1
|
Bagni G, Biancalana E, Chiara E, Costanzo I, Malandrino D, Lastraioli E, Palmerini M, Silvestri E, Urban ML, Emmi G. Epigenetics in autoimmune diseases: Unraveling the hidden regulators of immune dysregulation. Autoimmun Rev 2025; 24:103784. [PMID: 40043893 DOI: 10.1016/j.autrev.2025.103784] [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: 10/31/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Autoimmune diseases result from complex interactions between genetic and environmental factors. Recent advances in epigenetic research shed light on the intricate regulatory mechanisms that contribute to the development and progression of such conditions. The present review aims to explore the role of epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNAs, in the context of autoimmune diseases. We discuss the current understanding of epigenetic alterations associated with various autoimmune disorders, their impact on immune cell function, and their potential as innovative therapeutic targets. Additionally, we highlight the main future directions in the field of epigenetics in autoimmunity.
Collapse
Affiliation(s)
- Giacomo Bagni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Edoardo Biancalana
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Emanuele Chiara
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Iole Costanzo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Danilo Malandrino
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Elena Lastraioli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Miki Palmerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Maria Letizia Urban
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Giacomo Emmi
- Department of Medical, Surgery and Health Sciences, University of Trieste, and Clinical Medicine and Rheumatology Unit, Cattinara University Hospital, Trieste, Italy; Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Melbourne, Australia.
| |
Collapse
|
2
|
Zhou X, Shan NN. The intersection of epigenetics and immune thrombocytopenia: new insights into disease mechanisms and treatments. Mol Biol Rep 2025; 52:257. [PMID: 39982580 DOI: 10.1007/s11033-025-10363-z] [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: 01/04/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune hematologic disorder that arises from an imbalance in immune responses, disrupting the delicate equilibrium of the immune system. An increasing body of research has indicated that immune-related genes hold promise as biomarkers for diagnosis and prognosis, with a particular focus on the roles of B and T cells in ITP pathogenesis. Despite these advancements, a deeper understanding of the underlying regulatory mechanisms governing these immune-related genes remains essential. This review aims to integrate the current body of evidence and provide further insights into the epigenetic regulation of immune pathways involved in ITP development. The problem statement section highlights the complexity of ITP and its intricate connections with immune pathways. It also compares the epigenetic differences between pediatric and adult ITP based on existing evidence. Decoding epigenetic processes could potentially open up new avenues for improving diagnostic methods and therapeutic strategies for ITP.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing Wu Rd, Jinan, Shandong, 250021, China
| | - Ning-Ning Shan
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing Wu Rd, Jinan, Shandong, 250021, China.
| |
Collapse
|
3
|
Vahidi Z, Saghi E, Mahmoudi M, RezaieYazdi Z, Esmaeili SA, Zemorshidi F, Samadi M, Rastin M. Lactobacillus rhamnosus and Lactobacillus delbrueckii Ameliorate the Expression of miR-125a and miR-146a in Systemic Lupus Erythematosus Patients. Appl Biochem Biotechnol 2024; 196:6330-6341. [PMID: 38351428 DOI: 10.1007/s12010-023-04827-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 11/29/2024]
Abstract
The microRNAs are non-coding RNA molecules involved in physiological and pathological processes, causing autoimmune diseases such as systemic lupus erythematosus (SLE). Probiotics are living microorganisms that possess beneficial effects on the host immune system and modulate it. The effect of Lactobacillus rhamnosus and Lactobacillus delbrueckii on the expression of miR-125a and miR-146a was studied in peripheral blood mononuclear cells (PBMCs) from newly diagnosed lupus patients in this in vitro study. During this study, 20 recently diagnosed SLE patients and 20 healthy individuals participated. Ficoll method was used to isolate the PBMCs from whole blood, which were cultured for 48 h with Lactobacillus rhamnosus and Lactobacillus delbrueckii. In the next step, total RNA containing microRNA was extracted. cDNA was synthesized for miR-125a and miR-146a genes and analyzed by real-time PCR. Results were presented as fold changes. As compared to healthy controls, SLE patients expressed lower levels of miR-125a and miR-146a. PBMCs treated with Lactobacillus rhamnosus, Lactobacillus delbrueckii, or both probiotics had significantly higher levels of miR-125a and miR-146a compared to the untreated group. Treatment of PBMCs with both L. rhamnosus and L. delbrueckii upregulated the expression of miR-125a and miR-146a in treated cells compared with untreated cells in SLE patients (p = 0.02, p = 0.001). Lactobacillus rhamnosus and Lactobacillus delbrueckii modify lupus patients' immune responses and disease effects by regulating miR-125a and miR-146a.
Collapse
Affiliation(s)
- Zohreh Vahidi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Effat Saghi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Departments, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra RezaieYazdi
- Rheumatic Diseases Research Center, Ghaem Hospital, Internal Medicine Section, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Departments, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Zemorshidi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morteza Samadi
- Department of Immunology, Faculty of Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Rastin
- Immunology Departments, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Danieli MG, Casciaro M, Paladini A, Bartolucci M, Sordoni M, Shoenfeld Y, Gangemi S. Exposome: Epigenetics and autoimmune diseases. Autoimmun Rev 2024; 23:103584. [PMID: 39097180 DOI: 10.1016/j.autrev.2024.103584] [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: 07/14/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
Systemic autoimmune diseases are complex conditions characterized by an immune system dysregulation and an aberrant activation against self-antigens, leading to tissue and organ damage. Even though genetic predisposition plays a role, it cannot fully explain the onset of these diseases, highlighting the significant impact of non-heritable influences such as environment, hormones and infections. The exposome represents all those factors, ranging from chemical pollutants and dietary components to psychological stressors and infectious agents. Epigenetics, which studies changes in gene expression without altering the DNA sequence, is a crucial link between exposome and the development of autoimmune diseases. Key epigenetic mechanisms include DNA methylation, histone modifications, and non-coding RNAs. These epigenetic modifications could provide a potential piece of the puzzle in understanding systemic autoimmune diseases and their connection with the exposome. In this work we have collected the most important and recent evidence in epigenetic changes linked to systemic autoimmune diseases (systemic lupus erythematosus, idiopathic inflammatory myopathies, ANCA-associated vasculitis, and rheumatoid arthritis), emphasizing the roles these changes may play in disease pathogenesis, their potential as diagnostic biomarkers and their prospective in the development of targeted therapies.
Collapse
Affiliation(s)
- Maria Giovanna Danieli
- SOS Immunologia delle Malattie Rare e dei Trapianti, AOU delle Marche & Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, via Tronto 10/A, 60126 Torrette di Ancona, Italy; Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Marco Casciaro
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| | - Alberto Paladini
- PostGraduate School of Internal Medicine, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Martina Bartolucci
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Martina Sordoni
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat Gan 52621, Israel; Reichman University, Herzelia 46101, Israel.
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
5
|
Zare Moghaddam M, Mousavi MJ, Ghotloo S. Stem cell-based therapy for systemic lupus erythematous. J Transl Autoimmun 2024; 8:100241. [PMID: 38737817 PMCID: PMC11087996 DOI: 10.1016/j.jtauto.2024.100241] [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: 01/11/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Systemic lupus erythematosus (SLE), an autoimmune disease, is among the most prevalent rheumatic autoimmune disorders. It affects autologous connective tissues caused by the breakdown of self-tolerance mechanisms. During the last two decades, stem cell therapy has been increasingly considered as a therapeutic option in various diseases, including parkinson's disease, alzheimer, stroke, spinal cord injury, multiple sclerosis, inflammatory bowel disease, liver disease, diabete, heart disease, bone disease, renal disease, respiratory diseases, and hematological abnormalities such as anemia. This is due to the unique properties of stem cells that divide and differentiate to the specialized cells in the damaged tissues. Moreover, they impose immunomodulatory properties affecting the diseases caused by immunological abnormalities such as rheumatic autoimmune disorders. In the present manuscript, efficacy of stem cell therapy with two main types of stem cells, including mesenchymal stem cell (MSC), and hematopoietic stem cells (HSC) in animal models or human patients of SLE, has been reviewed. Taken together, MSC and HSC therapies improved the disease activity, and severity in kidney, lung, liver, and bone (improvement in the clinical manifestation). In addition, a change in the immunological parameters occurred (improvement in immunological parameters). The level of autoantibodies, including antinuclear antibody (ANA), and anti-double-stranded deoxyribonucleic acid antibodies (dsDNA Abs) reduced. A conversion of Th1/Th2 ratio (in favor of Th2), and Th17/Treg (in favor of Treg) was also detected. In spite of many advantages of MSC and HSC transplantations, including efficacy, safety, and increased survival rate of SLE patients, some complications, including recurrence of the disease, occurrence of infections, and secondary autoimmune diseases (SAD) were observed after transplantation that should be addressed in the next studies.
Collapse
Affiliation(s)
- Maryam Zare Moghaddam
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Somayeh Ghotloo
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
6
|
Omer MH, Shafqat A, Ahmad O, Nadri J, AlKattan K, Yaqinuddin A. Urinary Biomarkers for Lupus Nephritis: A Systems Biology Approach. J Clin Med 2024; 13:2339. [PMID: 38673612 PMCID: PMC11051403 DOI: 10.3390/jcm13082339] [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: 03/19/2024] [Revised: 04/12/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is the prototypical systemic autoimmune disorder. Kidney involvement, termed lupus nephritis (LN), is seen in 40-60% of patients with systemic lupus erythematosus (SLE). After the diagnosis, serial measurement of proteinuria is the most common method of monitoring treatment response and progression. However, present treatments for LN-corticosteroids and immunosuppressants-target inflammation, not proteinuria. Furthermore, subclinical renal inflammation can persist despite improving proteinuria. Serial kidney biopsies-the gold standard for disease monitoring-are also not feasible due to their inherent risk of complications. Biomarkers that reflect the underlying renal inflammatory process and better predict LN progression and treatment response are urgently needed. Urinary biomarkers are particularly relevant as they can be measured non-invasively and may better reflect the compartmentalized renal response in LN, unlike serum studies that are non-specific to the kidney. The past decade has overseen a boom in applying cutting-edge technologies to dissect the pathogenesis of diseases at the molecular and cellular levels. Using these technologies in LN is beginning to reveal novel disease biomarkers and therapeutic targets for LN, potentially improving patient outcomes if successfully translated to clinical practice.
