1
|
Mascolo R, Bizzi E, Martelli M, Facoetti C, Colazzo G, Barone F, Brucato A. Old and New Biomarkers in Idiopathic Recurrent Acute Pericarditis (IRAP): Prognosis and Outcomes. Curr Cardiol Rep 2025; 27:17. [PMID: 39798014 PMCID: PMC11724782 DOI: 10.1007/s11886-024-02170-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 01/13/2025]
Abstract
PURPOSE OF REVIEW To outline the latest discoveries regarding the utility and reliability of serum biomarkers in idiopathic recurrent acute pericarditis (IRAP), considering recent findings on its pathogenesis. The study highlights the predictive role of these biomarkers in potential short- (cardiac tamponade, recurrences) and long-term complications (constrictive pericarditis, death). RECENT FINDINGS The pathogenesis of pericarditis has been better defined in recent years, focusing on the autoinflammatory pathway. New studies have demonstrated the pivotal role of the classical inflammatory biomarkers in distinguishing pericarditis phenotypes (high-grade vs. low-grade inflammation) and in defining outcomes of this condition. Pericarditis involves intense inflammatory activity, which causes elevation of different markers, such as C-reactive protein, erythrocyte sedimentation rate, neutrophils and platelets, serum amyloid A and D-Dimer. Conversely, lymphocytes are often reduced, as well as hemoglobin during the acute phase. Cardiac troponins T and I are elevated in up to 30% of cases. A Biomarker for CRP-negative cases is needed. Other markers have been proposed for diagnosis and prognosis in IRAP, such as anti-heart antibodies and anti-intercalated disk antibodies, but we need further studies to validate them.
Collapse
Affiliation(s)
- Ruggiero Mascolo
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy.
| | - Emanuele Bizzi
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Martina Martelli
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Chiara Facoetti
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Giulia Colazzo
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Fabio Barone
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
| | - Antonio Brucato
- Division of Internal Medicine, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, University of Milan, Piazzale Principessa Clotilde, 3, Milan, 20121, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, 74, Milan, 20157, Italy
| |
Collapse
|
2
|
Habib I, Jawed JJ, Nasrin T, Shaikh S. Briefing of pulmonary sarcoidosis: Reduction-oxidation, misleading and possibilities. Indian J Tuberc 2025; 72:103-111. [PMID: 39890360 DOI: 10.1016/j.ijtb.2024.07.003] [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: 03/15/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 02/03/2025]
Abstract
Sarcoidosis is an inflammatory disease with limited treatment strategies and is characterized by the presence of abnormal lumps (granulomas) of the inflammatory cells. Among the types, pulmonary sarcoidosis most commonly occurs (about 90%), affecting the lungs and intrathoracic lymph nodes. Although the cause of its occurrence is still unknown, perhaps microbes and chemical exposures, as well as genetic history, may trigger the disease occurrence. The updated scenario also depicted the interconnection between oxidative stress and pulmonary sarcoidosis. Thus, the therapeutic value of the genetic consequences, as well as the redox status of pulmonary sarcoidosis, are under consideration. In addition, sarcoidosis complexity has been associated with tumor malignancy and tuberculosis. Therefore, in this review, we summarized the current status of pulmonary sarcoidosis, interference of lung cancer and tuberculosis complications, understanding of the role of reactive species in disease occurrence, and how they are associated with genetic features.
Collapse
Affiliation(s)
- Irfan Habib
- Department of Internal Medicine, College of Medicine and JNM Hospital, WB, India
| | - Junaid Jibran Jawed
- Institute of Health Sciences, Presidency University-2nd Campus, DG/02/02, New Town, Rajarhat, Kolkata, 700156, India
| | - Tina Nasrin
- Dept. of Electrical and Computer Engineering, University of Cyprus, Nicosia, Cyprus
| | - Soni Shaikh
- Laboratory of Histopathology, TATA MEDICAL CENTER 14, MAR (E-W), New Town, Rajarhat, Kolkata, 700160, India.
