1
|
Młynarska E, Badura K, Kurciński S, Sinkowska J, Jakubowska P, Rysz J, Franczyk B. The Role of MicroRNA in the Pathophysiology and Diagnosis of Viral Myocarditis. Int J Mol Sci 2024; 25:10933. [PMID: 39456716 PMCID: PMC11507602 DOI: 10.3390/ijms252010933] [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: 07/23/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Myocarditis is a non-ischemic condition with a heterogeneous etiology, clinical course and prognosis. The most common etiology of myocarditis are viral infections, whereas the most severe complications are acute and chronic heart failure and sudden cardiac death. The heterogeneous clinical course of the disease, as well as the availability and costs of diagnostic tools such as cardiac magnetic resonance and endomyocardial biopsy, hinder the diagnosis of myocarditis and its underlying cause. Non-coding RNAs such as micro-RNAs (miRNAs; miR) have been shown to be involved in the disease's pathophysiology; however, their potential in disease diagnosis and treatment should also be considered. Non-coding RNAs are RNAs that are not translated into proteins, and they have the ability to regulate several intracellular pathways. MiRNAs regulate gene expression by binding with their targets and inhibiting protein synthesis by interfering with the translation of coding genes or causing the degradation of messenger RNA. Several miRNAs, such as miR-1, -133, -21, -15, -98, -126, -155, -148, -203, -208, -221, -222, -203 and -590, have been shown to be involved in the pathophysiology of viral myocarditis (VMC), and some of them have been shown to have diagnostic abilities. This article summarizes the available data on miRNAs and their associations with VMC.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Krzysztof Badura
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Szymon Kurciński
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Julia Sinkowska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Paulina Jakubowska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
2
|
Grodzka O, Procyk G, Wrzosek M. A Narrative Review of Preclinical In Vitro Studies Investigating microRNAs in Myocarditis. Curr Issues Mol Biol 2024; 46:1413-1423. [PMID: 38392209 PMCID: PMC10887635 DOI: 10.3390/cimb46020091] [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: 01/03/2024] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
According to the World Health Organization's statement, myocarditis is an inflammatory myocardium disease. Although an endometrial biopsy remains the diagnostic gold standard, it is an invasive procedure, and thus, cardiac magnetic resonance imaging has become more widely used and is called a non-invasive diagnostic gold standard. Myocarditis treatment is challenging, with primarily symptomatic therapies. An increasing number of studies are searching for novel diagnostic biomarkers and potential therapeutic targets. Microribonucleic acids (miRNAs) are small, non-coding RNA molecules that decrease gene expression by inhibiting the translation or promoting the degradation of complementary mRNAs. Their role in different fields of medicine has been recently extensively studied. This review discusses all relevant preclinical in vitro studies regarding microRNAs in myocarditis. We searched the PubMed database, and after excluding unsuitable studies and clinical and preclinical in vivo trials, we included and discussed 22 preclinical in vitro studies in this narrative review. Several microRNAs presented altered levels in myocarditis patients in comparison to healthy controls. Moreover, microRNAs influenced inflammation, cell apoptosis, and viral replication. Finally, microRNAs were also found to determine the level of myocardial damage. Further studies may show the vital role of microRNAs as novel therapeutic agents or diagnostic/prognostic biomarkers in myocarditis management.
Collapse
Affiliation(s)
- Olga Grodzka
- Department of Neurology, Faculty of Medicine and Dentistry, Medical University of Warsaw, 80 Ceglowska St., 01-809 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 81 Żwirki i Wigury St., 02-091 Warsaw, Poland
| | - Grzegorz Procyk
- Doctoral School, Medical University of Warsaw, 81 Żwirki i Wigury St., 02-091 Warsaw, Poland
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1A Banacha St., 02-097 Warsaw, Poland
| | - Małgorzata Wrzosek
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha St., 02-097 Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, 1B Banacha St., 02-097 Warsaw, Poland
| |
Collapse
|
3
|
Procyk G, Grodzka O, Procyk M, Gąsecka A, Głuszek K, Wrzosek M. MicroRNAs in Myocarditis-Review of the Preclinical In Vivo Trials. Biomedicines 2023; 11:2723. [PMID: 37893097 PMCID: PMC10604573 DOI: 10.3390/biomedicines11102723] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Myocarditis is an inflammatory heart disease with viruses as the most common cause. Regardless of multiple studies that have recently been conducted, the diagnostic options still need to be improved. Although endomyocardial biopsy is known as a diagnostic gold standard, it is invasive and, thus, only sometimes performed. Novel techniques of cardiac magnetic resonance are not readily available. Therapy in viral infections is based mainly on symptomatic treatment, while steroids and intravenous immunoglobulins are used in autoimmune myocarditis. The effectiveness of neither of these methods has been explicitly proven to date. Therefore, novel diagnostic and therapeutic strategies are highly needed. MiRNAs are small, non-coding molecules that regulate fundamental cell functions, including differentiation, metabolism, and apoptosis. They present altered levels in different diseases, including myocarditis. Numerous studies investigating the role of miRNAs in myocarditis have already been conducted. In this review, we discussed only the original preclinical in vivo research. We eventually included 30 studies relevant to the discussed area. The altered miRNA levels have been observed, including upregulation and downregulation of different miRNAs in the mice models of myocarditis. Furthermore, the administration of mimics or inhibitors of particular miRNAs was shown to significantly influence inflammation, morphology, and function of the heart and overall survival. Finally, some studies presented prospective advantages in vaccine development.