Collapse
Affiliation(s)
- Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff CF14 4YS, UK;
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Omar Ahmad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Juzer Nadri
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| |
Collapse
|
7
|
Guzeldemir-Akcakanat E, Sunnetci-Akkoyunlu D, Balta-Uysal VM, Özer T, Işik EB, Cine N. Differentially expressed miRNAs associated with generalized aggressive periodontitis. Clin Oral Investig 2023; 28:7. [PMID: 38123758 DOI: 10.1007/s00784-023-05404-5] [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: 07/22/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE This study aimed to investigate miRNA expression profiles in individuals with periodontitis which is a chronic inflammatory condition affecting the integrity of the periodontal attachment. miRNAs play a crucial role in gene regulation through various mechanisms, making them potential diagnostic markers and therapeutic targets for various diseases. MATERIALS AND METHODS A total of 25 individuals with aggressive periodontitis and 25 controls were included in the study. Gingival tissues were collected for miRNA isolation and cDNA synthesis. miRNAs associated with periodontitis, including hsa-miR-185-5p, hsa-miR-17, hs-miR-146a, hs-miR-146b, hs-miR-155, hs-miR-203, hs-miR-205, hs-miR-223, and hsa-miR-21-3p, were analyzed using a combination of miRTarBase database analysis and literature mining was performed. Real-time PCR was used to assess the expression patterns of the target miRNAs, and the data were analyzed using the REST program. RESULTS The study revealed upregulated expression levels of hsa-miR-223-3p, hsa-miR-203b-5p, hsa-miR-146a-5p, hsa-miR-146b-5p, and hsa-miR-155-5p in individuals with periodontitis. Conversely, downregulated expression was observed for hsa-miR-185-5p, hsa-miR-21-3p, and hsa-miR-17-3p. CONCLUSION The findings suggest significant differences in the expression of specific miRNAs associated with inflammation in periodontitis. MZB1 acts as a hormone-regulated adipokine/pro-inflammatory cytokine, driving chronic inflammation and influencing cellular expansion. Predominantly expressed in marginal zone and B1 B cells, specialized subsets that respond rapidly to infections, MZB1 impacts immune protein synthesis and immune cell maturation, notably targeting microRNA-185 to potentially impede T cell development. Further research is needed to elucidate the functional significance and potential implications of these miRNAs. CLINICAL RELEVANCE miRNAs regulate the expression of target genes by finely tuning protein expression levels. The current findings provide compelling evidence of notable variations in the expression levels of specific miRNAs associated with inflammation in individuals affected by periodontitis; hence, miRNAs hold promise as potential therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Esra Guzeldemir-Akcakanat
- Department of Periodontology, Faculty of Dentistry, Kocaeli University, 41190 Basiskele, Kocaeli, Turkey.
| | | | - V Merve Balta-Uysal
- Department of Periodontology, Faculty of Dentistry, Kocaeli University, 41190 Basiskele, Kocaeli, Turkey
| | - Tolgahan Özer
- Department of Medical Genetics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Elif Büşra Işik
- Department of Medical Genetics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Naci Cine
- Department of Medical Genetics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
8
|
Huang R, Tang J, Wang S, Liu Y, Zhang M, Jin M, Qin H, Qian W, Lu Y, Yang Y, Lu B, Yao Y, Yan P, Huang J, Zhang W, Lu J, Gu M, Zhu Y, Guo X, Xian S, Liu X, Huang Z. Sequencing technology as a major impetus in the advancement of studies into rheumatism: A bibliometric study. Front Immunol 2023; 14:1067830. [PMID: 36875117 PMCID: PMC9982012 DOI: 10.3389/fimmu.2023.1067830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
Background Rheumatism covers a wide range of diseases with complex clinical manifestations and places a tremendous burden on humans. For many years, our understanding of rheumatism was seriously hindered by technology constraints. However, the increasing application and rapid advancement of sequencing technology in the past decades have enabled us to study rheumatism with greater accuracy and in more depth. Sequencing technology has made huge contributions to the field and is now an indispensable component and powerful tool in the study of rheumatism. Methods Articles on sequencing and rheumatism, published from 1 January 2000 to 25 April 2022, were retrieved from the Web of Science™ (Clarivate™, Philadelphia, PA, USA) database. Bibliometrix, the open-source tool, was used for the analysis of publication years, countries, authors, sources, citations, keywords, and co-words. Results The 1,374 articles retrieved came from 62 countries and 350 institutions, with a general increase in article numbers during the last 22 years. The leading countries in terms of publication numbers and active cooperation with other countries were the USA and China. The most prolific authors and most popular documents were identified to establish the historiography of the field. Popular and emerging research topics were assessed by keywords and co-occurrence analysis. Immunological and pathological process in rheumatism, classification, risks and susceptibility, and biomarkers for diagnosis were among the hottest themes for research. Conclusions Sequencing technology has been widely applied in the study of rheumatism and propells research in the area of discovering novel biomarkers, related gene patterns and physiopathology. We suggest that further efforts be made to advance the study of genetic patterns related to rheumatic susceptibility, pathogenesis, classification and disease activity, and novel biomarkers.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jieling Tang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siqiao Wang
- Tongji University School of Medicine, Shanghai, China
| | - Yifan Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyi Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghao Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengwei Qin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijin Qian
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuwei Lu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Yang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingnan Lu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuntao Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Penghui Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Zhang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Minyi Gu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shuyuan Xian
- Department of Orthopedics, Shibei Hospital, Shanghai, China
| | - Xin Liu
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Wang H, Geng G, Zhang D, Han F, Ye S. Analysis of microRNA-199a-3p expression in CD4 + T cells of systemic lupus erythematosus. Clin Rheumatol 2023; 42:1683-1694. [PMID: 36763225 DOI: 10.1007/s10067-023-06534-7] [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: 05/09/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
OBJECTIVES Accumulating evidence have suggested microRNAs (miRNAs) play important roles in the pathogenesis of systemic lupus erythematosus (SLE). Here we aimed to explore aberrant expression of miRNAs in CD4+ T cells from SLE patients and their potential function in SLE pathogenesis. METHODS First, next-generation sequencing was performed on CD4+ T cells from four SLE patients and three healthy controls (HCs). Candidate miRNAs were then validated in CD4+ T cells from 97 patients with SLE, 16 patients with rheumatoid arthritis, and 12 HCs using qRT-PCR. Then the relationship between the candidate miRNA and clinical characteristics was analyzed. Bioinformatics analysis and validation of the target genes of the candidate miRNA were performed. RESULTS A total of 66 upregulated miRNAs and 70 downregulated miRNAs were found between SLE and normal CD4+ T cells samples. miR-199a-3p was identified significant upregulation in the CD4+ T cells of lupus patients. High expression of miR-199a-3p was correlated with several clinical characteristics including low C3 level, positive anti-dsDNA antibody, high ESR level, active lupus nephritis, and active disease activity. When distinguishing active LN from non-LN or active lupus from stable lupus, the AUCs of miR-199a-3p were 0.68 and 0.70, respectively. And the expression of miR-199a-3p, involved in JAK-STAT signaling pathway, was negatively correlated with the STAM expression in CD4+ T cells of SLE. CONCLUSION Our study suggested a novel and promising role of miR-199a-3p in CD4+ T cells for SLE. Further studies are needed to precisely determine the function of miR-199a-3p in this disease. Key Points • Aberrant expression of miRNAs in CD4+ T cells and their potential function in SLE pathogenesis remained unclear. • miR-199a-3p in CD4+ T cells plays a novel role in the pathogenesis of SLE and serves as a potential target for SLE.
Collapse
Affiliation(s)
- Huijing Wang
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Institute of Nephrology, Zhejiang University, Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
| | - Guannan Geng
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gene Editing Core Facility, Center for Excellence in Brain Science and intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Danting Zhang
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Institute of Nephrology, Zhejiang University, Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
| | - Shuang Ye
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Gamal-Eldeen AM, Fahmy CA, Raafat BM, Althobaiti F, Bassyouni IH, Talaat RM. Circulating Levels of Hypoxia-regulating MicroRNAs in Systemic Lupus Erythematosus Patients with Hemolytic Anemia. Curr Med Sci 2022; 42:1231-1239. [PMID: 36469203 DOI: 10.1007/s11596-022-2644-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE MicroRNAs are fine regulators for gene expression during the post-transcriptional stage in many autoimmune diseases. HypoxamiRs (miR-210 and miR-21) play an important role in hypoxia and in inflammation-associated hypoxia. Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease that would potentiate many pathological complications, including hemolytic anemia. This study aimed to investigate the role of hypoxamiRs in SLE/hemolytic anemia patients. METHODS This work was designed to analyze the circulating levels of↱ the miR-210 and miR-21 expressions and hypoxia-inducible factor-1α (HIF-α) in SLE/hemolytic anemia patients. SLE activity was evaluated for all patients by SLE Disease Activity Index (SLEDAI). Clinical manifestations/complications and serological/hematological investigations were reported. HIF-α concentration was assayed by ELISA and expression of miR-21 and miR-210 was analyzed by qRT-PCR. RESULTS The results indicated that the fold change of the miR-210/miR-21 expressions in plasma was significantly elevated in SLE/hemolytic anemia patients. A strong positive correlation between the miR-210 and miR-21 expression levels was also recorded. Among the associated-disease complications, hypertension, arthritis, oral ulcers, and serositis were associated with a high circulating miR-210 expression, while the occurrence of renal disorders was associated with the increased miR-21 expression. Furthermore, the HIF-α level was remarkably elevated in SLE/hemolytic anemia patients. A high positive correlation was recorded between the HIF-α concentration and miR-210/miR-21 expression levels. The occurrence of oral ulcers, arthritis, and hypertension was associated with the increased HIF-α concentration. On the other hand, SLEDAI and white blood cell count were positively correlated with miR-21/ miR-210. The erythrocyte sedimentation rate was positively correlated with miR-21. CONCLUSION The dysregulation of the circulating miR-210/miR-210/HIF-1α levels in SLE/hemolytic anemia patients advocated that the hypoxia pathway might have an essential role in the pathogenesis and complications of these diseases.
Collapse
Affiliation(s)
- Amira M Gamal-Eldeen
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia. .,High Altitude Research Center, Prince Sultan Medical Complex, Al-Hawiyah, Taif University, Taif, 21944, Saudi Arabia.
| | - Cinderella A Fahmy
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, 12622, Egypt.,Biochemistry Department, National Research Centre, Cairo, 12622, Egypt
| | - Bassem M Raafat
- Radiological Sciences Department, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Fayez Althobaiti
- High Altitude Research Center, Prince Sultan Medical Complex, Al-Hawiyah, Taif University, Taif, 21944, Saudi Arabia.,Biotechnology Department, Faculty of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Iman H Bassyouni
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Cairo University, El-Kasr El-Aini Hospital, Cairo, 12613, Egypt
| | - Roba M Talaat
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), Sadat City University, Sadat City, 32897, Egypt
| |
Collapse
|
11
|
Chaudhari S, D'Souza BM, Morales JY, Young-Stubbs CM, Shimoura CG, Ma R, Mathis KW. Renal TLR-7/TNF-α pathway as a potential female-specific mechanism in the pathogenesis of autoimmune-induced hypertension. Am J Physiol Heart Circ Physiol 2022; 323:H1331-H1342. [PMID: 36367687 PMCID: PMC9744658 DOI: 10.1152/ajpheart.00286.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
Hypertension is prevalent in patients with systemic lupus erythematosus (SLE). The goal of the current study is to track the pathogenesis of hypertension and renal injury in SLE, identify contributory mechanisms, and highlight differences in disease development among sexes. Mean arterial pressure was measured in conscious male and female SLE (NZBWF1) and control (NZW) mice at 34-35 wk of age using indwelling arterial catheters. Measures of renal injury, renal inflammation, and renal hemodynamics were used to monitor the potential contributors to latent sex differences. Both male and female SLE mice were hypertensive at 35 wk of age, and the hypertension was linked to renal injury in females, but not in males. A known contributor of renal pathology in SLE, Toll-like receptor (TLR)-7, and its downstream effector, the proinflammatory cytokine tumor necrosis factor (TNF)-α, were lower in male SLE mice than in females. Male SLE mice also had higher glomerular filtration rate (GFR) and lower renal vascular resistance (RVR) than females. Our data suggest that although hypertension in female SLE mice is associated with renal mechanisms, hypertension in male SLE mice may develop independent of renal changes. Future studies will continue to dissect sex-specific factors that should be considered when treating patients with hypertension with underlying chronic inflammation and/or autoimmunity.NEW & NOTEWORTHY There is a high prevalence of hypertension in male and female SLE; however, male SLE mice are hypertensive without renal involvement. The development of hypertension in female SLE mice is renocentric and strongly associated with injurious renal mechanisms like the TLR-7→TNF-α pathway. This clear difference in the pathogenesis among the sexes could have a significant impact on how we treat patients with hypertension with underlying chronic autoimmune/inflammatory diseases.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Bradley M D'Souza
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Jessica Y Morales
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Cassandra M Young-Stubbs
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Caroline G Shimoura
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Keisa W Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
12
|
Elghzaly AA, Sun C, Looger LL, Hirose M, Salama M, Khalil NM, Behiry ME, Hegazy MT, Hussein MA, Salem MN, Eltoraby E, Tawhid Z, Alwasefy M, Allam W, El-Shiekh I, Elserafy M, Abdelnaser A, Hashish S, Shebl N, Shahba AA, Elgirby A, Hassab A, Refay K, El-Touchy HM, Youssef A, Shabacy F, Hashim AA, Abdelzaher A, Alshebini E, Fayez D, El-Bakry SA, Elzohri MH, Abdelsalam EN, El-Khamisy SF, Ibrahim S, Ragab G, Nath SK. Genome-wide association study for systemic lupus erythematosus in an egyptian population. Front Genet 2022; 13:948505. [PMID: 36324510 PMCID: PMC9619055 DOI: 10.3389/fgene.2022.948505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/30/2022] [Indexed: 04/11/2024] Open
Abstract
Systemic lupus erythematosus (SLE) susceptibility has a strong genetic component. Genome-wide association studies (GWAS) across trans-ancestral populations show both common and distinct genetic variants of susceptibility across European and Asian ancestries, while many other ethnic populations remain underexplored. We conducted the first SLE GWAS on Egyptians-an admixed North African/Middle Eastern population-using 537 patients and 883 controls. To identify novel susceptibility loci and replicate previously known loci, we performed imputation-based association analysis with 6,382,276 SNPs while accounting for individual admixture. We validated the association analysis using adaptive permutation tests (n = 109). We identified a novel genome-wide significant locus near IRS1/miR-5702 (Pcorrected = 1.98 × 10-8) and eight novel suggestive loci (Pcorrected < 1.0 × 10-5). We also replicated (Pperm < 0.01) 97 previously known loci with at least one associated nearby SNP, with ITGAM, DEF6-PPARD and IRF5 the top three replicated loci. SNPs correlated (r 2 > 0.8) with lead SNPs from four suggestive loci (ARMC9, DIAPH3, IFLDT1, and ENTPD3) were associated with differential gene expression (3.5 × 10-95 < p < 1.0 × 10-2) across diverse tissues. These loci are involved in cellular proliferation and invasion-pathways prominent in lupus and nephritis. Our study highlights the utility of GWAS in an admixed Egyptian population for delineating new genetic associations and for understanding SLE pathogenesis.