| |
Collapse
|
3
|
de Oliveira JCC, Barbosa EDS, Silva NB, Silva TDC, Matos AGDM, Pinho JD. Non-coding rnas in Turner syndrome: a systematic review. REVISTA PAULISTA DE PEDIATRIA : ORGAO OFICIAL DA SOCIEDADE DE PEDIATRIA DE SAO PAULO 2024; 43:e2024029. [PMID: 39630788 PMCID: PMC11606598 DOI: 10.1590/1984-0462/2025/43/2024029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/24/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE The aim of this study was to summarize the main findings of non-coding RNA (ncRNAs) in Turner syndrome (TS), correlating these biomolecules with the clinical manifestations in affected patients. DATA SOURCE Searches were conducted in the databases of the United States National Library of Medicine (PubMed), Scientific Electronic Library Online (SciELO), and ScienceDirect, covering original English articles published from 2014 to 2023. Descriptors used included "lncRNAs and Turner Syndrome," "miRNAs and Turner Syndrome," and "circRNAs and Turner Syndrome." The studies that were included addressed the role of ncRNAs in the clinical characteristics of patients with TS. Exclusion criteria comprised texts in abstracts, reports, reviews, and monographs. DATA SYNTHESIS We identified 147 studies, of which seven were included. In the analysis of microRNAs, miR-486-5p and miR-320a stood out, being associated with ovarian development; miR-126-3p and miR-126-5p were related to greater aortic stiffness. Regarding long non-coding RNAs, the downregulation of XIST indicated dysfunctions in X chromosome inactivation. Concerning circular RNAs, circPPP2R3B, circCSF2RA, and circPCTN were related to immunological functions, while circ_0090421, circ_0090392, and circ_0089945 were linked to cardiac development. CONCLUSIONS The data from these studies demonstrate that these biomolecules play crucial roles in processes related to specific characteristics observed in TS patients. Besides being suggested as potential biomarkers, they may be useful in clinical practice.
Collapse
|
4
|
Tepebasi MY, Selcuk E, Taner R, Tasan S, Asci H, Gunes AB, Sarisahin B, Aydın B. Potential ameliorative effect of Dapagliflozin on systemic inflammation-induced cardiovascular injury via endoplasmic reticulum stress and autophagy pathway. Mol Biol Rep 2024; 51:1080. [PMID: 39432138 DOI: 10.1007/s11033-024-09990-9] [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: 08/28/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Dapagliflozin (DPG) is a sodium-glucose cotransporter-2 inhibitor and is used in the treatment of diabetes. In this study, we aimed to investigate the effect of DPG on cardiotoxicity caused by systemic inflammation via endoplasmic reticulum (ER) stress and autophagy. METHODS AND RESULTS Four groups of thirty-two Wistar Albino rats were created: Control (1 ml oral physiological saline for five days and intraperitoneal saline on the 5th day), LPS (1 ml oral physiological saline for five days and intraperitoneal 5 mg/kg of LPS on the 5th day), LPS + DPG (10 mg/kg of DPG orally for five days and 5 mg/kg of LPS intraperitoneally on the 5th day), and DPG (10 mg/kg of DPG orally for five days and 5 mg/kg of SF intraperitoneally on the 5th day). Histopathological and immunohistochemical analyses were performed on heart and aorta tissues. ER stress and autophagy gene markers in heart tissues were evaluated by RT-qPCR. Oxidative stress in heart tissues and serum cardiac enzymes were analyzed by spectrophotometric method. The heart and aortic tissues of the LPS group showed increased expressions of Tumor Necrosis Factor-α (TNF-α) and Caspase-3 (Cas-3), along with mild hyperemia, slight inflammatory cell infiltrations, and myocardial cell damage. The heart tissues also showed genetically increased expressions of include binding immunoglobulin protein (BiP/ GRP78), protein kinase RNA-like ER Kinase (PERK), inositol-requiring enzyme 1 (IRE-1), activating transcription factors 4 (ATF-4), activating transcription factors 4 (ATF6), C/EBP homologous protein (CHOP), and BECLIN 1. Furthermore, Creatine kinase-MB (CK-MB) and Lactate dehydrogenase (LDH) levels in blood tissue significantly increased, according to biochemical analysis. With DPG therapy, all of these findings were reversed. CONCLUSION In conclusion, DPG protects against the cardiotoxic effect of systemic inflammation with its antioxidant and anti-inflammatory properties by regulating ER stress and autophagy pathways.
