoid dendritic cells and monocytes, resulting in impaired overall clearance of ICs and increased complement C5a generation. Importantly, ex vivo derived activated neutrophils from SLE patients demonstrated a similar cleavage of FcgRIIA that was correlated with markers of disease activity, as well as complement activation. Therapeutic approaches aimed at blocking TLR7/8 activation would be predicted to increase phagocytosis of circulating ICs, while disarming their inflammatory potential.
Collapse
Affiliation(s)
- Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Sabine Arve
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Jeffrey Ledbetter
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Keith B Elkon
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA 98109
| |
Collapse
|
115
|
Abstract
RIG-I-like receptors (RLRs) are cytosolic innate immune sensors that detect pathogenic RNA and induce a systemic antiviral response. During the last decade, many studies focused on their molecular characterization and the identification of RNA agonists. Therefore, it became more and more clear that RLR activation needs to be carefully regulated, because constitutive signaling or detection of endogenous RNA through loss of specificity is detrimental. Here, we review the current understanding of RLR activation and selectivity. We specifically focus upon recent findings on the function of the helicase domain in discriminating between different RNAs, and whose malfunctioning causes serious autoimmune diseases.
Collapse
Affiliation(s)
- Charlotte Lässig
- From the Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, 81377 Munich and
| | - Karl-Peter Hopfner
- From the Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, 81377 Munich and
- the Center for Integrated Protein Sciences, 81377 Munich, Germany
| |
Collapse
|
116
|
The A946T variant of the RNA sensor IFIH1 mediates an interferon program that limits viral infection but increases the risk for autoimmunity. Nat Immunol 2017; 18:744-752. [PMID: 28553952 PMCID: PMC5697900 DOI: 10.1038/ni.3766] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022]
Abstract
The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.
Collapse
|
117
|
Chen D, Long M, Xiao B, Xiong Y, Chen H, Chen Y, Kuang Z, Li M, Wu Y, Rock DL, Gong D, Wang Y, He H, Liu F, Luo S, Hao W. Transcriptomic profiles of human foreskin fibroblast cells in response to orf virus. Oncotarget 2017; 8:58668-58685. [PMID: 28938587 PMCID: PMC5601683 DOI: 10.18632/oncotarget.17417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/20/2017] [Indexed: 11/25/2022] Open
Abstract
Orf virus has been utilized as a safe and efficient viral vector against not only diverse infectious diseases, but also against tumors. However, the nature of the genes triggered by the vector in human cells is poorly characterized. Using RNA sequencing technology, we compared specific changes in the transcriptomic profiles in human foreskin fibroblast cells following infection by the orf virus. The results indicated that orf virus upregulates or downregulates expression of a variety of genes, including genes involved in antiviral immune response, apoptosis, cell cycle and a series of signaling pathways, such as the IFN and p53-signaling pathways. The orf virus stimulates or inhibits immune gene expression such as chemokines, chemokine receptors, cytokines, cytokine receptors, and molecules involved in antigen uptake and processing after infection. Expression of pro-apoptotic genes increased at 8 hours post-infection. The p53 signaling pathway was activated to induce apoptosis at the same time. However, the cell cycle program was promoted after infection, which may be due to the immunomodulatory genes of the orf virus. This presents the first description of transcription profile changes in human foreskin fibroblast cells after orf virus infection and provides an in-depth analysis of the interaction between the host and orf virus. These data offer new insights into the understanding of the mechanisms of infection by orf virus and identify potential targets for future studies.
Collapse
Affiliation(s)
- Daxiang Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Mingjian Long
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Bin Xiao
- Department of Laboratory Medicine, Guangzhou General Hospital of Guangzhou Military Command of PLA, Guangzhou, 510010, P.R. China
| | - Yufeng Xiong
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Huiqin Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Yu Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Zhenzhan Kuang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Ming Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Yingsong Wu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Daniel L Rock
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Champaign-Urbana, Urbana, IL 61802 USA
| | - Daoyuan Gong
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Yong Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Haijian He
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Fang Liu
- Department of Pathophysiology, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, P.R. China
| |
Collapse
|
118
|
Hypermethylation of Interferon Regulatory Factor 8 (IRF8) Confers Risk to Vogt-Koyanagi-Harada Disease. Sci Rep 2017; 7:1007. [PMID: 28432342 PMCID: PMC5430771 DOI: 10.1038/s41598-017-01249-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/27/2017] [Indexed: 12/15/2022] Open
Abstract
Aberrant methylation change of IRF8 confers risk to various tumors, and abnormal expression of IRF8 is involved in many autoimmune diseases, including ocular Behcet’s disease. However, whether the methylation change of IRF8 is associated with Vogt-Koyanagi-Harada (VKH) disease remains unknown. In the present study, we found a decreased IRF8 mRNA expression in association with a higher methylation level in monocyte-derived dendritic cells (DCs) from active VKH patients compared with the normal and inactive subjects. DCs incubated with cyclosporin a (CsA) or dexamethasone (DEX) showed a lower methylation and higher mRNA expression of IRF8 in active VKH patients. A demethylation reagent, 5-Aza-2′-deoxycytidine (DAC) showed a notable demethylation effect as evidenced by increasing the mRNA expression and reducing the methylation level of IRF8. It also suppressed the Th1 and Th17 responses through down-regulating the expression of co-stimulatory molecules (CD86, CD80, CD40), and reducing the production of pro-inflammatory cytokines (IL-6, IL-1β, IL-23, IL-12) produced by DCs. These findings shows that hypermethylation of IRF8 in DCs confers risk to VKH disease. Demethylation of IRF8 may offer a novel therapeutic strategy protect against VKH disease.
Collapse
|
119
|
Matyskiela ME, Zhang W, Man HW, Muller G, Khambatta G, Baculi F, Hickman M, LeBrun L, Pagarigan B, Carmel G, Lu CC, Lu G, Riley M, Satoh Y, Schafer P, Daniel TO, Carmichael J, Cathers BE, Chamberlain PP. A Cereblon Modulator (CC-220) with Improved Degradation of Ikaros and Aiolos. J Med Chem 2017; 61:535-542. [DOI: 10.1021/acs.jmedchem.6b01921] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mary E. Matyskiela
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Weihong Zhang
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Hon-Wah Man
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - George Muller
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Godrej Khambatta
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Frans Baculi
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Matthew Hickman
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Laurie LeBrun
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Barbra Pagarigan
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Gilles Carmel
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Chin-Chun Lu
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Gang Lu
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Mariko Riley
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Yoshitaka Satoh
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Peter Schafer
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Thomas O. Daniel
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - James Carmichael
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Brian E. Cathers
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| | - Philip P. Chamberlain
- 1Celgene Corporation, 10300 Campus Point Drive, Suite
100, San Diego, California 92121, United States
| |
Collapse
|
120
|
Qiu Y, Zhu Y, Yu H, Yi S, Su W, Cao Q, Yuan G, Kijlstra A, Yang P. Ocular Behcet's disease is associated with aberrant methylation of interferon regulatory factor 8 (IRF8) in monocyte-derived dendritic cells. Oncotarget 2017; 8:51277-51287. [PMID: 28881647 PMCID: PMC5584248 DOI: 10.18632/oncotarget.17235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/28/2017] [Indexed: 01/23/2023] Open
Abstract
Aberrant methylation of interferon regulatory factor 8 (IRF8) has been noted in various tumors. IRF8 has also been reported to be involved in many autoimmune diseases, including Behcet's disease (BD). However, the methylation status of IRF8 in BD has not been reported. To address this issue, we investigated whether the degree of methylation of IRF8 in dendritic cells (DCs) plays a role in the development of BD. We found a lower mRNA expression and a higher methylation level of IRF8 in active ocular BD patients as compared to normal subjects and inactive patients. Treatment with a demethylation agent, 5-Aza-2'-deoxycytidine (DAC) resulted in an increase of mRNA expression and a reduction of the IRF8 methylation level. It also down-regulated the expression of the co-stimulatory molecules CD86, CD80, CD40, and reduced the production of IL-6, IL-1β, IL-23 and IL-12. An inhibition of Th1/Th17 responses was observed as evidenced by a decreased production of IFN-γ, IL-17, and a reduction of IFN-γ/IL-17- producing CD4+ T cells following treatment with DAC. This study shows that active ocular BD patients have an aberrant IRF8 methylation status. These findings suggest that epigenetic control of IRF8 expression may offer a future target in the treatment of ocular BD.