Collapse
Affiliation(s)
- Ashraf A. Elghzaly
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, El-Mansoura, Egypt
| | - Celi Sun
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Loren L. Looger
- Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, San Diego, CA, United States
| | - Misa Hirose
- Division of Genetics, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Mohamed Salama
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo, Egypt
| | - Noha M. Khalil
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mervat Essam Behiry
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Tharwat Hegazy
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Ahmed Hussein
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamad Nabil Salem
- Department of Internal Medicine, Faculty of Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Ehab Eltoraby
- Department of Internal Medicine, Faculty of Medicine, Mansoura University, El-Mansoura, Egypt
| | - Ziyad Tawhid
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, El-Mansoura, Egypt
| | - Mona Alwasefy
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, El-Mansoura, Egypt
| | - Walaa Allam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Iman El-Shiekh
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Menattallah Elserafy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo, Egypt
| | - Sara Hashish
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo, Egypt
| | - Nourhan Shebl
- Institute of Global Health and Human Ecology, The American University in Cairo, New Cairo, Egypt
| | | | - Amira Elgirby
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Bab Sharqi, Egypt
| | - Amina Hassab
- Department of Clinical Pathology, Faculty of Medicine, Alexandria University, Bab Sharqi, Egypt
| | - Khalida Refay
- Department of Internal Medicine, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | | | - Ali Youssef
- Department of Rheumatology and Immunology, Faculty of Medicine, Benha University Hospital, Benha, Egypt
| | - Fatma Shabacy
- Department of Rheumatology and Immunology, Faculty of Medicine, Benha University Hospital, Benha, Egypt
| | | | - Asmaa Abdelzaher
- Department of Clinical Pathology, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Emad Alshebini
- Department of Internal Medicine, Faculty of Medicine, Menoufia University, Al Minufiyah, Egypt
| | - Dalia Fayez
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Samah A. El-Bakry
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona H. Elzohri
- Department of Internal Medicine, Faculty of Medicine, Assiut University, Asyut, Egypt
| | | | - Sherif F. El-Khamisy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- The Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
- The Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Saleh Ibrahim
- Division of Genetics, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Gaafar Ragab
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Swapan K. Nath
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| |
Collapse
|
13
|
Coit P, Roopnarinesingh X, Ortiz-Fernández L, McKinnon-Maksimowicz K, Lewis EE, Merrill JT, McCune WJ, Wren JD, Sawalha AH. Hypomethylation of miR-17-92 cluster in lupus T cells and no significant role for genetic factors in the lupus-associated DNA methylation signature. Ann Rheum Dis 2022; 81:1428-1437. [PMID: 35710306 PMCID: PMC10259175 DOI: 10.1136/annrheumdis-2022-222656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/07/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Lupus T cells demonstrate aberrant DNA methylation patterns dominated by hypomethylation of interferon-regulated genes. The objective of this study was to identify additional lupus-associated DNA methylation changes and determine the genetic contribution to epigenetic changes characteristic of lupus. METHODS Genome-wide DNA methylation was assessed in naïve CD4+ T cells from 74 patients with lupus and 74 age-matched, sex-matched and race-matched healthy controls. We applied a trend deviation analysis approach, comparing methylation data in our cohort with over 16 500 samples. Methylation quantitative trait loci (meQTL) analysis was performed by integrating methylation profiles with genome-wide genotyping data. RESULTS In addition to the previously reported epigenetic signature in interferon-regulated genes, we observed hypomethylation in the promoter region of the miR-17-92 cluster in patients with lupus. Members of this microRNA cluster play an important role in regulating T cell proliferation and differentiation. Expression of two microRNAs in this cluster, miR-19b1 and miR-18a, showed a significant positive correlation with lupus disease activity. Among miR-18a target genes, TNFAIP3, which encodes a negative regulator of nuclear factor kappa B, was downregulated in lupus CD4+ T cells. MeQTL identified in lupus patients showed overlap with genetic risk loci for lupus, including CFB and IRF7. The lupus risk allele in IRF7 (rs1131665) was associated with significant IRF7 hypomethylation. However, <1% of differentially methylated CpG sites in patients with lupus were associated with an meQTL, suggesting minimal genetic contribution to lupus-associated epigenotypes. CONCLUSION The lupus defining epigenetic signature, characterised by robust hypomethylation of interferon-regulated genes, does not appear to be determined by genetic factors. Hypomethylation of the miR-17-92 cluster that plays an important role in T cell activation is a novel epigenetic locus for lupus.
Collapse
Affiliation(s)
- Patrick Coit
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiavan Roopnarinesingh
- Graduate Program, Department of Biochemistry and Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Lourdes Ortiz-Fernández
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Emily E Lewis
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joan T Merrill
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - W Joseph McCune
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Department of Biochemistry and Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Lamana A, Castro-Vázquez D, de la Fuente H, Triguero-Martínez A, Martínez-Hernández R, Revenga M, Villanueva-Romero R, Llamas-Velasco M, Chicharro P, Juarranz Y, Marazuela M, Sales-Sanz M, García-Vicuña R, Tomero E, González-Álvaro I, Martínez C, Gomariz RP. VIP/VPAC Axis Expression in Immune-Mediated Inflammatory Disorders: Associated miRNA Signatures. Int J Mol Sci 2022; 23:ijms23158578. [PMID: 35955723 PMCID: PMC9369218 DOI: 10.3390/ijms23158578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 12/04/2022] Open
Abstract
Few studies have considered immune-mediated inflammatory disorders (IMID) together, which is necessary to adequately understand them given they share common mechanisms. Our goal was to investigate the expression of vasoactive intestinal peptide (VIP) and its receptors VPAC1 and VPAC2 in selected IMID, analyze the effect of biological therapies on them, and identify miRNA signatures associated with their expression. Serum VIP levels and mRNA of VPAC and miRNA expression in peripheral blood mononuclear cells were analyzed from 52 patients with psoriasis, rheumatoid arthritis, Graves’ disease, or spondyloarthritis and from 38 healthy subjects. IMID patients showed higher levels of VIP and increased expression of VPAC2 compared to controls (p < 0.0001 and p < 0.0192, respectively). Receiver operating characteristic curve analysis showed that the levels of VIP or VPAC2 expression were adequate discriminators capable of identifying IMID. Treatment of IMID patients with anti-TNFα and anti-IL12/23 significantly affected serum VIP levels. We identified miRNA signatures associated with levels of serum VIP and VPAC2 expression, which correlated with IMID diagnosis of the patients. The results indicate that the expression of VIP/VPAC2 is able of identify IMIDs and open up a line of research based on the association between the VIP/VPAC axis and miRNA signatures in immune-mediated diseases.
Collapse
Affiliation(s)
- Amalia Lamana
- Department of Cell Biology, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.L.); (D.C.-V.); (R.V.-R.); (Y.J.)
| | - David Castro-Vázquez
- Department of Cell Biology, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.L.); (D.C.-V.); (R.V.-R.); (Y.J.)
| | - Hortensia de la Fuente
- Department of Immunology, Instituto de Investigación Princesa, Hospital Universitario de La Princesa, 28006 Madrid, Spain;
| | - Ana Triguero-Martínez
- Department of Rheumatology, Instituto de Investigación Princesa Madrid, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (A.T.-M.); (R.G.-V.); (E.T.); (I.G.-Á.)
| | - Rebeca Martínez-Hernández
- Department of Endocrinology, Instituto de Investigación Princesa, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (R.M.-H.); (M.M.)
| | - Marcelino Revenga
- Department of Rheumatology, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
- Department of Medicine and Medical Specialties, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
| | - Raúl Villanueva-Romero
- Department of Cell Biology, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.L.); (D.C.-V.); (R.V.-R.); (Y.J.)
| | - Mar Llamas-Velasco
- Department of Dermatology, Instituto de Investigación Princesa, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (M.L.-V.); (P.C.)
| | - Pablo Chicharro
- Department of Dermatology, Instituto de Investigación Princesa, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (M.L.-V.); (P.C.)
| | - Yasmina Juarranz
- Department of Cell Biology, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.L.); (D.C.-V.); (R.V.-R.); (Y.J.)
| | - Mónica Marazuela
- Department of Endocrinology, Instituto de Investigación Princesa, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (R.M.-H.); (M.M.)
| | - Marco Sales-Sanz
- Department of Ophthalmology, Hospital Universitario Ramón y Cajal-IRYCIS, 28034 Madrid, Spain;
| | - Rosario García-Vicuña
- Department of Rheumatology, Instituto de Investigación Princesa Madrid, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (A.T.-M.); (R.G.-V.); (E.T.); (I.G.-Á.)
| | - Eva Tomero
- Department of Rheumatology, Instituto de Investigación Princesa Madrid, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (A.T.-M.); (R.G.-V.); (E.T.); (I.G.-Á.)
| | - Isidoro González-Álvaro
- Department of Rheumatology, Instituto de Investigación Princesa Madrid, Hospital Universitario de La Princesa, 28006 Madrid, Spain; (A.T.-M.); (R.G.-V.); (E.T.); (I.G.-Á.)
| | - Carmen Martínez
- Department of Cell Biology, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.L.); (D.C.-V.); (R.V.-R.); (Y.J.)
- Correspondence: (C.M.); (R.P.G.); Tel.: +34-91-3944971 (R.P.G.)
| | - Rosa P. Gomariz
- Department of Cell Biology, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.L.); (D.C.-V.); (R.V.-R.); (Y.J.)
- Correspondence: (C.M.); (R.P.G.); Tel.: +34-91-3944971 (R.P.G.)
| |
Collapse
|
15
|
Bose M, Jefferies C. Sex bias in systemic lupus erythematosus: a molecular insight. IMMUNOMETABOLISM (COBHAM, SURREY) 2022; 4:e00004. [PMID: 35966636 PMCID: PMC9358995 DOI: 10.1097/in9.0000000000000004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/24/2022]
Abstract
Acknowledging sex differences in immune response is particularly important when we consider the differences between men and women in the incidence of disease. For example, over 80% of autoimmune disease occurs in women, whereas men have a higher incidence of solid tumors compared to women. In general women have stronger innate and adaptive immune responses than men, explaining their ability to clear viral and bacterial infections faster, but also contributing to their increased susceptibility to autoimmune disease. The autoimmune disease systemic lupus erythematosus (SLE) is the archetypical sexually dimorphic disease, with 90% of patients being women. Various mechanisms have been suggested to account for the female prevalence of SLE, including sex hormones, X-linked genes, and epigenetic regulation of gene expression. Here, we will discuss how these mechanisms contribute to pathobiology of SLE and how type I interferons work with them to augment sex specific disease pathogenesis in SLE.