Collapse
Affiliation(s)
| | - Esma Selcuk
- Department of Medical Biology, Faculty of Medicine, University of Suleyman Demirel, Isparta, Turkey
| | - Rumeysa Taner
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, Isparta, Turkey
| | - Serife Tasan
- Department of Pathology, Faculty of Veterinary Medicine, University of Mehmet Akif Ersoy, Burdur, Turkey
| | - Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, Isparta, Turkey
| | - Ali Baran Gunes
- Undergraduate student, Faculty of Medicine, University of Suleyman Demirel, Isparta, Turkey
| | - Berkehan Sarisahin
- Undergraduate student, Faculty of Medicine, University of Suleyman Demirel, Isparta, Turkey
| | - Bunyamin Aydın
- Kutahya Evliya Celebi Training and Research Hospital, Division of Endocrinology and Metabolism, Kutahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
5
|
Huang X, Fei Y, Qiu X, Qian T, Shang Q, Cui J, Song Y, Sheng S, Xiao W, Yu Q, Wang T, Wang X. MiR-625-5p is a potential therapeutic target in sepsis by regulating CXCL16/CXCR6 axis and endothelial barrier. Int Immunopharmacol 2024; 137:112508. [PMID: 38889512 DOI: 10.1016/j.intimp.2024.112508] [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: 01/01/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND MicroRNA plays an important role in the progression of sepsis. We found a significant increase of in miR-625-5p expression in the blood of patients with sepsis, and lipopolysaccharide (LPS)-stimulated EA.hy926 cells. To date, little is known about the specific biological function of miR-625-5p in sepsis. METHODS Changes in miR-625-5p expression were verified through quantitative real-time polymerase chain reaction in 45 patients with sepsis or septic shock and 30 healthy subjects. In vitro, EA.hy926 cells were treated with LPS. Transendothelial electrical resistance assay and FITC-dextran were used in evaluating endothelial barrier function. RESULTS Herein, patients with sepsis or septic shock had significantly higher miR-625-5p expression levels, chemokine (C-X-C motif) ligand 16 (CXCL16) levels, and glycocalyx components than the healthy controls, and miR-625-5p level was positively correlated with disease. Kaplan-Meier analysis demonstrated a strong association between miR-625-5p level and 28-day mortality. Furthermore, the miR-625-5p inhibitor significantly alleviated LPS-induced endothelial barrier injury in vitro. Then, miR-625-5p positively regulated CXCL16 and down-regulated miR-625-5p attenuated CXCL16 transcription and expression in EA.hy926 cells. CXCL16 knockout significantly alleviated vascular barrier dysfunction in the LPS-induced EA.hy926 cells. sCXCL16 treatment in EA.hy926 cells significantly increased endothelial hyperpermeability by disrupting endothelial glycocalyx, tight junction proteins, and adherens junction proteins through the modulation of C-X-C chemokine receptor type 6 (CXCR6). CONCLUSIONS Increase in miR-625-5p level may be an effective biomarker for predicting 28-day mortality in patients with sepsis/septic shock. miR-625-5p is a critical pathogenic factor for endothelial barrier dysfunction in LPS-induced EA.hy926 cells because it activates the CXCL16/CXCR6 axis.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuxin Fei
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoyu Qiu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Tiantian Qian
- Department of Respiratory Medicine, Ji'nan Zhangqiu District People's Hospital, No. 1920 Mingshuihuiquan Road, Ji'nan, 250200, Shandong, China
| | - Quanmei Shang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinfeng Cui
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yutong Song
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shurui Sheng
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wenhan Xiao
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qilin Yu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tao Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xiaozhi Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
6
|
Wang N, Chen C, Ren J, Dai D. MicroRNA delivery based on nanoparticles of cardiovascular diseases. Mol Cell Biochem 2024; 479:1909-1923. [PMID: 37542599 DOI: 10.1007/s11010-023-04821-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/24/2023] [Indexed: 08/07/2023]
Abstract
Cardiovascular disease, especially myocardial infarction, is a serious threat to human health. Many drugs currently used cannot achieve the desired therapeutic effect due to the lack of selectivity. With the in-depth understanding of the role of microRNA (miRNA) in cardiovascular disease and the wide application of nanotechnology, loading drugs into nanoparticles with the help of nano-delivery system may have a better effect in the treatment of cardiomyopathy. In this review, we highlight the latest research on miRNAs in the treatment of cardiovascular disease in recent years and discuss the possibilities and challenges of using miRNA to treat cardiomyopathy. Secondly, we discuss the delivery of miRNA through different nano-carriers, especially inorganic, polymer and liposome nano-carriers. The preparation of miRNA nano-drugs by encapsulating miRNA in these nano-materials will provide a new treatment option. In addition, the research status of miRNA in the treatment of cardiomyopathy based on nano-carriers is summarized. The use of this delivery tool cannot only realize therapeutic potential, but also greatly improve drug targeting and reduce side effects.
Collapse
Affiliation(s)
- Nan Wang
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, Zhejiang, China
| | - Chunyan Chen
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, Zhejiang, China
| | - Jianmin Ren
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, Zhejiang, China
| | - Dandan Dai
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
7
|
Jing Y, Qi L, Zhang X, Zheng L, Yang P, Yin J, Shi Y, Yan S. miR-let-7a inhibits sympathetic nerve remodeling after myocardial infarction by downregulating the expression of nerve growth factor. Open Med (Wars) 2024; 19:20240975. [PMID: 38883335 PMCID: PMC11179384 DOI: 10.1515/med-2024-0975] [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: 01/05/2024] [Revised: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 06/18/2024] Open
Abstract
Objective Sympathetic hyperinnervation following myocardial infarction (MI) is one of the primary causes of ventricular arrhythmias (VAs) after MI. Nerve growth factor (NGF) is a key molecule that induces sympathetic nerve remodeling. Previous studies have confirmed that microRNA (miR)-let-7a interacts with NGF. However, whether miR-let-7a is involved in sympathetic remodeling after MI remains unknown. We aimed to investigate whether miR-let-7a was associated with the occurrence of VA after MI. Methods and results A rat model of myocardial infarction was established using left coronary artery ligation. miR-let-7a expression levels were analyzed by reverse transcription-quantitative PCR. Western blotting was also used to examine NGF expression levels in vivo and in M1 macrophages in vitro. The relationship between miR-let-7a and NGF levels was investigated using a luciferase reporter assay. The results revealed that the expression of miR-let-7a decreased significantly after MI, while NGF expression was significantly upregulated. In addition, overexpression of miR-let-7a effectively inhibited NGF expression in rats, which was also verified in M1 macrophages. Tyrosine hydroxylase and growth-associated protein 43 immunofluorescence results revealed that the administration of a miR-let-7a overexpression lentivirus to rats inhibited sympathetic remodeling after MI. Programmed electrical stimulation, renal sympathetic nerve activity recording, and heart rate variability measurements showed that miR-let-7a overexpression decreased sympathetic activity. Conclusions These findings provide novel insights into the molecular mechanisms by which miR-let-7a and NGF contribute to the progression of sympathetic nerve remodeling after MI. Therefore, miR-let-7a may be a promising therapeutic target to reduce the incidence of arrhythmia following MI.