Collapse
Affiliation(s)
- Yiguo Qiu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Yunyun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Hongsong Yu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Shenglan Yi
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Wencheng Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qingfeng Cao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Gangxiang Yuan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| |
Collapse
|
121
|
Abstract
Self-reactive B cells are tolerized at various stages of B-cell development and differentiation, including the immature B-cell stage (central tolerance) and the germinal center (GC) B-cell stage, and B-cell tolerance involves various mechanisms such as deletion, anergy, and receptor editing. Self-reactive B cells generated by random immunoglobulin variable gene rearrangements are tolerized by central tolerance and anergy in the periphery, and these processes involve apoptosis regulated by Bim, a pro-apoptotic member of the Bcl-2 family, and regulation of B-cell signaling by various phosphatases, including SHIP-1 and SHP-1. Self-reactive B cells generated by somatic mutations during GC reaction are also eliminated. Fas is not directly involved in this process but prevents persistence of GC reaction that allows generation of less stringently regulated B cells, including self-reactive B cells. Defects in self-tolerance preferentially cause lupus-like disease with production of anti-nuclear antibodies, probably due to the presence of a large potential B-cell repertoire reactive to nucleic acids and the presence of nucleic acid-induced activation mechanisms in various immune cells, including B cells and dendritic cells. A feed-forward loop composed of anti-nuclear antibodies produced by B cells and type 1 interferons secreted from nucleic acid-activated dendritic cells plays a crucial role in the development of systemic lupus erythematosus.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| |
Collapse
|
122
|
Matta B, Song S, Li D, Barnes BJ. Interferon regulatory factor signaling in autoimmune disease. Cytokine 2017; 98:15-26. [PMID: 28283223 DOI: 10.1016/j.cyto.2017.02.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
Interferon regulatory factors (IRFs) play critical roles in pathogen-induced innate immune responses and the subsequent induction of adaptive immune response. Dysregulation of IRF signaling is therefore thought to contribute to autoimmune disease pathogenesis. Indeed, numerous murine in vivo studies have documented protection from or enhanced susceptibility to particular autoimmune diseases in Irf-deficient mice. What has been lacking, however, is replication of these in vivo observations in primary immune cells from patients with autoimmune disease. These types of studies are essential as the majority of in vivo data support a protective role for IRFs in Irf-deficient mice, yet IRFs are often found to be overexpressed in patient immune cells. A significant body of work is beginning to emerge from both of these areas of study - mouse and human.
Collapse
Affiliation(s)
- Bharati Matta
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, NY 11030, United States
| | - Su Song
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, NY 11030, United States
| | - Dan Li
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, NY 11030, United States
| | - Betsy J Barnes
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, Manhasset, NY 11030, United States.
| |
Collapse
|
123
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with complex genetic underpinnings. This review attempts to assemble the myriad of genomic findings to build a clearer picture of the pathobiology of SLE to serve as a guide for therapeutics. Over 100 genes are now known for SLE, and several more penetrant ones have led to the emergence of more defined lupus phenotypes. Also discussed here are the targeted therapies that have come up on the horizon and the specific biologic mechanisms of more traditional therapies which have only recently been explored. The diagnostic toolbox has been enhanced by the addition of new antibodies, gene expression signatures, and mutation panels. This provides an opportunity to piece together the lupus puzzle and even revisit the clinical classification of SLE.
Collapse
|
124
|
Hirahara K, Schwartz D, Gadina M, Kanno Y, O'Shea JJ. Targeting cytokine signaling in autoimmunity: back to the future and beyond. Curr Opin Immunol 2016; 43:89-97. [PMID: 27821272 DOI: 10.1016/j.coi.2016.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 12/16/2022]
Abstract
Cytokines represent structurally diverse soluble factors with critical roles in normal immune function and the pathogenesis of autoimmunity. The emergence of many successful biological therapies targeting cytokines and cytokine receptors exemplifies the importance of cytokines in driving human autoimmune disease; unsurprisingly, there is no paucity of reviews on this subject. Nonetheless, many patients with autoimmune disease do not respond to biologicals, and cure remains an unmet goal. Thus, targeting the intracellular pathways employed by cytokines provides new therapeutic opportunities. A subset of cytokines utilizes the Janus kinase-signal transducer of activators of transcription (JAK-STAT) pathway as a mode of signal transduction. First generation JAK inhibitors (jakinibs) are used to treat rheumatologic disease, and second-generation jakinibs are being developed. Simultaneously, rapid advances are being made in our understanding of the genomic and epigenomic impact of cytokines. In this review, we will briefly review the role of JAK-STAT-dependent cytokines in immune-mediated disease, the current status of Jakinibs, and future possibilities for therapeutic intervention using genomic insights.
Collapse
Affiliation(s)
- Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku, Chiba 260-8670, Japan
| | - Daniella Schwartz
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Massimo Gadina
- Translational Immunology Section, Office of Science Technology, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuka Kanno
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
125
|
Bronson PG, Chang D, Bhangale T, Seldin MF, Ortmann W, Ferreira RC, Urcelay E, Pereira LF, Martin J, Plebani A, Lougaris V, Friman V, Freiberger T, Litzman J, Thon V, Pan-Hammarström Q, Hammarström L, Graham RR, Behrens TW. Common variants at PVT1, ATG13-AMBRA1, AHI1 and CLEC16A are associated with selective IgA deficiency. Nat Genet 2016; 48:1425-1429. [PMID: 27723758 PMCID: PMC5086090 DOI: 10.1038/ng.3675] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/24/2016] [Indexed: 12/18/2022]
Abstract
Selective immunoglobulin A deficiency (IgAD) is the most common primary immunodeficiency in Europeans. Our genome-wide association study (GWAS) meta-analysis of 1,635 patients with IgAD and 4,852 controls identified four new significant (P < 5 × 10-8) loci and association with a rare IFIH1 variant (p.Ile923Val). Peak new variants (PVT1, P = 4.3 × 10-11; ATG13-AMBRA1, P = 6.7 × 10-10; AHI1, P = 8.4 × 10-10; CLEC16A, P = 1.4 × 10-9) overlapped with autoimmune markers (3/4) and correlated with 21 putative regulatory variants, including expression quantitative trait loci (eQTLs) for AHI1 and DEXI and DNase hypersensitivity sites in FOXP3+ regulatory T cells. Pathway analysis of the meta-analysis results showed striking association with the KEGG pathway for IgA production (pathway P < 0.0001), with 22 of the 30 annotated pathway genes containing at least one variant with P ≤ 0.05 in the IgAD meta-analysis. These data suggest that a complex network of genetic effects, including genes known to influence the biology of IgA production, contributes to IgAD.
