1
|
Prajjwal P, Marsool MDM, Yadav V, Kanagala RSD, Reddy YB, John J, Lam JR, Karra N, Amiri B, Islam MU, Nithya V, Marsool ADM, Gadam S, Vora N, Hussin OA. Neurological, cardiac, musculoskeletal, and renal manifestations of scleroderma along with insights into its genetics, pathophysiology, diagnostic, and therapeutic updates. Health Sci Rep 2024; 7:e2072. [PMID: 38660003 PMCID: PMC11040569 DOI: 10.1002/hsr2.2072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Background Scleroderma, also referred to as systemic sclerosis, is a multifaceted autoimmune condition characterized by abnormal fibrosis and impaired vascular function. Pathologically, it encompasses the persistent presence of inflammation, abnormal collagen buildup, and restructuring of blood vessels in various organs, resulting in a wide range of clinical symptoms. This review incorporates the most recent scientific literature on scleroderma, with a particular emphasis on its pathophysiology, clinical manifestations, diagnostic approaches, and treatment options. Methodology A comprehensive investigation was carried out on numerous databases, such as PubMed, MEDLINE, Scopus, Web of Science, and Google Scholar, to collect pertinent studies covering diverse facets of scleroderma research. Results Scleroderma presents with a range of systemic manifestations, such as interstitial lung disease, gastrointestinal dysmotility, Raynaud's phenomenon, pulmonary arterial hypertension, renal complications, neurological symptoms, and cardiac abnormalities. Serological markers, such as antinuclear antibodies, anti-centromere antibodies, and anti-topoisomerase antibodies, are important for classifying diseases and predicting their outcomes. Discussion The precise identification of scleroderma is crucial for promptly and correctly implementing effective treatment plans. Treatment approaches aim to improve symptoms, reduce complications, and slow down the progression of the disease. An integrated approach that combines pharmacological agents, including immunosuppressants, endothelin receptor antagonists, and prostanoids, with nonpharmacological interventions such as physical and occupational therapy is essential for maximizing patient care. Conclusion Through the clarification of existing gaps in knowledge and identification of emerging trends, our goal is to improve the accuracy of diagnosis, enhance the effectiveness of therapeutic interventions, and ultimately enhance the overall quality of life for individuals suffering from scleroderma. Ongoing cooperation and creative research are necessary to advance the field and achieve improved patient outcomes and new therapeutic discoveries.
Collapse
Affiliation(s)
| | | | - Vikas Yadav
- Department of Internal MedicinePt. B. D. S. Postgraduate Institute of Medical SciencesRohtakIndia
| | | | | | - Jobby John
- Department of Internal MedicineDr. Somervell Memorial CSI Medical College and HospitalNeyyāttinkaraIndia
| | - Justin Riley Lam
- Department of Internal MedicineCebu Institute of MedicineCebuPhilippines
| | - Nanditha Karra
- Department of Internal MedicineOsmania Medical CollegeHyderabadTelanganaIndia
| | - Bita Amiri
- Cardiovascular Research CenterTabriz University of Medical SciencesTabrizIran
| | - Moiz Ul Islam
- Department of Internal MedicinePunjab Medical CollegeFaisalabadPakistan
| | - Venkatesh Nithya
- Department of Internal MedicineS. D. Asfendiyarov Kazakh National Medical UniversityAlmatyKazakhstan
| | | | | | | | - Omniat Amir Hussin
- Department of MedicineAlmanhal University Academy of ScienceKhartoumSudan
| |
Collapse
|
2
|
Sabet P, Karimi S, Dehghan A, Bijani M. Effect of Spirituality-Based Palliative Care on Pain, Nausea, Vomiting, and the Quality of Life in Women with Colon Cancer: A Clinical Trial in Southern Iran. JOURNAL OF RELIGION AND HEALTH 2023; 62:1985-1997. [PMID: 36809520 DOI: 10.1007/s10943-023-01742-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 06/18/2023]
Abstract
This randomized controlled clinical trial aimed to examine the effect of spirituality-based palliative care on pain, nausea, vomiting, and the quality of life in 80 Iranian colon cancer inpatients from January to June 2020 in southern Iran. Patients were randomly assigned to an intervention group and a control group. The intervention group took part in four 120-min sessions while the control group received standard care. Pain, nausea, vomiting, and quality of life were assessed before the intervention and one month after the intervention. Data were analyzed using paired t-test and independent t-test. Between-groups differences analysis showed a significant difference in the quality of life scores, pain score, as well as nausea and vomiting scores following the one-month intervention. In conclusion, this group spirituality-based palliative care intervention might be beneficial in improving quality of life and reducing symptoms.
Collapse
Affiliation(s)
- Parisa Sabet
- Department of Medical Surgical Nursing, School of Nursing, Fasa University of Medical Sciences, Fasa, Iran
| | - Shahnaz Karimi
- Department of Medical Education, Medical Education Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| | - Azizallah Dehghan
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mostafa Bijani
- Department of Medical Surgical Nursing, School of Nursing, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
3
|
Műzes G, Sipos F. Autoimmunity and Carcinogenesis: Their Relationship under the Umbrella of Autophagy. Biomedicines 2023; 11:biomedicines11041130. [PMID: 37189748 DOI: 10.3390/biomedicines11041130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system and autophagy share a functional relationship. Both innate and adaptive immune responses involve autophagy and, depending on the disease’s origin and pathophysiology, it may have a detrimental or positive role on autoimmune disorders. As a “double-edged sword” in tumors, autophagy can either facilitate or impede tumor growth. The autophagy regulatory network that influences tumor progression and treatment resistance is dependent on cell and tissue types and tumor stages. The connection between autoimmunity and carcinogenesis has not been sufficiently explored in past studies. As a crucial mechanism between the two phenomena, autophagy may play a substantial role, though the specifics remain unclear. Several autophagy modifiers have demonstrated beneficial effects in models of autoimmune disease, emphasizing their therapeutic potential as treatments for autoimmune disorders. The function of autophagy in the tumor microenvironment and immune cells is the subject of intensive study. The objective of this review is to investigate the role of autophagy in the simultaneous genesis of autoimmunity and malignancy, shedding light on both sides of the issue. We believe our work will assist in the organization of current understanding in the field and promote additional research on this urgent and crucial topic.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
4
|
Maritati F, Provenzano M, Lerario S, Corradetti V, Bini C, Busutti M, Grandinetti V, Cuna V, La Manna G, Comai G. Kidney transplantation in systemic sclerosis: Advances in graft, disease, and patient outcome. Front Immunol 2022; 13:878736. [PMID: 35958558 PMCID: PMC9360313 DOI: 10.3389/fimmu.2022.878736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Systemic sclerosis (SSc) is an immune-mediated rheumatic disease characterized by vascular abnormalities, tissue fibrosis, and inflammation. Renal disease occurring in patients with SSc may have a variable clinicopathological picture. However, the most specific renal condition associated with this disease is the scleroderma renal crisis (SRC), characterized by acute onset of renal failure and severe hypertension. SRC develops in about 20% of cases of SSc, especially in those patients with diffuse cutaneous disease. The prognosis of this condition is often negative, with a rapid progression to end-stage renal disease (ESRD). The advent of the antihypertensive angiotensin-converting enzyme inhibitors in 1980 was associated with a significant improvement in patients’ survival and recovery of renal function. However, the prognosis of these patients can still be improved. The dialytic condition is associated with early death, and mortality is significantly higher than among patients undergoing renal replacement therapy (RRT) due to other conditions. Patients with SRC who show no signs of renal functional recovery despite timely blood pressure control are candidates for kidney transplantation (KT). In this review, we reported the most recent advances in KT in patients with ESRD due to SSc, with a particular overview of the risk of disease recurrence after transplantation and the evolution of other disease manifestations.