Collapse
Affiliation(s)
- Yanyan Jing
- Department of Cardiology, Yantai Yuhuangding Hospital, Shandong University, Yantai, China
- Department of Cardiology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Qi
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong, 250014, China
| | - Xueli Zhang
- Department of Cardiology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu Zheng
- Department of Cardiology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peijin Yang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong, 250014, China
| | - Jie Yin
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong, 250014, China
| | - Yugen Shi
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Shandong, 250014, China
| | - Suhua Yan
- Department of Cardiology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| |
Collapse
|
8
|
Chatterjee B, Sarkar M, Bose S, Alam MT, Chaudhary AA, Dixit AK, Tripathi PP, Srivastava AK. MicroRNAs: Key modulators of inflammation-associated diseases. Semin Cell Dev Biol 2024; 154:364-373. [PMID: 36670037 DOI: 10.1016/j.semcdb.2023.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/06/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023]
Abstract
Inflammation is a multifaceted biological and pathophysiological response to injuries, infections, toxins, and inflammatory mechanisms that plays a central role in the progression of various diseases. MicroRNAs (miRNAs) are tiny, 19-25 nucleotides long, non-coding RNAs that regulate gene expression via post-transcriptional repression. In this review, we highlight the recent findings related to the significant roles of miRNAs in regulating various inflammatory cascades and immunological processes in the context of many lifestyle-related diseases such as diabetes, cardiovascular diseases, cancer, etc. We also converse on how miRNAs can have a dual impact on inflammatory responses, suggesting that regulation of their functions for therapeutic purposes may be disease-specific.
Collapse
Affiliation(s)
- Bilash Chatterjee
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mrinmoy Sarkar
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Subhankar Bose
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Md Tanjim Alam
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSUI), Riyadh, Saudi Arabia
| | | | - Prem Prakash Tripathi
- Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amit Kumar Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
9
|
Jafari N, Shahabi Rabori V, Zolfi Gol A, Saberiyan M. Crosstalk of NLRP3 inflammasome and noncoding RNAs in cardiomyopathies. Cell Biochem Funct 2023; 41:1060-1075. [PMID: 37916887 DOI: 10.1002/cbf.3882] [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: 08/02/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
Cardiovascular diseases (CVDs) identified as a serious public health problem. Although there is a lot of evidence that inflammatory processes play a significant role in the progression of CVDs, however, the precise mechanism is not fully understood. Nevertheless, recent studies have focused on inflammation and its related agents. Nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) is a type of pattern recognition receptor (PRR) that can recognize pathogen-associated molecular patterns and trigger innate immune response. NLRP3 is a component of the NOD-like receptor (NLR) family and have a pivotal role in detecting damage to cardiovascular tissue. It is suggested that activation of NLRP3 inflammasome leads to initiating and propagating the inflammatory response in cardiomyopathy. So, late investigations have highlighted the NLRP3 inflammasome activation in various forms of cardiomyopathy. On the other side, it was shown that noncoding RNAs (ncRNAs), particularly, microRNAs, lncRNAs, and circRNAs possess a regulatory function in the immune system's inflammatory response, implicating their involvement in various inflammatory disorders. In addition, their role in different cardiomyopathies was indicated in recent studies. This review article provides a summary of recent advancements focusing on the function of the NLRP3 inflammasome in common CVDs, especially cardiomyopathy, while also discussing the therapeutic potential of inhibiting the NLRP3 inflammasome regulated by ncRNAs.