Collapse
Affiliation(s)
- Paola G. Bronson
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Diana Chang
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Tushar Bhangale
- Department of Bioinformatics and Computational Biology,
Genentech, Inc., South San Francisco, CA, USA
| | - Michael F. Seldin
- Department of Biochemistry, School of Medicine, University
of California, Davis, CA, USA
| | - Ward Ortmann
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Ricardo C. Ferreira
- Juvenile Diabetes Research Foundation/Wellcome Trust
Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research,
Cambridge, UK
| | - Elena Urcelay
- Department of Immunology, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | | | - Javier Martin
- Instituto de Parasitología y Biomedicina
López-Neyra, CSIC, Granada, Spain
| | - Alessandro Plebani
- Pediatrics Clinic, Department of Clinical and Experimental
Sciences, University of Brescia, Spedali Civili di Brescia, Italy
- Institute for Molecular Medicine, A. Nocivelli, Department
of Clinical and Experimental Sciences, University of Brescia, Spedali Civili di
Brescia, Italy
| | - Vassilios Lougaris
- Pediatrics Clinic, Department of Clinical and Experimental
Sciences, University of Brescia, Spedali Civili di Brescia, Italy
- Institute for Molecular Medicine, A. Nocivelli, Department
of Clinical and Experimental Sciences, University of Brescia, Spedali Civili di
Brescia, Italy
| | - Vanda Friman
- Department of Infectious Diseases, University of
Gothenburg, Gothenburg, Sweden
| | - Tomáš Freiberger
- Molecular Genetics Laboratory, Centre for Cardiovascular
Surgery and Transplantation, Brno, Czech Republic
- Central European Institute of Technology, Masaryk
University, Brno, Czech Republic
| | - Jiri Litzman
- Department of Clinical Immunology and Allergy, Faculty of
Medicine, Masaryk University, St. Anne’s Univ. Hospital, Brno, Czech
Republic
| | - Vojtech Thon
- Department of Clinical Immunology and Allergy, Faculty of
Medicine, Masaryk University, St. Anne’s Univ. Hospital, Brno, Czech
Republic
- Research Centre for Toxic Compounds in the Environment,
Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Qiang Pan-Hammarström
- Division of Clinical Immunology & Transfusion
Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lennart Hammarström
- Division of Clinical Immunology & Transfusion
Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert R. Graham
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Timothy W. Behrens
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| |
Collapse
|
126
|
USP15 regulates type I interferon response and is required for pathogenesis of neuroinflammation. Nat Immunol 2016; 18:54-63. [DOI: 10.1038/ni.3581] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/25/2016] [Indexed: 12/21/2022]
|
127
|
Abstract
The ability to distinguish between self and nonself is the fundamental basis of the immune system in all organisms. The conceptual distinction between self and nonself, however, breaks down when it comes to endogenous retroviruses and other retroelements. While some retroelements retain the virus-like features including the capacity to replicate and reinvade the host genome, most have become inactive through mutations or host epigenetic silencing. And yet, accumulating evidence suggests that endogenous retroelements, both active and inactive, play important roles not only in pathogenesis of immune disorders, but also in proper functioning of the immune system. This review discusses the recent development in our understanding of the interaction between retroelements and the host innate immune system. In particular, it focuses on the impact of retroelement transcripts on the viral RNA sensors such as Toll-like receptors, RIG-I-like receptors, protein kinase R, and the inflammasomes.
Collapse
Affiliation(s)
- X Mu
- Harvard Medical School, Boston, MA, United States; Boston Children's Hospital, Boston, MA, United States
| | - S Ahmad
- Harvard Medical School, Boston, MA, United States; Boston Children's Hospital, Boston, MA, United States
| | - S Hur
- Harvard Medical School, Boston, MA, United States; Boston Children's Hospital, Boston, MA, United States.
| |
Collapse
|
128
|
Zhang YM, Zhou XJ, Cheng FJ, Qi YY, Hou P, Zhao MH, Zhang H. Association of the IKZF1 5ʹ UTR variant rs1456896 with lupus nephritis in a northern Han Chinese population. Scand J Rheumatol 2016; 46:210-214. [PMID: 27684961 DOI: 10.1080/03009742.2016.1194458] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Y-M Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - X-J Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - F-J Cheng
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Y-Y Qi
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - P Hou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - M-H Zhao
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - H Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, and Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| |
Collapse
|
129
|
Teruel M, Alarcón-Riquelme ME. The genetic basis of systemic lupus erythematosus: What are the risk factors and what have we learned. J Autoimmun 2016; 74:161-175. [PMID: 27522116 DOI: 10.1016/j.jaut.2016.08.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 08/02/2016] [Indexed: 12/19/2022]
Abstract
The genome-wide association study is a free-hypothesis approach based on screening of thousands or even millions of genetic variants distributed throughout the whole human genome in relation to a phenotype. The relevant role of the genome-wide association studies in the last decade is undisputed because it has permitted to elucidate multiple risk genetic factors associated with the susceptibility to several human complex diseases. Regarding systemic lupus erythematosus (SLE) this approach has allowed to identify more than 60 risk loci for SLE susceptibility across populations to date, increasing our understanding on the pathogenesis of this disease. We present the latest findings in the genetic of SLE across populations using genome-wide approaches. These studies revealed that most of the genetic risk is shared across borders and ethnicities. Finally, we focus on describing the most important risk loci for SLE attempting to cover the genetic findings in relation to functional polymorphisms, such as missense single nucleotide polymorphisms (SNPs) or regulatory variants involved in the development of the disease. The functional studies try to identify the causality of some GWAS-associated variants, many of which fall in non-coding regions of the genome, suggesting a regulatory role. Many loci show an environmental interaction, another aspect revealed by the studies of epigenetic modifications and those associated with genetic variants. Finally, new-generation sequencing technologies can open other paths in the research on SLE genetics, the role of rare variants and the detailed identification of causal regulatory variation. The clinical relevance of the genetic factors will be shown when we are able to use them or in combination with other molecular measurements to re-classify a heterogeneous disease such as SLE.
Collapse
Affiliation(s)
- Maria Teruel
- Center for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Government, PTS, Granada, 18016, Spain.
| | - Marta E Alarcón-Riquelme
- Center for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Government, PTS, Granada, 18016, Spain; Institute of Environmental Medicine, Karolinska Institute, Stockholm, 171 67, Sweden.
| |
Collapse
|
130
|
López-Isac E, Campillo-Davo D, Bossini-Castillo L, Guerra SG, Assassi S, Simeón CP, Carreira P, Ortego-Centeno N, García de la Peña P, Beretta L, Santaniello A, Bellocchi C, Lunardi C, Moroncini G, Gabrielli A, Riemekasten G, Witte T, Hunzelmann N, Kreuter A, Distler JH, Voskuyl AE, de Vries-Bouwstra J, Herrick A, Worthington J, Denton CP, Fonseca C, Radstake TR, Mayes MD, Martín J. Influence of TYK2 in systemic sclerosis susceptibility: a new locus in the IL-12 pathway. Ann Rheum Dis 2016; 75:1521-6. [PMID: 26338038 PMCID: PMC7228811 DOI: 10.1136/annrheumdis-2015-208154] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/13/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVES TYK2 is a common genetic risk factor for several autoimmune diseases. This gene encodes a protein kinase involved in interleukin 12 (IL-12) pathway, which is a well-known player in the pathogenesis of systemic sclerosis (SSc). Therefore, we aimed to assess the possible role of this locus in SSc. METHODS This study comprised a total of 7103 patients with SSc and 12 220 healthy controls of European ancestry from Spain, USA, Germany, the Netherlands, Italy and the UK. Four TYK2 single-nucleotide polymorphisms (V362F (rs2304256), P1104A (rs34536443), I684S (rs12720356) and A928V (rs35018800)) were selected for follow-up based on the results of an Immunochip screening phase of the locus. Association and dependence analyses were performed by the means of logistic regression and conditional logistic regression. Meta-analyses were performed using the inverse variance method. RESULTS Genome-wide significance level was reached for TYK2 V362F common variant in our pooled analysis (p=3.08×10(-13), OR=0.83), while the association of P1104A, A928V and I684S rare and low-frequency missense variants remained significant with nominal signals (p=2.28×10(-3), OR=0.80; p=1.27×10(-3), OR=0.59; p=2.63×10(-5), OR=0.83, respectively). Interestingly, dependence and allelic combination analyses showed that the strong association observed for V362F with SSc, corresponded to a synthetic association dependent on the effect of the three previously mentioned TYK2 missense variants. CONCLUSIONS We report for the first time the association of TYK2 with SSc and reinforce the relevance of the IL-12 pathway in SSc pathophysiology.