Collapse
|
5
|
Kowalska-Kępczyńska A. Systemic Scleroderma-Definition, Clinical Picture and Laboratory Diagnostics. J Clin Med 2022; 11:2299. [PMID: 35566425 PMCID: PMC9100749 DOI: 10.3390/jcm11092299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Scleroderma (Sc) is a rare connective tissue disease classified as an autoimmune disorder. The pathogenesis of this disease is not fully understood. (2) Methods: This article reviews the literature on systemic scleroderma (SSc). A review of available scientific articles was conducted using the PubMed database with a time range of January 1985 to December 2021. (3) Results and Conclusions: The article is a review of information on epidemiology, criteria for diagnosis, pathogenesis, a variety of clinical pictures and the possibility of laboratory diagnostic in the diagnosis and monitoring of systemic scleroderma.
Collapse
Affiliation(s)
- Anna Kowalska-Kępczyńska
- Department of Biochemical Diagnostics, Chair of Laboratory Diagnostics, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
6
|
Huang B, Li J, Zhao J. Screening and identification of potential biomarkers and therapeutic targets for systemic sclerosis-associated interstitial lung disease. Arch Rheumatol 2022; 36:548-559. [PMID: 35382367 PMCID: PMC8957772 DOI: 10.46497/archrheumatol.2021.8625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/18/2021] [Indexed: 11/03/2022] Open
Abstract
Objectives This study aims to analyze gene expression in lung tissue and lung fibroblasts of patients with systemic sclerosis-associated interstitial lung disease (SSc-ILD) to identify potential biomarkers and therapeutic targets and to examine its possible role in the pathogenesis of SSc-ILD. Patients and methods We obtained datasets from Gene Expression Omnibus (GEO) database, and used Robust Rank Aggregation to calculate the co-expressed differentially-expressed-genes (DEGs) in three chips, then analyzed the function, signaling pathways and the protein-protein interaction network of the DEGs. Finally, we verified the DEGs related to SSc-ILD by three databases of Comparative Toxicogenomics Database (CTD), GENE, and DisGeNET, respectively. Results There were 16 co-expressed DEGs related to SSc-ILD in three GEO series, of which six genes were upregulated, and 10 genes were downregulated. The CTD included 29,936 genes related to SSc, and the GENE and DisGeNET databases had 429 genes related to SSc. Conclusion The results of gene differential expression analysis suggest that interleukin-6, chemokine ligand 2, intercellular adhesion molecule 1, tumor necrosis factor alpha-induced protein 3, pentraxin 3, and cartilage oligomeric matrix protein may be implicated in the pathogenesis of SSc-ILD and are expected to be potential biomarkers and therapeutic targets for SSc-ILD.
Collapse
Affiliation(s)
- Biqing Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
7
|
Shen CY, Lu CH, Wu CH, Li KJ, Kuo YM, Hsieh SC, Yu CL. Molecular Basis of Accelerated Aging with Immune Dysfunction-Mediated Inflammation (Inflamm-Aging) in Patients with Systemic Sclerosis. Cells 2021; 10:cells10123402. [PMID: 34943909 PMCID: PMC8699891 DOI: 10.3390/cells10123402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic connective tissue disorder characterized by immune dysregulation, chronic inflammation, vascular endothelial cell dysfunction, and progressive tissue fibrosis of the skin and internal organs. Moreover, increased cancer incidence and accelerated aging are also found. The increased cancer incidence is believed to be a result of chromosome instability. Accelerated cellular senescence has been confirmed by the shortening of telomere length due to increased DNA breakage, abnormal DNA repair response, and telomerase deficiency mediated by enhanced oxidative/nitrative stresses. The immune dysfunctions of SSc patients are manifested by excessive production of proinflammatory cytokines IL-1, IL-6, IL-17, IFN-α, and TNF-α, which can elicit potent tissue inflammation followed by tissue fibrosis. Furthermore, a number of autoantibodies including anti-topoisomerase 1 (anti-TOPO-1), anti-centromere (ACA or anti-CENP-B), anti-RNA polymerase enzyme (anti-RNAP III), anti-ribonuclear proteins (anti-U1, U2, and U11/U12 RNP), anti-nucleolar antigens (anti-Th/T0, anti-NOR90, anti-Ku, anti-RuvBL1/2, and anti-PM/Scl), and anti-telomere-associated proteins were also found. Based on these data, inflamm-aging caused by immune dysfunction-mediated inflammation exists in patients with SSc. Hence, increased cellular senescence is elicited by the interactions among excessive oxidative stress, pro-inflammatory cytokines, and autoantibodies. In the present review, we will discuss in detail the molecular basis of chromosome instability, increased oxidative stress, and functional adaptation by deranged immunome, which are related to inflamm-aging in patients with SSc.
Collapse
Affiliation(s)
- Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan
| | - Cheng-Hsun Lu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
- Correspondence: (S.-C.H.); (C.-L.Y.); Tel.: +886-2-23123456 (S.-C.H. & C.-L.Y.)
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-H.W.); (K.-J.L.); (Y.-M.K.)
- Correspondence: (S.-C.H.); (C.-L.Y.); Tel.: +886-2-23123456 (S.-C.H. & C.-L.Y.)
| |
Collapse
|
8
|
Hinchcliff M, Garcia-Milian R, Di Donato S, Dill K, Bundschuh E, Galdo FD. Cellular and Molecular Diversity in Scleroderma. Semin Immunol 2021; 58:101648. [PMID: 35940960 DOI: 10.1016/j.smim.2022.101648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the increasing armamentarium of high-throughput tools available at manageable cost, it is attractive and informative to determine the molecular underpinnings of patient heterogeneity in systemic sclerosis (SSc). Given the highly variable clinical outcomes of patients labelled with the same diagnosis, unravelling the cellular and molecular basis of disease heterogeneity will be crucial to predicting disease risk, stratifying management and ultimately informing a patient-centered precision medicine approach. Herein, we summarise the findings of the past several years in the fields of genomics, transcriptomics, and proteomics that contribute to unraveling the cellular and molecular heterogeneity of SSc. Expansion of these findings and their routine integration with quantitative analysis of histopathology and imaging studies into clinical care promise to inform a scientifically driven patient-centred personalized medicine approach to SSc in the near future.
Collapse
Affiliation(s)
- Monique Hinchcliff
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA.
| | | | - Stefano Di Donato
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK
| | | | - Elizabeth Bundschuh
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA
| | - Francesco Del Galdo
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK.
| |
Collapse
|
9
|
Expression of macrophage migration inhibitory factor and its receptor CD74 in systemic sclerosis. Cent Eur J Immunol 2021; 46:375-383. [PMID: 34764810 PMCID: PMC8574103 DOI: 10.5114/ceji.2021.109756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 05/10/2021] [Indexed: 11/24/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) has been associated with the
pathogenesis of several rheumatic diseases. In systemic sclerosis (SSc) it has
been shown that MIF expression is dysregulated in serum and skin. However, the
MIF receptor, CD74, has been poorly investigated and its potential role in the
pathogenesis of SSc remains unknown. This study aimed to analyze mRNA, tissue,
and serum expression of MIF and CD74 in patients with limited (lcSSc) and
diffuse (dcSSc) systemic sclerosis. A case-control study in 20 SSc patients and
20 control subjects (CS) from southern México was conducted. MIF and CD74
mRNA expression levels were quantified by real-time PCR, MIF serum levels were
measured by an ELISA kit, and MIF and its receptor CD74 were evaluated by
immunohistochemistry of skin biopsies. MIF mRNA expression was significantly
higher in CS than in SSc patients (p = 0.02), while CD74 showed no differences
between patients and CS. MIF serum levels were similar between SSc patients and
CS: dcSSc = 3.82 ng/ml, lcSSc = 3.57 ng/ml, and CS = 3.28 ng/ml. In skin
biopsies of SSc, MIF and CD74 were enhanced in keratinocytes, while they showed
decreased expression in endothelial cells. On the other hand, the staining of
CD74 was high in fibroblasts of dcSSc patients. Our findings show MIF and CD74
deregulation at the transcriptional and translational levels in SSc, which might
be associated with the proinflammatory process leading to tissue remodeling and
excessive fibrosis in SSc.