Collapse
Affiliation(s)
- Moumita Bose
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Caroline Jefferies
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
16
|
Czaja AJ. Examining micro-ribonucleic acids as diagnostic and therapeutic prospects in autoimmune hepatitis. Expert Rev Clin Immunol 2022; 18:591-607. [PMID: 35510750 DOI: 10.1080/1744666x.2022.2074839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Micro-ribonucleic acids modulate the immune response by affecting the post-transcriptional expression of genes that influence the proliferation and function of activated immune cells, including regulatory T cells. Individual expressions or patterns in peripheral blood and liver tissue may have diagnostic value, reflect treatment response, or become therapeutic targets. The goals of this review are to present the properties and actions of micro-ribonucleic acids, indicate the key individual expressions in autoimmune hepatitis, and describe prospective clinical applications in diagnosis and management. AREAS COVERED Abstracts were identified in PubMed using the search words "microRNAs", "microRNAs in liver disease", and "microRNAs in autoimmune hepatitis". The number of abstracts reviewed exceeded 2000, and the number of full-length articles reviewed was 108. EXPERT OPINION Individual micro-ribonucleic acids, miR-21, miR-122, and miR-155, have been associated with biochemical severity, histological grade of inflammation, and pivotal pathogenic mechanisms in autoimmune hepatitis. Antisense oligonucleotides that down-regulate deleterious individual gene expressions, engineered molecules that impair targeting of gene products, and drugs that non-selectively up-regulate the biogenesis of potentially deficient gene regulators are feasible treatment options. Micro-ribonucleic acids constitute an under-evaluated area in autoimmune hepatitis that promises to improve diagnosis, pathogenic concepts, and therapy.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
17
|
Chinniah R, Adimulam T, Nandlal L, Arumugam T, Ramsuran V. The Effect of miRNA Gene Regulation on HIV Disease. Front Genet 2022; 13:862642. [PMID: 35601502 PMCID: PMC9117004 DOI: 10.3389/fgene.2022.862642] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/13/2022] [Indexed: 12/24/2022] Open
Abstract
Over many years, research on HIV/AIDS has advanced with the introduction of HAART. Despite these advancements, significant gaps remain with respect to aspects in HIV life cycle, with specific attention to virus-host interactions. Investigating virus-host interactions may lead to the implementation of novel therapeutic strategies against HIV/AIDS. Notably, host gene silencing can be facilitated by cellular small non-coding RNAs such as microRNAs paving the way for epigenetic anti-viral therapies. Numerous studies have elucidated the importance of microRNAs in HIV pathogenesis. Some microRNAs can either promote viral infection, while others can be detrimental to viral replication. This is accomplished by targeting the HIV-proviral genome or by regulating host genes required for viral replication and immune responses. In this review, we report on 1) the direct association of microRNAs with HIV infection; 2) the indirect association of known human genetic factors with HIV infection; 3) the regulation of human genes by microRNAs in other diseases that can be explored experimentally to determine their effect on HIV-1 infection; and 4) therapeutic interactions of microRNA against HIV infection.
Collapse
Affiliation(s)
- Romona Chinniah
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Theolan Adimulam
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Louansha Nandlal
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Thilona Arumugam
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Veron Ramsuran
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
18
|
Banerjee N, Wang H, Wang G, Boor PJ, Khan MF. Differential Expression of miRNAs in Trichloroethene-Mediated Inflammatory/Autoimmune Response and Its Modulation by Sulforaphane: Delineating the Role of miRNA-21 and miRNA-690. Front Immunol 2022; 13:868539. [PMID: 35422807 PMCID: PMC9001960 DOI: 10.3389/fimmu.2022.868539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Trichloroethene (TCE), an occupational and ubiquitous environmental contaminant, is associated with the induction of autoimmune diseases (ADs). Although oxidative stress plays a major role in TCE-mediated autoimmunity, the underlying molecular mechanisms still need to be delineated. Altered non-coding RNAs, including the expression of microRNAs (miRNAs), can influence target genes, especially related to apoptosis and inflammation, and contribute to ADs. Therefore, the objective of this study was to delineate the contribution of miRNAs in TCE-mediated inflammatory and autoimmune response. To achieve this, we treated female MRL+/+ mice with TCE (10 mmol/kg in corn oil, i.p., every fourth day) with/without antioxidant sulforaphane (SFN; 8 mg/kg in corn oil, i.p., every other day) for 6 weeks. With the use of miRNA microarray, 293 miRNAs were analyzed, which included 35 miRNAs that were relevant to inflammation and ADs. Among those 35 miRNAs, 8 were modulated by TCE and/or TCE+SFN exposure. TCE treatment led to increased expression of 3 miRNAs and also decreased expression of 3 miRNAs. Interestingly, among the 35 differentially expressed miRNAs, antioxidant SFN modulated the expression of 6 miRNAs. Based on the microarray findings, we subsequently focused on two miRNAs (miRNA-21 and miRNA-690), which are known to be involved in inflammation and autoimmune response. The increases in miRNA-21 and miR-690 (observed using miRNA microarray) were further validated by RT-PCR, and the TCE-mediated increases in miR-21 and miR-690 were ameliorated by SFN treatment. Modulating miR-21 and miR-690 by respective inhibitors or mimics suppressed the expression of NF-κB (p65) and IL-12 in RAW 264.7 cells. Our findings suggest a contributory role of miR-21 and miR-690 in TCE-mediated and its metabolite dichloroacetyl chloride (DCAC)-mediated inflammation and autoimmune response and support that antioxidant SFN could be a potential therapeutic candidate for inflammatory responses and ADs.
Collapse
Affiliation(s)
- Nivedita Banerjee
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Paul J Boor
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
19
|
Pinacchio C, Scordio M, Santinelli L, Frasca F, Sorrentino L, Bitossi C, Oliveto G, Viscido A, Ceci FM, Celani L, Ceccarelli G, Antonelli G, Mastroianni CM, d’Ettorre G, Scagnolari C. Analysis of serum microRNAs and rs2910164 GC single-nucleotide polymorphism of miRNA-146a in COVID-19 patients. J Immunoassay Immunochem 2022; 43:347-364. [DOI: 10.1080/15321819.2022.2035394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Claudia Pinacchio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Mirko Scordio
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
| | - Letizia Santinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Federica Frasca
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
| | - Leonardo Sorrentino
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
| | - Camilla Bitossi
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
| | - Giuseppe Oliveto
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
| | - Agnese Viscido
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
| | - Flavio Maria Ceci
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Luigi Celani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Guido Antonelli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
- Microbiology and Virology Unit, Sapienza University, Hospital Policlinico Umberto I, Rome, Italy
- Department of Molecular Medicine, Pasteur Institute Italy, Cenci Bolognetti Foundation, Rome, Italy
| | | | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy
- Department of Molecular Medicine, Pasteur Institute Italy, Cenci Bolognetti Foundation, Rome, Italy
| |
Collapse
|
20
|
Zhao Y, Cui S, Wang Y, Xu R. The Extensive Regulation of MicroRNA in Immune Thrombocytopenia. Clin Appl Thromb Hemost 2022; 28:10760296221093595. [PMID: 35536600 PMCID: PMC9096216 DOI: 10.1177/10760296221093595] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
MicroRNA (miRNA) is a small, single-stranded, non-coding RNA molecule that plays
a variety of key roles in different biological processes through
post-transcriptional regulation of gene expression. MiRNA has been proved to be
a variety of cellular processes involved in development, differentiation, signal
transduction, and is an important regulator of immune and autoimmune diseases.
Therefore, it may act as potent modulators of the immune system and play an
important role in the development of several autoimmune diseases. Immune
thrombocytopenia (ITP) is an autoimmune systemic disease characterized by a low
platelet count. Several studies suggest that like other autoimmune disorders,
miRNAs are deeply involved in the pathogenesis of ITP, interacting with the
function of innate and adaptive immune responses. In this review, we discuss
emerging knowledge about the function of miRNAs in ITP and describe miRNAs in
terms of their role in the immune system and autoimmune response. These findings
suggest that miRNA may be a useful therapeutic target for ITP by regulating the
immune system. In the future, we need to have a more comprehensive understanding
of miRNAs and how they regulate the immune system of patients with ITP.
Collapse
Affiliation(s)
- Yuerong Zhao
- 74738Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siyuan Cui
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.,Institute of Hematology, 74738Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruirong Xu
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.,Institute of Hematology, 74738Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
21
|
Fava A, Rao DA. Cellular and molecular heterogeneity in systemic lupus erythematosus. Semin Immunol 2021; 58:101653. [PMID: 36184357 DOI: 10.1016/j.smim.2022.101653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Andrea Fava
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA.
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Host miRNA and immune cell interactions: relevance in nano-therapeutics for human health. Immunol Res 2021; 70:1-18. [PMID: 34716546 DOI: 10.1007/s12026-021-09247-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Around 2200 miRNA (microRNA) genes were found in the human genome. miRNAs are arranged in clusters within the genome and share the same transcriptional regulatory units. It has been revealed that approximately 50% of miRNAs elucidated in the genome are transcribed from non-protein-coding genes, and the leftover miRNAs are present in the introns of coding sequences. We are now approaching a stage in which miRNA diagnostics and therapies can be established confidently, and several commercial efforts are underway to carry these innovations from the bench to the clinic. MiRNAs control many of the significant cellular activities such as production, differentiation, growth, and metabolism. Particularly in the immune system, miRNAs have emerged as a crucial biological component during diseased state and homeostasis. miRNAs have been found to regulate inflammatory responses and autoimmune disorders. Moreover, each miRNA targets multiple genes simultaneously, making miRNAs promising tools as diagnostic biomarkers and as remedial targets. Still, one of the major obstacles in miRNA-based approaches is the achievement of specific and efficient systemic delivery of miRNAs. To overcome these challenges, nanoformulations have been synthesized to protect miRNAs from degradation and enhance cellular uptake. The current review deals with the miRNA-mediated regulation of the recruitment and activation of immune cells, especially in the tumor microenvironment, viral infection, inflammation, and autoimmunity. The nano-based miRNA delivery modes are also discussed here, especially in the context of immune modulation.
Collapse
|
23
|
Chi M, Ma K, Li Y, Quan M, Han Z, Ding Z, Liang X, Zhang Q, Song L, Liu C. Immunological Involvement of MicroRNAs in the Key Events of Systemic Lupus Erythematosus. Front Immunol 2021; 12:699684. [PMID: 34408748 PMCID: PMC8365877 DOI: 10.3389/fimmu.2021.699684] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an archetype autoimmune disease characterized by a myriad of immunoregulatory abnormalities that drives injury to multiple tissues and organs. Due to the involvement of various immune cells, inflammatory cytokines, and related signaling pathways, researchers have spent a great deal of effort to clarify the complex etiology and pathogenesis of SLE. Nevertheless, current understanding of the pathogenesis of SLE is still in the early stages, and available nonspecific treatment options for SLE patients remain unsatisfactory. First discovered in 1993, microRNAs (miRNAs) are small RNA molecules that control the expression of 1/3 of human genes at the post-transcriptional level and play various roles in gene regulation. The aberrant expression of miRNAs in SLE patients has been intensively studied, and further studies have suggested that these miRNAs may be potentially relevant to abnormal immune responses and disease progression in SLE. The aim of this review was to summarize the specific miRNAs that have been observed aberrantly expressed in several important pathogenetic processes in SLE, such as DCs abnormalities, overactivation and autoantibody production of B cells, aberrant activation of CD4+ T cells, breakdown of immune tolerance, and abnormally increased production of inflammatory cytokines. Our summary highlights a novel perspective on the intricate regulatory network of SLE, which helps to enrich our understanding of this disorder and ignite future interest in evaluating the molecular regulation of miRNAs in autoimmunity SLE.
