1
|
Ruggeri E, Klohonatz K, Durrant B, Sirard MA. Identification and Preliminary Analysis of Granulosa Cell Biomarkers to Predict Oocyte In Vitro Maturation Outcome in the Southern White Rhinoceros ( Ceratotherium simum simum). Animals (Basel) 2024; 14:3538. [PMID: 39682503 DOI: 10.3390/ani14233538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In recent years, biomarkers in granulosa cells (GC) have been determined and associated in several species with oocyte maturation, in vitro fertilization success, and embryo development outcomes. The identification of biomarkers of oocyte competence can aid in improving assisted reproductive technologies (ARTs) in the southern white rhino (SWR). This study aimed to identify biomarkers present in SWR GC associated with oocytes that either did or did not mature in vitro. We evaluated follicle development (FD), meiotic competence (MC), cell death and atresia (CDA), and embryonic genome activation (EGA). Our objective was to design biomarkers to predict oocyte in vitro maturation results in the SWR. RNA was isolated from GC obtained during ovum pick up (OPU) for qPCR analysis. Overall, 22 genes were assessed, and nine were differentially expressed between GC from oocytes that did or did not mature in vitro (FD-GDF9 and mTOR; MC-GGPS1, JMY, and NPR2; CDA-COL4A1, MACIR, and TMPO; EGA-NFYA). From these data, we determined that GC can be used as a predictor for oocyte in vitro maturation outcome in the SWR. Our results provide crucial information needed to improve in vitro maturation and ARTs in this species.
Collapse
Affiliation(s)
- Elena Ruggeri
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA 92027, USA
| | - Kristin Klohonatz
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Barbara Durrant
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA 92027, USA
| | - Marc-André Sirard
- Département des Sciences Animales, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
2
|
Klohonatz K, Durrant B, Sirard MA, Ruggeri E. Granulosa cells provide transcriptomic information on ovarian follicle dynamics in southern white rhinoceros. Sci Rep 2024; 14:19321. [PMID: 39164442 PMCID: PMC11336098 DOI: 10.1038/s41598-024-70235-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Much remains unknown about the reproductive physiology of southern white rhinoceros (SWR) and the effect of ovarian stimulation prior to ovum pickup (OPU) have not been fully elucidated. Granulosa cells (GC) provide valuable insight into follicle growth and oocyte maturation status. The goals of this study were to evaluate transcriptomic changes in GC from three stages of follicle development and to identify biomarkers possibly associated with follicular growth and maturation as a result of ovarian stimulation. GC collected from SWRs following OPU were assigned stages based upon follicle size. Total RNA was isolated, and cDNA libraries were prepared and sequenced on a NovaSeq 6000. All bioinformatics analyses were performed utilizing the Galaxy web platform. Reads were aligned to CerSimCot1.0, and the manual curation was performed with EquCab3.0. Overall, 39,455 transcripts (21,612 genes) were identified across follicle stages, and manual curation yielded a 61% increase in gene identification from the original annotation. Granulosa cells from preovulatory follicles expressed the highest number of unique transcripts. The following seven biomarkers were determined based upon cluster analysis and patterns of expression: COL1A1, JMY, FBXW11, NRG1, TMPO, MACIR and COL4A1. These data can be used to potentially evaluate the effects of different ovarian stimulation protocols on follicle dynamics, improve OPU results, and support conservation efforts in this species.
Collapse
Affiliation(s)
- Kristin Klohonatz
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara Durrant
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA, USA
| | - Marc-André Sirard
- Département des Sciences Animales, Université Laval, Québec City, Québec, Canada
| | - Elena Ruggeri
- Reproductive Sciences, Conservation Science Wildlife Health, San Diego Zoo Wildlife Alliance, Escondido, CA, USA.
| |
Collapse
|
3
|
Yamada S, Nagafuchi Y, Fujio K. Pathophysiology and stratification of treatment-resistant rheumatoid arthritis. Immunol Med 2024; 47:12-23. [PMID: 37462450 DOI: 10.1080/25785826.2023.2235734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/09/2023] [Indexed: 02/23/2024] Open
Abstract
Early diagnosis and timely therapeutic intervention are clinical challenges of rheumatoid arthritis (RA), especially for treatment-resistant or difficult-to-treat patients. Little is known about the immunological mechanisms involved in refractory RA. In this review, we summarize previous research findings on the immunological mechanisms of treatment-resistant RA. Genetic prediction of treatment-resistant RA is challenging. Patients with and without anti-cyclic citrullinated peptide autoantibodies are considered part of distinct subgroups, especially regarding long-term clinical prognosis and treatment responses. B cells, T cells and other immune cells and fibroblasts are of pathophysiological importance and are associated with treatment responses. Finally, we propose a new hypothesis that stratifies patients with RA into two subgroups with distinct immunological pathologies based on our recent immunomics analysis of RA. One RA subgroup with a favorable prognosis is characterized by increased interferon signaling. Another subgroup with a worse prognosis is characterized by enhanced acquired immune responses. Increases in dendritic cell precursors and diversified autoreactive anti-modified protein antibodies may have pathophysiological roles, especially in the latter subgroup. These findings that improve treatment response predictions might contribute to future precision medicine for RA.