Collapse
|
10
|
Nakano M, Ayano M, Kushimoto K, Kawano S, Higashioka K, Inokuchi S, Mitoma H, Kimoto Y, Akahoshi M, Ono N, Arinobu Y, Akashi K, Horiuchi T, Niiro H. Association of elevated serum soluble CD226 levels with the disease activity and flares of systemic lupus erythematosus. Sci Rep 2021; 11:16162. [PMID: 34373559 PMCID: PMC8352936 DOI: 10.1038/s41598-021-95711-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/23/2021] [Indexed: 11/09/2022] Open
Abstract
CD226 is an activating receptor expressed on the cell surface of natural killer cells and T cells. Although CD226 polymorphism is known to be involved in systemic lupus erythematosus (SLE), the involvement of soluble CD226 (sCD226) in SLE is still unknown. In the present study, we measured serum sCD226 levels using an enzyme-linked immunosorbent assay in 58 SLE patients and 33 healthy controls (HCs) and evaluated their associations with SLE Disease Activity Index 2000 (SLEDAI-2K), clinical manifestations, laboratory data, and the cumulative probability of flare. Serum sCD226 levels showed no significant differences between SLE patients and HCs. However, sCD226 levels were significantly elevated in active SLE patients with a SLEDAI-2K score of ≥ 20 compared with HCs. In SLE patients, sCD226 levels were significantly correlated with SLEDAI-2K scores and anti-dsDNA antibody titers. Moreover, the cumulative probability of flare was markedly higher in patients with high sCD226 than in those with low sCD226. In patients with neuropsychiatric involvement, sCD226 levels were elevated and reflected neuropsychiatric disease activity. These findings indicate that serum sCD226 levels are associated with disease activity and flares of SLE. Thus, it may be a useful biomarker for SLE, and its monitoring allows for more precise SLE management.
Collapse
Affiliation(s)
- Miki Nakano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masahiro Ayano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Department of Cancer Stem Cell Research, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Kazuo Kushimoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shotaro Kawano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazuhiko Higashioka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shoichiro Inokuchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasutaka Kimoto
- Department of Internal Medicine, Kyushu University Beppu Hospital, 4546 Tsurumibaru, Tsurumi, Beppu, 874-0838, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nobuyuki Ono
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, 4546 Tsurumibaru, Tsurumi, Beppu, 874-0838, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
11
|
Xu X, Ramanujam M, Visvanathan S, Assassi S, Liu Z, Li L. Transcriptional insights into pathogenesis of cutaneous systemic sclerosis using pathway driven meta-analysis assisted by machine learning methods. PLoS One 2020; 15:e0242863. [PMID: 33253326 PMCID: PMC7703909 DOI: 10.1371/journal.pone.0242863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/10/2020] [Indexed: 11/19/2022] Open
Abstract
Pathophysiology of systemic sclerosis (SSc, Scleroderma), an autoimmune rheumatic disease, comprises of mechanisms that drive vasculopathy, inflammation and fibrosis. Understanding of the disease and associated clinical heterogeneity has advanced considerably in the past decade, highlighting the necessity of more specific targeted therapy. While many of the recent trials in SSc failed to meet the primary end points that predominantly relied on changes in modified Rodnan skin scores (MRSS), sub-group analysis, especially those focused on the basal skin transcriptomic data have provided insights into patient subsets that respond to therapies. These findings suggest that deeper understanding of the molecular changes in pathways is very important to define disease drivers in various patient subgroups. In view of these challenges, we performed meta-analysis on 9 public available SSc microarray studies using a novel pathway pivoted approach combining consensus clustering and machine learning assisted feature selection. Selected pathway modules were further explored through cluster specific topological network analysis in search of novel therapeutic concepts. In addition, we went beyond previously described SSc class divisions of 3 clusters (e.g. inflammation, fibro-proliferative, normal-like) and expanded into a much finer stratification in order to profile SSc patients more accurately. Our analysis unveiled an important 80 pathway signatures that differentiated SSc patients into 8 unique subtypes. The 5 pathway modules derived from such signature successfully defined the 8 SSc subsets and were validated by in-silico cellular deconvolution analysis. Myeloid cells and fibroblasts involvement in different clusters were confirmed and linked to corresponding pathway activities. Collectively, our findings revealed more complex disease subtypes in SSc; Key gene mediators such as IL6, FGFR1, TLR7, PLCG2, IRK2 identified by network analysis underscored the scientific rationale for exploring additional targets in treatment of SSc.
Collapse
Affiliation(s)
- Xiao Xu
- Computational Biology, Boehringer-Ingelheim Pharmaceuticals Inc, Ridgefield, CT, United States of America
| | - Meera Ramanujam
- Immunology and Respiratory Diseases Research, Boehringer-Ingelheim Pharmaceuticals Inc, Ridgefield, CT, United States of America
| | - Sudha Visvanathan
- Translational Medicine and Clinical Pharmacology, Boehringer-Ingelheim Pharmaceuticals Inc, Ridgefield, CT, United States of America
| | - Shervin Assassi
- Division of Rheumatology, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Zheng Liu
- Computational Biology, Boehringer-Ingelheim Pharmaceuticals Inc, Ridgefield, CT, United States of America
| | - Li Li
- Computational Biology, Boehringer-Ingelheim Pharmaceuticals Inc, Ridgefield, CT, United States of America
- * E-mail:
| |
Collapse
|
12
|
Cheikhi AM, Johnson ZI, Julian DR, Wheeler S, Feghali-Bostwick C, Conley YP, Lyons-Weiler J, Yates CC. Prediction of severity and subtype of fibrosing disease using model informed by inflammation and extracellular matrix gene index. PLoS One 2020; 15:e0240986. [PMID: 33095822 PMCID: PMC7584227 DOI: 10.1371/journal.pone.0240986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/06/2020] [Indexed: 11/19/2022] Open
Abstract
Fibrosis is a chronic disease with heterogeneous clinical presentation, rate of progression, and occurrence of comorbidities. Systemic sclerosis (scleroderma, SSc) is a rare rheumatic autoimmune disease that encompasses several aspects of fibrosis, including highly variable fibrotic manifestation and rate of progression. The development of effective treatments is limited by these variabilities. The fibrotic response is characterized by both chronic inflammation and extracellular remodeling. Therefore, there is a need for improved understanding of which inflammation-related genes contribute to the ongoing turnover of extracellular matrix that accompanies disease. We have developed a multi-tiered method using Naïve Bayes modeling that is capable of predicting level of disease and clinical assessment of patients based on expression of a curated 60-gene panel that profiles inflammation and extracellular matrix production in the fibrotic disease state. Our novel modeling design, incorporating global and parametric-based methods, was highly accurate in distinguishing between severity groups, highlighting the importance of these genes in disease. We refined this gene set to a 12-gene index that can accurately identify SSc patient disease state subsets and informs knowledge of the central regulatory pathways in disease progression.