Collapse
Affiliation(s)
- Mingxuan Chi
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University, Suita, Japan
| | - Yunlong Li
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Quan
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhaolun Ding
- Department of Emergency Surgery, Shannxi Provincial People's Hospital, Xi'an, China
| | - Xin Liang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinxiu Zhang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China
| |
Collapse
|
24
|
Abstract
Chronic kidney disease (CKD), which is characterized by the gradual loss of kidney function, is a growing worldwide problem due to CKD-related morbidity and mortality. There are no reliable and early biomarkers enabling the monitoring, the stratification of CKD progression and the estimation of the risk of CKD-related complications, and therefore, the search for such molecules is still going on. Numerous studies have provided evidence that miRNAs are potentially important particles in the CKD field. Studies indicate that some miRNA levels can be increased in patients with CKD stages III–V and hemodialysis and decreased in renal transplant recipients (miR-143, miR-145 and miR-223) as well as elevated in patients with CKD stages III–V, decreased in hemodialysis patients and even more markedly decreased in renal transplant recipients (miR-126 and miR-155). miRNA have great potential of being sensitive and specific biomarkers in kidney diseases as they are tissue specific and stable in various biological materials. Some promising non-invasive miRNA biomarkers have already been recognized in renal disease with the potential to enhance diagnostic accuracy, predict prognosis and monitor the course of disease. However, large-scale clinical trials enrolling heterogeneous patients are required to evaluate the clinical value of miRNAs.
Collapse
|
25
|
Sampath P, Periyasamy KM, Ranganathan UD, Bethunaickan R. Monocyte and Macrophage miRNA: Potent Biomarker and Target for Host-Directed Therapy for Tuberculosis. Front Immunol 2021; 12:667206. [PMID: 34248945 PMCID: PMC8267585 DOI: 10.3389/fimmu.2021.667206] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
The end TB strategy reinforces the essentiality of readily accessible biomarkers for early tuberculosis diagnosis. Exploration of microRNA (miRNA) and pathway analysis opens an avenue for the discovery of possible therapeutic targets. miRNA is a small, non-coding oligonucleotide characterized by the mechanism of gene regulation, transcription, and immunomodulation. Studies on miRNA define their importance as an immune marker for active disease progression and as an immunomodulator for innate mechanisms, such as apoptosis and autophagy. Monocyte research is highly advancing toward TB pathogenesis and biomarker efficiency because of its innate and adaptive response connectivity. The combination of monocytes/macrophages and their relative miRNA expression furnish newer insight on the unresolved mechanism for Mycobacterium survival, exploitation of host defense, latent infection, and disease resistance. This review deals with miRNA from monocytes, their relative expression in different disease stages of TB, multiple gene regulating mechanisms in shaping immunity against tuberculosis, and their functionality as biomarker and host-mediated therapeutics. Future collaborative efforts involving multidisciplinary approach in various ethnic population with multiple factors (age, gender, mycobacterial strain, disease stage, other chronic lung infections, and inflammatory disease criteria) on these short miRNAs from body fluids and cells could predict the valuable miRNA biosignature network as a potent tool for biomarkers and host-directed therapy.
Collapse
Affiliation(s)
- Pavithra Sampath
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | | - Uma Devi Ranganathan
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | |
Collapse
|
26
|
Zhao M, He X, Yang J, Feng Y, Wang H, Shao Z, Xing L. Aberrant microRNA expression in B lymphocytes from patients with primary warm autoimmune haemolytic anaemia. Autoimmunity 2021; 54:264-274. [PMID: 34044675 DOI: 10.1080/08916934.2021.1931842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To screen and analyze the micro-Ribonucleic Acid (miRNA) expression profile in B lymphocytes from patients with autoimmune haemolytic anaemia (AIHA) using high-throughput sequencing. METHODS Twelve patients with warm autoimmune haemolytic anaemia (wAIHA) and twelve healthy controls (HCs) were enrolled. CD19+ B lymphocytes were isolated and purified using magnetic activated cell sorting (MACS). RNA was subsequently extracted from these cells and a small RNA library was created. The miRNA expression profile was analyzed using Beijing Genomics Institute Sequencing 500 (BGISEQ-500), and stem-loop real-time quantitative PCR (stem-loop qRT-PCR) was used to verify the sequencing results. Downstream target genes of the differentially expressed miRNAs were predicted using miRanda and TargetScan online software, and GO functional enrichment and pathway enrichment analyses were performed on these genes. RESULTS Compared with HCs, 178 upregulated and 143 downregulated miRNAs were identified in wAIHA patients, and stem-loop qRT-PCR of four randomly selected differentially expressed miRNAs verified the sequencing results. Ninety-five significantly enriched GO terms and eighty-five significantly enriched pathways were identified. Genes targeted by differentially expressed miRNAs were found to be mainly involved in the regulation of signal transduction, metabolic processes, immune reactions, and neoplastic disease development. CONCLUSION The expression of miRNAs in B lymphocytes from patients with primary wAIHA was deregulated, and this phenomenon may be involved in the pathogenesis of wAIHA.
Collapse
Affiliation(s)
- Manjun Zhao
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| | - Xin He
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| | - Jin Yang
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| | - Yingying Feng
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| | - Huaquan Wang
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| | - Zonghong Shao
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| | - Limin Xing
- Department of Hematology, General Hospital Tianjin Medical University, Tianjin, China
| |
Collapse
|
27
|
Liu F, Liang Y, Zhao Y, Chen L, Wang X, Zhang C. Meta-analysis of association of microRNAs genetic variants with susceptibility to rheumatoid arthritis and systemic lupus erythematosus. Medicine (Baltimore) 2021; 100:e25689. [PMID: 33907143 PMCID: PMC8084041 DOI: 10.1097/md.0000000000025689] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 02/28/2021] [Accepted: 04/07/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND An increasing body of studies has investigated that genetic polymorphisms in microRNA (miRNA) may be related to susceptibility to rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). However, some results remain controversial. Thus, a meta-analysis was embarked on assessing whether some miRNA polymorphisms are associated with the risk of RA and SLE. METHODS Relevant studies were acquired on PubMed, Web of Science, Cochrane Library, CNKI, and Embase electronic databases from inception to December 2019. The strength of the association of miRNA polymorphisms with the risk of RA and SLE was assessed by odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Eligible 20 articles (36 studies) involving 5 miRNAs were enrolled in the meta-analysis. For RA, the polled result showed that there was no significant relationship between miR-146a rs2910164 and RA, but subgroup analysis based on ethnicity demonstrated that CC genotype may be a genetic protect factor for RA in Caucasians (CC vs CG+GG, OR = 0.825, 95% CI: 0.684-0.996, Pz = .045, Ph = .166). Besides, statistical significance of miR-499 rs3746444 (T/C) with susceptibility to RA was observed as well in the overall population, and the association was only significant in Caucasians but not Asians. For SLE, the associations of miR-146a rs2431697 T allele/T-carrier with increased risk of SLE were observed. CONCLUSIONS Our results highlight that miR-499 rs3746444 may contribute to RA susceptibility, particularly in Caucasians. In addition, CC genotype in miR-146a rs2910164 may act as a protector of RA in Caucasians. For SLE, miR-146a rs2431697 (C/T) is most likely to the increased the risk of SLE. These findings do not support the genetic association between miR-196a2 rs11614913 and RA/SLE susceptibility, as well as the association of miR-146a rs2910164, miR-146a rs57095329, miR-499 rs3746444 with SLE.
Collapse
Affiliation(s)
- Fengzhen Liu
- Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi
| | - Yahang Liang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medical, Shandong University, Jinan, Shandong, China
| | - Yu Zhao
- Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi
| | - Lili Chen
- Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi
| | - Xiaolin Wang
- Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi
| | - Chunquan Zhang
- Department of Ultrasound, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi
| |
Collapse
|
28
|
Gachpazan M, Akhlaghipour I, Rahimi HR, Saburi E, Mojarrad M, Abbaszadegan MR, Moghbeli M. Genetic and molecular biology of systemic lupus erythematosus among Iranian patients: an overview. AUTO- IMMUNITY HIGHLIGHTS 2021; 12:2. [PMID: 33516274 PMCID: PMC7847600 DOI: 10.1186/s13317-020-00144-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a clinicopathologically heterogeneous chronic autoimmune disorder affecting different organs and tissues. It has been reported that there is an increasing rate of SLE incidence among Iranian population. Moreover, the Iranian SLE patients have more severe clinical manifestations compared with other countries. Therefore, it is required to introduce novel methods for the early detection of SLE in this population. Various environmental and genetic factors are involved in SLE progression. MAIN BODY In present review we have summarized all of the reported genes which have been associated with clinicopathological features of SLE among Iranian patients. CONCLUSIONS Apart from the reported cytokines and chemokines, it was interestingly observed that the apoptosis related genes and non-coding RNAs were the most reported genetic abnormalities associated with SLE progression among Iranians. This review clarifies the genetics and molecular biology of SLE progression among Iranian cases. Moreover, this review paves the way of introducing an efficient panel of genetic markers for the early detection and better management of SLE in this population.
Collapse
Affiliation(s)
- Meisam Gachpazan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Wu Y, Li Q, Zhang R, Dai X, Chen W, Xing D. Circulating microRNAs: Biomarkers of disease. Clin Chim Acta 2021; 516:46-54. [PMID: 33485903 DOI: 10.1016/j.cca.2021.01.008] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
MicroRNAs are a class of endogenous noncoding single-stranded RNA molecules with approximately 20-24 nucleotides and are associated with a broad range of biological processes. Researchers found that microRNAs are abundant in tissues, and more importantly, there are also trace circulating microRNAs that exist in biological fluids. In recent years, circulating microRNAs had emerged as promising diagnostic and prognostic biomarkers for the noninvasive detection of diseases with high specificity and sensitivity. More importantly, specific microRNA expression signatures reflect not only the existence of early-stage diseases but also the dynamic development of advanced-stage diseases, disease prognosis prediction, and drug resistance. To date, an increasing number of potential miRNA biomarkers have been reported, but their practical application prospects are still unclear. Therefore, microRNAs, as potential diagnostic and prognostic biomarkers in a variety of diseases, need to be updated, as they are of great importance in the diagnosis, prognosis and prediction of therapeutic responses. In this review, we summary our current understanding of microRNAs as potential biomarkers in the major diseases (e.g., cancers and cardio-cerebrovascular diseases), which provide the basis for the design of diagnosis and treatment plan and the improvement of the cure rate.
Collapse
Affiliation(s)
- Yudong Wu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Qian Li
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Renshuai Zhang
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Xiaoli Dai
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Wujun Chen
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
30
|
Nijakowski K, Surdacka A. Salivary Biomarkers for Diagnosis of Inflammatory Bowel Diseases: A Systematic Review. Int J Mol Sci 2020; 21:ijms21207477. [PMID: 33050496 PMCID: PMC7589027 DOI: 10.3390/ijms21207477] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Saliva as a biological fluid has a remarkable potential in the non-invasive diagnostics of several systemic disorders. Inflammatory bowel diseases are chronic inflammatory disorders of the gastrointestinal tract. This systematic review was designed to answer the question “Are salivary biomarkers reliable for the diagnosis of inflammatory bowel diseases?”. Following the inclusion and exclusion criteria, eleven studies were included (according to PRISMA statement guidelines). Due to their heterogeneity, the potential salivary markers for IBD were divided into four groups: oxidative status markers, inflammatory cytokines, microRNAs and other biomarkers. Active CD patients manifest decreased activity of antioxidants (e.g., glutathione, catalase) and increased lipid peroxidation. Therefore, malondialdehyde seems to be a good diagnostic marker of CD. Moreover, elevated concentrations of proinflammatory cytokines (such as interleukin 1β, interleukin 6 or tumour necrosis factor α) are associated with the activity of IBD. Additionaly, selected miRNAs are altered in saliva (overexpressed miR-101 in CD; overexpressed miR-21, miR-31, miR-142-3p and underexpressed miR-142-5p in UC). Among other salivary biomarkers, exosomal PSMA7, α-amylase and calprotectin are detected. In conclusion, saliva contains several biomarkers which can be used credibly for the early diagnosis and regular monitoring of IBD. However, further investigations are necessary to validate these findings, as well as to identify new reliable salivary biomarkers.