Collapse
Affiliation(s)
- Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Olga Grodzka
- Department of Neurology, Faculty of Medicine and Dentistry, Medical University of Warsaw, Ceglowska 80, 01-809 Warsaw, Poland
| | - Marcelina Procyk
- Faculty of Biology and Biotechnology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Katarzyna Głuszek
- Collegium Medicum, Jan Kochanowski University of Kielce, 25-406 Kielce, Poland
| | - Małgorzata Wrzosek
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
4
|
Hu Z, Hua X, Mo X, Chang Y, Chen X, Xu Z, Tao M, Hu G, Song J. Inhibition of NETosis via PAD4 alleviated inflammation in giant cell myocarditis. iScience 2023; 26:107162. [PMID: 37534129 PMCID: PMC10391931 DOI: 10.1016/j.isci.2023.107162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 04/11/2023] [Accepted: 06/13/2023] [Indexed: 08/04/2023] Open
Abstract
Giant cell myocarditis (GCM) is a rare, usually rapidly progressive, and potentially fatal disease. Detailed inflammatory responses remain unknown, in particular the formation of multinucleate giant cells. We performed single-cell RNA sequencing analysis on 15,714 Cd45+ cells extracted from the hearts of GCM rats and normal rats. NETosis has been found to contribute to the GCM process. An inhibitor of NETosis, GSK484, alleviated GCM inflammation in vivo. MPO (a marker of neutrophils) and H3cit (a marker of NETosis) were expressed at higher levels in patients with GCM than in patients with DCM and healthy controls. Imaging mass cytometry analysis revealed that immune cell types within multinucleate giant cells included CD4+ T cells, CD8+ T cells, neutrophils, and macrophages but not B cells. We elucidated the role of NETosis in GCM pathogenesis, which may serve as a potential therapeutic target in the clinic.
Collapse
Affiliation(s)
- Zhan Hu
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Xiuxue Mo
- School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Zhenyu Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Pathology Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Mengtao Tao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| | - Gang Hu
- School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Tianjin 300071, China
| |
Collapse
|
5
|
Guo L, Cai Y, Wang B, Zhang F, Zhao H, Liu L, Tao L. Characterization of the circulating transcriptome expression profile and identification of novel miRNA biomarkers in hypertrophic cardiomyopathy. Eur J Med Res 2023; 28:205. [PMID: 37391825 PMCID: PMC10314611 DOI: 10.1186/s40001-023-01159-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM), one of the most common genetic cardiovascular diseases, but cannot be explained by single genetic factors. Circulating microRNAs (miRNAs) are stable and highly conserved. Inflammation and immune response participate in HCM pathophysiology, but whether the miRNA profile changes correspondingly in human peripheral blood mononuclear cells (PBMCs) with HCM is unclear. Herein, we aimed to investigate the circulating non-coding RNA (ncRNA) expression profile in PBMCs and identify potential miRNAs for HCM biomarkers. METHODS A Custom CeRNA Human Gene Expression Microarray was used to identify differentially expressed (DE) mRNAs, miRNAs, and ncRNAs (including circRNA and lncRNA) in HCM PBMCs. Weighted correlation network analysis (WGCNA) was used to identify HCM-related miRNA and mRNA modules. The mRNAs and miRNAs from the key modules were used to construct a co-expression network. Three separate machine learning algorithms (random forest, support vector machine, and logistic regression) were applied to identify potential biomarkers based on miRNAs from the HCM co-expression network. Gene Expression Omnibus (GEO) database (GSE188324) and experimental samples were used for further verification. Gene set enrichment analysis (GSEA) and competing endogenous RNA (ceRNA) network was used to determine the potential functions of the selected miRNAs in HCM. RESULTS We identified 1194 DE-mRNAs, 232 DE-miRNAs and 7696 DE-ncRNAs in HCM samples compared with normal controls from the microarray data sets. WGCNA identified key miRNA modules and mRNA modules evidently associated with HCM. We constructed a miRNA‒mRNA co-expression network based on these modules. A total of three hub miRNAs (miR-924, miR-98 and miR-1) were identified by random forest, and the areas under the receiver operator characteristic curves of miR-924, miR-98 and miR-1 were 0.829, 0.866, and 0.866, respectively. CONCLUSIONS We elucidated the transcriptome expression profile in PBMCs and identified three hub miRNAs (miR-924, miR-98 and miR-1) as potential biomarkers for HCM detection.
Collapse
Affiliation(s)
- Lanyan Guo
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China
| | - Yue Cai
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China
| | - Bo Wang
- Department of Ultrasound, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China
| | - Hang Zhao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China
| | - Liwen Liu
- Department of Ultrasound, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China.
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaan Xi, China.
| |
Collapse
|
6
|
Chimenti C, Magnocavallo M, Vetta G, Alfarano M, Manguso G, Ajmone F, Ballatore F, Costantino J, Ciaramella P, Severino P, Miraldi F, Lavalle C, Vizza CD. The Role of MicroRNA in the Myocarditis: a Small Actor for a Great Role. Curr Cardiol Rep 2023:10.1007/s11886-023-01888-5. [PMID: 37269474 DOI: 10.1007/s11886-023-01888-5] [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] [Accepted: 04/24/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE OF REVIEW Myocarditis is an inflammation of the myocardium secondary to a variety of agents such as infectious pathogens, toxins, drugs, and autoimmune disorders. In our review, we provide an overview of miRNA biogenesis and their role in the etiology and pathogenesis of myocarditis, evaluating future directions for myocarditis management. RECENT FINDINGS Advances in genetic manipulation techniques allowed to demonstrate the important role of RNA fragments, especially microRNAs (miRNAs), in cardiovascular pathogenesis. miRNAs are small non-coding RNA molecules that regulate the post-transcriptional gene expression. Advances in molecular techniques allowed to identify miRNA's role in pathogenesis of myocarditis. miRNAs are related to viral infection, inflammation, fibrosis, and apoptosis of cardiomyocytes, making them not only promising diagnostic markers but also prognostics and therapeutic targets in myocarditis. Of course, further real-world studies will be needed to assess the diagnostic accuracy and applicability of miRNA in the myocarditis diagnosis.