Collapse
Affiliation(s)
- Elena López-Isac
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| | - Diana Campillo-Davo
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| | - Lara Bossini-Castillo
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| | - Sandra G Guerra
- Centre for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Shervin Assassi
- Division of Rheumatology and Clinical Immunogenetics, The University of Texas Health Science Center-Houston, Houston, USA
| | | | - Patricia Carreira
- Department of Rheumatology, 12 de Octubre University Hospital, Madrid, Spain
| | | | | | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Alessandro Santaniello
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Chiara Bellocchi
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Claudio Lunardi
- Department of Medicine, Università degli Studi di Verona, Verona, Italy
| | - Gianluca Moroncini
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche and Ospedali Riuniti, Ancona, Italy
| | - Armando Gabrielli
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche and Ospedali Riuniti, Ancona, Italy
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Torsten Witte
- Department of Clinical Immunology, Hannover Medical School, Hannover, Germany
| | | | - Alexander Kreuter
- Department of Dermatology, Venereology, and Allergology, HELIOS St Elisabeth Hospital Oberhausen, Germany
| | - Jörg Hw Distler
- Department of Internal Medicine, Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexandre E Voskuyl
- Department of Rheumatology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Ariane Herrick
- Centre for Musculoskeletal Research and NIHR Manchester Musculoskeletal Biomedical Research Unit, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jane Worthington
- Centre for Musculoskeletal Research and NIHR Manchester Musculoskeletal Biomedical Research Unit, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Christopher P Denton
- Centre for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Carmen Fonseca
- Centre for Rheumatology, Royal Free and University College Medical School, London, UK
| | - Timothy Rdj Radstake
- Department of Rheumatology & Clinical Immunology, Laboratory of Translational Immunology, department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maureen D Mayes
- Division of Rheumatology and Clinical Immunogenetics, The University of Texas Health Science Center-Houston, Houston, USA
| | - Javier Martín
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| |
Collapse
|
131
|
Lee YH, Bae SC. Association between TYK2 polymorphisms and susceptibility to autoimmune rheumatic diseases: a meta-analysis. Lupus 2016; 25:1307-14. [DOI: 10.1177/0961203316638933] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/22/2016] [Indexed: 01/14/2023]
Abstract
Objective This study aimed to explore whether TYK2 polymorphisms are associated with susceptibility to autoimmune rheumatic diseases. Methods We conducted a meta-analysis on the association between TYK2 polymorphisms and autoimmune rheumatic diseases. Results Twelve studies with a total of 16,335 patients and 30,065 controls were included in the meta-analysis. Meta-analysis revealed an association between rheumatic diseases and the 2 allele of the TYK2 rs2304256 (OR = 0.885, 95% CI = 0.802–0.978, p = 0.016). Furthermore, stratification by ethnicity identified a significant association between this polymorphism and rheumatic diseases in Caucasians (OR = 0.822, 95% CI = 0.706–0.889, p = 9.5 × 10−7), but not in Asians (OR = 1.127, 95% CI = 0.835–1.522, p = 0.434). Meta-analysis by rheumatic disease type revealed a significant association between the 2 allele of the TYK2 rs2304256 and SLE in Caucasians (OR = 0.737, 95% CI = 0.673–0.808, p < 1.0 × 10−8) but not in Asians (OR = 1.211, 95% CI = 0.813–1.804, p = 0.347). Meta-analysis revealed that the rs12720356 polymorphism was associated with susceptibility to rheumatic diseases in Caucasians (OR = 0.812, 95% CI = 0.661–0.997, p = 0.046) but not in Asians. Interestingly, the rs280519 polymorphism was significantly associated with susceptibility to SLE both in Caucasians and Asians. However, no associations were found between the rs12720270, rs280500, rs280523 and rs8108236 polymorphisms and susceptibility to rheumatic diseases. Conclusions This meta-analysis demonstrates that the TYK2 rs2304256 and rs12720356 polymorphisms are associated with susceptibility to rheumatic diseases, rs2304256 polymorphism is associated with SLE in Caucasians, and rs280519 polymorphism is associated with SLE in Caucasians and Asians.
Collapse
Affiliation(s)
- Y H Lee
- Division of Rheumatology, Department of Internal Medicine, Korea University College of Medicine, Korea
| | - S-C Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| |
Collapse
|
132
|
Several Critical Cell Types, Tissues, and Pathways Are Implicated in Genome-Wide Association Studies for Systemic Lupus Erythematosus. G3-GENES GENOMES GENETICS 2016; 6:1503-11. [PMID: 27172182 PMCID: PMC4889647 DOI: 10.1534/g3.116.027326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We aimed to elucidate the cell types, tissues, and pathways influenced by common variants in systemic lupus erythematosus (SLE). We applied a nonparameter enrichment statistical approach, termed SNPsea, in 181 single nucleotide polymorphisms (SNPs) that have been identified to be associated with the risk of SLE through genome-wide association studies (GWAS) in Eastern Asian and Caucasian populations, to manipulate the critical cell types, tissues, and pathways. In the two most significant cells’ findings (B lymphocytes and CD14+ monocytes), we subjected the GWAS association evidence in the Han Chinese population to an enrichment test of expression quantitative trait locus (QTL) sites and DNase I hypersensitivity, respectively. In both Eastern Asian and Caucasian populations, we observed that the expression level of SLE GWAS implicated genes was significantly elevated in xeroderma pigentosum B cells (P ≤ 1.00 × 10−6), CD14+ monocytes (P ≤ 2.74 × 10−4) and CD19+ B cells (P ≤ 2.00 × 10−6), and plasmacytoid dendritic cells (pDCs) (P ≤ 9.00 × 10−6). We revealed that the SLE GWAS-associated variants were more likely to reside in expression QTL in B lymphocytes (q1/q0 = 2.15, P = 1.23 × 10−44) and DNase I hypersensitivity sites (DHSs) in CD14+ monocytes (q1/q0 = 1.41, P = 0.08). We observed the common variants affected the risk of SLE mostly through by regulating multiple immune system processes and immune response signaling. This study sheds light on several immune cells and responses, as well as the regulatory effect of common variants in the pathogenesis of SLE.
Collapse
|
133
|
Buers I, Nitschke Y, Rutsch F. Novel interferonopathies associated with mutations in RIG-I like receptors. Cytokine Growth Factor Rev 2016; 29:101-7. [PMID: 26993858 DOI: 10.1016/j.cytogfr.2016.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Type I interferonopathies are a relatively new class of inherited autoimmune disorders associated with an inborn elevated interferon response. Activation of cytosolic receptors which recognize viral double stranded RNA including the RIG-I (retinoic acid-inducible gene I) like receptors RIG-I and MDA5 (melanoma differentiation-associated gene 5) has been shown to induce the transcription of type I interferon genes. Within recent years, with the help of next generation sequencing techniques in syndromic families, mutations in the genes encoding for RIG-I and MDA5 have been identified to cause rare diseases including Aicardi-Goutières syndrome, Systemic Lupus Erythematosus in certain individuals as well as classic and atypical Singleton-Merten syndrome. Patients carrying mono-allelic mutations in MDA5 and RIG-I show constitutive activation of the RIG-I receptors and downstream signalling associated with increased type I interferon production. Although differing in the degree of phenotypic expression and severity, the phenotype of these "novel" diseases shows a considerable overlap reflecting their common pathogenetic pathway.