Collapse
Affiliation(s)
- Saeko Yamada
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Serwin K, Kozak M, Mazurek-Mochol M, Piotrowska K, Czerewaty M, Pawlik A. Human macrophage immunometabolism regulator (MACIR) in patients with periodontitis. Immunobiology 2023; 228:152760. [PMID: 37976770 DOI: 10.1016/j.imbio.2023.152760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/02/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Periodontitis is a local inflammatory reaction caused by bacterial infection in which immune cells, including macrophages, are involved. Recent studies have shown that an important regulator of macrophage function is the human macrophage immunometabolism regulator (MACIR). This gene has been shown to play a key role in modulating the immune response by affecting the activity of fibroblasts and macrophages. In this study, we investigated the expression of MACIR in the gingival tissues of patients with periodontal disease, as well as the effect of IL-1β and TNF-α on the expression of MACIR gene and protein in human gingival fibroblasts. METHODS MACIR mRNA expression in gingival tissue samples was determined using Real-time PCR. Expression of MACIR protein was determined using immunofluorescent staining and western blotting. RESULTS The MACIR mRNA expression in gingival tissue samples in patients with periodontitis was statistically significantly lower than in gingival tissue samples from healthy controls (p = 0.009). The stimulation of human gingival fibroblasts with IL-1β and TNF-α resulted in a statistically significant decrease of MACIR gene mRNA expression. In western blotting and immunofluorescent analysis, we confirmed that the stimulation of the primary culture of human gingival fibroblasts by both IL-1β and TNF-α decreases the expression of MACIR protein. CONCLUSION The results of the study suggest that MACIR is an important regulator of the inflammatory process in patients with periodontitis. Decreased expression of the MACIR gene may activate macrophages to secrete mediators that increase inflammation and cause periodontal tissue destruction.
Collapse
Affiliation(s)
- Karol Serwin
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, Arkońska 4, 71-455 Szczecin, Poland
| | - Małgorzata Kozak
- Chair and Department of Dental Prosthetics, Pomeranian Medical University, Powstańców Wlkp 72, 70-111 Szczecin, Poland
| | - Małgorzata Mazurek-Mochol
- Department of Periodontology, Pomeranian Medical University, Powstańców Wlkp 72, 70-111 Szczecin, Poland
| | - Katarzyna Piotrowska
- Department of Physiology, Pomeranian Medical University, Powstańców Wlkp 72, 70-111 Szczecin, Poland
| | - Michał Czerewaty
- Department of Physiology, Pomeranian Medical University, Powstańców Wlkp 72, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Powstańców Wlkp 72, 70-111 Szczecin, Poland.
| |
Collapse
|
5
|
Yuan G, Yang ST, Yang S. Regulator of G protein signaling 12 drives inflammatory arthritis by activating synovial fibroblasts through MYCBP2/KIF2A signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:197-210. [PMID: 36700049 PMCID: PMC9843488 DOI: 10.1016/j.omtn.2022.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Synovial fibroblasts are the active and aggressive drivers in the progression of arthritis, but the cellular and molecular mechanisms remain unknown. Here, our results showed that regulator of G protein signaling 12 (RGS12) maintained ciliogenesis in synovial fibroblasts, which is critical for the development of inflammatory arthritis. Deletion of RGS12 led to a significant decrease in ciliogenesis, adhesion, migration, and secretion of synovial fibroblasts. Mechanistically, RGS12 overexpression in synovial fibroblasts increased the length and number of cilia but decreased the protein level of kinesin family member 2A (KIF2A). The results of LC-MS analyses showed that RGS12 interacted with MYC binding protein 2 to enhance its ubiquitination activity, through which the KIF2A protein was degraded in synovial fibroblasts. Moreover, overexpression of KIF2A blocked the increases in cilia length and number. Mice with RGS12 deficiency or treatment of RGS12 shRNA nanoparticles significantly decreased the clinical score, paw swelling, synovitis, and cartilage destruction during inflammatory arthritis by inhibiting the activation of synovial fibroblasts. Therefore, this study provides evidence that RGS12 activates synovial fibroblasts' pathological function via promoting MCYBP2-mediated degradation of KIF2A and ciliogenesis. Our data suggest that RGS12 may be a potential drug target for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Gongsheng Yuan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shu-ting Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuying Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Penn Center for Musculoskeletal Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
6
|
Li K, Wang M, Zhao L, Liu Y, Zhang X. ACPA-negative rheumatoid arthritis: From immune mechanisms to clinical translation. EBioMedicine 2022; 83:104233. [PMID: 36027873 PMCID: PMC9404277 DOI: 10.1016/j.ebiom.2022.104233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022] Open
Abstract
The presence of anti-citrullinated protein autoantibodies (ACPA) is a hallmark feature of rheumatoid arthritis (RA), which causes chronic joint destruction and systemic inflammation. Based on ACPA status, RA patients can be sub-grouped into two major subsets: ACPA-positive RA (ACPA+ RA) and ACPA-negative RA (ACPA– RA). Accumulating evidence have suggested that ACPA+ RA and ACPA– RA are two distinct disease entities with different underlying pathophysiology. In contrast to the well-characterized pathogenic mechanisms of ACPA+ RA, the etiology of ACPA– RA remains largely unknown. In this review, we summarized current knowledge about the primary drivers of ACPA– RA, particularly focusing on the serological, cellular, and molecular aspects of immune mechanisms. A better understanding of the immunopathogenesis in ACPA– RA will help in designing more precisely targeting strategies, and paving the road to personalized treatment. In addition, identification of novel biomarkers in ACPA– RA will substantially promote early treatment and improve the outcomes.
Collapse
Affiliation(s)
- Ketian Li
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China; The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, PR China.