Collapse
Affiliation(s)
- Cristina Chimenti
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy.
| | - Michele Magnocavallo
- Cardiology Division, Arrhythmology Unit, S. Giovanni Calibita Hospital, Isola Tiberina, Rome, Italy
| | - Giampaolo Vetta
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Mesina, Italy
| | - Maria Alfarano
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Giulia Manguso
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Francesco Ajmone
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Federico Ballatore
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Jacopo Costantino
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Piera Ciaramella
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Paolo Severino
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Fabio Miraldi
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Carlo Lavalle
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Carmine Dario Vizza
- Clinical, Anestesiologic and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| |
Collapse
|
7
|
AbdelMassih A, Agha H, El-Saiedi S, El-Sisi A, El Shershaby M, Gaber H, Ismail HA, El-Husseiny N, Amin AR, ElBoraie A, Ayad A, Menshawey E, Sefein F, Osman II, Moursi M, Hanafy M, Abdelaziz MS, Arsanyous MB, Khaled-Ibn-El-Walid M, Tawfik MG, Habib M, Mansour ME, Ashraf M, Khattab MA, Alshehry N, Hafez N, ElDeeb NE, Ashraf N, Khalil N, AbdElSalam NI, Shebl N, Hafez NGA, Youssef NH, Bahnan O, Ismail P, Kelada P, Menshawey R, Saeed R, Husseiny RJ, Yasser R, Sharaf S, Adel V, Naeem Y, Nicola YNF, Kamel A, Hozaien R, Fouda R. The role of miRNAs in viral myocarditis, and its possible implication in induction of mRNA-based COVID-19 vaccines-induced myocarditis. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:267. [PMID: 36415483 PMCID: PMC9672617 DOI: 10.1186/s42269-022-00955-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Several reports of unheeded complications secondary to the current mass international rollout of SARS-COV-2 vaccines, one of which is myocarditis occurring with the FDA fully approved vaccine, Pfizer, and others. MAIN BODY OF THE ABSTRACT Certain miRNAs (non-coding RNA sequences) are involved in the pathogenesis in viral myocarditis, and those miRNAs are interestingly upregulated in severe COVID-19. We hypothesize that the use of mRNA-based vaccines may be triggering the release of host miRNAs or that trigger the occurrence of myocarditis. This is based on the finding of altered host miRNA expression promoting virus-induced myocarditis. SHORT CONCLUSION In conclusion, miRNAs are likely implicated in myocarditis associated with mRNA vaccines. Our hypothesis suggests the use of miRNA as a biomarker for the diagnosis of mRNA vaccine-induced myocarditis. Additionally, the interplay between viral miRNA and the host immune system could alter inflammatory profiles, hence suggesting the use of therapeutic inhibition to prevent such complications.
Collapse
Affiliation(s)
- Antoine AbdelMassih
- Pediatric Cardiology Unit, Pediatrics’ Department, Faculty of Medicine, Cairo University, P.O. Box 12411, Cairo, Egypt
- Pediatric Cardio-Oncology Clinic, Children Cancer Hospital of Egypt, Cairo, Egypt
| | - Hala Agha
- Pediatric Cardiology Unit, Pediatrics’ Department, Faculty of Medicine, Cairo University, P.O. Box 12411, Cairo, Egypt
| | - Sonia El-Saiedi
- Pediatric Cardiology Unit, Pediatrics’ Department, Faculty of Medicine, Cairo University, P.O. Box 12411, Cairo, Egypt
| | - Amal El-Sisi
- Pediatric Cardiology Unit, Pediatrics’ Department, Faculty of Medicine, Cairo University, P.O. Box 12411, Cairo, Egypt
| | - Meryam El Shershaby
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hanya Gaber
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Habiba-Allah Ismail
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nadine El-Husseiny
- Faculty of Dentistry, Cairo University, Cairo, Egypt
- Pixagon Graphic Design Agency, Cairo, Egypt
| | - Abeer Reda Amin
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Aly ElBoraie
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Aya Ayad
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Esraa Menshawey
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Fady Sefein
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ibrahim Ihab Osman
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mai Moursi
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maram Hanafy
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mariam Sherif Abdelaziz
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mariem Badr Arsanyous
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mariam Khaled-Ibn-El-Walid
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Marwa Gamal Tawfik
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Menna Habib
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mina Ehab Mansour
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mirette Ashraf
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Ayman Khattab
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nada Alshehry
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nada Hafez
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Naheel Essam ElDeeb
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nirvana Ashraf
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noha Khalil
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noheir Ismail AbdElSalam
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noura Shebl
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nouran Gamal Ali Hafez
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nourhan Hatem Youssef
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Odette Bahnan
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Passant Ismail
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Peter Kelada
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rahma Menshawey
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rana Saeed
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Reem Jalal Husseiny
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Reem Yasser
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Safa Sharaf
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Veronia Adel
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Youstina Naeem
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Youstina Nagy Farid Nicola
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Aya Kamel
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rafeef Hozaien
- Student and Internship Research Program (Research Accessibility Team), Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Raghda Fouda
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Deep Learning-Based Medical Data Association Rules to Explore the Connectivity and Regulation Mechanism of miRNA-mRNA Network in Myocarditis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9272709. [PMID: 36193199 PMCID: PMC9525760 DOI: 10.1155/2022/9272709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022]
Abstract
Acute, chronic myocarditis as myocardial localized or diffuse inflammation lesions is usually involving cardiac function in patients with severe adverse outcomes such as heart failure, sudden death, and no unified, but its pathogenesis clinical is mainly composed of a number of factors including infection and autoimmune defects, such as physical and chemical factors; therefore, it is of great significance to explore the regulation mechanism of myocarditis-related miRNA network connectivity and temperament for in-depth understanding of the pathogenesis of myocarditis and the direction of targeted therapy. Based on this, this study explored the miRNA network related to the pathogenesis of myocarditis through deep learning medical data association rules and analyzed its specific mechanism. The results showed that 39 upregulated miRNAs, 88 downregulated miRNAs, 109 upregulated differentially expressed miRNAs, and 589 downregulated mRNAs were obtained by data association through GSE126677 and GSE4172 databases. GO enrichment and KRGG enrichment analysis showed that the differentially expressed mRNAs were involved in the regulation of a variety of biological processes, cellular components, and molecular functions. At the same time, the miRNA with differentially expressed miRNAs and their corresponding mRNAs were connected to further clarify the specific molecular mechanism of the pathological changes of myocarditis by constructing miRNA-mRNA network. It provides effective potential molecular targets for subsequent treatment and diagnosis.