Collapse
Affiliation(s)
- Amin M. Cheikhi
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - Zariel I. Johnson
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - Dana R. Julian
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, PA, United States of America
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Carol Feghali-Bostwick
- Department of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Yvette P. Conley
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - James Lyons-Weiler
- Genomic and Proteomic Core Laboratories, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Cecelia C. Yates
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, PA, United States of America
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
13
|
Li L, Zuo X, Liu D, Luo H, Zhu H. The profiles of miRNAs and lncRNAs in peripheral blood neutrophils exosomes of diffuse cutaneous systemic sclerosis. J Dermatol Sci 2020; 98:88-97. [PMID: 32147195 DOI: 10.1016/j.jdermsci.2020.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Diffuse cutaneous systemic sclerosis (dSSc) is a systemic autoimmune disease with skin fibrosis. Neutrophils display important roles in autoimmunity, inflammation, vasculopathy and fibrosis. Exosomes (EXOs) are cell-derived vesicles contained various noncoding RNAs, mRNA and proteins with biological roles. OBJECTIVE To investigate the roles of miRNAs and lncRNAs from dSSc neutrophils EXOs. METHODS EXOs were isolated from cultured neutrophils supernatants and identified by transmission electron microscopy. Global expression of miRNAs and lncRNAs in neutrophils EXOs were sequenced by Illumina HiSeq 3000 and bioinformatic analyses were performed by R/Bioconductor. Genes were validated by real-time quantitative PCR. RESULTS In profiles of neutrophils EXOs, we identified 22 dysregulated miRNAs and 281 dysregulated lncRNAs. Predicted target genes of them were enriched in GO, KEGG and Reactome pathways, Wnt, AMPK, IL-23 and NOTCH signaling pathways were selected for further analysis. Widely interactions among them were also found. Human dermal microvascular endothelial cells and human primary skin fibroblasts were stimulated with dSSc neutrophils EXOs, these fibrosis related genes were detected and some changes were found, such as ENST00000533886.1-hsa-miR-1268a-CAMK2G in Wnt and IL-23 signaling pathways, ENST00000610091.1-hsa-miR-299-3p, 512-3p-CPT1A in IL-23 and AMPK signaling pathways, NR_001564.2, ENST00000520562.1, ENST00000596567.1-hsa-miR-299-3p, 512-3p -TFDP2 in IL-23, AMPK and NOTCH signaling pathways. CONCLUSIONS The profiles of miRNAs and lncRNAs of neutrophils EXOs provided novel clues for dSSc pathogenesis. We identified several gene pairs in the Wnt, AMPK, IL-23 and NOTCH signaling pathways, which could be potential biomarkers and therapeutic targets in dSSc.
Collapse
Affiliation(s)
- Liya Li
- Department of Rheumatology and immunology, Xiangya Hospital of Central South University, Changsha, China; The Institute of Rheumatology and Immunology, Central South University, Changsha, China.
| | - Xiaoxia Zuo
- Department of Rheumatology and immunology, Xiangya Hospital of Central South University, Changsha, China; The Institute of Rheumatology and Immunology, Central South University, Changsha, China.
| | - Di Liu
- Department of Rheumatology and immunology, Xiangya Hospital of Central South University, Changsha, China; The Institute of Rheumatology and Immunology, Central South University, Changsha, China.
| | - Hui Luo
- Department of Rheumatology and immunology, Xiangya Hospital of Central South University, Changsha, China; The Institute of Rheumatology and Immunology, Central South University, Changsha, China.
| | - Honglin Zhu
- Department of Rheumatology and immunology, Xiangya Hospital of Central South University, Changsha, China; The Institute of Rheumatology and Immunology, Central South University, Changsha, China.
| |
Collapse
|
14
|
Sobolewski P, Maślińska M, Wieczorek M, Łagun Z, Malewska A, Roszkiewicz M, Nitskovich R, Szymańska E, Walecka I. Systemic sclerosis - multidisciplinary disease: clinical features and treatment. Reumatologia 2019; 57:221-233. [PMID: 31548749 PMCID: PMC6753596 DOI: 10.5114/reum.2019.87619] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/19/2019] [Indexed: 12/17/2022] Open
Abstract
Systemic sclerosis is a chronic autoimmune disease of still not fully understood pathogenesis. Fibrosis, vascular wall damage, and disturbances of innate and acquired immune responses with autoantibody production are prominent features. Systemic sclerosis has specific subsets with different autoantibodies, and differences in the affected skin areas. The suspicion of systemic sclerosis and establishing the diagnosis will be facilitated by the criteria created by EULAR/ACR experts. The treatment of this autoimmune disease remains a challenge for clinicians and new therapeutic options are constantly sought. The occurrence of various symptoms and the involvement of many organs and systems make systemic sclerosis a multidisciplinary disease and require a holistic approach. The present article summarizes different clinical features of systemic sclerosis and the profile of autoantibodies and discusses recent rules and future perspectives in disease management.
Collapse
Affiliation(s)
- Piotr Sobolewski
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | - Maria Maślińska
- Clinic of Early Arthritis, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Marta Wieczorek
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | - Zuzanna Łagun
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | - Aleksandra Malewska
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | - Marek Roszkiewicz
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | | | - Elżbieta Szymańska
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | - Irena Walecka
- Clinic of Dermatology, Centre of Postgraduate Medical Education, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| |
Collapse
|
15
|
Wang Q, Shi G, Zhang Y, Lu F, Xie D, Wen C, Huang L. Deciphering the Potential Pharmaceutical Mechanism of GUI-ZHI-FU-LING-WAN on Systemic Sclerosis based on Systems Biology Approaches. Sci Rep 2019; 9:355. [PMID: 30674993 PMCID: PMC6344516 DOI: 10.1038/s41598-018-36314-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
Systemic sclerosis (SSc; scleroderma) is a complicated idiopathic connective tissue disease with seldom effective treatment. GUI-ZHI-FU-LING-WAN (GFW) is a classic Traditional Chinese Medicine (TCM) formula widely used for the treatment of SSc. However, the mechanism of how the GFW affects SSc remains unclear. In this study, the system biology approach was utilized to analyze herb compounds and related targets to get the general information of GFW. The KEGG enrichment analysis of 1645 related targets suggested that the formula is involved in the VEGF signaling pathway, the Toll-like receptor signaling pathway, etc. Quantitative and qualitative analysis of the relationship among the 3 subsets (formula targets, drug targets and disease genes) showed that the formula targets overlapped with 38.0% drug targets and 26.0% proteins encoded by disease genes. Through the analysis of SSc related microarray statistics from the GEO database, we also validated the consistent expression behavior among the 3 subsets before and after treatment. To further reveal the mechanism of prescription, we constructed a network among 3 subsets and decomposed it into 24 modules to decipher how GFW interfere in the progress of SSc. The modules indicated that the intervention may come into effect through following pathogenic processes: vasculopathy, immune dysregulation and tissue fibrosis. Vitro experiments confirmed that GFW could suppress the proliferation of fibroblasts and decrease the Th1 cytokine (TNF-α, MIP-2 and IL-6) expression for lipopolysaccharide (LPS) and bleomycin (BLM) stimulation in macrophages, which is consistent with previous conclusion that GFW is able to relieve SSc. The systems biology approach provides a new insight for deepening understanding about TCM.
Collapse
Affiliation(s)
- Qiao Wang
- TCM Clinical Basis Institute, Zhejiang Chinese Medicine University, 548 Binwen Road, Hangzhou, Zhejiang, 310000, China
| | - Guoshan Shi
- Department of Integrative Traditional & Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Yun Zhang
- TCM Clinical Basis Institute, Zhejiang Chinese Medicine University, 548 Binwen Road, Hangzhou, Zhejiang, 310000, China
| | - Feilong Lu
- TCM Clinical Basis Institute, Zhejiang Chinese Medicine University, 548 Binwen Road, Hangzhou, Zhejiang, 310000, China
| | - Duoli Xie
- TCM Clinical Basis Institute, Zhejiang Chinese Medicine University, 548 Binwen Road, Hangzhou, Zhejiang, 310000, China
| | - Chengping Wen
- TCM Clinical Basis Institute, Zhejiang Chinese Medicine University, 548 Binwen Road, Hangzhou, Zhejiang, 310000, China.
| | - Lin Huang
- TCM Clinical Basis Institute, Zhejiang Chinese Medicine University, 548 Binwen Road, Hangzhou, Zhejiang, 310000, China.