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, PR China.
| |
Collapse
|
7
|
Honda S, Ikari K, Yano K, Terao C, Tanaka E, Harigai M, Kochi Y. Polygenic risk scores are associated with radiographic progression in patients with rheumatoid arthritis. Arthritis Rheumatol 2022; 74:791-800. [PMID: 35048562 DOI: 10.1002/art.42051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/18/2021] [Accepted: 12/09/2021] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To investigate whether polygenic risk score (PRS) using data from a genome-wide association study (GWAS) for rheumatoid arthritis (RA) susceptibility can be a predictor for radiographic progression. METHODS We constructed the PRS using GWAS summary data for disease susceptibility to predict Sharp/van der Heijde score (SHS) changes in first five years from the onset (the top quartile of SHS changes was defined as severe progression and the remaining as non-severe progression). We selected the best model in a training set (n = 500) and validated it in a testing set (n = 740). We evaluated the performance of PRS in univariable and multivariable analyses with other factors to predict severe progression. RESULTS PRS constructed of 43,784 SNPs significantly differed between severe and non-severe progression in both training (P = 0.0064) and testing sets (P = 0.017). The patients with the top quintile PRS had a higher risk for severe progression compared to those with the bottom quintile (odds ratio (OR) 1.90, P = 0.0022), which was higher when restricted to younger-onset patients (OR 5.06, P = 0.00038). The top quintile PRS and ACPAs positive groups had significantly higher proportion of patients with severe progression compared to the remaining groups (P = 0.00052, and 0.0022, respectively). Multivariable analysis showed that PRS (P = 0.00019) as well as sex (female) (P = 0.0033), ACPAs (P = 0.0023), and BMI (P = 0.031) were independent risk factors. CONCLUSION PRS using GWAS data for RA susceptibility is associated with radiographic progression.
Collapse
Affiliation(s)
- Suguru Honda
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.,Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Katsunori Ikari
- Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan.,Department of Orthopedic Surgery, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.,Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Koichiro Yano
- Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan.,Department of Orthopedic Surgery, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan.,The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Eiichi Tanaka
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.,Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Masayoshi Harigai
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.,Institute of Rheumatology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Yuta Kochi
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Ge X, Frank-Bertoncelj M, Klein K, McGovern A, Kuret T, Houtman M, Burja B, Micheroli R, Shi C, Marks M, Filer A, Buckley CD, Orozco G, Distler O, Morris AP, Martin P, Eyre S, Ospelt C. Functional genomics atlas of synovial fibroblasts defining rheumatoid arthritis heritability. Genome Biol 2021; 22:247. [PMID: 34433485 PMCID: PMC8385949 DOI: 10.1186/s13059-021-02460-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genome-wide association studies have reported more than 100 risk loci for rheumatoid arthritis (RA). These loci are shown to be enriched in immune cell-specific enhancers, but the analysis so far has excluded stromal cells, such as synovial fibroblasts (FLS), despite their crucial involvement in the pathogenesis of RA. Here we integrate DNA architecture, 3D chromatin interactions, DNA accessibility, and gene expression in FLS, B cells, and T cells with genetic fine mapping of RA loci. RESULTS We identify putative causal variants, enhancers, genes, and cell types for 30-60% of RA loci and demonstrate that FLS account for up to 24% of RA heritability. TNF stimulation of FLS alters the organization of topologically associating domains, chromatin state, and the expression of putative causal genes such as TNFAIP3 and IFNAR1. Several putative causal genes constitute RA-relevant functional networks in FLS with roles in cellular proliferation and activation. Finally, we demonstrate that risk variants can have joint-specific effects on target gene expression in RA FLS, which may contribute to the development of the characteristic pattern of joint involvement in RA. CONCLUSION Overall, our research provides the first direct evidence for a causal role of FLS in the genetic susceptibility for RA accounting for up to a quarter of RA heritability.
Collapse
Affiliation(s)
- Xiangyu Ge
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Mojca Frank-Bertoncelj
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kerstin Klein
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Amanda McGovern
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Tadeja Kuret
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia
| | - Miranda Houtman
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Blaž Burja
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia
| | - Raphael Micheroli
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Chenfu Shi
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | | | - Andrew Filer
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| | - Christopher D Buckley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, UK
| | - Gisela Orozco
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Foundation Trust, Manchester, UK
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Paul Martin
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Foundation Trust, Manchester, UK
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stephen Eyre
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Foundation Trust, Manchester, UK
| | - Caroline Ospelt
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Kwan A, Winder N, Atkinson E, Al-Janabi H, Allen RJ, Hughes R, Moamin M, Louie R, Evans D, Hutchinson M, Capper D, Cox K, Handley J, Wilshaw A, Kim T, Tazzyman SJ, Srivastava S, Ottewell P, Vadakekolathu J, Pockley G, Lewis CE, Brown JE, Danson SJ, Conner J, Muthana M. Macrophages Mediate the Antitumor Effects of the Oncolytic Virus HSV1716 in Mammary Tumors. Mol Cancer Ther 2021; 20:589-601. [PMID: 33298589 DOI: 10.1158/1535-7163.mct-20-0748] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 12/04/2020] [Indexed: 11/16/2022]
Abstract
Oncolytic viruses (OV) have been shown to activate the antitumor functions of specific immune cells like T cells. Here, we show OV can also reprogram tumor-associated macrophage (TAM) to a less immunosuppressive phenotype. Syngeneic, immunocompetent mouse models of primary breast cancer were established using PyMT-TS1, 4T1, and E0771 cell lines, and a metastatic model of breast cancer was established using the 4T1 cell line. Tumor growth and overall survival was assessed following intravenous administration of the OV, HSV1716 (a modified herpes simplex virus). Infiltration and function of various immune effector cells was assessed by NanoString, flow cytometry of dispersed tumors, and immunofluorescence analysis of tumor sections. HSV1716 administration led to marked tumor shrinkage in primary mammary tumors and a decrease in metastases. This was associated with a significant increase in the recruitment/activation of cytotoxic T cells, a reduction in the presence of regulatory T cells and the reprograming of TAMs towards a pro-inflammatory, less immunosuppressive phenotype. These findings were supported by in vitro data demonstrating that human monocyte-derived macrophages host HSV1716 replication, and that this led to immunogenic macrophage lysis. These events were dependent on macrophage expression of proliferating cell nuclear antigen (PCNA). Finally, the antitumor effect of OV was markedly diminished when TAMs were depleted using clodronate liposomes. Together, our results show that TAMs play an essential role in support of the tumoricidal effect of the OV, HSV1716-they both host viral replication via a novel, PCNA-dependent mechanism and are reprogramed to express a less immunosuppressive phenotype.