Collapse
|
9
|
Ghafouri-Fard S, Khoshbakht T, Hussen BM, Taheri M, Jamali E. The emerging role non-coding RNAs in B cell-related disorders. Cancer Cell Int 2022; 22:91. [PMID: 35193592 PMCID: PMC8862212 DOI: 10.1186/s12935-022-02521-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/10/2022] [Indexed: 12/17/2022] Open
Abstract
Long non-coding RNAs and microRNAs have recently attained much attention regarding their role in the development of B cell lineage as well as participation in the lymphomagenesis. These transcripts have a highly cell type specific signature which endows them the potential to be used as biomarkers for clinical situations. Aberrant expression of several non-coding RNAs has been linked with B cell malignancies and immune related disorders such as rheumatoid arthritis, systemic lupus erythematous, asthma and graft-versus-host disease. Moreover, these transcripts can alter response of immune system to infectious conditions. miR-7, miR-16-1, miR-15a, miR-150, miR-146a, miR-155, miR-212 and miR-132 are among microRNAs whose role in the development of B cell-associated disorders has been investigated. Similarly, SNHG14, MALAT1, CRNDE, AL133346.1, NEAT1, SMAD5-AS1, OR3A4 and some other long non-coding RNAs participate in this process. In the current review, we describe the role of non-coding RNAs in B cell malignancies.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Elena Jamali
- Department of Pathology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Mirna M, Paar V, Topf A, Kraus T, Sotlar K, Aigner A, Ewe A, Watzinger S, Podesser BK, Hackl M, Pistulli R, Hoppe UC, Kiss A, Lichtenauer M. A new player in the game: treatment with antagomiR-21a-5p significantly attenuates histological and echocardiographic effects of experimental autoimmune myocarditis. Cardiovasc Res 2022; 118:556-572. [PMID: 33483746 DOI: 10.1093/cvr/cvab015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/09/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Myocarditis is associated with formidable symptoms and increased risk of adverse outcomes. Current approaches mostly rely on symptomatic treatments, warranting novel concepts for clinical practice. The aim of this study was to investigate the microRNA (miRNA) expression profile of Balb/c mice with experimental autoimmune myocarditis (EAM), choose a representative miRNA to antagonize after review of available literature and test its effects on myocardial inflammation in vitro and in vivo. METHODS AND RESULTS Phase 1: EAM was induced in 12 male Balb/c mice, 10 animals served as controls. After sacrifice, next-generation sequencing (NGS) of the miRNA expression profile was performed. Based on these results, H9C2 cells and human ventricular cardiac fibroblasts exposed to lipopolysaccharide (LPS) were treated with the selected candidate antagomiR-21a-5p. Phase 2: EAM was induced in 48 animals. Thereof, 24 animals were either treated with antagomiR-21a-5p or negative control oligonucleotide in a nanoparticle formulation. Transthoracic echocardiography (TTE) was performed on Days 0, 7, 14, and 21. Histopathological examination was performed after sacrifice. Phase 1: EAM resulted in a significant up-regulation of 27 miRNAs, including miR-21a-5p (log2FC: 2.23, adj. P = 0.0026). Transfection with antagomiR-21a-5p resulted in a significant reduction of TNFα, IL-6, and collagen I in vitro. Phase 2: Treatment with antagomiR-21a-5p, formulated in polymeric nanoparticles for systemic injection, significantly attenuated myocardial inflammation (P = 0.001) and fibrosis (P = 0.013), as well as myocardial 'hypertrophy' on TTE. CONCLUSIONS Silencing of miR-21a-5p results in a significant reduction of the expression of pro-inflammatory cytokines in vitro, as well as a significant attenuation of inflammation, fibrosis and echocardiographic effects of EAM in vivo.
Collapse
Affiliation(s)
- Moritz Mirna
- Department of Cardiology, University Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Vera Paar
- Department of Cardiology, University Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Albert Topf
- Department of Cardiology, University Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Theo Kraus
- University Institute of Pathology, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Karl Sotlar
- University Institute of Pathology, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Achim Aigner
- Rudolf-Boehm-Institut for Pharmacology und Toxicology, Clinical Pharmacology, University of Leipzig, Faculty of Medicine, Haertelstraße 16-18, 04107 Leipzig, Germany
| | - Alexander Ewe
- Rudolf-Boehm-Institut for Pharmacology und Toxicology, Clinical Pharmacology, University of Leipzig, Faculty of Medicine, Haertelstraße 16-18, 04107 Leipzig, Germany
| | - Simon Watzinger
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | | | - Rudin Pistulli
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Muenster, Albert-Schweitzer-Strasse 33, 48149 Muenster, Germany
| | - Uta C Hoppe
- Department of Cardiology, University Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Michael Lichtenauer
- Department of Cardiology, University Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Muellner Hauptstrasse 48, 5020 Salzburg, Austria
| |
Collapse
|
11
|
Chang Y, Li X, Cheng Q, Hu Y, Chen X, Hua X, Fan X, Tao M, Song J, Hu S. Single-cell transcriptomic identified HIF1A as a target for attenuating acute rejection after heart transplantation. Basic Res Cardiol 2021; 116:64. [PMID: 34870762 DOI: 10.1007/s00395-021-00904-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/04/2021] [Accepted: 11/20/2021] [Indexed: 10/19/2022]
Abstract
Acute rejection (AR) is an important contributor to graft failure, which remains a leading cause of death after heart transplantation (HTX). The regulation of immune metabolism has become a new hotspot in the development of immunosuppressive drugs. In this study, Increased glucose metabolism of cardiac macrophages was found in patients with AR. To find new therapeutic targets of immune metabolism regulation for AR, CD45+ immune cells extracted from murine isografts, allografts, and untransplanted donor hearts were explored by single-cell RNA sequencing. Total 20 immune cell subtypes were identified among 46,040 cells. The function of immune cells in AR were illustrated simultaneously. Cardiac resident macrophages were substantially replaced by monocytes and proinflammatory macrophages during AR. Monocytes/macrophages in AR allograft were more active in antigen presentation and inflammatory recruitment ability, and glycolysis. Based on transcription factor regulation analysis, we found that the increase of glycolysis in monocytes/macrophages was mainly regulated by HIF1A. Inhibition of HIF1A could alleviate inflammatory cells infiltration in AR. To find out the effect of HIF1A on AR, CD45+ immune cells extracted from allografts after HIF1A inhibitor treatment were explored by single-cell RNA sequencing. HIF1A inhibitor could reduce the antigen presenting ability and pro-inflammatory ability of macrophages, and reduce the infiltration of Cd4+ and Cd8a+ T cells in AR. The expression of Hif1α in AR monocytes/macrophages was regulated by pyruvate kinase 2. Higher expression of HIF1A in macrophages was also detected in human hearts with AR. These indicated HIF1A may serve as a potential target for attenuating AR.