Collapse
Affiliation(s)
- Negar Jafari
- Department of Cardiology, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Ali Zolfi Gol
- Department of Pediatrics Cardiology, Shahid Motahari Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
10
|
Han X, Liu X, Zhao X, Wang X, Sun Y, Qu C, Liang J, Yang B. Dapagliflozin ameliorates sepsis-induced heart injury by inhibiting cardiomyocyte apoptosis and electrical remodeling through the PI3K/Akt pathway. Eur J Pharmacol 2023; 955:175930. [PMID: 37479014 DOI: 10.1016/j.ejphar.2023.175930] [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: 03/26/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Sepsis-induced heart injury is one of the leading causes of circulation disorders worldwide. Dapagliflozin, a sodium-glucose cotransporter 2 inhibitor mainly used for controlling blood glucose, has been shown to exert a protective effect on cardiomyocytes. However, the protective effect of dapagliflozin against sepsis-induced cardiac injury and the underlying mechanism needs to be studied. AIM This study aims to investigate the effect of dapagliflozin on sepsis-induced cardiomyopathy and the potential mechanisms involved. METHODS The rat model of sepsis was constructed by intraperitoneal injection of lipopolysaccharide. Echocardiography and electrophysiological studies were performed to detect changes in cardiac function and electrical activity. Cardiac pathological alternation and cardiomyocyte apoptosis were measured by H&E staining, serological analysis, immunohistochemical, immunofluorescence, and TUNEL assays. Western blot and qRT-PCR were performed to elucidate the underlying mechanism of dapagliflozin. Additionally, corresponding experiments in H9c2 cells were performed to further validate the mechanisms in vitro. RESULTS Dapagliflozin improved cardiac dysfunction and reduced the susceptibility to ventricular arrhythmias in sepsis rats by ameliorating cardiac inflammation, suppressing cardiomyocyte apoptosis, and alleviating ventricular electrical remodeling. The PI3K/Akt signaling pathway inhibitor inhibited the anti-apoptotic effect of dapagliflozin, indicating that the protective effect was related to the activation of the PI3K/Akt pathway. CONCLUSION Dapagliflozin ameliorated sepsis-induced cardiac injury by suppressing electrical remodeling and cardiomyocyte apoptosis, which could be attributed to the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Xin Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Yazhou Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Jinjun Liang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| |
Collapse
|
11
|
Chu X, Hou Y, Zhang X, Li M, Ma D, Tang Y, Yuan C, Sun C, Liang M, Liu J, Wei Q, Chang Y, Wang C, Zhang J. Hepatic Glucose Metabolism Disorder Induced by Adipose Tissue-Derived miR-548ag via DPP4 Upregulation. Int J Mol Sci 2023; 24:ijms24032964. [PMID: 36769291 PMCID: PMC9917501 DOI: 10.3390/ijms24032964] [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: 01/06/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to explore the molecular mechanism underlying the regulation of glucose metabolism by miR-548ag. For the first time, we found that miR-548ag expression was elevated in the abdominal adipose tissue and serum of subjects with obesity and type 2 diabetes mellitus (T2DM). The conditional knockout of adipose tissue Dicer notably reduced the expression and content of miR-548ag in mouse adipose tissue, serum, and liver tissue. The combined use of RNAseq, an miRNA target gene prediction software, and the dual luciferase reporter assay confirmed that miR-548ag exerts a targeted regulatory effect on DNMT3B and DPP4. miR-548ag and DPP4 expression was increased in the adipose tissue, serum, and liver tissue of diet-induced obese mice, while DNMT3B expression was decreased. It was subsequently confirmed both in vitro and in vivo that adipose tissue-derived miR-548ag impaired glucose tolerance and insulin sensitivity by inhibiting DNMT3B and upregulating DPP4. Moreover, miR-548ag inhibitors significantly improved the adverse metabolic phenotype in both obese mice and db/db mice. These results revealed that the expression of the adipose tissue-derived miR-548ag increased in obese subjects, and that this could upregulate the expression of DPP4 by targeting DNMT3B, ultimately leading to glucose metabolism disorder. Therefore, miR-548ag could be utilized as a potential target in the treatment of T2DM.
Collapse
Affiliation(s)
- Xiaolong Chu
- Medical College, Shihezi University, Shihezi 832000, China
- Department of Medical Genetics, Medical College of Tarim University, Alaer 843300, China
| | - Yanting Hou
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Xueting Zhang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Menghuan Li
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Dingling Ma
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Yihan Tang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Chenggang Yuan
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Chaoyue Sun
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Maodi Liang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Jie Liu
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Qianqian Wei
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Yongsheng Chang
- Medical College, Shihezi University, Shihezi 832000, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Cuizhe Wang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
- Correspondence: (C.W.); (J.Z.); Tel./Fax: +86-993-205-5801 (C.W. & J.Z.)
| | - Jun Zhang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
- Correspondence: (C.W.); (J.Z.); Tel./Fax: +86-993-205-5801 (C.W. & J.Z.)