Collapse
Affiliation(s)
- Amy Kwan
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Natalie Winder
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Emer Atkinson
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Haider Al-Janabi
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Richard J Allen
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Russell Hughes
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Mohammed Moamin
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Rikah Louie
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Dhanajay Evans
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Matthew Hutchinson
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Drew Capper
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Katie Cox
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Joshua Handley
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Adam Wilshaw
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Taewoo Kim
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Simon J Tazzyman
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Sanjay Srivastava
- Department of Immunotherapeutics and Biotechnology and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, Texas
| | - Penelope Ottewell
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Centre for Health and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Centre for Health and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Claire E Lewis
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
- Sheffield ECMC, Cancer Clinical Trials Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - Janet E Brown
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
- Sheffield ECMC, Cancer Clinical Trials Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - Sarah J Danson
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
- Sheffield ECMC, Cancer Clinical Trials Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - Joe Conner
- Virttu Biologics Ltd., BioCity Scotland, Newhouse, United Kingdom
| | - Munitta Muthana
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom.
- Sheffield ECMC, Cancer Clinical Trials Centre, Weston Park Hospital, Sheffield, United Kingdom
| |
Collapse
|
10
|
McGauran G, Dorris E, Borza R, Morgan N, Shields DC, Matallanas D, Wilson AG, O'Connell DJ. Resolving the Interactome of the Human Macrophage Immunometabolism Regulator (MACIR) with Enhanced Membrane Protein Preparation and Affinity Proteomics. Proteomics 2020; 20:e2000062. [PMID: 32864787 DOI: 10.1002/pmic.202000062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/17/2020] [Indexed: 11/10/2022]
Abstract
Expression of the macrophage immunometabolism regulator gene (MACIR) is associated with severity of autoimmune disease pathology and with the regulation of macrophage biology through unknown mechanisms. The encoded 206 amino acid protein lacks homology to any characterized protein sequence and is a disordered protein according to structure prediction algorithms. To identify interactions of MACIR with proteins from all subcellular compartments, a membrane solubilization buffer is employed, that together with a high affinity EF hand based pull down method, increases the resolution of quantitative mass spectrometry analysis with significant enrichment of interactions from membrane bound nuclear and mitochondrial compartments compared to samples prepared with radioimmunoprecipitation assay buffer. A total of 63 significant interacting proteins are identified and interaction with the nuclear transport receptor TNPO1 and the trafficking proteins UNC119 homolog A and B are validated by immunoprecipitation. Mutational analysis in two candidate nuclear localization signal motifs in the MACIR amino acid sequence shows the interaction with TNPO1 is likely via a non-classical proline/tyrosine-nuclear localization signal motif (aa98-117). It is shown that employing a highly specific and high affinity pull down method that performs efficiently in this glycerol and detergent rich buffer is a powerful approach for the analysis of uncharacterized protein interactomes.
Collapse
Affiliation(s)
- Gavin McGauran
- School of Biomolecular & Biomedical Science, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| | - Emma Dorris
- School of Medicine, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Razvan Borza
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Niamh Morgan
- School of Medicine, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Denis C Shields
- School of Medicine, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - David Matallanas
- School of Medicine, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.,Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Anthony G Wilson
- School of Medicine, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - David J O'Connell
- School of Biomolecular & Biomedical Science, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland.,BiOrbic Bioeconomy Research Centre, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| |
Collapse
|
11
|
Feng X, Hao X, Shi R, Xia Z, Huang L, Yu Q, Zhou F. Detection and Comparative Analysis of Methylomic Biomarkers of Rheumatoid Arthritis. Front Genet 2020; 11:238. [PMID: 32292416 PMCID: PMC7119472 DOI: 10.3389/fgene.2020.00238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/28/2020] [Indexed: 01/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disorder influenced by both genetic and environmental factors. To investigate possible contributions of DNA methylation to the etiology of RA with minimum confounding genetic heterogeneity, we investigated genome-wide DNA methylation in disease-discordant monozygotic twin pairs. This study hypothesized that methylomic biomarkers might facilitate accurate RA detection. A comprehensive series of biomarker detection algorithms were utilized to find the best methylomic biomarkers for detecting RA patients using the methylomic data of the peripheral blood samples. The best model achieved 100.00% in accuracy (Acc) with 81 methylomic biomarkers and a 10-fold cross-validation (10FCV) strategy. Some of the methylomic biomarkers were experimentally confirmed to be associated with the onset or development of RA. It is also interesting to observe that many of the detected biomarkers were from chromosome Y, supporting the knowledge that RA has a significant gender discrepancy.
Collapse
Affiliation(s)
- Xin Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China.,Jilin Institute of Chemical Technology, Jilin, China.,BioKnow Health Informatics Lab, College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Xubing Hao
- BioKnow Health Informatics Lab, College of Software, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Ruoyao Shi
- BioKnow Health Informatics Lab, College of Life Sciences, Jilin University, Changchun, China
| | - Zhiqiang Xia
- BioKnow Health Informatics Lab, College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Lan Huang
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Fengfeng Zhou
- BioKnow Health Informatics Lab, College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, China
| |
Collapse
|
12
|
Scaravaggi I, Borel N, Romer R, Imboden I, Ulbrich SE, Zeng S, Bollwein H, Bauersachs S. Cell type-specific endometrial transcriptome changes during initial recognition of pregnancy in the mare. Reprod Fertil Dev 2019; 31:496-508. [PMID: 30253121 DOI: 10.1071/rd18144] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022] Open
Abstract
Previous endometrial gene expression studies during the time of conceptus migration did not provide final conclusions on the mechanisms of maternal recognition of pregnancy (MRP) in the mare. This called for a cell type-specific endometrial gene expression analysis in response to embryo signals to improve the understanding of gene expression regulation in the context of MRP. Laser capture microdissection was used to collect luminal epithelium (LE), glandular epithelium and stroma from endometrial biopsies from Day 12 of pregnancy and Day 12 of the oestrous cycle. RNA sequencing (RNA-Seq) showed greater expression differences between cell types than between pregnant and cyclic states; differences between the pregnant and cyclic states were mainly found in LE. Comparison with a previous RNA-Seq dataset for whole biopsy samples revealed the specific origin of gene expression differences. Furthermore, genes specifically differentially expressed (DE) in one cell type were found that were not detectable as DE in biopsies. Overall, this study revealed spatial information about endometrial gene expression during the phase of initial MRP. The conceptus induced changes in the expression of genes involved in blood vessel development, specific spatial regulation of the immune system, growth factors, regulation of prostaglandin synthesis, transport prostaglandin receptors, specifically prostaglandin F receptor (PTGFR) in the context of prevention of luteolysis.