| |
Collapse
|
16
|
Hayden LP, Cho MH, Raby BA, Beaty TH, Silverman EK, Hersh CP. Childhood asthma is associated with COPD and known asthma variants in COPDGene: a genome-wide association study. Respir Res 2018; 19:209. [PMID: 30373671 PMCID: PMC6206739 DOI: 10.1186/s12931-018-0890-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/12/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Childhood asthma is strongly influenced by genetics and is a risk factor for reduced lung function and chronic obstructive pulmonary disease (COPD) in adults. This study investigates self-reported childhood asthma in adult smokers from the COPDGene Study. We hypothesize that childhood asthma is associated with decreased lung function, increased risk for COPD, and that a genome-wide association study (GWAS) will show association with established asthma variants. METHODS We evaluated current and former smokers ages 45-80 of non-Hispanic white (NHW) or African American (AA) race. Childhood asthma was defined by self-report of asthma, diagnosed by a medical professional, with onset at < 16 years or during childhood. Subjects with a history of childhood asthma were compared to those who never had asthma based on lung function, development of COPD, and genetic variation. GWAS was performed in NHW and AA populations, and combined in meta-analysis. Two sets of established asthma SNPs from published literature were examined for association with childhood asthma. RESULTS Among 10,199 adult smokers, 730 (7%) reported childhood asthma and 7493 (73%) reported no history of asthma. Childhood asthmatics had reduced lung function and increased risk for COPD (OR 3.42, 95% CI 2.81-4.18). Genotype data was assessed for 8031 subjects. Among NHWs, 391(7%) had childhood asthma, and GWAS identified one genome-wide significant association in KIAA1958 (rs59289606, p = 4.82 × 10- 8). Among AAs, 339 (12%) had childhood asthma. No SNPs reached genome-wide significance in the AAs or in the meta-analysis combining NHW and AA subjects; however, potential regions of interest were identified. Established asthma SNPs were examined, seven from the NHGRI-EBI database and five with genome-wide significance in the largest pediatric asthma GWAS. Associations were found in the current childhood asthma GWAS with known asthma loci in IL1RL1, IL13, LINC01149, near GSDMB, and in the C11orf30-LRRC32 region (Bonferroni adjusted p < 0.05 for all comparisons). CONCLUSIONS Childhood asthmatics are at increased risk for COPD. Defining asthma by self-report is valid in populations at risk for COPD, identifying subjects with clinical and genetic characteristics known to associate with childhood asthma. This has potential to improve clinical understanding of asthma-COPD overlap (ACO) and enhance future research into ACO-specific treatment regimens. TRIAL REGISTRATION ClinicalTrials.gov, NCT00608764 (Active since January 28, 2008).
Collapse
Affiliation(s)
- Lystra P. Hayden
- Division of Respiratory Diseases, Boston Children’s Hospital, Boston, MA USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Benjamin A. Raby
- Division of Respiratory Diseases, Boston Children’s Hospital, Boston, MA USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Terri H. Beaty
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| |
Collapse
|
17
|
Integration of Genome-Wide DNA Methylation and Transcription Uncovered Aberrant Methylation-Regulated Genes and Pathways in the Peripheral Blood Mononuclear Cells of Systemic Sclerosis. Int J Rheumatol 2018; 2018:7342472. [PMID: 30245726 PMCID: PMC6139224 DOI: 10.1155/2018/7342472] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/16/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
Objective. Systemic sclerosis (SSc) is a systemic connective tissue disease of unknown etiology. Aberrant gene expression and epigenetic modifications in circulating immune cells have been implicated in the pathogenesis of SSc. This study is to delineate the interaction network between gene transcription and DNA methylation in PBMC of SSc patients and to identify methylation-regulated genes which are involved in the pathogenesis of SSc. Methods. Genome-wide mRNA transcription and global DNA methylation analysis were performed on PBMC from 18 SSc patients and 19 matched normal controls (NC) using Illumina BeadChips. Differentially expressed genes (DEGs) and differentially methylated positions (DMPs) were integrative analyzed to identify methylation-regulated genes and associated molecular pathways. Results. Transcriptome analysis distinguished 453 DEGs (269 up- and 184 downregulated) in SSc from NC. Global DNA methylation analysis identified 925 DMPs located on 618 genes. Integration of the two lists revealed only 20 DEGs which harbor inversely correlated DMPs, including 12 upregulated (ELANE, CTSG, LTBR, C3AR1, CSTA, SPI1, ODF3B, SAMD4A, PLAUR, NFE2, ZYX, and CTSZ) and eight downregulated genes (RUNX3, PRF1, PRKCH, PAG1, RASSF5, FYN, CXCR6, and F2R). These potential methylation-regulated DEGs (MeDEGs) are enriched in the pathways related to immune cell migration, proliferation, activation, and inflammation activities. Using a machine learning algorism, we identified six out of the 20 MeDEGs, including F2R, CXCR6, FYN, LTBR, CTSG, and ELANE, which distinguished SSc from NC with 100% accuracy. Four genes (F2R, FYN, PAG1, and PRKCH) differentially expressed in SSc with interstitial lung disease (ILD) compared to SSc without ILD. Conclusion. The identified MeDEGs may represent novel candidate factors which lead to the abnormal activation of immune regulatory pathways in the pathogenesis of SSc. They may also be used as diagnostic biomarkers for SSc and clinical complications.
Collapse
|
18
|
Ingegnoli F, Ughi N, Mihai C. Update on the epidemiology, risk factors, and disease outcomes of systemic sclerosis. Best Pract Res Clin Rheumatol 2018; 32:223-240. [DOI: 10.1016/j.berh.2018.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022]
|
19
|
Langenmayer MC, Jung S, Majzoub-Altweck M, Trefz FM, Seifert C, Knubben-Schweizer G, Fries R, Hermanns W, Gollnick NS. Zinc Deficiency-Like Syndrome in Fleckvieh Calves: Clinical and Pathological Findings and Differentiation from Bovine Hereditary Zinc Deficiency. J Vet Intern Med 2018; 32:853-859. [PMID: 29424482 PMCID: PMC5866964 DOI: 10.1111/jvim.15040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/17/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022] Open
Abstract
Background Zinc deficiency‐like (ZDL) syndrome is an inherited defect of Fleckvieh calves, with striking similarity to bovine hereditary zinc deficiency (BHZD). However, the causative mutation in a phospholipase D4 encoding gene (PLD4) shows no connection to zinc metabolism. Objectives To describe clinical signs, laboratory variables, and pathological findings of ZDL syndrome and their utility to differentiate ZDL from BHZD and infectious diseases with similar phenotype. Animals Nine hospitalized calves with crusting dermatitis and confirmed mutation in PLD4 and medical records from 25 calves with crusting dermatitis or suspected zinc deficiency. Methods Prospective and retrospective case series. Results The 9 calves (age: 5–53 weeks) displayed a moderate to severe crusting dermatitis mainly on the head, ventrum, and joints. Respiratory and digestive tract inflammations were frequently observed. Zinc supplementation did not lead to remission of clinical signs in 4 calves. Laboratory variables revealed slight anemia in 8 calves, hypoalbuminemia in 6 calves, but reduced serum zinc concentrations in only 3 calves. Mucosal erosions/ulcerations were present in 7 calves and thymus atrophy or reduced thymic weights in 8 calves. Histologically, skin lesions were indistinguishable from BHZD. Retrospective analysis of medical records revealed the presence of this phenotype since 1988 and pedigree analysis revealed a common ancestor of several affected calves. Conclusions and Clinical Importance ZDL syndrome should be suspected in Fleckvieh calves with crusting dermatitis together with diarrhea or respiratory tract inflammations without response to oral zinc supplementation. Definite diagnosis requires molecular genetic confirmation of the PLD4 mutation.