| |
Collapse
|
12
|
Yan S, Wang M. HCG11 inhibits salivary adenoid cystic carcinoma by upregulating EphA2 via binding to miR-1297. Oral Surg Oral Med Oral Pathol Oral Radiol 2023; 135:257-267. [PMID: 36396591 DOI: 10.1016/j.oooo.2022.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/04/2022] [Accepted: 08/28/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Ephrin receptor A2 (EphA2) was reported to be related to the tumorigenesis of salivary adenoid cystic carcinoma (SACC), which is a rare malignancy accounting for less than 1% of all oral and maxillofacial tumors. This research aimed to assess the molecular mechanisms of EphA2 in SACC. STUDY DESIGN The expression of long non-coding RNA human leukocyte antigen complex group 11 (HCG11), microRNA-1297 (miR-1297), and EphA2 in SACC cell lines compared with normal human salivary gland (HSG) cell line was measured by reverse transcription-quantitative polymerase chain reaction. EphA2 protein level was detected by western blot. 5-ethynyl-2'-deoxyuridine (EdU), colony formation, Transwell, and wounding healing experiments were applied to evaluate SACC cell proliferation, migration, and invasion. The relationship among HCG11, miR-1297, and EphA2 was confirmed by luciferase reporter, RNA pulldown, and RNA immunoprecipitation experiments. RESULTS HCG11 and EphA2 were downregulated while miR-1297 was upregulated in SACC cells. EphA2 overexpression suppressed SACC cell proliferation, migration, and invasion. HCG11 bound to miR-1297 to reduce the inhibition of miR-1297 on EphA2 expression. EphA2 knockdown reversed the suppression of HCG11 overexpression on SACC cell phenotypes. CONCLUSION This study identified the HCG11/miR-1297/EphA2 regulatory axis in SACC, which might provide novel therapeutic targets for SACC.
Collapse
Affiliation(s)
- Shujuan Yan
- Department of Oral and Maxillofacial Surgery, Changyi People's Hospital, Weifang 261300, Shandong, China
| | - Meng Wang
- Health Management Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong, China.
| |
Collapse
|
13
|
Lin C, Zeng S, Li M. miR-424-5p combined with miR-17-5p has high diagnostic efficacy for endometriosis. Arch Gynecol Obstet 2023; 307:169-177. [PMID: 35366691 DOI: 10.1007/s00404-022-06492-6] [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: 12/01/2021] [Accepted: 02/25/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE Endometriosis (EMT) is a chronic benign disease with high prevalence. This study investigated the diagnostic value of serum miR-17-5p, miR-424-5p, and their combined expressions for EMT. METHODS Total 80 EMT patients of reproductive age who underwent laparoscopy or laparotomy and were confirmed by pathological examination were included as the study subjects, and another 80 healthy women of reproductive age receiving gynecological examination and ultrasonography with no pelvic abnormalities were selected as the control group. The whole blood samples of enrolled subjects were collected and clinical characteristics were recorded. The miR-17-5p, miR-424-5p, VEGFA, IL-4, and IL-6 levels in the serum were measured. ROC curve was used to evaluate the diagnostic efficacy of miR-17-5p and miR-424-5p expressions for EMT. Pearson correlation was performed to analyze the correlation of miR-17-5p and miR-424-5p with clinical indexes in EMT patients. RESULTS miR-17-5p and miR-424-5p were downregulated in EMT patients. For diagnosing EMT, the AUC of miR-17-5p was 0.865 and cutoff value was 0.890 (91.3% sensitivity and 85% specificity), the AUC of miR-424-5p was 0.737, and cutoff value was 0.915 (98.8% sensitivity and 61.2% specificity), and the AUC of miR-424-5p combined with miR-17-5p was 0.938 and cutoff value was 2.205 (93.8% sensitivity and 88.7% specificity), with the diagnostic efficacy higher than miR-424-5p or miR-17-5p alone. miR-17-5p and miR-424-5p expressions were negatively correlated with dysmenorrhea, infertility, pelvic pain, and rASRM stage, but not with age, BMI, menstrual disorder, and nulliparity. VEGFA, IL-4, IL-6, and CA-125 were increased in EMT patients and were inversely associated with miR-17-5p and miR-424-5p. CONCLUSION miR-424-5p combined with miR-17-5p has high diagnostic efficacy for EMT.
Collapse
Affiliation(s)
- Chunli Lin
- Department of Gynecology, Hunan Province Maternal and Child Health Care Hospital, 53 Xiangchun Road, Kaifu District, Changsha, 410008, Hunan, China.
| | - Saili Zeng
- Department of Respiratory Medicine, The Second Hospital of University of South China, 30 Jiefang Road, Shigu District, Hengyang, 421000, Hunan, China.
| | - Miaojie Li
- Department of Gynecology, People's Hospital of Yuxi City, Yuxi, 653100, Yunnan, China
| |
Collapse
|
14
|
Ilieva M, Panella R, Uchida S. MicroRNAs in Cancer and Cardiovascular Disease. Cells 2022; 11:3551. [PMID: 36428980 PMCID: PMC9688578 DOI: 10.3390/cells11223551] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Although cardiac tumor formation is rare, accumulating evidence suggests that the two leading causes of deaths, cancers, and cardiovascular diseases are similar in terms of pathogenesis, including angiogenesis, immune responses, and fibrosis. These similarities have led to the creation of new exciting field of study called cardio-oncology. Here, we review the similarities between cancer and cardiovascular disease from the perspective of microRNAs (miRNAs). As miRNAs are well-known regulators of translation by binding to the 3'-untranslated regions (UTRs) of messenger RNAs (mRNAs), we carefully dissect how a specific set of miRNAs are both oncomiRs (miRNAs in cancer) and myomiRs (muscle-related miRNAs). Furthermore, from the standpoint of similar pathogenesis, miRNAs categories related to the similar pathogenesis are discussed; namely, angiomiRs, Immune-miRs, and fibromiRs.