Collapse
|
31
|
Mathias LM, Stohl W. Systemic lupus erythematosus (SLE): emerging therapeutic targets. Expert Opin Ther Targets 2020; 24:1283-1302. [PMID: 33034541 DOI: 10.1080/14728222.2020.1832464] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a heterogeneous clinical presentation whose etiologies are multifactorial. A myriad of genetic, hormonal, immunologic, and environmental factors contribute to its pathogenesis, and its diverse biological basis and phenotypic presentations make development of therapeutics difficult. In the past decade, tens of therapeutic targets with hundreds of individual candidate therapeutics have been investigated. AREAS COVERED We used a PUBMED database search through April 2020 to review the relevant literature. This review discusses therapeutic targets in the adaptive and innate immune systems, specifically: B cell surface antigens, B cell survival factors, Bruton's tyrosine kinase, costimulators, IL-12/IL-23, the calcineurin pathway, the JAK/STAT pathway, and interferons. EXPERT OPINION Our ever-improving understanding of SLE pathophysiology in the past decade has allowed us to identify new therapeutic targets. Multiple new drugs are on the horizon that target different elements of the adaptive and innate immune systems. SLE research remains challenging due to the heterogenous clinical presentation of SLE, confounding from background immunosuppressives being taken by SLE patients, animal models that inadequately recapitulate human disease, and imperfect and complicated outcome measures. Despite these limitations, research is promising and ongoing. The search for new therapies that target specific elements of SLE pathophysiology are discussed as well as key findings, pitfalls, and questions surrounding these targets.
Collapse
Affiliation(s)
- Lauren M Mathias
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine , Los Angeles, CA, USA
| | - William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine , Los Angeles, CA, USA
| |
Collapse
|
32
|
Yilmaz HO, Cebi AH, Kocak M, Ersoz HO, Ikbal M. MicroRNA Expression Levels in Patients with Hashimoto Thyroiditis: A Single Centre Study. Endocr Metab Immune Disord Drug Targets 2020; 21:1066-1072. [PMID: 32957899 DOI: 10.2174/1871530320999200918142429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To determine the circulatory miRNA expression levels in patients with Hashimoto thyroiditis (HT) at the time of diagnosis and follow-up period compared with healthy controls. METHODS We collected blood samples from 34 patients with HT (4 males and 30 females) at the time of first diagnosis (Group P) and euthyroid period (Group E). Thirty-three healthy controls (Group H) blood samples were also included in the study. Expression levels of five different circulating miRNAs (miR-22, miR-141, miR-155, miR-375, miR-451) were evaluated using real-time polymerase chain reaction. RESULTS There was a significant difference in miR-375 levels between the groups P and H. Also, for miR-451, there was a significant difference between the P and E groups. Finally, there was a moderate positive correlation between thyroid-stimulating hormone values and miR-22 expression levels for the P group. CONCLUSION miRNAs have important roles at all stages of the diseases. More studies must be performed in all thyroid diseases and autoimmune diseases, including HT.
Collapse
Affiliation(s)
- Hale O Yilmaz
- Department of Medical Genetics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Alper H Cebi
- Department of Medical Genetics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mustafa Kocak
- Department of Endocrinology and Metabolism, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Halil O Ersoz
- Department of Endocrinology and Metabolism, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mevlit Ikbal
- Department of Medical Genetics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
33
|
Omidi F, Hosseini SA, Ahmadi A, Hassanzadeh K, Rajaei S, Cesaire HM, Hosseini V. Discovering the signature of a lupus-related microRNA profile in the Gene Expression Omnibus repository. Lupus 2020; 29:1321-1335. [PMID: 32723063 DOI: 10.1177/0961203320944473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lupus is one of the most prevalent systemic autoimmune diseases. It is a multifactorial disease in which genetic, epigenetic and environmental factors play significant roles. The pathogenesis of lupus is not yet well understood. However, deregulation of microRNAs (miRNAs) - one of the post-transcriptional regulators of genes - can contribute to the development of autoimmune diseases. Over the last two decades, advances in the profiling of miRNA using microarray have received much attention, and it has been demonstrated that miRNAs play a regulatory role in the pathogenesis of lupus. Therefore, dysregulated miRNAs can be considered as promising diagnostic biomarkers for lupus. This article is an overview of lupus-related miRNA profiling studies and arrays in the Gene Expression Omnibus (GEO) database. The aims of our study were to widen current knowledge of known dysregulated miRNAs as potential biomarkers of SLE and to introduce a bioinformatics approach to using microarray data and finding novel miRNA and gene candidates for further study. We identified hsa-miR-4709-5p, hsa-miR-140, hsa-miR-145, hsa-miR-659, hsa-miR-134, hsa-miR-150, hsa-miR-584, hsa-miR-409 and hsa-miR-152 as potential biomarkers by integrated bioinformatics analysis.
Collapse
Affiliation(s)
- Forouzan Omidi
- Department of Immunology, School of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sayed Abdolhakim Hosseini
- Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Hosseini Nasab Medical Laboratory, Sanandaj, Iran
| | - Abbas Ahmadi
- Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Cellular and Molecular Research Centre, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Kambiz Hassanzadeh
- Cellular and Molecular Research Centre, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Shima Rajaei
- HelthWeX Clinical Research Co., Ltd, Tehran, Iran
| | | | - Vahedeh Hosseini
- Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Cellular and Molecular Research Centre, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
34
|
Yang Y, Hou Y, Li J, Zhang F, Du Q. Characterization of antiapoptotic microRNAs in primary Sjögren's syndrome. Cell Biochem Funct 2020; 38:1111-1118. [PMID: 32575162 DOI: 10.1002/cbf.3569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 02/02/2023]
Abstract
During the development of primary Sjögren's syndrome (pSS), aberrant expression of autoantigen is a hallmark event. To explore the regulation of autoantigen tripartite motif containing 21 (Ro/SSA, TRIM21), microRNA profiling was performed in our previous study. In which, two TRIM21-targeting microRNAs were identified, namely miR-1207-5p and miR-4695-3p. To further pursue their roles in the development of pSS, assays were performed with cultured human submandibular gland (HSG) cells, and salivary gland tissues. Results showed that transfection of miR-1207-5p or miR-4695-3p mimics down-regulated not only the expression of TRIM21, but also the levels of pro-apoptotic genes B cell lymphoma 2 associated X (BAX), Caspase 9 (CASP-9) and Caspase 8 (CASP-8). This finally led to antiapoptotic phenotypes in HSG cells. Consistent with the antiapoptotic activity, transfection of microRNA inhibitors up-regulated the expression of TRIM21 and led to a pro-apoptotic phenotype. These therefore propose miR-1207-5p and miR-4695-3p as two antiapoptotic microRNAs functioning through apoptosis pathway. Supporting this speculation, assays performed with salivary gland tissues revealed down-regulation of miR-1207-5p and miR-4695-3p, as well as up-regulation of TRIM21 and pro-apoptotic CASP-8 gene in pSS samples. SIGNIFICANCE OF THE STUDY: For pSS patients, apoptosis of acinar and ductal epithelial cells has been proposed to be a potential mechanism that impairs the secretion of salivary glands. In our study, two autoantigen-targeting microRNAs were characterized as antiapoptotic microRNAs functioning through apoptosis pathway, which may be potential targets for the treatment of pSS.
Collapse
Affiliation(s)
- Ying Yang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingzi Hou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jinghui Li
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fangming Zhang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Quan Du
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
35
|
Hong SM, Liu C, Yin Z, Wu L, Qu B, Shen N. MicroRNAs in Systemic Lupus Erythematosus: a Perspective on the Path from Biological Discoveries to Clinical Practice. Curr Rheumatol Rep 2020; 22:17. [PMID: 32405712 DOI: 10.1007/s11926-020-00895-7] [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] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW MicroRNAs (miRNAs) play essential roles in immune abnormalities and organ damage of systemic lupus erythematosus (SLE). Current findings have indicated potential clinical applications of miRNAs for combating SLE. Here, we review recent evidence which support the notions that miRNAs can be novel biomarkers and therapeutic agents for SLE. RECENT FINDINGS Following years of the studies of the expression patterns of miRNAs in both peripheral blood cells and body fluids, such as plasma and urine, several miRNAs or miRNA combinations have been associated with disease activity and specific organ damage. In depth analysis reveals complex and multiple roles of certain miRNAs in the pathogenesis of SLE. Manipulating miRNA expression shows in vivo therapeutic effects in lupus mouse models. MiRNAs contribute to the immune disorders and organ damage in SLE. MiRNA based biomarkers and therapies have the potential to be viable options for the treatment of SLE.
Collapse
Affiliation(s)
- Soon-Min Hong
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, China
| | - Can Liu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, 518040, China
| | - Lingling Wu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, China
| | - Bo Qu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, China. .,Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, 518040, China.
| | - Nan Shen
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, China. .,Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, 518040, China. .,Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA. .,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200032, China.
| |
Collapse
|
36
|
Berlinberg A, Kuhn KA. Molecular Biology Approaches to Understanding Spondyloarthritis. Rheum Dis Clin North Am 2020; 46:203-211. [PMID: 32340696 DOI: 10.1016/j.rdc.2020.01.001] [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] [Indexed: 12/12/2022]
Abstract
New and emerging molecular techniques are expanding understanding of the pathophysiology of spondyloarthritis (SpA). Genome-wide association studies identified novel pathways in antigen processing and presentation as well as helper T cell type 17 (TH17) immunity associated with SpA. Immune cell profiling techniques have supported TH17 immune responses and increasingly are revealing intestinal mucosal immune cells as associated with disease. Emerging technologies in epigenetics, transcriptomics, microbiome, and proteomics/metabolomics are adding to these, refining disease pathways and potentially identifying biomarkers for diagnosis and treatment responses. This review describes many of the new molecular techniques that are being utilized to investigate SpA.
Collapse
Affiliation(s)
- Adam Berlinberg
- Division of Rheumatology, University of Colorado School of Medicine, 1775 Aurora Court Mail Stop B115, Aurora, CO 80045, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado School of Medicine, 1775 Aurora Court Mail Stop B115, Aurora, CO 80045, USA.
| |
Collapse
|
37
|
Kotyla PJ, Islam MA. MicroRNA (miRNA): A New Dimension in the Pathogenesis of Antiphospholipid Syndrome (APS). Int J Mol Sci 2020; 21:ijms21062076. [PMID: 32197340 PMCID: PMC7139820 DOI: 10.3390/ijms21062076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded, endogenous RNA molecules that play a significant role in the regulation of gene expression as well as cell development, differentiation, and function. Recent data suggest that these small molecules are responsible for the regulation of immune responses. Therefore, they may act as potent modulators of the immune system and play an important role in the development of several autoimmune diseases. Antiphospholipid syndrome (APS) is an autoimmune systemic disease characterized by venous and/or arterial thromboses and/or recurrent fetal losses in the presence of antiphospholipid antibodies (aPLs). Several lines of evidence suggest that like other autoimmune disorders, miRNAs are deeply involved in the pathogenesis of APS, interacting with the function of innate and adaptive immune responses. In this review, we characterize miRNAs in the light of having a functional role in the immune system and autoimmune responses focusing on APS. In addition, we also discuss miRNAs as potential biomarkers and target molecules in treating APS.