Collapse
Affiliation(s)
- Insa Buers
- Department of General Pediatrics, Muenster University Children's Hospital, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany.
| | - Yvonne Nitschke
- Department of General Pediatrics, Muenster University Children's Hospital, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany.
| | - Frank Rutsch
- Department of General Pediatrics, Muenster University Children's Hospital, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany.
| |
Collapse
|
134
|
Budde P, Zucht HD, Vordenbäumen S, Goehler H, Fischer-Betz R, Gamer M, Marquart K, Rengers P, Richter J, Lueking A, Schulz-Knappe P, Schneider M. Multiparametric detection of autoantibodies in systemic lupus erythematosus. Lupus 2016; 25:812-22. [DOI: 10.1177/0961203316641770] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous disease with respect to disease manifestations, disease progression and treatment response. Therefore, strategies to identify biomarkers that help distinguishing SLE subgroups are a major focus of biomarker research. We reasoned that a multiparametric autoantibody profiling approach combined with data mining tools could be applied to identify SLE patient clusters. We used a bead-based array containing 86 antigens including diverse nuclear and immune defense pathway proteins. Sixty-four autoantibodies were significantly ( p < 0.05) increased in SLE ( n = 69) compared to healthy controls (HC, n = 59). Using binary cut-off thresholds (95% quantile of HC), hierarchical clustering of SLE patients yields five clusters, which differ qualitatively and in their total number of autoantibodies. In two patient clusters the overall accumulated autoantibody reactivity of all antigens tested was 31% and 48%, respectively. We observed a positive association between the autoantibody signature present in these two patient clusters and the clinical manifestation of glomerulonephritis (GLMN). In addition, groups of autoantibodies directed against distinct intracellular compartments and/or biological motifs characterize the different SLE subgroups. Our findings highlight the relevant potential of multiparametric autoantibody detection and may contribute to a deeper understanding of the clinical and serological diversity of SLE.
Collapse
Affiliation(s)
| | | | - S Vordenbäumen
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| | | | - R Fischer-Betz
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| | | | | | | | - J Richter
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| | | | | | - M Schneider
- Heinrich-Heine-University, Medical Faculty, Policlinic of Rheumatology, Düsseldorf, Germany
| |
Collapse
|
135
|
Koff JL, Flowers CR. B cells gone rogue: the intersection of diffuse large B cell lymphoma and autoimmune disease. Expert Rev Hematol 2016; 9:553-61. [PMID: 27098121 DOI: 10.1080/17474086.2016.1180972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Diffuse large B cell lymphoma (DLBCL) is characterized by genetic, genomic and clinical heterogeneity. Autoimmune diseases (AIDs) have recently been shown to represent significant risk factors for development of DLBCL. AREAS COVERED Studies that examined the relationships between AIDs and lymphoma in terms of pathogenesis, genetic lesions, and treatment were identified in the MEDLINE database using combinations of medical subject heading (MeSH) terms. Co-authors independently performed study selection for inclusion based on appropriateness of the study question and nature of the study design and sample size. Expert commentary: Identification of AID as a substantial risk factor for DLBCL raises new questions regarding how autoimmunity influences lymphomagenesis and disease behavior. It will be important to identify whether DLBCL cases arising in the setting of AID harbor inferior prognoses, and, if so, whether they also exhibit certain molecular abnormalities that may be targeted to overcome such a gap in clinical outcomes.
Collapse
Affiliation(s)
- Jean L Koff
- a Department of Hematology and Medical Oncology, Winship Cancer Institute , Emory University , Atlanta , GA , USA
| | - Christopher R Flowers
- a Department of Hematology and Medical Oncology, Winship Cancer Institute , Emory University , Atlanta , GA , USA
| |
Collapse
|
136
|
A survey of endogenous retrovirus (ERV) sequences in the vicinity of multiple sclerosis (MS)-associated single nucleotide polymorphisms (SNPs). Mol Biol Rep 2016; 43:827-36. [PMID: 27169423 DOI: 10.1007/s11033-016-4004-0] [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] [Received: 02/02/2016] [Accepted: 05/04/2016] [Indexed: 12/18/2022]
Abstract
Although multiple sclerosis (MS) is one of the most common central nervous system diseases in young adults, little is known about its etiology. Several human endogenous retroviruses (ERVs) are considered to play a role in MS. We are interested in which ERVs can be identified in the vicinity of MS associated genetic marker to find potential initiators of MS. We analysed the chromosomal regions surrounding 58 single nucleotide polymorphisms (SNPs) that are associated with MS identified in one of the last major genome wide association studies. We scanned these regions for putative endogenous retrovirus sequences with large open reading frames (ORFs). We observed that more retrovirus-related putative ORFs exist in the relatively close vicinity of SNP marker indices in multiple sclerosis compared to control SNPs. We found very high homologies to HERV-K, HCML-ARV, XMRV, Galidia ERV, HERV-H/env62 and XMRV-like mouse endogenous retrovirus mERV-XL. The associated genes (CYP27B1, CD6, CD58, MPV17L2, IL12RB1, CXCR5, PTGER4, TAGAP, TYK2, ICAM3, CD86, GALC, GPR65 as well as the HLA DRB1*1501) are mainly involved in the immune system, but also in vitamin D regulation. The most frequently detected ERV sequences are related to the multiple sclerosis-associated retrovirus, the human immunodeficiency virus 1, HERV-K, and the Simian foamy virus. Our data shows that there is a relation between MS associated SNPs and the number of retroviral elements compared to control. Our data identifies new ERV sequences that have not been associated with MS, so far.
Collapse
|
137
|
Genetic association study of systemic lupus erythematosus and disease subphenotypes in European populations. Clin Rheumatol 2016; 35:1161-8. [DOI: 10.1007/s10067-016-3235-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 03/11/2016] [Accepted: 03/12/2016] [Indexed: 01/10/2023]
|
138
|
Langlais D, Barreiro LB, Gros P. The macrophage IRF8/IRF1 regulome is required for protection against infections and is associated with chronic inflammation. J Exp Med 2016; 213:585-603. [PMID: 27001747 PMCID: PMC4821649 DOI: 10.1084/jem.20151764] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/10/2016] [Indexed: 12/26/2022] Open
Abstract
IRF8 and IRF1 are transcriptional regulators that play critical roles in the development and function of myeloid cells, including activation of macrophages by proinflammatory signals such as interferon-γ (IFN-γ). Loss of IRF8 or IRF1 function causes severe susceptibility to infections in mice and in humans. We used chromatin immunoprecipitation sequencing and RNA sequencing in wild type and inIRF8andIRF1mutant primary macrophages to systematically catalog all of the genes bound by (cistromes) and transcriptionally activated by (regulomes) IRF8, IRF1, PU.1, and STAT1, including modulation of epigenetic histone marks. Of the seven binding combinations identified, two (cluster 1 [IRF8/IRF1/STAT1/PU.1] and cluster 5 [IRF1/STAT1/PU.1]) were found to have a major role in controlling macrophage transcriptional programs both at the basal level and after IFN-γ activation. They direct the expression of a set of genes, the IRF8/IRF1 regulome, that play critical roles in host inflammatory and antimicrobial defenses in mouse models of neuroinflammation and of pulmonary tuberculosis, respectively. In addition, this IRF8/IRF1 regulome is enriched for genes mutated in human primary immunodeficiencies and with loci associated with several inflammatory diseases in humans.