Collapse
Affiliation(s)
- M C Langenmayer
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany.,Institute for Infectious Diseases and Zoonoses, LMU Munich, Munich, Germany
| | - S Jung
- Chair of Animal Breeding, Technische Universität München, Freising-Weihenstephan, Germany
| | - M Majzoub-Altweck
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - F M Trefz
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| | - C Seifert
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| | - G Knubben-Schweizer
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| | - R Fries
- Chair of Animal Breeding, Technische Universität München, Freising-Weihenstephan, Germany
| | - W Hermanns
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - N S Gollnick
- Clinic for Ruminants with Ambulatory and Herd Health Services at the Centre for Clinical Veterinary Medicine, LMU Munich, Oberschleißheim, Germany
| |
Collapse
|
20
|
Gorlova OY, Li Y, Gorlov I, Ying J, Chen WV, Assassi S, Reveille JD, Arnett FC, Zhou X, Bossini-Castillo L, Lopez-Isac E, Acosta-Herrera M, Gregersen PK, Lee AT, Steen VD, Fessler BJ, Khanna D, Schiopu E, Silver RM, Molitor JA, Furst DE, Kafaja S, Simms RW, Lafyatis RA, Carreira P, Simeon CP, Castellvi I, Beltran E, Ortego N, Amos CI, Martin J, Mayes MD. Gene-level association analysis of systemic sclerosis: A comparison of African-Americans and White populations. PLoS One 2018; 13:e0189498. [PMID: 29293537 PMCID: PMC5749683 DOI: 10.1371/journal.pone.0189498] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/27/2017] [Indexed: 12/15/2022] Open
Abstract
Gene-level analysis of ImmunoChip or genome-wide association studies (GWAS) data has not been previously reported for systemic sclerosis (SSc, scleroderma). The objective of this study was to analyze genetic susceptibility loci in SSc at the gene level and to determine if the detected associations were shared in African-American and White populations, using data from ImmunoChip and GWAS genotyping studies. The White sample included 1833 cases and 3466 controls (956 cases and 2741 controls from the US and 877 cases and 725 controls from Spain) and the African American sample, 291 cases and 260 controls. In both Whites and African Americans, we performed a gene-level analysis that integrates association statistics in a gene possibly harboring multiple SNPs with weak effect on disease risk, using Versatile Gene-based Association Study (VEGAS) software. The SNP-level analysis was performed using PLINK v.1.07. We identified 4 novel candidate genes (STAT1, FCGR2C, NIPSNAP3B, and SCT) significantly associated and 4 genes (SERBP1, PINX1, TMEM175 and EXOC2) suggestively associated with SSc in the gene level analysis in White patients. As an exploratory analysis we compared the results on Whites with those from African Americans. Of previously established susceptibility genes identified in Whites, only TNFAIP3 was significant at the nominal level (p = 6.13x10-3) in African Americans in the gene-level analysis of the ImmunoChip data. Among the top suggestive novel genes identified in Whites based on the ImmunoChip data, FCGR2C and PINX1 were only nominally significant in African Americans (p = 0.016 and p = 0.028, respectively), while among the top novel genes identified in the gene-level analysis in African Americans, UNC5C (p = 5.57x10-4) and CLEC16A (p = 0.0463) were also nominally significant in Whites. We also present the gene-level analysis of SSc clinical and autoantibody phenotypes among Whites. Our findings need to be validated by independent studies, particularly due to the limited sample size of African Americans.
Collapse
Affiliation(s)
- Olga Y. Gorlova
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States of America
| | - Yafang Li
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States of America
| | - Ivan Gorlov
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States of America
| | - Jun Ying
- Department of Internal Medicine, Division of Rheumatology, University of Texas McGovern Medical School, Houston, TX, United States of America
| | - Wei V. Chen
- Department of Biostatistics, UT MD Anderson Cancer Center, Houston, TX, United States of America
| | - Shervin Assassi
- Department of Internal Medicine, Division of Rheumatology, University of Texas McGovern Medical School, Houston, TX, United States of America
| | - John D. Reveille
- Department of Internal Medicine, Division of Rheumatology, University of Texas McGovern Medical School, Houston, TX, United States of America
| | - Frank C. Arnett
- Department of Internal Medicine, Division of Rheumatology, University of Texas McGovern Medical School, Houston, TX, United States of America
| | - Xiaodong Zhou
- Department of Internal Medicine, Division of Rheumatology, University of Texas McGovern Medical School, Houston, TX, United States of America
| | | | - Elena Lopez-Isac
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, Granada, Spain
| | | | - Peter K. Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, Manhasset, NY, United States of America
| | - Annette T. Lee
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, Manhasset, NY, United States of America
| | - Virginia D. Steen
- Division of Rheumatology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Barri J. Fessler
- Division of Rheumatology, University of Alabama—Birmingham, Birmingham, AL, United States of America
| | - Dinesh Khanna
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, United States of America
| | - Elena Schiopu
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, United States of America
| | - Richard M. Silver
- Division of Rheumatology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Jerry A. Molitor
- Division of Rheumatic and Autoimmune Diseases, University of Minnesota, Minneapolis, MN, United States of America
| | - Daniel E. Furst
- Division of Rheumatology, University of California—Los Angeles, Los Angeles, CA, United States of America
- University of Washington, Seattle, WA, United States of America
- University of Florence, Florence, Italy
| | - Suzanne Kafaja
- Division of Rheumatology, University of California—Los Angeles, Los Angeles, CA, United States of America
| | - Robert W. Simms
- Division of Rheumatology, Boston University, Boston, MA, United States of America
| | | | | | | | | | - Emma Beltran
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Christopher I. Amos
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States of America
| | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, Granada, Spain
| | - Maureen D. Mayes
- Department of Internal Medicine, Division of Rheumatology, University of Texas McGovern Medical School, Houston, TX, United States of America
| |
Collapse
|
21
|
Profiling Celiac Disease-Related Transcriptional Changes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:149-174. [DOI: 10.1016/bs.ircmb.2017.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
22
|
Activation of STAT3 integrates common profibrotic pathways to promote fibroblast activation and tissue fibrosis. Nat Commun 2017; 8:1130. [PMID: 29066712 PMCID: PMC5654983 DOI: 10.1038/s41467-017-01236-6] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/29/2017] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is phosphorylated by various kinases, several of which have been implicated in aberrant fibroblast activation in fibrotic diseases including systemic sclerosis (SSc). Here we show that profibrotic signals converge on STAT3 and that STAT3 may be an important molecular checkpoint for tissue fibrosis. STAT3 signaling is hyperactivated in SSc in a TGFβ-dependent manner. Expression profiling and functional studies in vitro and in vivo demonstrate that STAT3 activation is mediated by the combined action of JAK, SRC, c-ABL, and JNK kinases. STAT3-deficient fibroblasts are less sensitive to the pro-fibrotic effects of TGFβ. Fibroblast-specific knockout of STAT3, or its pharmacological inhibition, ameliorate skin fibrosis in experimental mouse models. STAT3 thus integrates several profibrotic signals and might be a core mediator of fibrosis. Considering that several STAT3 inhibitors are currently tested in clinical trials, STAT3 might be a candidate for molecular targeted therapies of SSc. STAT3 is a transcription factor that is activated in fibrotic diseases such as systemic sclerosis. Here the authors show that STAT3 is the converging point for multiple pro-fibrotic signalling pathways, and that its genetic ablation or inhibition ameliorate skin fibrosis in mouse models.