Collapse
Affiliation(s)
- Yuan Chang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.,The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xiangjie Li
- School of Statistics and Data Science, Nankai University, Tianjin, 300371, China.,The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Qi Cheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Ministry of Education, National Health Commission, Wuhan, 430000, China
| | - Yiqing Hu
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xiao Chen
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xiumeng Hua
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xuexin Fan
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Menghao Tao
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Jiangping Song
- The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China.
| | - Shengshou Hu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.,The Cardiomyopathy Research Group at Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| |
Collapse
|
12
|
Roberts LB, Kapoor P, Howard JK, Shah AM, Lord GM. An update on the roles of immune system-derived microRNAs in cardiovascular diseases. Cardiovasc Res 2021; 117:2434-2449. [PMID: 33483751 PMCID: PMC8562329 DOI: 10.1093/cvr/cvab007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVD) are a leading cause of human death worldwide. Over the past two decades, the emerging field of cardioimmunology has demonstrated how cells of the immune system play vital roles in the pathogenesis of CVD. MicroRNAs (miRNAs) are critical regulators of cellular identity and function. Cell-intrinsic, as well as cell-extrinsic, roles of immune and inflammatory cell-derived miRNAs have been, and continue to be, extensively studied. Several 'immuno-miRNAs' appear to be specifically expressed or demonstrate greatly enriched expression within leucocytes. Identification of miRNAs as critical regulators of immune system signalling pathways has posed the question of whether and how targeting these molecules therapeutically, may afford opportunities for disease treatment and/or management. As the field of cardioimmunology rapidly continues to advance, this review discusses findings from recent human and murine studies which contribute to our understanding of how leucocytes of innate and adaptive immunity are regulated-and may also regulate other cell types, via the actions of the miRNAs they express, in the context of CVD. Finally, we focus on available information regarding miRNA regulation of regulatory T cells and argue that targeted manipulation of miRNA regulated pathways in these cells may hold therapeutic promise for the treatment of CVD and associated risk factors.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Puja Kapoor
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Jane K Howard
- School of Life Course Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- Faculty of Biology, Medicine and Health, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
13
|
Exosomal microRNA-98-5p from hypoxic bone marrow mesenchymal stem cells inhibits myocardial ischemia-reperfusion injury by reducing TLR4 and activating the PI3K/Akt signaling pathway. Int Immunopharmacol 2021; 101:107592. [PMID: 34715573 DOI: 10.1016/j.intimp.2021.107592] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) are essential biomarkers during development of human diseases. We aimed to explore the role of hypoxia-induced bone marrow mesenchymal stem cells (BMSCs)-derived exosomal miR-98-5p in myocardial ischemia-reperfusion injury (MI/RI). METHODS BMSCs were isolated, cultured, stimulated by hypoxia and transfected with adenovirus expressing miR-98-5p. The exosomes were extracted from BMSCs and named as BMSC-exos. The rat MI/RI models were established by ligation of left anterior descending artery and were respectively injected. Then, hemodynamic indices, myocardial enzymes, oxidative stress factors, inflammatory factors, macrophage infiltration and infarct size in these rats were determined. Expression of miR-98-5p, toll-like receptor 4 (TLR4) and the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) signaling pathway-related proteins was assessed. The target relation between miR-98-5p and TLR4 was confirmed by bioinformatic method and dual luciferase report gene assay. RESULTS MiR-98-5p was downregulated, TLR4 was upregulated and the PI3K/Akt signaling pathway was inactivated in MI/RI rat myocardial tissues. Exosomal miR-98-5p from hypoxic BMSCs promoted cardiac function and suppressed myocardial enzyme levels, oxidative stress, inflammation response, macrophage infiltration and infarct size in I/R myocardial tissues. Moreover, TRL4 was targeted by miR-98-5p and miR-98-5p activated PI3K/Akt signaling pathway. CONCLUSION Hypoxia-induced BMSC-exos elevated miR-98-5p to protect against MI/RI. This study may be helpful for treatment of MI/RI.
Collapse
|
14
|
Pan Y, Pan YM, Liu FT, Xu SL, Gu JT, Hang PZ, Du ZM. MicroRNA-98 ameliorates doxorubicin-induced cardiotoxicity via regulating caspase-8 dependent Fas/RIP3 pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103624. [PMID: 33617954 DOI: 10.1016/j.etap.2021.103624] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/13/2021] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
Cardiotoxicity is one of the primary limitations in the clinical use of the anticancer drug doxorubicin (DOX). However, the role of microRNAs (miRNAs) in DOX-induced cardiomyocyte death has not yet been covered. To investigate this, we observed a significant increase in miR-98 expression in neonatal rat ventricular myocytes after DOX treatment, and MTT, LIVE/Dead and Viability/Cytotoxicity staining showed that miR-98 mimic inhibited DOX-induced cell death. This was also confirmed by Flow cytometry and Annexin V-FITC/PI staining. Interestingly, the protein expression of caspase-8 was upregulated by miR-98 mimics during this process, whereas Fas and RIP3 were downregulated. In addition, the effect of miR-98 against the expression of Fas and RIP3 were restored by the specific caspase-8 inhibitor Z-IETD-FMK. Thus, we demonstrate that miR-98 protects cardiomyocytes from DOX-induced injury by regulating the caspase-8-dependent Fas/RIP3 pathway. Our findings enhance understanding of the therapeutic role of miRNAs in the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yang Pan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yu-Miao Pan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Fang-Tong Liu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Si-Lun Xu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jin-Tao Gu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Peng-Zhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhi-Min Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Heilongjiang Province, Harbin, 150086, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150081, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China.