Collapse
Affiliation(s)
- Iside Scaravaggi
- Clinic of Reproductive Medicine, Department for Farm Animals, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Nicole Borel
- Institute of Veterinary Pathology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Rebekka Romer
- Clinic of Reproductive Medicine, Department for Farm Animals, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Isabel Imboden
- Clinic of Reproductive Medicine, Department for Farm Animals, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Susanne E Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Universitätstrasse 2, 8092 Zurich, Switzerland
| | - Shuqin Zeng
- Clinic of Reproductive Medicine, Department for Farm Animals, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Heinrich Bollwein
- Clinic of Reproductive Medicine, Department for Farm Animals, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Stefan Bauersachs
- Clinic of Reproductive Medicine, Department for Farm Animals, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| |
Collapse
|
13
|
Machaj F, Rosik J, Szostak B, Pawlik A. The evolution in our understanding of the genetics of rheumatoid arthritis and the impact on novel drug discovery. Expert Opin Drug Discov 2019; 15:85-99. [PMID: 31661990 DOI: 10.1080/17460441.2020.1682992] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Rheumatoid arthritis (RA) is an autoimmune disease that is characterized by chronic inflammation of the joints and affects 1% of the population. Polymorphisms of genes that encode proteins that primarily participate in inflammation may influence RA occurrence or become useful biomarkers for certain types of anti-rheumatic treatment.Areas covered: The authors summarize the recent progress in our understanding of the genetics of RA. In the last few years, multiple variants of genes that are associated with RA risk have been identified. The development of new technologies and the detection of new potential therapeutic targets that contribute to novel drug discovery are also described.Expert opinion: There is still the need to search for new genes which may be a potential target for RA therapy. The challenge is to develop appropriate strategies for achieving insight into the molecular pathways involved in RA pathogenesis. Understanding the genetics, immunogenetics, epigenetics and immunology of RA could help to identify new targets for RA therapy. The development of new technologies has enabled the detection of a number of new genes, particularly genes associated with proinflammatory cytokines and chemokines, B- and T-cell activation pathways, signal transducers and transcriptional activators, which might be potential therapeutic targets in RA.
Collapse
Affiliation(s)
- Filip Machaj
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
14
|
Dorris ER, Linehan E, Trenkmann M, Veale DJ, Fearon U, Wilson AG. Association of the Rheumatoid Arthritis Severity Variant rs26232 with the Invasive Activity of Synovial Fibroblasts. Cells 2019; 8:cells8101300. [PMID: 31652652 PMCID: PMC6829881 DOI: 10.3390/cells8101300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
rs26232, located in intron one of C5orf30, is associated with the susceptibility to and severity of rheumatoid arthritis (RA). Here, we investigate the relationship between this variant and the biological activities of rheumatoid arthritis synovial fibroblasts (RASFs). RASFs were isolated from the knee joints of 33 RA patients. The rs26232 genotype was determined and cellular migration, invasion, and apoptosis were compared using in vitro techniques. The production of adhesion molecules, chemokines, and proteases was measured by ELISA or flow cytometry. Cohort genotypes were CC n = 16; CT n = 14; TT n = 3. In comparison with the RASFs of the CT genotype, the CC genotype showed a 1.48-fold greater invasiveness in vitro (p = 0.02), 1.6-fold higher expression intracellular adhesion molecule (ICAM)-1 (p = 0.001), and 5-fold IFN-γ inducible protein-10 (IP-10) (p = 0.01). There was no association of the rs26232 genotype with the expression levels of either total C5orf30 mRNA or any of the three transcript variants. The rs26232 C allele, which has previously been associated with both the risk and severity of RA, is associated with greater invasive activity of RASFs in vitro, and with higher expression of ICAM-1 and IP-10. In resting RASFs, rs26232 is not a quantitative trait locus for C5orf30 mRNA, indicating a more complex mechanism underlying the genotype‒phenotype relationship.
Collapse
Affiliation(s)
- Emma R Dorris
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland.
| | - Eimear Linehan
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland.
| | - Michelle Trenkmann
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland.
| | - Douglas J Veale
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland.
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D06 R590, Ireland.
| | - Anthony G Wilson
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland.
| |
Collapse
|
15
|
Karami J, Aslani S, Jamshidi A, Garshasbi M, Mahmoudi M. Genetic implications in the pathogenesis of rheumatoid arthritis; an updated review. Gene 2019; 702:8-16. [PMID: 30904715 DOI: 10.1016/j.gene.2019.03.033] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/09/2019] [Accepted: 03/17/2019] [Indexed: 01/11/2023]
Abstract
Three important factors, including genetics, environment factors and autoimmunity play a role in the pathogenesis of rheumatoid arthritis (RA). The heritability of RA has been accounted to be 50-60%, while the HLA involvement in heritability of the disease has been accounted to be 10-40%. It has been documented that shared epitope (SE) alleles, such as HLA-DRB1*01 and DRB1*04, some HLA alleles like HLA-DRB1*13 and DRB1*15 are connected to RA susceptibility. An advanced classification of SE categorizes SE alleles into four main groups namely, S1, S2, S3D, and S3P. The S2 and S3P groups have been linked to susceptibility of seropositive RA. Various genome-wide association studies (GWAS) have discovered many susceptibility loci implicated in pathogenesis of RA. Some of the important single nucleotide polymorphisms (SNPs) linked to RA are TRAF1, STAT4, CTLA4, IRF5, CCR6, PTPN22, IL23R, and PADI4. HLA and non-HLA genes may discriminate anti-cyclic citrullinated peptide (anti-CCP) antibody-positive and anti-CCP-negative RA groups. Furthermore, risk of the disease has also been linked to environmental agents, mainly cigarette smoking. Pharmacogenomics has also confirmed SNPs or genetic patterns that might be linked to drugs responses. Different aspects of genetic involvement in the pathogenesis, etiology, and RA complications are reviewed in this article.