Collapse
Affiliation(s)
| | | | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen SV, Denmark
| |
Collapse
|
15
|
Kang Q, Luo A. The efficacy of mindfulness-based intervention for heart diseases: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2022; 101:e29649. [PMID: 36181030 PMCID: PMC9524963 DOI: 10.1097/md.0000000000029649] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION The efficacy of mindfulness-based intervention for heart diseases remains controversial. We conduct a systematic review and meta-analysis to explore the impact of mindfulness-based intervention on heart diseases. METHODS We have search PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through August 2020 for randomized controlled trials (RCTs) assessing the effect of mindfulness-based intervention versus usual care on heart diseases. This meta-analysis is performed using the random-effect model. RESULTS Five RCTs involving 458 patients are included in the meta-analysis. Overall, compared with control group for heart diseases, mindfulness-based intervention is associated with significantly increased 6 minute walking test [mean difference (MD) = 14.74; 95% confidence interval (95% CI) = 2.50-26.97; P = .02], decreased heart rate (MD = -2.54; 95% CI = -4.76 to -0.31; P = .03) and stress score (MD = -2.31; 95% CI = -4.23 to -0.38; P = .02), but shows no obvious impact on anxiety score (MD = -3.48; 95% CI = -7.98 to 1.03; P = .13) or respiratory rate (MD = -0.42; 95% CI = -1.31 to 0.46; P = .35). CONCLUSIONS Mindfulness-based intervention can provide additional benefits to heart diseases.
Collapse
Affiliation(s)
- Qingxia Kang
- Department of Cardiology, Yongchuan Hospital of Chongqing Medical University, Chongqing City, China
| | - Aihua Luo
- Department of Cardiology, Yongchuan Hospital of Chongqing Medical University, Chongqing City, China
- *Correspondence: Aihua Luo, Department of Cardiology, Yongchuan Hospital of Chongqing Medical University, No. 1 Youyi Street, Yongchuan District, Chongqing City 400453, China (e-mail: )
| |
Collapse
|
16
|
Li J, Sun G, Ma H, Wu X, Li C, Ding P, Lu S, Li Y, Yang P, Li C, Yang J, Peng Y, Meng Z, Wang L. Identification of immune-related hub genes and miRNA-mRNA pairs involved in immune infiltration in human septic cardiomyopathy by bioinformatics analysis. Front Cardiovasc Med 2022; 9:971543. [PMID: 36204577 PMCID: PMC9530044 DOI: 10.3389/fcvm.2022.971543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract Septic cardiomyopathy (SCM) is a serious complication caused by sepsis that will further exacerbate the patient's prognosis. However, immune-related genes (IRGs) and their molecular mechanism during septic cardiomyopathy are largely unknown. Therefore, our study aims to explore the immune-related hub genes (IRHGs) and immune-related miRNA-mRNA pairs with potential biological regulation in SCM by means of bioinformatics analysis and experimental validation. Method Firstly, screen differentially expressed mRNAs (DE-mRNAs) from the dataset GSE79962, and construct a PPI network of DE-mRNAs. Secondly, the hub genes of SCM were identified from the PPI network and the hub genes were overlapped with immune cell marker genes (ICMGs) to further obtain IRHGs in SCM. In addition, receiver operating characteristic (ROC) curve analysis was also performed in this process to determine the disease diagnostic capability of IRHGs. Finally, the crucial miRNA-IRHG regulatory network of IRHGs was predicted and constructed by bioinformatic methods. Real-time quantitative reverse transcription-PCR (qRT-PCR) and dataset GSE72380 were used to validate the expression of the key miRNA-IRHG axis. Result The results of immune infiltration showed that neutrophils, Th17 cells, Tfh cells, and central memory cells in SCM had more infiltration than the control group; A total of 2 IRHGs were obtained by crossing the hub gene with the ICMGs, and the IRHGs were validated by dataset and qRT-PCR. Ultimately, we obtained the IRHG in SCM: THBS1. The ROC curve results of THBS1 showed that the area under the curve (AUC) was 0.909. Finally, the miR-222-3p/THBS1 axis regulatory network was constructed. Conclusion In summary, we propose that THBS1 may be a key IRHG, and can serve as a biomarker for the diagnosis of SCM; in addition, the immune-related regulatory network miR-222-3p/THBS1 may be involved in the regulation of the pathogenesis of SCM and may serve as a promising candidate for SCM therapy.