| |
Collapse
|
15
|
Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, Aravena O. Immunosuppressive Mechanisms of Regulatory B Cells. Front Immunol 2021; 12:611795. [PMID: 33995344 PMCID: PMC8118522 DOI: 10.3389/fimmu.2021.611795] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory B cells (Bregs) is a term that encompasses all B cells that act to suppress immune responses. Bregs contribute to the maintenance of tolerance, limiting ongoing immune responses and reestablishing immune homeostasis. The important role of Bregs in restraining the pathology associated with exacerbated inflammatory responses in autoimmunity and graft rejection has been consistently demonstrated, while more recent studies have suggested a role for this population in other immune-related conditions, such as infections, allergy, cancer, and chronic metabolic diseases. Initial studies identified IL-10 as the hallmark of Breg function; nevertheless, the past decade has seen the discovery of other molecules utilized by human and murine B cells to regulate immune responses. This new arsenal includes other anti-inflammatory cytokines such IL-35 and TGF-β, as well as cell surface proteins like CD1d and PD-L1. In this review, we examine the main suppressive mechanisms employed by these novel Breg populations. We also discuss recent evidence that helps to unravel previously unknown aspects of the phenotype, development, activation, and function of IL-10-producing Bregs, incorporating an overview on those questions that remain obscure.
Collapse
Affiliation(s)
- Diego Catalán
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Miguel Andrés Mansilla
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Ashley Ferrier
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Unidad de Dolor, Hospital Clínico, Universidad de Chile (HCUCH), Santiago, Chile
| | | | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Octavio Aravena
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
16
|
Chen X, Zhao MZ, Miao BP, Liu ZQ, Yang G, Liu JQ, Yang PC, Song JP. Inhibition of Bcl2L12 Attenuates Eosinophilia-Related Inflammation in the Heart. Front Immunol 2020; 11:1955. [PMID: 33013849 PMCID: PMC7516035 DOI: 10.3389/fimmu.2020.01955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/20/2020] [Indexed: 11/25/2022] Open
Abstract
Background: The eosinophilic inflammation plays a critical role in myocarditis (Mcd); its underlying mechanism remains to be further elucidated. This study aims to investigate the role of Bcl2-like protein 12 (Bcl2L12) in inducing the defects of apoptosis in eosinophils (Eos) of the heart tissues. Methods: Human explant heart samples were collected. Eosinophilia and myocarditis (Mcd)-like inflammation were induced in the mouse heart by immunizing with murine cardiac α-myosin heavy chain (MyHCα) peptides. Results: Markedly more Eos were observed in heart tissues from patients with Mcd than those from patients with dilated cardiomyopathy. Eos isolated from Mcd hearts showed the signs of apoptosis defects. The Eo counts in the Mcd heart tissues were positively correlated with the Bcl2L12 expression in Eos isolated from the heart tissues. Exposure to interleukin 5 in the culture induced the expression of Bcl2L12 in Eos. Bcl2L12 bound c-Myc, the transcription factor of Fas ligand (FasL), to prevent c-Myc from binding to the FasL promoter, to restrict the FasL gene transcription in Eos. Inhibition of Bcl2L12 prevented the induction of eosinophilia and Mcd-like inflammation in the mouse heart. Conclusions: The Bcl2L12 expression contributes to apoptosis defects in Eos of the Mcd heart. Blocking Bcl2L12 prevents the eosinophilia induction and alleviates Mcd-like inflammation in mice.
Collapse
Affiliation(s)
- Xiao Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei-Zhen Zhao
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Bei-Ping Miao
- Department of Otolaryngology, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhi-Qiang Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital, Shenzhen, China
| | - Jiang-Qi Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Ping-Chang Yang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
| | - Jiang-Ping Song
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Zheng Z, Huang G, Gao T, Huang T, Zou M, Zou Y, Duan S. Epigenetic Changes Associated With Interleukin-10. Front Immunol 2020; 11:1105. [PMID: 32582189 PMCID: PMC7287023 DOI: 10.3389/fimmu.2020.01105] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
IL-10 is a regulator of inflammation and immunosuppression. IL-10 regulates a variety of immune cells to limit and stop the inflammatory response, and thus plays an important role in autoimmune diseases, inflammatory diseases and cancer. IL-10 is closely related to epigenetic modification, in which changes in DNA methylation of IL-10 gene can affect mRNA and protein levels of IL-10. In addition, changes in histone modifications, especially histone acetylation, can also lead to abnormal expression of IL-10 mRNA. At the same time, a handful of IL-10 related microRNAs (miRNAs) are found to be aberrantly expressed in multiple diseases. Besides, long non-coding RNA (lncRNA) growth arrest specific transcript 5 (GAS5) also inhibits IL-10 expression. Here, we reviewed the epigenetic changes related to IL-10 in various diseases, as well as the regulation of IL-10 gene expression in various diseases by epigenetic modifications such as DNA methylation, histone modification, miRNA, and lncRNA.