Collapse
Affiliation(s)
- David Langlais
- Department of Biochemistry, McGill University, H3G 0B1 Montreal, Quebec, Canada Complex Traits Group, McGill University, H3G 0B1 Montreal, Quebec, Canada
| | - Luis B Barreiro
- Sainte Justine Hospital Research Centre, H3T 1C5 Montreal, Quebec, Canada Department of Pediatrics, Faculty of Medicine, University of Montreal, H3T 1J4 Montreal, Quebec, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, H3G 0B1 Montreal, Quebec, Canada Complex Traits Group, McGill University, H3G 0B1 Montreal, Quebec, Canada
| |
Collapse
|
139
|
Klaeschen AS, Wenzel J. Upcoming therapeutic targets in cutaneous lupus erythematous. Expert Rev Clin Pharmacol 2016; 9:567-578. [DOI: 10.1586/17512433.2016.1145543] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
140
|
Raj P, Rai E, Song R, Khan S, Wakeland BE, Viswanathan K, Arana C, Liang C, Zhang B, Dozmorov I, Carr-Johnson F, Mitrovic M, Wiley GB, Kelly JA, Lauwerys BR, Olsen NJ, Cotsapas C, Garcia CK, Wise CA, Harley JB, Nath SK, James JA, Jacob CO, Tsao BP, Pasare C, Karp DR, Li QZ, Gaffney PM, Wakeland EK. Regulatory polymorphisms modulate the expression of HLA class II molecules and promote autoimmunity. eLife 2016; 5:e12089. [PMID: 26880555 PMCID: PMC4811771 DOI: 10.7554/elife.12089] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/13/2016] [Indexed: 12/15/2022] Open
Abstract
Targeted sequencing of sixteen SLE risk loci among 1349 Caucasian cases and controls produced a comprehensive dataset of the variations causing susceptibility to systemic lupus erythematosus (SLE). Two independent disease association signals in the HLA-D region identified two regulatory regions containing 3562 polymorphisms that modified thirty-seven transcription factor binding sites. These extensive functional variations are a new and potent facet of HLA polymorphism. Variations modifying the consensus binding motifs of IRF4 and CTCF in the XL9 regulatory complex modified the transcription of HLA-DRB1, HLA-DQA1 and HLA-DQB1 in a chromosome-specific manner, resulting in a 2.5-fold increase in the surface expression of HLA-DR and DQ molecules on dendritic cells with SLE risk genotypes, which increases to over 4-fold after stimulation. Similar analyses of fifteen other SLE risk loci identified 1206 functional variants tightly linked with disease-associated SNPs and demonstrated that common disease alleles contain multiple causal variants modulating multiple immune system genes.
Collapse
Affiliation(s)
- Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ekta Rai
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Ran Song
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Shaheen Khan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Benjamin E Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kasthuribai Viswanathan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Carlos Arana
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chaoying Liang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Bo Zhang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Igor Dozmorov
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ferdicia Carr-Johnson
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Mitja Mitrovic
- Department of Neurology, Yale School of Medicine, New Haven, United States
| | - Graham B Wiley
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Jennifer A Kelly
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Bernard R Lauwerys
- Pole de pathologies rhumatismales, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Nancy J Olsen
- Division of Rheumatology, Department of Medicine, Penn State Medical School, Hershey, United States
| | - Chris Cotsapas
- Department of Neurology, Yale School of Medicine, New Haven, United States
| | - Christine K Garcia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, United States
| | - Carol A Wise
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, United States
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, United States
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - John B Harley
- Cincinnati VA Medical Center, Cincinnati, United States
- Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Swapan K Nath
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Judith A James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Chaim O Jacob
- Department of Medicine, University of Southern California, Los Angeles, United States
| | - Betty P Tsao
- Department of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Chandrashekhar Pasare
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - David R Karp
- Rheumatic Diseases Division, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Quan Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
141
|
Macias-Garcia A, Heizmann B, Sellars M, Marchal P, Dali H, Pasquali JL, Muller S, Kastner P, Chan S. Ikaros Is a Negative Regulator of B1 Cell Development and Function. J Biol Chem 2016; 291:9073-86. [PMID: 26841869 DOI: 10.1074/jbc.m115.704239] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Indexed: 12/19/2022] Open
Abstract
B1 B cells secrete most of the circulating natural antibodies and are considered key effector cells of the innate immune response. However, B1 cell-associated antibodies often cross-react with self-antigens, which leads to autoimmunity, and B1 cells have been implicated in cancer. How B1 cell activity is regulated remains unclear. We show that the Ikaros transcription factor is a major negative regulator of B1 cell development and function. Using conditional knock-out mouse models to delete Ikaros at different locations, we show that Ikaros-deficient mice exhibit specific and significant increases in splenic and bone marrow B1 cell numbers, and that the B1 progenitor cell pool is increased ∼10-fold in the bone marrow. Ikaros-null B1 cells resemble WT B1 cells at the molecular and cellular levels, but show a down-regulation of signaling components important for inhibiting proliferation and immunoglobulin production. Ikaros-null B1 cells hyper-react to TLR4 stimulation and secrete high amounts of IgM autoantibodies. These results indicate that Ikaros is required to limit B1 cell homeostasis in the adult.
Collapse
Affiliation(s)
- Alejandra Macias-Garcia
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Beate Heizmann
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France,
| | - MacLean Sellars
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Patricia Marchal
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Hayet Dali
- Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, 67000 Strasbourg, France
| | - Jean-Louis Pasquali
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, 67000 Strasbourg, France, UFR Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Sylviane Muller
- Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, 67000 Strasbourg, France, Institut d'Etudes Avancées, Université de Strasbourg, 67000 Strasbourg, France, and
| | - Philippe Kastner
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France, Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
| | - Susan Chan
- From the Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France,
| |
Collapse
|
142
|
Yogo T, Nagamiya H, Seto M, Sasaki S, Shih-Chung H, Ohba Y, Tokunaga N, Lee GN, Rhim CY, Yoon CH, Cho SY, Skene R, Yamamoto S, Satou Y, Kuno M, Miyazaki T, Nakagawa H, Okabe A, Marui S, Aso K, Yoshida M. Structure-Based Design and Synthesis of 3-Amino-1,5-dihydro-4H-pyrazolopyridin-4-one Derivatives as Tyrosine Kinase 2 Inhibitors. J Med Chem 2016; 59:733-49. [PMID: 26701356 DOI: 10.1021/acs.jmedchem.5b01857] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report herein the discovery and optimization of 3-amino-1,5-dihydro-4H-pyrazolopyridin-4-one TYK2 inhibitors. High-throughput screening against TYK2 and JAK1-3 provided aminoindazole derivative 1 as a hit compound. Scaffold hopping of the aminoindazole core led to the discovery of 3-amino-1,5-dihydro-4H-pyrazolopyridin-4-one derivative 3 as a novel chemotype of TYK2 inhibitors. Interestingly, initial SAR study suggested that this scaffold could have a vertically flipped binding mode, which prompted us to introduce a substituent at the 7-position as a moiety directed toward the solvent-exposed region. Introduction of a 1-methyl-3-pyrazolyl moiety at the 7-position resulted in a dramatic increase in TYK2 inhibitory activity, and further optimization led to the discovery of 20. Compound 20 inhibited IL-23-induced IL-22 production in a rat PD assay, as well as inhibited IL-23 signaling in human PBMC. Furthermore, 20 showed selectivity for IL-23 signaling inhibition against GM-CSF, demonstrating the unique cytokine selectivity of the novel TYK2 inhibitor.