Collapse
Affiliation(s)
- Jafar Karami
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Cavalli M, Baltzer N, Umer HM, Grau J, Lemnian I, Pan G, Wallerman O, Spalinskas R, Sahlén P, Grosse I, Komorowski J, Wadelius C. Allele specific chromatin signals, 3D interactions, and motif predictions for immune and B cell related diseases. Sci Rep 2019; 9:2695. [PMID: 30804403 PMCID: PMC6389883 DOI: 10.1038/s41598-019-39633-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Several Genome Wide Association Studies (GWAS) have reported variants associated to immune diseases. However, the identified variants are rarely the drivers of the associations and the molecular mechanisms behind the genetic contributions remain poorly understood. ChIP-seq data for TFs and histone modifications provide snapshots of protein-DNA interactions allowing the identification of heterozygous SNPs showing significant allele specific signals (AS-SNPs). AS-SNPs can change a TF binding site resulting in altered gene regulation and are primary candidates to explain associations observed in GWAS and expression studies. We identified 17,293 unique AS-SNPs across 7 lymphoblastoid cell lines. In this set of cell lines we interrogated 85% of common genetic variants in the population for potential regulatory effect and we identified 237 AS-SNPs associated to immune GWAS traits and 714 to gene expression in B cells. To elucidate possible regulatory mechanisms we integrated long-range 3D interactions data to identify putative target genes and motif predictions to identify TFs whose binding may be affected by AS-SNPs yielding a collection of 173 AS-SNPs associated to gene expression and 60 to B cell related traits. We present a systems strategy to find functional gene regulatory variants, the TFs that bind differentially between alleles and novel strategies to detect the regulated genes.
Collapse
Affiliation(s)
- Marco Cavalli
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nicholas Baltzer
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden
| | - Husen M Umer
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden
| | - Jan Grau
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ioana Lemnian
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Gang Pan
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ola Wallerman
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rapolas Spalinskas
- Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Pelin Sahlén
- Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ivo Grosse
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany.,German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Jan Komorowski
- Department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, Uppsala, Sweden.,Institute of Computer Science, Polish Academy of Sciences, Warszawa, Poland
| | - Claes Wadelius
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
17
|
Yu YP, Liu P, Nelson J, Hamilton RL, Bhargava R, Michalopoulos G, Chen Q, Zhang J, Ma D, Pennathur A, Luketich J, Nalesnik M, Tseng G, Luo JH. Identification of recurrent fusion genes across multiple cancer types. Sci Rep 2019; 9:1074. [PMID: 30705370 PMCID: PMC6355770 DOI: 10.1038/s41598-019-38550-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/27/2018] [Indexed: 01/21/2023] Open
Abstract
Chromosome changes are one of the hallmarks of human malignancies. Chromosomal rearrangement is frequent in human cancers. One of the consequences of chromosomal rearrangement is gene fusions in the cancer genome. We have previously identified a panel of fusion genes in aggressive prostate cancers. In this study, we showed that 6 of these fusion genes are present in 7 different types of human malignancies with variable frequencies. Among them, the CCNH-C5orf30 and TRMT11-GRIK2 gene fusions were found in breast cancer, colon cancer, non-small cell lung cancer, esophageal adenocarcinoma, glioblastoma multiforme, ovarian cancer and liver cancer, with frequencies ranging from 12.9% to 85%. In contrast, four other gene fusions (mTOR-TP53BP1, TMEM135-CCDC67, KDM4-AC011523.2 and LRRC59-FLJ60017) are less frequent. Both TRMT11-GRIK2 and CCNH-C5orf30 are also frequently present in lymph node metastatic cancer samples from the breast, colon and ovary. Thus, detecting these fusion transcripts may have significant biological and clinical implications in cancer patient management.
Collapse
Affiliation(s)
- Yan-Ping Yu
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Peng Liu
- Departments of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Joel Nelson
- Departments of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Ronald L Hamilton
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Rohit Bhargava
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - George Michalopoulos
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Qi Chen
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas, Kansas City, KS, 66160, USA
| | - Jun Zhang
- Department of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Deqin Ma
- Department of Pathology, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Arjun Pennathur
- Departments of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - James Luketich
- Departments of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Michael Nalesnik
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - George Tseng
- Departments of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Jian-Hua Luo
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
18
|
Dorris ER, Tazzyman SJ, Moylett J, Ramamoorthi N, Hackney J, Townsend M, Muthana M, Lewis MJ, Pitzalis C, Wilson AG. The Autoimmune Susceptibility Gene C5orf30 Regulates Macrophage-Mediated Resolution of Inflammation. THE JOURNAL OF IMMUNOLOGY 2019; 202:1069-1078. [PMID: 30659109 DOI: 10.4049/jimmunol.1801155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Genetic variants in C5orf30 have been associated with development of the autoimmune conditions primary biliary cirrhosis and rheumatoid arthritis. In rheumatoid arthritis, C5orf30 expression is cell-specific, with highest expression found in macrophages and synovial fibroblasts. C5orf30 is highly expressed in inflamed joints and is a negative regulator of tissue damage in a mouse model of inflammatory arthritis. Transcriptomic analysis from ultrasound-guided synovial biopsy of inflamed joints in a well characterized clinical cohort of newly diagnosed, disease-modifying antirheumatic drugs-naive rheumatoid arthritis patients was used to determine the clinical association of C5orf30 expression with disease activity. A combined molecular and computational biology approach was used to elucidate C5orf30 function in macrophages both in vitro and in vivo. Synovial expression of C5orf30 is inversely correlated with both clinical measures of rheumatoid arthritis disease activity and with synovial TNF mRNA expression. C5orf30 plays a role in regulating macrophage phenotype and is differentially turned over in inflammatory and anti-inflammatory macrophages. Inhibition of C5orf30 reduces wound healing/repair-associated functions of macrophages, reduces signaling required for resolution of inflammation, and decreases secretion of anti-inflammatory mediators. In an animal model of wound healing (zebrafish), C5orf30 inhibition increases the recruitment of macrophages to the wound site. Finally, we demonstrate that C5orf30 skews macrophage immunometabolism, demonstrating a mechanism for C5orf30-mediated immune regulation.