Collapse
Affiliation(s)
- Przemysław J. Kotyla
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Faculty in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
- Correspondence: (P.J.K.); (M.A.I.)
| | - Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Correspondence: (P.J.K.); (M.A.I.)
| |
Collapse
|
38
|
Lopez-Pedrera C, Barbarroja N, Patiño-Trives AM, Luque-Tévar M, Torres-Granados C, Aguirre-Zamorano MA, Collantes-Estevez E, Pérez-Sánchez C. Role of microRNAs in the Development of Cardiovascular Disease in Systemic Autoimmune Disorders. Int J Mol Sci 2020; 21:E2012. [PMID: 32188016 PMCID: PMC7139533 DOI: 10.3390/ijms21062012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid Arthritis (RA), Systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS) are the systemic autoimmune diseases (SADs) most associated with an increased risk of developing cardiovascular (CV) events. Cardiovascular disease (CVD) in SADs results from a complex interaction between traditional CV-risk factors, immune deregulation and disease activity. Oxidative stress, dyslipidemia, endothelial dysfunction, inflammatory/prothrombotic mediators (cytokines/chemokines, adipokines, proteases, adhesion-receptors, NETosis-derived-products, and intracellular-signaling molecules) have been implicated in these vascular pathologies. Genetic and genomic analyses further allowed the identification of signatures explaining the pro-atherothrombotic profiles in RA, SLE and APS. However, gene modulation has left significant gaps in our understanding of CV co-morbidities in SADs. MicroRNAs (miRNAs) are emerging as key post-transcriptional regulators of a suite of signaling pathways and pathophysiological effects. Abnormalities in high number of miRNA and their associated functions have been described in several SADs, suggesting their involvement in the development of atherosclerosis and thrombosis in the setting of RA, SLE and APS. This review focusses on recent insights into the potential role of miRNAs both, as clinical biomarkers of atherosclerosis and thrombosis in SADs, and as therapeutic targets in the regulation of the most influential processes that govern those disorders, highlighting the potential diagnostic and therapeutic properties of miRNAs in the management of CVD.
Collapse
|
39
|
Song W, Tang D, Chen D, Zheng F, Huang S, Xu Y, Yu H, He J, Hong X, Yin L, Liu D, Dai W, Dai Y. Advances in applying of multi-omics approaches in the research of systemic lupus erythematosus. Int Rev Immunol 2020; 39:163-173. [PMID: 32138562 DOI: 10.1080/08830185.2020.1736058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Wencong Song
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Deheng Chen
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Fengping Zheng
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Shaoying Huang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Yong Xu
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Haiyan Yu
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Jingquan He
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Xiaoping Hong
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongzhou Liu
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, TX, USA
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
40
|
Xu H, Chen W, Zheng F, Tang D, Liu D, Wang G, Xu Y, Yin L, Zhang X, Dai Y. Reconstruction and analysis of the aberrant lncRNA-miRNA-mRNA network in systemic lupus erythematosus. Lupus 2020; 29:398-406. [PMID: 32070185 DOI: 10.1177/0961203320908927] [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] [Indexed: 12/13/2022]
Abstract
OBJECTIVE A new perspective of determining the pathophysiology of systemic lupus erythematosus (SLE) development is required. The current study explores the aberrant expression of long non-coding RNAs (lncRNA), microRNA (miRNA) and mRNA. The study further constructs and analyses the lncRNA-miRNA-mRNA network to elucidate their gene regulation roles in SLE. METHOD We extracted mRNA, lncRNA and miRNA from the whole venous blood of 20 SLE patients and 20 normal control (NC) healthy individuals. A lncRNA-mRNA-miRNA network in SLE was constructed using a bioinformatics approach. Subsequently, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed using the Cytoscape plug-in BinGo, the DAVID database and Cytoscape software to explore the function of mRNAs in this network. RESULT A total of 855 mRNA, 7311 lncRNA and 134 miRNA with differentially expressed profiles were identified. Meanwhile, we established a competing endogenous RNA (ceRNA) subnetwork composed of 52 differentially expressed lncRNAs (DElncRNAs), seven differentially expressed miRNAs and 10 differentially expressed mRNAs. We extracted the subnetwork from the ceRNA network and found that three novel miRNAs were key: hsa-miR-145, hsa-miR-17 and hsa-miR-143. We also deduced that the DElncRNAs MIAT and NEAT1 might play crucial roles in the pathogenesis of SLE. The results were verified by bioinformatics analysis. CONCLUSION Our results provide a novel perspective for studying lncRNA-related and miRNA-related ceRNA networks in SLE.
Collapse
Affiliation(s)
- H Xu
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - W Chen
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - F Zheng
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - D Tang
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - D Liu
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - G Wang
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - Y Xu
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| | - L Yin
- Division of Nephrology, The First Affiliated Hospital, Jinan University, Guangzhou, PR China
| | - X Zhang
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, PR China
| | - Y Dai
- Clinical Medical Research Centre, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, PR China
| |
Collapse
|
41
|
Kim YJ, Yeon Y, Lee WJ, Shin YU, Cho H, Sung YK, Kim DR, Lim HW, Kang MH. Comparison of MicroRNA Expression in Tears of Normal Subjects and Sjögren Syndrome Patients. Invest Ophthalmol Vis Sci 2020; 60:4889-4895. [PMID: 31752018 DOI: 10.1167/iovs.19-27062] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Deregulated expression of several microRNAs (miRNAs) in sera or salivary glands of patients with Sjögren syndrome (SS) has been reported. However, none have investigated miRNAs in samples that can represent lacrimal glands. We compared the miRNAs expression in the tears of SS patients and healthy controls. Moreover, we investigated the correlation between miRNAs expression and ocular staining score (OSS). Methods Individual tear samples were collected from 18 SS patients and 8 age-matched controls. Clinical ophthalmologic assessments included Schirmer I test, tear film breakup time (tBUT), and OSS. The expression of 43 different miRNAs in tears was measured using real-time polymerase chain reaction, and compared between the SS patients and controls. And we also compared between the three groups of control, primary SS, and secondary SS patients. The correlation between the miRNA expression and OSS was evaluated. Results The expression levels of miR-16-5p, miR-34a-5p, miR-142-3p, and miR-223-3p were significantly upregulated in patients with SS when compared with those in the control group (P < 0.05). The expression of 10 miRNAs (miR-30b-5p, miR-30c-5p, miR-30d-5p, miR-92a-3p, miR-134-5p, miR-137, miR-302d-5p, miR-365b-3p, miR-374c-5p, miR-487b-3p) was significantly downregulated in the SS patients (P < 0.05). Eight miRNAs showed statistically significant differences between the three groups of control, primary SS and secondary SS. All 14 miRNAs with significant differences in SS patients and control group were not significantly correlated with OSSs. Conclusions The 14 differentially expressed miRNAs may be involved in the pathogenesis of SS, in particular, related to autoimmunity and neuropathy.
Collapse
Affiliation(s)
- Yu Jeong Kim
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yeji Yeon
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Won June Lee
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yong Un Shin
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Heeyoon Cho
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yoon-Kyoung Sung
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Republic of Korea
| | - Du Roo Kim
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Han Woong Lim
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Min Ho Kang
- Department of Ophthalmology, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea.,Wilmer Eye Institute, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| |
Collapse
|
42
|
MicroRNA Expression in Cutaneous Lupus: A New Window to Understand Its Pathogenesis. Mediators Inflamm 2019; 2019:5049245. [PMID: 32082077 PMCID: PMC7012207 DOI: 10.1155/2019/5049245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/28/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023] Open
Abstract
Background The role of miRNAs in the pathogenesis of cutaneous lupus has not been studied. Objective It was to assess the levels of a selected panel of circulating miRNAs that could be involved in the regulation of the immune response, inflammation, and fibrosis in cutaneous lupus. Methods It was a cross-sectional study. We included 22 patients with subacute (SCLE) and 20 with discoid (DLE) lesions, and 19 healthy donors (HD). qRT-PCR for miRNA analysis, flow cytometry in peripheral blood, and skin immunohistochemistry were performed to determine the distribution of CD4 T cells and regulatory cells and their correlation with circulating miRNAs. Results miR-150, miR-1246, miR-21, miR-23b, and miR-146 levels were downregulated in SCLE vs. HD. miR-150, miR-1246, and miR-21 levels were downregulated in DLE vs. HD. miR-150, miR-1246, and miR-21 levels were downregulated in DLE γ+ with miR-1246 in SCLE, whereas CD123+/CD196+/IDO+ cells were positively associated with miR-150 in DLE. In the tissue, CD4+/IL-4+ and CD20+/IL-10+ cells were positively associated with miR-21 and CD4+/IFN-γ+ with miR-1246 in SCLE, whereas CD123+/CD196+/IDO+ cells were positively associated with miR-150 in DLE. In the tissue, CD4+/IL-4+ and CD20+/IL-10+ cells were positively associated with miR-21 and CD4+/IFN-β, thyroid hormone, and cancer signaling pathways were shared between miR-21, miR-31, miR-23b, miR-146a, miR-1246, and miR-150. Conclusions A downregulation of miR-150, miR-1246, and miR-21 in both CLE varieties vs. HD. miR-150, miR-1246, and miR-21 levels were downregulated in DLE
Collapse
|
43
|
Huang W, Wu Y, Cheng D, He Z. Mechanism of epithelial‑mesenchymal transition inhibited by miR‑203 in non‑small cell lung cancer. Oncol Rep 2019; 43:437-446. [PMID: 31894278 PMCID: PMC6967097 DOI: 10.3892/or.2019.7433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 10/25/2019] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate whether miR-203 can inhibit transforming growth factor-β (TGF-β)-induced epithelial-mesenchymal transition (EMT), and the migration and invasion ability of non-small cell lung cancer (NSCLC) cells by targeting SMAD3. In the present study, the expression levels of miR-203, SMAD3 mRNA and protein in NSCLC tissues were examined, as well as their corresponding paracancerous samples. The miR-203 mimics and miR-203 inhibitor were transfected into the H226 cell line. RT-qPCR was used to assess the expression levels of E-cadherin, Snail, N-cadherin and vimentin mRNA, and western blotting was performed to detect the expression levels of p-SMAD2, SMAD2, p-SMAD3, SMAD3 and SMAD4. The cell migration and invasion abilities were detected by Transwell assays. The target site of SMAD3 was predicted by the combined action between miR-203 and dual luciferase. The results revealed that the RNA levels of miR-203, compared with paracancerous tissues, were decreased in NSCLC tissues, while SMAD3 mRNA and protein levels were upregulated, and miR-203 inhibited SMAD3 expression. Induction of TGF-β led to decreased E-cadherin mRNA levels, upregulation of Snail, N-cadherin and vimentin mRNA levels (P<0.05), and significant increase in cell migration and invasion, whereas transfection of miR-203 mimics reversed the aforementioned results (P<0.05). Conversely, miR-203 inhibitor could further aggravate the aforementioned results (P<0.05). Western blot results revealed that transfection of miR-203 mimics significantly reduced the protein expression of SMAD3 and p-SMAD3 (P<0.05). Furthermore, the results of the Dual-Luciferase assay revealed that miR-203 inhibited SMAD3 expression by interacting with specific regions of its 3′-UTR. Overall, a novel mechanism is revealed, in which, miR-203 can inhibit SMAD3 by interacting with specific regions of the 3′-UTR of SMAD3, thereby restraining TGF-β-induced EMT progression and migration and invasion of NSCLC cells.