Collapse
Affiliation(s)
- Zhonghua Zheng
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Gang Huang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Tong Gao
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Tianyi Huang
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Mengsha Zou
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Yuhao Zou
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
18
|
Wang L, Zhang Y, Zhu G, Ma Y, Zuo H, Tian X. miR-16 exhibits protective function in LPS-treated cardiomyocytes by targeting DOCK2 to repress cell apoptosis and exert anti-inflammatory effect. Cell Biol Int 2020; 44:1760-1768. [PMID: 32369253 DOI: 10.1002/cbin.11371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/23/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022]
Abstract
This study aims to investigate the effects of microRNA (miR)-16/dedicator of cytokinesis 2 (DOCK2) on myocarditis. The differences in the expression of genes in acute myocarditis were filtered out across Gene Expression Omnibus (GEO) database. Myocarditis cell model was established by lipopolysaccharide (LPS) stimulation in cardiomyocytes. The association between miR-16 and DOCK2 was predicted by bioinformatics software and confirmed by dual-luciferase assay. Polymerase chain reaction and western blot analysis were employed to assess the expression levels of miR-16 and DOCK2 under different conditions. Cells viability, apoptosis, and inflammatory reaction were evaluated by Cell Counting Kit-8, flow cytometry, and enzyme-linked immunosorbent assays. miR-16, as an upstream regulator of DOCK2, exhibited lower expression in LPS-induced myocarditis model. More importantly, we revealed that a marked augmentation of miR-16 promoted the growth of LPS-stimulated cardiomyocytes, and attenuated cell apoptosis and inflammatory response. However, an increasing expression of DOCK2 inhibited the remission of LPS-induced myocardial injury caused by miR-16 mimic. Herein, our results highlighted that upregulation of miR-16 resulted in the protective effects on LPS-induced myocardial injury by reducing DOCK2 expression, affording a pair of novel target molecules for ameliorating the symptoms of myocarditis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Yangyang Zhang
- Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Guangfu Zhu
- Intervention Room, Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Yuncong Ma
- Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| | - Huan Zuo
- Department of Neurology, Tengzhou Traditional Chinese Medicine Hospital, Tengzhou, Shandong, China
| | - Xia Tian
- Intervention Room, Department of Cardiology, Tengzhou Central People's Hospital Affiliated to Jining Medical College, Tengzhou, Shandong, China
| |
Collapse
|
19
|
Su T, Yang B, Gao T, Liu T, Li J. Polymer nanoparticle-assisted chemotherapy of pancreatic cancer. Ther Adv Med Oncol 2020; 12:1758835920915978. [PMID: 32426046 PMCID: PMC7222269 DOI: 10.1177/1758835920915978] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is a lethal disease characterized by highly dense stroma fibrosis. Only 15-20% of patients with pancreatic cancer have resectable tumors, and only around 20% of them survive to 5 years. Traditional cancer treatments have little effect on their prognosis, and successful surgical resection combined with effective perioperative therapy is the main method for maximizing long-term survival. For this reason, chemotherapy is an adjunct treatment for resectable cancer and is the main therapy for incurable pancreatic cancer, including metastatic pancreatic adenocarcinoma. However, there are various side effects of chemotherapeutic medicine and low drug penetration because the complex tumor microenvironment limits the application of chemotherapy. As a novel strategy, polymer nanoparticles make it possible to target the tumor microenvironment, release cytotoxic agents through various responsive reactions, and thus overcome the treatment barrier. As drug carriers, polymer nanoparticles show marked advantages, such as increased drug delivery and efficiency, controlled drug release, decreased side effects, prolonged half-life, and evasion of immunogenic blockade. In this review, we discuss the factors that cause chemotherapy obstacles in pancreatic cancer, and introduce the application of polymer nanoparticles to treat pancreatic cancer.
Collapse
Affiliation(s)
- Tianqi Su
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Bo Yang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Tianren Gao
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Tongjun Liu
- Department of General Surgery, Second Hospital of Jilin University, Changchun 130041, People’s Republic of China
| | - Jiannan Li
- Department of General Surgery, Second Hospital of Jilin University, Changchun 130041, People’s Republic of China
| |
Collapse
|
20
|
Wang J, Han B. Dysregulated CD4+ T Cells and microRNAs in Myocarditis. Front Immunol 2020; 11:539. [PMID: 32269577 PMCID: PMC7109299 DOI: 10.3389/fimmu.2020.00539] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 03/09/2020] [Indexed: 12/14/2022] Open
Abstract
Myocarditis is a polymorphic disease complicated with indeterminate etiology and pathogenesis, and represents one of the most challenging clinical problems lacking specific diagnosis and effective therapy. It is caused by a complex interplay of environmental and genetic factors, and causal links between dysregulated microribonucleic acids (miRNAs) and myocarditis have also been supported by recent epigenetic researches. Both dysregulated CD4+ T cells and miRNAs play critical roles in the pathogenesis of myocarditis, and the classic triphasic model of its pathogenesis consists of the acute infectious, subacute immune, and recovery/chronic myopathic phase. CD4+ T cells are key pathogenic factors underlying the development and progression of myocarditis, and the effector and regulatory subsets, respectively, promote and inhibit autoimmune responses. Furthermore, the reciprocal interplay of these subsets influences the pathogenesis as well. Dysregulated miRNAs along with their mRNA and protein targets have been identified in heart biopsies (intracellular miRNAs) and body fluids (circulating miRNAs) during myocarditis. These miRNAs show phase-dependent changes, and correlate with viral infection, immune status, fibrosis, destruction of cardiomyocytes, arrhythmias, cardiac functions, and outcomes. Thus, miRNAs are promising diagnostic markers and therapeutic targets in myocarditis. In this review, we review myocarditis with an emphasis on its pathogenesis, and present a summary of current knowledge of dysregulated CD4+ T cells and miRNAs in myocarditis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
21
|
Shi S, Zhang S, Zhang H, Jin Q, Wu D. Silencing circANKRD36 protects H9c2 cells against lipopolysaccharide-induced injury via up-regulating miR-138. Exp Mol Pathol 2019; 111:104300. [DOI: 10.1016/j.yexmp.2019.104300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/26/2019] [Accepted: 08/20/2019] [Indexed: 12/17/2022]
|
22
|
miR-98 Modulates Cytokine Production from Human PBMCs in Systemic Lupus Erythematosus by Targeting IL-6 mRNA. J Immunol Res 2019; 2019:9827574. [PMID: 31886314 PMCID: PMC6914974 DOI: 10.1155/2019/9827574] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/02/2019] [Indexed: 02/07/2023] Open
Abstract
Objective There is evidence that interleukin-6 (IL-6) upregulation plays a critical role in immunopathology of systemic lupus erythematosus (SLE). MicroRNA- (miRNA-) 98 was predicted to bind with the 3′-untranslated region (3′-UTR) of IL-6 gene. We hypothesized miR-98 through its regulation of IL-6 gene expression to influence cytokine production from peripheral blood mononuclear cells (PBMCs) in SLE. Methods The expression of miR-98 and IL-6 mRNA in the PBMCs of 41 SLE patients and 20 healthy controls (HC) was detected by quantitative reverse transcription PCR (qRT-PCR). The correlations between miR-98 expression and clinical features were evaluated. Luciferase reporter assay was performed to identify miR-98 targets. miR-98 mimics, miR-98 inhibitor, and IL-6 overexpression vector were generated. Cell viability of PBMCs was assessed using MTT assay. Gene expression and protein level were determined by qRT-PCR and Western blotting. TNF-α, IL-8, IL-1β, and IL-10 levels in cultured supernatants were quantified using ELISA. Results The expression of miR-98 was downregulated in PBMCs of SLE patients, and its expression is negatively associated with IL-6 levels. miR-98 expression was correlated with disease activity, lupus nephritis, and anti-dsDNA antibody. IL-6 mRNA was a target gene of miR-98. IL-6 overexpression promoted the proliferation of PBMCs and increased the levels of TNF-α, IL-8, IL-1β, and IL-10. Those effects were further enhanced by miR-98 inhibitor, while were suppressed by miR-98 mimics. miR-98 regulated the levels of STAT3 phosphorylation via its target gene IL-6. Conclusion The current study revealed that miR-98 could ameliorate STAT3-mediated cell proliferation and inflammatory cytokine production via its target gene IL-6 in patients with SLE. These results suggest that miR-98 might serve as a potential target for SLE treatment and other IL-6-mediated diseases.