Collapse
Affiliation(s)
- Emma R Dorris
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland;
| | | | - John Moylett
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland
| | - Nandhini Ramamoorthi
- Biomarker Discovery OMNI, Genentech Research and Early Development, San Francisco, CA 94080; and
| | - Jason Hackney
- Biomarker Discovery OMNI, Genentech Research and Early Development, San Francisco, CA 94080; and
| | - Michael Townsend
- Biomarker Discovery OMNI, Genentech Research and Early Development, San Francisco, CA 94080; and
| | | | - Myles J Lewis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry and Barts Health National Health Service Trust, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry and Barts Health National Health Service Trust, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Anthony G Wilson
- University College Dublin Centre for Arthritis Research, Conway Institute, University College Dublin, Dublin D04 W6F6, Ireland
| |
Collapse
|
19
|
Sun HN, Luo YH, Meng LQ, Piao XJ, Wang Y, Wang JR, Wang H, Zhang Y, Li JQ, Xu WT, Liu Y, Zhang Y, Zhang T, Han YH, Jin MH, Shen GN, Zang YQ, Cao LK, Zhang DJ, Jin CH. Cryptotanshinone induces reactive oxygen species‑mediated apoptosis in human rheumatoid arthritis fibroblast‑like synoviocytes. Int J Mol Med 2018; 43:1067-1075. [PMID: 30535477 DOI: 10.3892/ijmm.2018.4012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/16/2018] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the mechanisms of apoptosis induced by cryptotanshinone (CT) in human rheumatoid arthritis fibroblast‑like synoviocytes (RA‑FLSs). Cell Counting kit‑8 assay was performed to determine the cytotoxic effects of CT in human RA‑FLSs, including primary RA‑FLS, HFLS‑RA and MH7A cells, and in HFLS cells derived from normal synovial tissue. Annexin V‑FITC/PI staining was used to detect the apoptotic effects of CT in HFLS‑RA and MH7A cells. Flow cytometry was performed to detect the apoptotic and reactive oxygen species (ROS) levels induced by CT in HFLS‑RA cells. Western blotting was used to assess the expression levels of proteins associated with apoptosis and with the mitogen‑activated protein kinase (MAPK), protein kinase B (Akt), and signal transducer and activator of transcription‑3 (STAT3) signaling pathways. The results demonstrated that CT treatment significantly suppressed HFLS‑RA and MH7A cell growth, whereas no clear inhibitory effect was observed in normal HFLS cells. CT exposure downregulated the expression levels of B‑cell lymphoma 2 (Bcl‑2), p‑Akt, p‑extracellular signal‑related kinase and p‑STAT3, while it upregulated the expression levels of Bcl‑2‑associated death promoter (Bad), caspase‑3, poly (ADP‑ribose) polymerase (PARP), p‑p38 and p‑c‑Jun N‑terminal kinase. Following ROS scavenging, the CT‑induced apoptosis and altered expression levels of Bcl‑2, Bad, cleaved caspase‑3 and cleaved PARP were restored. Furthermore, the Akt, MAPK and STAT3 signaling pathways were regulated by intracellular ROS. These results suggest that ROS‑mediated Akt, MAPK and STAT3 signaling pathways serve important roles in the CT‑induced apoptosis of RA‑FLSs.
Collapse
Affiliation(s)
- Hu-Nan Sun
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ling-Qi Meng
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163316, P.R. China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hao Han
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Mei-Hua Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yan-Qing Zang
- Department of Food Science and Engineering, College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Long-Kui Cao
- Department of Food Science and Engineering, College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Dong-Jie Zhang
- Department of Food Science and Engineering, College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
20
|
Chaudhry M, Wilson AG. The role of genetic analysis for predicting outcome of rheumatoid arthritis. Expert Rev Mol Diagn 2017; 17:809-814. [PMID: 28707487 DOI: 10.1080/14737159.2017.1355732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Rheumatoid Arthritis (RA) varies from a mild to a severe, unremitting illness characterized by uncontrolled inflammation with consequent damage to cartilage and bone of joints. Individualized therapeutic approaches based on likely outcome would facilitate a personalized therapeutic approach. Areas covered: Genetics is known to contribute a significant component of the variability in RA outcome, estimated at 45-60%. A number of candidate gene studies have been associated with variability in radiologically assessed joint damage; however a more comprehensive genome wide analysis is required to more fully characterize the genetic basis of RA severity. Expert commentary: Genetic profiling of patient presenting with RA has the potential to aid stratification based on predicted prognosis, this would inform the clinical development of a personalized therapeutic approach. It will also result in the identification of novel mediators of tissue damage in RA.