Collapse
Affiliation(s)
- Takatoshi Yogo
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiroyuki Nagamiya
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masaki Seto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Satoshi Sasaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Huang Shih-Chung
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited , 40 Landsdowne Street, Cambridge, Massachusetts 02139, United States
| | - Yusuke Ohba
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Norihito Tokunaga
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Gil Nam Lee
- Chemizon , 3F Dongbang Rental Building, 333-1 Sangdaewon-Dong, Jungwon-Gu, Seongnam-Si, Kyunggi-Do 462-120, Korea
| | - Chul Yun Rhim
- Chemizon , 3F Dongbang Rental Building, 333-1 Sangdaewon-Dong, Jungwon-Gu, Seongnam-Si, Kyunggi-Do 462-120, Korea
| | - Cheol Hwan Yoon
- Chemizon , 3F Dongbang Rental Building, 333-1 Sangdaewon-Dong, Jungwon-Gu, Seongnam-Si, Kyunggi-Do 462-120, Korea
| | - Suk Young Cho
- Chemizon , 3F Dongbang Rental Building, 333-1 Sangdaewon-Dong, Jungwon-Gu, Seongnam-Si, Kyunggi-Do 462-120, Korea
| | - Robert Skene
- Takeda California , 10410 Science Center Drive, San Diego, California 92121, United States
| | - Syunsuke Yamamoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yousuke Satou
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masako Kuno
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takahiro Miyazaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hideyuki Nakagawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Atsutoshi Okabe
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shogo Marui
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazuyoshi Aso
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masato Yoshida
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
143
|
Jiang Y, Wang H, Yu H, Li L, Xu D, Hou S, Kijlstra A, Yang P. Two Genetic Variations in the IRF8 region are associated with Behçet's disease in Han Chinese. Sci Rep 2016; 6:19651. [PMID: 26794091 PMCID: PMC4726413 DOI: 10.1038/srep19651] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/12/2015] [Indexed: 12/22/2022] Open
Abstract
Several modulatory factors in the TLR signaling pathway including IRF3, IRF7, IRF8, TRIM20, MYD88 and NF-κB1 have been associated with autoimmune disease. In this study, we investigated the association of 13 SNPs for these genes with Behçet’s disease (BD) and Vogt-Koyanagi-Harada (VKH) syndrome using a polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) assay. Haplotype and linkage disequilibrium (LD) analysis were performed by Haploview4.2. IRF8 mRNA expression and cytokine production was tested by real-time PCR and ELISA. Two SNPs near IRF8 were associated with BD (for rs17445836 GG genotype, Pc = 9.56 × 10−8, OR = 2.044; for rs11642873 AA genotype, Pc = 9.24 × 10−7, OR = 1.776). No significant association was found for the 13 SNPs tested with VKH syndrome. Haplotype analysis of the two positive SNPs revealed that the AG haplotype was significantly increased in BD patients (Pc = 2.60 × 10−8, OR = 1.646). Functional studies revealed an increased mRNA expression of IRF8 and IFN-γ production and a decreased production of IL-10 in rs17445836 carriers with the GG genotype. Increased expression of IRF8 as well as IFN-γ production and a decreased production of IL-10 were found in individuals carrying the rs11642873/AA genotype. In conclusion, this study indicates that IRF8 may contribute to the genetic susceptibility of BD by regulating IRF8 expression and cytokine production.
Collapse
Affiliation(s)
- Yanni Jiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, P R China
| | - Hong Wang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Science, Beijing Tongren Hospital, Capital Medical University, Beijing, P R China
| | - Hongsong Yu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, P R China
| | - Lin Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, P R China
| | - Dengfeng Xu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, P R China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, P R China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, P R China
| |
Collapse
|
144
|
|
145
|
Yarwood A, Viatte S, Okada Y, Plenge R, Yamamoto K, BRAGGSS, RACI, Barton A, Symmons D, Raychaudhuri S, Klareskog L, Gregersen P, Worthington J, Eyre S. Loci associated with N-glycosylation of human IgG are not associated with rheumatoid arthritis: a Mendelian randomisation study. Ann Rheum Dis 2016; 75:317-20. [PMID: 26386125 PMCID: PMC4717396 DOI: 10.1136/annrheumdis-2014-207210] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 06/15/2015] [Accepted: 07/14/2015] [Indexed: 01/12/2023]
Abstract
OBJECTIVES A recent study identified 16 genetic variants associated with N-glycosylation of human IgG. Several of the genomic regions where these single nucleotide polymorphisms (SNPs) reside have also been associated with autoimmune disease (AID) susceptibility, suggesting there may be pleiotropy (genetic sharing) between loci controlling both N-glycosylation and AIDs. We investigated this by testing variants associated with levels of IgG N-glycosylation for association with rheumatoid arthritis (RA) susceptibility using a Mendelian randomisation study, and testing a subset of these variants in a less well-powered study of treatment response and severity. METHODS SNPs showing association with IgG N-glycosylation were analysed for association with RA susceptibility in 14 361 RA cases and 43 923 controls. Five SNPs were tested for association with response to anti-tumour necrosis factor (TNF) therapy in 1081 RA patient samples and for association with radiological disease severity in 342 patients. RESULTS Only one SNP (rs9296009) associated with N-glycosylation showed an association (p=6.92×10(-266)) with RA susceptibility, although this was due to linkage disequilibrium with causal human leukocyte antigen (HLA) variants. Four regions of the genome harboured SNPs associated with both traits (shared loci); although statistical analysis indicated that the associations observed for the two traits are independent. No SNPs showed association with response to anti-TNF therapy. One SNP rs12342831 was modestly associated with Larsen score (p=0.05). CONCLUSIONS In a large, well-powered cohort of RA patients, we show SNPs driving levels of N-glycosylation have no association with RA susceptibility, indicating colocalisation of associated SNPs are not necessarily indicative of a shared genetic background or a role for glycosylation in disease susceptibility.
Collapse
Affiliation(s)
- Annie Yarwood
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Sebastien Viatte
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Yukinori Okada
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, 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
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Robert Plenge
- Merck Research Laboratories, Merck & Co. Inc., Boston, Massachusetts, USA
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | - Anne Barton
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Deborah Symmons
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, Stopford Building, The University of Manchester, Manchester, UK
| | - Soumya Raychaudhuri
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Peter Gregersen
- The Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, New York, USA
| | - Jane Worthington
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Steve Eyre
- Faculty of Medical and Human Sciences, Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
146
|
Abstract
The interferon (IFN) response is a powerful system that was evolutionarily acquired by vertebrates including mammals to protect against viral infection. The cytoplasmic RNA helicases, RIG-I-like receptors (RLRs), were discovered in 2004 as viral sensors that trigger the antiviral IFN response by recognizing the nonself signatures of viral RNAs. The mechanisms underlying the recognition of viral RNAs and signal transduction leading to the production of type I IFN have been intensively studied following the discovery of RLRs. Moreover, a dysregulation in the expression of RLR or aberrant RLR signaling has been implicated in the development of a number of autoimmune diseases. We herein provide an overview of recent advances in RLR research and discussed future directions.
Collapse
|
147
|
Demirci FY, Wang X, Kelly JA, Morris DL, Barmada MM, Feingold E, Kao AH, Sivils KL, Bernatsky S, Pineau C, Clarke A, Ramsey-Goldman R, Vyse TJ, Gaffney PM, Manzi S, Kamboh MI. Identification of a New Susceptibility Locus for Systemic Lupus Erythematosus on Chromosome 12 in Individuals of European Ancestry. Arthritis Rheumatol 2016; 68:174-83. [PMID: 26316170 PMCID: PMC4747422 DOI: 10.1002/art.39403] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/18/2015] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) in individuals of European ancestry identified a number of systemic lupus erythematosus (SLE) susceptibility loci using earlier versions of high-density genotyping platforms. Followup studies on suggestive GWAS regions using larger samples and more markers identified additional SLE loci in subjects of European descent. This multistage study was undertaken to identify novel SLE loci. METHODS In stage 1, we conducted a new GWAS of SLE in a North American case-control sample of subjects of European ancestry (n = 1,166) genotyped on Affymetrix Genome-Wide Human SNP Array 6.0. In stage 2, we further investigated top new suggestive GWAS hits by in silico evaluation and meta-analysis using an additional data set of subjects of European descent (>2,500 individuals), followed by replication of top meta-analysis findings in another data set of subjects of European descent (>10,000 individuals) in stage 3. RESULTS As expected, our GWAS revealed the most significant associations at the major histocompatibility complex locus (6p21), which easily surpassed the genome-wide significance threshold (P < 5 × 10(-8)). Several other SLE signals/loci previously implicated in Caucasians and/or Asians were also confirmed in the stage 1 discovery sample, and the strongest signals were observed at 2q32/STAT4 (P = 3.6 × 10(-7)) and at 8p23/BLK (P = 8.1 × 10(-6)). Stage 2 meta-analyses identified a new genome-wide significant SLE locus at 12q12 (meta P = 3.1 × 10(-8)), which was replicated in stage 3. CONCLUSION Our multistage study identified and replicated a new SLE locus that warrants further followup in additional studies. Publicly available databases suggest that this newly identified SLE signal falls within a functionally relevant genomic region and near biologically important genes.