Collapse
|
23
|
Li Q, Xi J, Li B, Li N. MiR‐16, as a potential NF‐κB‐related miRNA, exerts anti‐inflammatory effects on LPS‐induced myocarditis via mediating CD40 expression: A preliminary study. J Biochem Mol Toxicol 2019; 34:e22426. [PMID: 31777165 DOI: 10.1002/jbt.22426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/10/2019] [Accepted: 11/14/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Qiang‐Qiang Li
- Department of Cardiology of Integrated Traditional Chinese and Western MedicineAnqiu People's Hospital Weifang Shandong China
| | - Jing Xi
- Department of CardiologyAnqiu People's Hospital Weifang Shandong China
| | - Bing‐Qiang Li
- Department of CardiologyAnqiu People's Hospital Weifang Shandong China
| | - Ning Li
- Department of CardiologyAnqiu People's Hospital Weifang Shandong China
| |
Collapse
|
24
|
Pan A, Tan Y, Wang Z, Xu G. STAT4 silencing underlies a novel inhibitory role of microRNA-141-3p in inflammation response of mice with experimental autoimmune myocarditis. Am J Physiol Heart Circ Physiol 2019; 317:H531-H540. [PMID: 31225989 DOI: 10.1152/ajpheart.00048.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As an inflammatory disease afflicting the heart muscle, autoimmune myocarditis (AM) represents one of the foremost causes of heart failure. Accumulating evidence has implicated microRNAs (miRNAs) in the process of inflammation and autoimmunity. Hence, the current study aimed to investigate the mechanism by which miR-141-3p influences experimental AM (EAM). An EAM mouse model was established using 6-wk old male BALB/c mice, after which the expression of miR-141-3p and STAT4 was measured. Gain-of-function and loss-of-function investigations were performed to identify the functional role of miR-141-3p and STAT4 in EAM. Heart weight-to-body weight ratio, cardiac function, and degree of inflammation, as well as the levels of inflammation factors (IFN-γ, TNF-α, IL-2, IL-6, and IL-17) in the serum were detected. STAT4 was subsequently verified to be upregulated, and miR-141-3p was downregulated in the EAM mice. Furthermore, the overexpression of miR-141-3p or silencing of STAT4 was observed to reduce the heart weight-to-body weight ratio of EAM mice and improve cardiac function, while alleviating the degree of inflammatory cell infiltration in the myocardial tissue. Meanwhile, the overexpression of miR-141-3p was identified to diminish serum inflammatory factor levels by downregulating STAT4. Additionally, miR-141-3p could bind to STAT4 to downregulate its expression, ultimately mitigating inflammation and inducing an anti-inflammatory effect in EAM mice. Taken together, upregulation of miR-141-3p alleviates the inflammatory response in EAM mice by inhibiting STAT4, providing a promising intervention target for the molecular treatment of AM.NEW & NOTEWORTHY miR-141-3p is poorly expressed, and STAT4 is upregulated in experimental autoimmune myocarditis (EAM) mice. Overexpressing miR-141-3p inhibits EAM. miR-141-3p binds to and suppresses STAT4 expression. miR-141-3p overexpression inhibits inflammatory factors by downregulating STAT4. This study provides new insights into the treatment of autoimmune myocarditis.
Collapse
Affiliation(s)
- Aiqun Pan
- Department of Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yuying Tan
- Department of Echocardiography, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Zhihao Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Guoliang Xu
- Department of Cardiovascular Medicine, The Eastern Division of the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
25
|
MicroRNA-98 interferes with thrombospondin 1 expression in peripheral B cells of patients with asthma. Biosci Rep 2017; 37:BSR20170149. [PMID: 28760845 PMCID: PMC5577176 DOI: 10.1042/bsr20170149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/09/2017] [Accepted: 07/31/2017] [Indexed: 01/31/2023] Open
Abstract
Thrombospondin 1 (TSP1)-producing B cells are an important immune regulatory cell fraction in the body, which are compromised in a number of immune diseases. miRs are involved in the immune regulation. The present study aims to elucidate the mechanism by which miR-98 interferes with the expression of TSP1 in B cells of the peripheral blood system. In the present study, peripheral blood samples were collected from patients with allergic asthma. The B cells were isolated from the blood samples to be analyzed for the expression of miR-98 and TSP1. The results showed that the levels of miR-98 were higher, the levels of TSP1 were lower, in B cells isolated from the peripheral blood in patients with asthma. A negative correlation was identified between the data of miR-98 and TSP1 in B cells. Exposure to T helper (Th) 2 (Th2) cytokine, interleukin (IL)-13, increased the expression of miR-98 and suppressed the expression of TSP1 in peripheral B cells, which was abolished by knocking down the miR-98 gene. In conclusion, miR-98 can suppress the expression of TSP1 in the peripheral B cells of patients with allergic asthma.
Collapse
|