Collapse
Affiliation(s)
- Mamoonah Chaudhry
- a School of Medicine , UCD Conway Institute of Biomolecular and Biomedical Research , Dublin , Ireland
| | - Anthony G Wilson
- a School of Medicine , UCD Conway Institute of Biomolecular and Biomedical Research , Dublin , Ireland
| |
Collapse
|
21
|
Viatte S, Barton A. Genetics of rheumatoid arthritis susceptibility, severity, and treatment response. Semin Immunopathol 2017; 39:395-408. [PMID: 28555384 PMCID: PMC5486781 DOI: 10.1007/s00281-017-0630-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/10/2017] [Indexed: 12/18/2022]
Abstract
A decade after the first genome-wide association study in rheumatoid arthritis (RA), a plethora of genetic association studies have been published on RA and its clinical or serological subtypes. We review the major milestones in the study of the genetic architecture of RA susceptibility, severity, and response to treatment. We set the scientific context necessary for non-geneticists to understand the potential clinical applications of human genetics and its significance for a stratified approach to the management of RA in the future.
Collapse
Affiliation(s)
- Sebastien Viatte
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| | - Anne Barton
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Grafton Street, Manchester, M13 9WL, UK
| |
Collapse
|
22
|
Kim K, Bang SY, Lee HS, Bae SC. Update on the genetic architecture of rheumatoid arthritis. Nat Rev Rheumatol 2016; 13:13-24. [PMID: 27811914 DOI: 10.1038/nrrheum.2016.176] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human genetic studies into rheumatoid arthritis (RA) have uncovered more than 100 genetic loci associated with susceptibility to RA and have refined the RA-association model for HLA variants. The majority of RA-risk variants are highly shared across multiple ancestral populations and are located in noncoding elements that might have allele-specific regulatory effects in relevant tissues. Emerging multi-omics data, high-density genotype data and bioinformatic approaches are enabling researchers to use RA-risk variants to identify functionally relevant cell types and biological pathways that are involved in impaired immune processes and disease phenotypes. This Review summarizes reported RA-risk loci and the latest insights from human genetic studies into RA pathogenesis, including how genetic data has helped to identify currently available drugs that could be repurposed for patients with RA and the role of genetics in guiding the development of new drugs.
Collapse
Affiliation(s)
- Kwangwoo Kim
- Department of Biology, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - So-Young Bang
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| |
Collapse
|
23
|
Genetic markers as therapeutic target in rheumatoid arthritis: A game changer in clinical therapy? Rheumatol Int 2016; 36:1601-1607. [PMID: 27638722 DOI: 10.1007/s00296-016-3563-7] [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: 06/06/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory, multi-systemic autoimmune disease unremitted by genetic and environmental factors. The factors are crucial but inadequate in the development of disease; however, these factors can be representative of potential therapeutic targets and response to clinical therapy. Insights into the contribution of genetic risk factors are currently in progress with studies querying the genetic variation, their role in gene expression of coding and non-coding genes and other mechanisms of disease. In this review, we describe the significance of genetic markers architecture of RA through genome-wide association studies and meta-analysis studies. Further, it also reveals the mechanism of disease pathogenesis investigated through the mutual findings of functional and genetic studies of individual RA-associated genes, which includes HLA-DRB1, HLA-DQB1, HLA-DPB1, PADI4, PTPN22, TRAF1-C5, STAT4 and C5orf30. However, the genetic background of RA remains to be clearly depicted. Prospective efforts of the post-genomic and functional genomic period can travel toward real possible assessment of the genetic effect on RA. The discovery of novel genes associated with the disease can be appropriate in identifying potential biomarkers, which could assist in early diagnosis and aggressive treatment.
Collapse
|
24
|
Peloquin JM, Goel G, Kong L, Huang H, Haritunians T, Sartor RB, Daly MJ, Newberry RD, McGovern DP, Yajnik V, Lira SA, Xavier RJ. Characterization of candidate genes in inflammatory bowel disease-associated risk loci. JCI Insight 2016; 1:e87899. [PMID: 27668286 DOI: 10.1172/jci.insight.87899] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
GWAS have linked SNPs to risk of inflammatory bowel disease (IBD), but a systematic characterization of disease-associated genes has been lacking. Prior studies utilized microarrays that did not capture many genes encoded within risk loci or defined expression quantitative trait loci (eQTLs) using peripheral blood, which is not the target tissue in IBD. To address these gaps, we sought to characterize the expression of IBD-associated risk genes in disease-relevant tissues and in the setting of active IBD. Terminal ileal (TI) and colonic mucosal tissues were obtained from patients with Crohn's disease or ulcerative colitis and from healthy controls. We developed a NanoString code set to profile 678 genes within IBD risk loci. A subset of patients and controls were genotyped for IBD-associated risk SNPs. Analyses included differential expression and variance analysis, weighted gene coexpression network analysis, and eQTL analysis. We identified 116 genes that discriminate between healthy TI and colon samples and uncovered patterns in variance of gene expression that highlight heterogeneity of disease. We identified 107 coexpressed gene pairs for which transcriptional regulation is either conserved or reversed in an inflammation-independent or -dependent manner. We demonstrate that on average approximately 60% of disease-associated genes are differentially expressed in inflamed tissue. Last, we identified eQTLs with either genotype-only effects on expression or an interaction effect between genotype and inflammation. Our data reinforce tissue specificity of expression in disease-associated candidate genes, highlight genes and gene pairs that are regulated in disease-relevant tissue and inflammation, and provide a foundation to advance the understanding of IBD pathogenesis.
Collapse
Affiliation(s)
- Joanna M Peloquin
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology
| | - Gautam Goel
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology
| | - Lingjia Kong
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - R Balfour Sartor
- Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dermot P McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vijay Yajnik
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease
| | - Sergio A Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease.,Center for Computational and Integrative Biology.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
25
|
Onuora S. Rheumatoid arthritis: C5orf30 regulates severity of tissue destruction in RA. Nat Rev Rheumatol 2015; 11:622. [PMID: 26439407 DOI: 10.1038/nrrheum.2015.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|