Collapse
|
23
|
Trivedi P, Kumar RK, Iyer A, Boswell S, Gerarduzzi C, Dadhania VP, Herbert Z, Joshi N, Luyendyk JP, Humphreys BD, Vaidya VS. Targeting Phospholipase D4 Attenuates Kidney Fibrosis. J Am Soc Nephrol 2017; 28:3579-3589. [PMID: 28814511 DOI: 10.1681/asn.2016111222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 07/11/2017] [Indexed: 01/13/2023] Open
Abstract
Phospholipase D4 (PLD4), a single-pass transmembrane glycoprotein, is among the most highly upregulated genes in murine kidneys subjected to chronic progressive fibrosis, but the function of PLD4 in this process is unknown. Here, we found PLD4 to be overexpressed in the proximal and distal tubular epithelial cells of murine and human kidneys after fibrosis. Genetic silencing of PLD4, either globally or conditionally in proximal tubular epithelial cells, protected mice from the development of fibrosis. Mechanistically, global knockout of PLD4 modulated innate and adaptive immune responses and attenuated the upregulation of the TGF-β signaling pathway and α1-antitrypsin protein (a serine protease inhibitor) expression and downregulation of neutrophil elastase (NE) expression induced by obstructive injury. In vitro, treatment with NE attenuated TGF-β-induced accumulation of fibrotic markers. Furthermore, therapeutic targeting of PLD4 using specific siRNA protected mice from folic acid-induced kidney fibrosis and inhibited the increase in TGF-β signaling, decrease in NE expression, and upregulation of mitogen-activated protein kinase signaling. Immunoprecipitation/mass spectrometry and coimmunoprecipitation experiments confirmed that PLD4 binds three proteins that interact with neurotrophic receptor tyrosine kinase 1, a receptor also known as TrkA that upregulates mitogen-activated protein kinase. PLD4 inhibition also prevented the folic acid-induced upregulation of this receptor in mouse kidneys. These results suggest inhibition of PLD4 as a novel therapeutic strategy to activate protease-mediated degradation of extracellular matrix and reverse fibrosis.
Collapse
Affiliation(s)
- Priyanka Trivedi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ramya K Kumar
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ashwin Iyer
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sarah Boswell
- Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Casimiro Gerarduzzi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Vivekkumar P Dadhania
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Zach Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nikita Joshi
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan
| | - Benjamin D Humphreys
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri; and
| | - Vishal S Vaidya
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; .,Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
24
|
Kamal A, Elgengehy FT, EL Dakrony ALHM. Study of urotensin-2 (T21M and S89N) gene polymorphisms in systemic sclerosis. Meta Gene 2017. [DOI: 10.1016/j.mgene.2017.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
25
|
Gong S, Xu C, Wang L, Liu Y, Owusu D, Bailey BA, Li Y, Wang K. Genetic association analysis of polymorphisms in PSD3 gene with obesity, type 2 diabetes, and HDL cholesterol. Diabetes Res Clin Pract 2017; 126:105-114. [PMID: 28237857 DOI: 10.1016/j.diabres.2017.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/02/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND The pleckstrin and Sec7 domain-containing 3 (PSD3) gene has been linked to immune diseases. We examined whether the genetic variants within the PSD3 gene are associated with obesity, type 2 diabetes (T2D), and high-density lipoprotein (HDL) cholesterol level. METHODS Multiple logistic regression model and linear regression model were used to examine the associations of 259 single nucleotide polymorphisms (SNPs) within the PSD3 gene with obesity and T2D as binary traits, and HDL level as a continuous trait using the Marshfield data, respectively. A replication study of obesity was conducted using the Health Aging and Body Composition (Health ABC) sample. RESULTS 23SNPs were associated with obesity (p<0.05) in the Marshfield sample and rs4921966 revealed the strongest association (p=3.97×10-6). Of the 23SNPs, 20 were significantly associated with obesity in the meta-analysis of two samples (p<0.05). Furthermore, 6SNPs revealed associations with T2D in the Marshfield data (top SNP rs12156368 with p=3.05×10-3); while two SNPs (rs6983992 and rs7843239) were associated with both obesity and T2D (p=0.0188 and 0.023 for obesity and p=8.47×10-3 and 0.0128 for T2D, respectively). Furthermore, 11SNPs revealed associations with HDL level (top SNP rs13254772 with p=2.79×10-3) in the Marshfield data; meanwhile rs7009615 was associated with both T2D (p=0.038) and HDL level (p=4.44×10-3). In addition, haplotype analyses further supported the results of single SNP analysis. CONCLUSIONS Common variants in PSD3 were associated with obesity, T2D and HDL level. These findings add important new insights into the pathogenesis of obesity, T2D and HDL cholesterol.
Collapse
Affiliation(s)
- Shaoqing Gong
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Chun Xu
- Department of Health and Biomedical Science, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Liang Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Daniel Owusu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Beth A Bailey
- Department of Family Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Yujing Li
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA.
| |
Collapse
|
26
|
Liu L, Zhou H, Yang J, Sun L. Association of a TNIP1 gene polymorphism with chronic primary immune thrombocytopenia in Chinese population. Meta Gene 2016. [DOI: 10.1016/j.mgene.2016.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
27
|
Kwiatkowska M, Rakowska A, Walecka I, Rudnicka L. The diagnostic value of trichoscopy in systemic sclerosis. J Dermatol Case Rep 2016; 10:21-25. [PMID: 27900061 DOI: 10.3315/jdcr.2016.1225] [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] [Received: 03/22/2016] [Accepted: 06/08/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND/OBJECTIVES Systemic sclerosis is a connective tissue disease, which is characterized by fibrosis of the skin and internal organs, presence of specific antibodies and vascular involvement. Capillaroscopy is a useful method for the diagnosis and follow-up of patients with systemic sclerosis. Trichoscopy is a rapid, non-invasive technique, which has become a standard procedure in differential diagnosis of scalp and hair diseases. The aim of this study was to assess whether trichoscopy may be applied in imaging microvessels in patients with systemic sclerosis. METHODS The study included 17 patients with systemic sclerosis, and 31 healthy patients. In every patient 10 trichoscopy images were taken with Fotofinder II. RESULTS In patients with systemic sclerosis trichoscopy of the frontal scalp area revealed polymorphic microvessels in 64,7% of patients, spider vessels (76,4%), capillary loops (52,9%), arborising vessels (41,1%) and avascular areas (35,2%). In healthy individuals these features were observed in polymorphic microvessels 6,4% of patients, spider vessels 6,4%, capillary loops 100%, arborising vessels 16,1%, avascular areas 9,6%, respectively. CONCLUSIONS In conclusion, the presence of polymorphic vessels in frontal area in trichoscopy is characteristic for systemic sclerosis.
Collapse
Affiliation(s)
| | - Adriana Rakowska
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | | | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland; ; Department of Neuropeptides, Mossakowski Medical Research Centre, Warsaw, Poland
| |
Collapse
|
28
|
Multiple genes, especially immune-regulating genes, contribute to disease susceptibility in systemic sclerosis. Curr Opin Rheumatol 2016; 28:595-605. [DOI: 10.1097/bor.0000000000000334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Chairta P, Nicolaou P, Christodoulou K. Genomic and genetic studies of systemic sclerosis: A systematic review. Hum Immunol 2016; 78:153-165. [PMID: 27984087 DOI: 10.1016/j.humimm.2016.10.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
Systemic sclerosis is an autoimmune rheumatic disease characterised by fibrosis, vasculopathy and inflammation. The exact aetiology of SSc remains unknown but evidences show that various genetic factors may be involved. This review aimed to assess HLA alleles/non-HLA polymorphisms, microsatellites and chromosomal abnormalities that have thus far been associated with SSc. PubMed, Embase and Scopus databases were searched up to July 29, 2015 using a combination of search-terms. Articles retrieved were evaluated based on set exclusion and inclusion criteria. A total of 150 publications passed the filters. HLA and non-HLA studies showed that particular alleles in the HLA-DRB1, HLA-DQB1, HLA-DQA1, HLA-DPB1 genes and variants in STAT4, IRF5 and CD247 are frequently associated with SSc. Non-HLA genes analysis was performed using the PANTHER and STRING10 databases. PANTHER classification revealed that inflammation mediated by chemokine and cytokine, interleukin and integrin signalling pathways are among the common extracted pathways associated with SSc. STRING10 analysis showed that NFKB1, CSF3R, STAT4, IFNG, PRL and ILs are the main "hubs" of interaction network of the non-HLA genes associated with SSc. This study gathers data of valid genetic factors associated with SSc and discusses the possible interactions of implicated molecules.