Collapse
Affiliation(s)
- Weicong Huang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuanbo Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Dezhi Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhifeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
44
|
The pathogenesis of systemic lupus erythematosus: Harnessing big data to understand the molecular basis of lupus. J Autoimmun 2019; 110:102359. [PMID: 31806421 DOI: 10.1016/j.jaut.2019.102359] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease that causes damage to multiple organ systems. Despite decades of research and available murine models that capture some aspects of the human disease, new treatments for SLE lag behind other autoimmune diseases such as Rheumatoid Arthritis and Crohn's disease. Big data genomic assays have transformed our understanding of SLE by providing important insights into the molecular heterogeneity of this multigenic disease. Gene wide association studies have demonstrated more than 100 risk loci, supporting a model of multiple genetic hits increasing SLE risk in a non-linear fashion, and providing evidence of ancestral diversity in susceptibility loci. Epigenetic studies to determine the role of methylation, acetylation and non-coding RNAs have provided new understanding of the modulation of gene expression in SLE patients and identified new drug targets and biomarkers for SLE. Gene expression profiling has led to a greater understanding of the role of myeloid cells in the pathogenesis of SLE, confirmed roles for T and B cells in SLE, promoted clinical trials based on the prominent interferon signature found in SLE patients, and identified candidate biomarkers and cellular signatures to further drug development and drug repurposing. Gene expression studies are advancing our understanding of the underlying molecular heterogeneity in SLE and providing hope that patient stratification will expedite new therapies based on personal molecular signatures. Although big data analyses present unique interpretation challenges, both computationally and biologically, advances in machine learning applications may facilitate the ability to predict changes in SLE disease activity and optimize therapeutic strategies.
Collapse
|
45
|
Yan F, Meng W, Ye S, Zhang X, Mo X, Liu J, Chen D, Lin Y. MicroRNA‑146a as a potential regulator involved in the pathogenesis of atopic dermatitis. Mol Med Rep 2019; 20:4645-4653. [PMID: 31545496 PMCID: PMC6797935 DOI: 10.3892/mmr.2019.10695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/23/2019] [Indexed: 01/27/2023] Open
Abstract
Previous studies have demonstrated that microRNA (miR)‑146a is involved in the inflammatory response of atopic dermatitis (AD). The aim of the present study was to investigate the expression of miR‑146a in the serum of patients with AD and in skin lesions of AD animal models. In addition, we aimed to predict and verify the target genes of miR‑146a. miR‑146a expression was measured in AD patient serum via reverse transcription‑quantitative PCR. T‑helper (Th)1 [CD4+; interferon (IFN)‑γ+] and Th2 [CD4+; interleukin (IL)‑4+] expression in peripheral blood mononuclear cells was evaluated using flow cytometry. Following the establishment of a 2,4‑dinitrofluorobenzene‑induced C57BL/6 mouse AD model, Th1 (CD4+IFN‑γ+) and Th2 (CD4+IL‑4+) expression was analyzed in murine spleen cells via flow cytometry. Plasmids were transfected into 293T cells and at 48 h post‑transfection, cells were analyzed using a luciferase assay system. The results revealed that the AD group had a significantly lower Th1/Th2 ratio and a significantly higher miR‑146a expression compared with the control group (P<0.05). Furthermore, a decreased Th1/Th2 ratio and a significantly increased miR‑146a expression were observed in the model group compared with the control group (P<0.01). We also conducted a dual‑luciferase assay to determine whether small ubiquitin‑related modifier 1 (SUMO1) if the target gene of miR‑146a. We observed a ~30% decrease in the relative luciferase activity in cells containing the 3'‑untranslated region of SUMO1 + miR‑146a). The results of the luciferase assay indicated that may be a direct mRNA target of miR‑146a; however, the quantification of band density of SUMO1 expression following western blotting did not significantly differ. The development of animal models in AD research is of vital importance. The results revealed that miR‑146a may be a potential regulator involved in the pathogenesis of AD. Furthermore, the current study determined that miR‑146a could be a valuable marker of AD and thus, may be applied in the development of therapeutic strategies for treating AD.
Collapse
Affiliation(s)
- Fenggen Yan
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Weiwei Meng
- Department of Dermatology, The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Provincial Hospital of Chinese Medicine, Henan, Zhengzhou 450000, P.R. China
| | - Siqi Ye
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xiumei Mo
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Junfeng Liu
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Dacan Chen
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Ying Lin
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
46
|
De Angelis MT, Santamaria G, Parrotta EI, Scalise S, Lo Conte M, Gasparini S, Ferlazzo E, Aguglia U, Ciampi C, Sgura A, Cuda G. Establishment and characterization of induced pluripotent stem cells (iPSCs) from central nervous system lupus erythematosus. J Cell Mol Med 2019; 23:7382-7394. [PMID: 31536674 PMCID: PMC6815917 DOI: 10.1111/jcmm.14598] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022] Open
Abstract
Involvement of the central nervous system (CNS) is an uncommon feature in systemic lupus erythematosus (SLE), making diagnosis rather difficult and challenging due to the poor specificity of neuropathic symptoms and neurological symptoms. In this work, we used human‐induced pluripotent stem cells (hiPSCs) derived from CNS‐SLE patient, with the aim to dissect the molecular insights underlying the disease by gene expression analysis and modulation of implicated pathways. CNS‐SLE‐derived hiPSCs allowed us to provide evidence of Erk and Akt pathways involvement and to identify a novel cohort of potential biomarkers, namely CHCHD2, IDO1, S100A10, EPHA4 and LEFTY1, never reported so far. We further extended the study analysing a panel of oxidative stress‐related miRNAs and demonstrated, under normal or stress conditions, a strong dysregulation of several miRNAs in CNS‐SLE‐derived compared to control hiPSCs. In conclusion, we provide evidence that iPSCs reprogrammed from CNS‐SLE patient are a powerful useful tool to investigate the molecular mechanisms underlying the disease and to eventually develop innovative therapeutic approaches.
Collapse
Affiliation(s)
- Maria Teresa De Angelis
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Sara Gasparini
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Edoardo Ferlazzo
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Clara Ciampi
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Antonella Sgura
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| |
Collapse
|
47
|
Gnanakkumaar P, Murugesan R, Ahmed SSSJ. Gene Regulatory Networks in Peripheral Mononuclear Cells Reveals Critical Regulatory Modules and Regulators of Multiple Sclerosis. Sci Rep 2019; 9:12732. [PMID: 31484947 PMCID: PMC6726613 DOI: 10.1038/s41598-019-49124-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/20/2019] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a complex, demyelinating disease with the involvement of autoimmunity and neurodegeneration. Increasing efforts have been made towards identifying the diagnostic markers to differentiate the classes of MS from other similar neurological conditions. Using a systems biology approach, we constructed four types of gene regulatory networks (GRNs) involved in peripheral blood mononuclear cells (PBMCs). The regulatory strength of each GRN across primary progressive MS (PPMS), relapsing-remitting MS (RRMS), secondary progressive MS (SPMS), and control were evaluated by an integrity algorithm. Among the constructed GRNs (referred as TF_gene_miRNA), POU3F2_CDK6_hsa-miR-590-3p, MEIS1_CASC3_hsa-miR-1261, STAT3_OGG1_hsa-miR-298, and TCF4_FMR1_hsa-miR-301b were top-ranked and differentially regulated in all classes of MS compared to control. These GRNs showed potential involvement in regulating various molecular pathways such as interleukin, integrin, glypican, sphingosine phosphate, androgen, and Wnt signaling pathways. For validation, the qPCR analysis of the GRN components (TFs, gene, and miRNAs) in PBMCs of healthy controls (n = 30), RRMS (n = 14), PPMS (n = 13) and SPMS (n = 12) were carried out. Real-time expression analysis of GRNs showed a similar regulatory pattern as derived from our systems biology approach. Also, our study provided several novel GRNs that regulate unique and common molecular mechanisms between MS conditions. Hence, these regulatory components of GRNs will help to understand the disease mechanism across MS classes and further insight may though light towards diagnosis.
Collapse
Affiliation(s)
- Perumal Gnanakkumaar
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Kelambakkam, 603103, India
| | - Ram Murugesan
- Drug Discovery Lab, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, 603103, India
| | - Shiek S S J Ahmed
- Drug Discovery Lab, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, 603103, India.
| |
Collapse
|
48
|
Stokes MB, D'Agati VD. Classification of Lupus Nephritis; Time for a Change? Adv Chronic Kidney Dis 2019; 26:323-329. [PMID: 31733716 DOI: 10.1053/j.ackd.2019.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/13/2019] [Indexed: 12/28/2022]
Abstract
Renal biopsy plays a critical role in the diagnosis and management of kidney disease in patients with systemic lupus erythematosus. The current pathologic classification of lupus nephritis is widely accepted but remains a work in progress. We discuss the key challenges in lupus nephritis classification and review new approaches to improve clinical utility and prognostic value.
Collapse
|
49
|
Tsai DY, Hung KH, Chang CW, Lin KI. Regulatory mechanisms of B cell responses and the implication in B cell-related diseases. J Biomed Sci 2019; 26:64. [PMID: 31472685 PMCID: PMC6717636 DOI: 10.1186/s12929-019-0558-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022] Open
Abstract
Terminally differentiated B cell, the plasma cell, is the sole cell type capable of producing antibodies in our body. Over the past 30 years, the identification of many key molecules controlling B cell activation and differentiation has elucidated the molecular pathways for generating antibody-producing plasma cells. Several types of regulation modulating the functions of the important key molecules in B cell activation and differentiation add other layers of complexity in shaping B cell responses following antigen exposure in the absence or presence of T cell help. Further understanding of the mechanisms contributing to the proper activation and differentiation of B cells into antibody-secreting plasma cells may enable us to develop new strategies for managing antibody humoral responses during health and disease. Herein, we reviewed the effect of different types of regulation, including transcriptional regulation, post-transcriptional regulation and epigenetic regulation, on B cell activation, and on mounting memory B cell and antibody responses. We also discussed the link between the dysregulation of the abovementioned regulatory mechanisms and B cell-related disorders.
Collapse
Affiliation(s)
- Dong-Yan Tsai
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan
| | - Kuo-Hsuan Hung
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan
| | - Chia-Wei Chang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan.,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 110, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan. .,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 110, Taiwan.
| |
Collapse
|
50
|
Zhou C, Zhao L, Wang K, Qi Q, Wang M, Yang L, Sun P, Mu H. MicroRNA-146a inhibits NF-κB activation and pro-inflammatory cytokine production by regulating IRAK1 expression in THP-1 cells. Exp Ther Med 2019; 18:3078-3084. [PMID: 31572547 PMCID: PMC6755493 DOI: 10.3892/etm.2019.7881] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
MicroRNA (miR)-146a levels are reduced in peripheral blood mononuclear cells of patients with systemic lupus erythematosus (SLE); however, its function is not well understood. The present study investigated the role of miR-146a in the regulation of lipopolysaccharide (LPS)-induced inflammation in THP-1 cells. A miR-146a mimic and an inhibitor were used to overexpress and downregulate miR-146a expression, respectively. Reverse transcription-quantitative PCR and western blot analyses were performed to evaluate interleukin (IL)-1 receptor-associated kinase 1 (IRAK1) expression, and western blot analysis was applied to assess nuclear factor-κB activation by analyzing p65 subunit levels in the nucleus. To investigate the effects of miR-146a on LPS-induced inflammation, IL-6 and tumor necrosis factor-α (TNF-α) levels were also measured using ELISA. The results of the present study revealed thatmiR-146a overexpression significantly reduced IRAK1 expression, reduced p65 levels in the nucleus and reduced IL-6 and TNF-α levels in the supernatant of the cell culture medium of THP-1 cells following LPS treatment. Luciferase assays confirmed IRAK1 to be a direct target of miR-146a in THP-1 cells. In conclusion, miR-146a may regulate IRAK1 expression and inhibit the activation of inflammatory signals and secretion of pro-inflammatory cytokines. The present study revealed, at least in part, the mechanisms by which miR-146a regulate the inflammatory response in SLE.
Collapse
Affiliation(s)
- Chunlei Zhou
- Department of Medical Laboratory, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Lan Zhao
- Laboratory of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China.,Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin 300193, P.R. China
| | - Kai Wang
- Department of Medical Laboratory, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Qianru Qi
- Department of Medical Laboratory, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| | - Meng Wang
- Department of Medical Laboratory, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Lei Yang
- Department of Medical Laboratory, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Ping Sun
- Department of Medical Laboratory, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Hong Mu
- Department of Medical Laboratory, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| |
Collapse
|