Collapse
MESH Headings
- Adult
- Case-Control Studies
- Casein Kinase II/genetics
- Cell Cycle Proteins/genetics
- Chromosomes, Human, Pair 12/genetics
- Chromosomes, Human, Pair 2
- Chromosomes, Human, Pair 6
- Chromosomes, Human, Pair 8
- Computer Simulation
- Female
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Genotype
- HLA-DQ alpha-Chains/genetics
- HLA-DQ beta-Chains/genetics
- Humans
- Lupus Erythematosus, Systemic/genetics
- Major Histocompatibility Complex/genetics
- Male
- Middle Aged
- Polymorphism, Single Nucleotide
- Quantitative Trait Loci
- STAT4 Transcription Factor/genetics
- Tenascin/genetics
- Transcriptome
- White People/genetics
- src-Family Kinases/genetics
Collapse
Affiliation(s)
- F. Yesim Demirci
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xingbin Wang
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jennifer A. Kelly
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - David L. Morris
- Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - M. Michael Barmada
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Eleanor Feingold
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Amy H. Kao
- Lupus Center of Excellence, Department of Medicine, Allegheny Health Network, Pittsburgh, PA 15224, USA
| | - Kathy L. Sivils
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sasha Bernatsky
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada
| | - Christian Pineau
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada
| | - Ann Clarke
- Division of Rheumatology, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Rosalind Ramsey-Goldman
- Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Timothy J. Vyse
- Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Patrick M. Gaffney
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Susan Manzi
- Lupus Center of Excellence, Department of Medicine, Allegheny Health Network, Pittsburgh, PA 15224, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
148
|
Bentham J, Morris DL, Graham DSC, Pinder CL, Tombleson P, Behrens TW, Martín J, Fairfax BP, Knight JC, Chen L, Replogle J, Syvänen AC, Rönnblom L, Graham RR, Wither JE, Rioux JD, Alarcón-Riquelme ME, Vyse TJ. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat Genet 2015; 47:1457-1464. [PMID: 26502338 PMCID: PMC4668589 DOI: 10.1038/ng.3434] [Citation(s) in RCA: 697] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 10/02/2015] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is a genetically complex autoimmune disease characterized by loss of immune tolerance to nuclear and cell surface antigens. Previous genome-wide association studies (GWAS) had modest sample sizes, reducing their scope and reliability. Our study comprised 7,219 cases and 15,991 controls of European ancestry, constituting a new GWAS, a meta-analysis with a published GWAS and a replication study. We have mapped 43 susceptibility loci, including ten new associations. Assisted by dense genome coverage, imputation provided evidence for missense variants underpinning associations in eight genes. Other likely causal genes were established by examining associated alleles for cis-acting eQTL effects in a range of ex vivo immune cells. We found an over-representation (n = 16) of transcription factors among SLE susceptibility genes. This finding supports the view that aberrantly regulated gene expression networks in multiple cell types in both the innate and adaptive immune response contribute to the risk of developing SLE.
Collapse
Affiliation(s)
- James Bentham
- Division of Genetics and Molecular Medicine, King's College London, UK
| | - David L Morris
- Division of Genetics and Molecular Medicine, King's College London, UK
| | | | | | - Philip Tombleson
- Division of Genetics and Molecular Medicine, King's College London, UK
| | | | - Javier Martín
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
| | - Benjamin P Fairfax
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julian C Knight
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lingyan Chen
- Division of Genetics and Molecular Medicine, King's College London, UK
| | | | - Ann-Christine Syvänen
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Joan E Wither
- Toronto Western Research Institute (TWRI), University Health Network, Toronto, Ontario, Canada
| | - John D Rioux
- Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Montreal, Quebec, Canada
| | - Marta E Alarcón-Riquelme
- Centro de Genómica e Investigación Oncológica (GENYO), Pfizer-Universidad de Granada-Junta de Andalucía, Granada, Spain
| | - Timothy J Vyse
- Division of Genetics and Molecular Medicine, King's College London, UK
- Division of Immunology, Infection and Inflammatory Disease, King's College London, UK
| |
Collapse
|
149
|
Kato H, Fujita T. RIG-I-like receptors and autoimmune diseases. Curr Opin Immunol 2015; 37:40-5. [PMID: 26530735 DOI: 10.1016/j.coi.2015.10.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 09/26/2015] [Accepted: 10/08/2015] [Indexed: 12/24/2022]
Abstract
Type I interferon (IFN) plays an essential role in antiviral innate immune responses and also in adaptive immune responses. Defects in the production of IFN markedly increase susceptibility to viral invasion and attenuate the acquired immunity. Recently an increased expression of type I IFN, also termed IFN signature, has been reported in patients with autoimmune diseases such as systemic lupus erythematosus (SLE) and Aicardi-Goutières syndrome (AGS). The evidence clearly shows that the initiation and termination of IFN production should be tightly controlled. RIG-I-like receptors (RLRs) are viral RNA sensors and are essential for type I IFN induction. We herein summarize recent reports on RLR mutations in patients and MDA5 mutant mice, and discuss possible mechanisms by which aberrant activation of RLRs can cause autoimmunity.
Collapse
Affiliation(s)
- Hiroki Kato
- Laboratory of Molecular Genetics, Institute for Virus Research, and Laboratory of Molecular and Cellular Immunology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; PRESTO, Science and Technology Agency (JST), Kawaguchi, Saitama 332-0012, Japan
| | - Takashi Fujita
- Laboratory of Molecular Genetics, Institute for Virus Research, and Laboratory of Molecular and Cellular Immunology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
150
|
O'Neill S, Brault J, Stasia MJ, Knaus UG. Genetic disorders coupled to ROS deficiency. Redox Biol 2015; 6:135-156. [PMID: 26210446 PMCID: PMC4550764 DOI: 10.1016/j.redox.2015.07.009] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/15/2015] [Accepted: 07/16/2015] [Indexed: 12/24/2022] Open
Abstract
Maintaining the redox balance between generation and elimination of reactive oxygen species (ROS) is critical for health. Disturbances such as continuously elevated ROS levels will result in oxidative stress and development of disease, but likewise, insufficient ROS production will be detrimental to health. Reduced or even complete loss of ROS generation originates mainly from inactivating variants in genes encoding for NADPH oxidase complexes. In particular, deficiency in phagocyte Nox2 oxidase function due to genetic variants (CYBB, CYBA, NCF1, NCF2, NCF4) has been recognized as a direct cause of chronic granulomatous disease (CGD), an inherited immune disorder. More recently, additional diseases have been linked to functionally altered variants in genes encoding for other NADPH oxidases, such as for DUOX2/DUOXA2 in congenital hypothyroidism, or for the Nox2 complex, NOX1 and DUOX2 as risk factors for inflammatory bowel disease. A comprehensive overview of novel developments in terms of Nox/Duox-deficiency disorders is presented, combined with insights gained from structure-function studies that will aid in predicting functional defects of clinical variants.
Collapse
Affiliation(s)
- Sharon O'Neill
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Julie Brault
- Université Grenoble Alpes, TIMC-IMAG Pôle Biologie, CHU de Grenoble, Grenoble, France; CGD Diagnosis and Research Centre, Pôle Biologie, CHU de Grenoble, Grenoble, France
| | - Marie-Jose Stasia
- Université Grenoble Alpes, TIMC-IMAG Pôle Biologie, CHU de Grenoble, Grenoble, France; CGD Diagnosis and Research Centre, Pôle Biologie, CHU de Grenoble, Grenoble, France
| | - Ulla G Knaus
- Conway Institute, University College Dublin, Dublin, Ireland.
| |
Collapse
|