Collapse
Affiliation(s)
- Paraskevi Chairta
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus
| | - Paschalis Nicolaou
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus
| | - Kyproula Christodoulou
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus.
| |
Collapse
|
30
|
Abstract
Purpose of review Large-scale and follow-up genetic association studies in systemic sclerosis (SSc) have implicated over 40 regions in disease risk, 15 of which with robust associations. Nevertheless, the causal variants and the functional mechanisms underlying the genetic associations remain elusive, and the reasons for the higher disease burden in African Americans unknown. Incorporating tools from diverse fields is beginning to unveil the role of genetic diversity and regulatory variation in SSc susceptibility. This review will summarize recent advances in SSc genetics, including autoimmune disease overlap, evidence of natural selection, and current progress towards the dissection of the functional role of associated risk variants. Recent findings In the past year, multiple large-scale studies reported novel strong and suggestive SSc associations. These results, coupled with the regions shared with other autoimmune diseases, emphasize the role of dysregulation of immune pathways as a key causative factor in SSc pathogenesis. Strong evidence implicates natural selection as a mechanism contributing to the maintenance of some of these SSc alleles in the population. Studies integrating genomic, transcriptomic, and epigenomic datasets in specific cell types to identify causal autoimmune disease variants are emerging. Summary The identification and comprehensive understanding of the factors and mechanisms contributing to SSc will contribute to improved diagnosis and disease management.
Collapse
|
31
|
Fuschiotti P. Current perspectives on the immunopathogenesis of systemic sclerosis. Immunotargets Ther 2016; 5:21-35. [PMID: 27529059 PMCID: PMC4970639 DOI: 10.2147/itt.s82037] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Systemic sclerosis (SSc or scleroderma) is a progressive and highly debilitating autoimmune disorder characterized by inflammation, vasculopathy, and extensive fibrosis. SSc is highly heterogeneous in its clinical presentation, extent and severity of skin and internal organ involvement, and clinical course and has the highest fatality rate among connective tissue diseases. While clinical outcomes have improved in recent years, no current therapy is able to reverse or slow the natural progression of SSc, a reflection of its complex pathogenesis. Although activation of the immune system has long been recognized, the mechanisms responsible for the initiation of autoimmunity and the role of immune effector pathways in the pathogenesis of SSc remain incompletely understood. This review summarizes recent progress in disease pathogenesis with particular focus on the immunopathogenetic mechanisms of SSc.
Collapse
Affiliation(s)
- Patrizia Fuschiotti
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Wei P, Yang Y, Guo X, Hei N, Lai S, Assassi S, Liu M, Tan F, Zhou X. Identification of an Association of TNFAIP3 Polymorphisms With Matrix Metalloproteinase Expression in Fibroblasts in an Integrative Study of Systemic Sclerosis-Associated Genetic and Environmental Factors. Arthritis Rheumatol 2016; 68:749-60. [PMID: 26474180 PMCID: PMC4767670 DOI: 10.1002/art.39476] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a fibrotic disease attributed to both genetic susceptibility and environmental factors. This study was undertaken to investigate the associations between SSc-associated genetic variants and the expression of extracellular matrix (ECM) genes in human fibroblasts stimulated with silica particles in time-course and dose-response experiments. METHODS A total of 200 fibroblast strains were examined for ECM gene expression after stimulation with silica particles. The fibroblasts were genetically profiled using Immunochip assays and then subjected to whole-genome genotype imputation. Associations of genotypes and gene expression were first analyzed in a Caucasian cohort and then validated in a meta-analysis combining the results from Caucasian, African American, and Hispanic subjects. A linear mixed model for longitudinal data analysis was used to identify genetic variants associated with the expression of ECM genes, and the associations were validated by using a haplotype-based longitudinal association test on regions that included the loci identified. RESULTS The single-nucleotide polymorphism rs58905141 in TNFAIP3 was consistently associated with time-course and/or dose-response expression of MMP3 and MMP1 in the fibroblasts stimulated with silica particles in both the analysis of Caucasian subjects only and the meta-analysis. Results of the haplotype-based analysis validated the association signals. CONCLUSION Our findings indicate that a genetic variant of TNFAIP3 is strongly associated with the silica-induced profibrotic response of fibroblasts. In silico functional analysis based on the ENCODE database revealed that rs58905141 might affect the binding activities of the transcription factors for TNFAIP3. This is the first genome-wide study of interactions between genetic and environmental factors in a complex SSc fibroblast model.
Collapse
Affiliation(s)
- Peng Wei
- Human Genetics Center and Department of Biostatistics, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yang Yang
- Human Genetics Center and Department of Biostatistics, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030
- Division of Rheumatology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Xinjian Guo
- Division of Rheumatology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Nainan Hei
- Human Genetics Center and Department of Biostatistics, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Syeling Lai
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030
| | - Shervin Assassi
- Division of Rheumatology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Mengyuan Liu
- Division of Rheumatology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Filemon Tan
- Division of Rheumatology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Xiaodong Zhou
- Division of Rheumatology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
33
|
Bayram Y, Karaca E, Coban Akdemir Z, Yilmaz EO, Tayfun GA, Aydin H, Torun D, Bozdogan ST, Gezdirici A, Isikay S, Atik MM, Gambin T, Harel T, El-Hattab AW, Charng WL, Pehlivan D, Jhangiani SN, Muzny DM, Karaman A, Celik T, Yuregir OO, Yildirim T, Bayhan IA, Boerwinkle E, Gibbs RA, Elcioglu N, Tuysuz B, Lupski JR. Molecular etiology of arthrogryposis in multiple families of mostly Turkish origin. J Clin Invest 2016; 126:762-78. [PMID: 26752647 DOI: 10.1172/jci84457] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/25/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Arthrogryposis, defined as congenital joint contractures in 2 or more body areas, is a clinical sign rather than a specific disease diagnosis. To date, more than 400 different disorders have been described that present with arthrogryposis, and variants of more than 220 genes have been associated with these disorders; however, the underlying molecular etiology remains unknown in the considerable majority of these cases. METHODS We performed whole exome sequencing (WES) of 52 patients with clinical presentation of arthrogryposis from 48 different families. RESULTS Affected individuals from 17 families (35.4%) had variants in known arthrogryposis-associated genes, including homozygous variants of cholinergic γ nicotinic receptor (CHRNG, 6 subjects) and endothelin converting enzyme-like 1 (ECEL1, 4 subjects). Deleterious variants in candidate arthrogryposis-causing genes (fibrillin 3 [FBN3], myosin IXA [MYO9A], and pleckstrin and Sec7 domain containing 3 [PSD3]) were identified in 3 families (6.2%). Moreover, in 8 families with a homozygous mutation in an arthrogryposis-associated gene, we identified a second locus with either a homozygous or compound heterozygous variant in a candidate gene (myosin binding protein C, fast type [MYBPC2] and vacuolar protein sorting 8 [VPS8], 2 families, 4.2%) or in another disease-associated genes (6 families, 12.5%), indicating a potential mutational burden contributing to disease expression. CONCLUSION In 58.3% of families, the arthrogryposis manifestation could be explained by a molecular diagnosis; however, the molecular etiology in subjects from 20 families remained unsolved by WES. Only 5 of these 20 unrelated subjects had a clinical presentation consistent with amyoplasia; a phenotype not thought to be of genetic origin. Our results indicate that increased use of genome-wide technologies will provide opportunities to better understand genetic models for diseases and molecular mechanisms of genetically heterogeneous disorders, such as arthrogryposis. FUNDING This work was supported in part by US National Human Genome Research Institute (NHGRI)/National Heart, Lung, and Blood Institute (NHLBI) grant U54HG006542 to the Baylor-Hopkins Center for Mendelian Genomics, and US National Institute of Neurological Disorders and Stroke (NINDS) grant R01NS058529 to J.R. Lupski.
Collapse
|