Collapse
Affiliation(s)
- Jingru Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guihu Sun
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haocheng Ma
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xinyu Wu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chaozhong Li
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Peng Ding
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Si Lu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanyan Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ping Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chaguo Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunzhu Peng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhaohui Meng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Zhaohui Meng
| | - Luqiao Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Luqiao Wang
| |
Collapse
|
17
|
Zhang G, Dong D, Wan X, Zhang Y. Cardiomyocyte death in sepsis: Mechanisms and regulation (Review). Mol Med Rep 2022; 26:257. [PMID: 35703348 PMCID: PMC9218731 DOI: 10.3892/mmr.2022.12773] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/20/2022] [Indexed: 11/06/2022] Open
Abstract
Sepsis‑induced cardiac dysfunction is one of the most common types of organ dysfunction in sepsis; its pathogenesis is highly complex and not yet fully understood. Cardiomyocytes serve a key role in the pathophysiology of cardiac function; due to the limited ability of cardiomyocytes to regenerate, their loss contributes to decreased cardiac function. The activation of inflammatory signalling pathways affects cardiomyocyte function and modes of cardiomyocyte death in sepsis. Prevention of cardiomyocyte death is an important therapeutic strategy for sepsis‑induced cardiac dysfunction. Thus, understanding the signalling pathways that activate cardiomyocyte death and cross‑regulation between death modes are key to finding therapeutic targets. The present review focused on advances in understanding of sepsis‑induced cardiomyocyte death pathways, including apoptosis, necroptosis, mitochondria‑mediated necrosis, pyroptosis, ferroptosis and autophagy. The present review summarizes the effect of inflammatory activation on cardiomyocyte death mechanisms, the diversity of regulatory mechanisms and cross‑regulation between death modes and the effect on cardiac function in sepsis to provide a theoretical basis for treatment of sepsis‑induced cardiac dysfunction.
Collapse
Affiliation(s)
- Geping Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Dan Dong
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xianyao Wan
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yongli Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
18
|
MicroRNAs (miRNAs) in Cardiovascular Complications of Rheumatoid Arthritis (RA): What Is New? Int J Mol Sci 2022; 23:ijms23095254. [PMID: 35563643 PMCID: PMC9101033 DOI: 10.3390/ijms23095254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 02/08/2023] Open
Abstract
Rheumatoid Arthritis (RA) is among the most prevalent and impactful rheumatologic chronic autoimmune diseases (AIDs) worldwide. Within a framework that recognizes both immunological activation and inflammatory pathways, the exact cause of RA remains unclear. It seems however, that RA is initiated by a combination between genetic susceptibility, and environmental triggers, which result in an auto-perpetuating process. The subsequently, systemic inflammation associated with RA is linked with a variety of extra-articular comorbidities, including cardiovascular disease (CVD), resulting in increased mortality and morbidity. Hitherto, vast evidence demonstrated the key role of non-coding RNAs such as microRNAs (miRNAs) in RA, and in RA-CVD related complications. In this descriptive review, we aim to highlight the specific role of miRNAs in autoimmune processes, explicitly on their regulatory roles in the pathogenesis of RA, and its CV consequences, their main role as novel biomarkers, and their possible role as therapeutic targets.
Collapse
|
19
|
Fang F, Zhang X, Li B, Gan S. miR-182-5p combined with brain-derived neurotrophic factor assists the diagnosis of chronic heart failure and predicts a poor prognosis. J Cardiothorac Surg 2022; 17:88. [PMID: 35501813 PMCID: PMC9063236 DOI: 10.1186/s13019-022-01802-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Chronic heart failure (CHF) is a general progressive disorder with high morbidity and poor prognosis. This study analyzed the serum expression and clinical value of miR-182-5p and brain-derived neurotrophic factor (BDNF) in CHF patients. Methods A total of 82 CHF patients were selected as the study subjects (15 cases in NYHA stage I, 29 cases in stage II, 27 cases in stage III, and 11 cases in stage IV), with another 78 healthy people as the controls. The expression of serum miR-182-5p was detected by RT-qPCR. BDNF expression was measured by ELISA. Furthermore, the Pearson coefficient was used to analyze the correlation of miR-182-5p/BDNF with BNP and LVEF. ROC curve was employed to assess the potential of miR-182-5p or/and BDNF for the diagnosis of CHF. Kaplan–Meier survival curve was implemented to evaluate the prognostic value of miR-182-5p and BDNF. Results Serum miR-182-5p level was elevated and BDNF expression was lowered in CHF patients. Serum miR-182-5p in CHF patients was positively-related with BNP and inversely-correlated with LVEF, while serum BDNF was negatively-linked with BNP and positively-correlated with LVEF. ROC curve indicated the diagnostic value of serum miR-182-5p and BDNF for CHF and the diagnostic accuracy of miR-182-5p combined with BDNF was improved. Kaplan–Meier analysis unveiled that miR-182-5p low expression and BDNF high expression could predict the overall survival in CHF patients. Conclusion miR-182-5p expression is increased and BDNF level is decreased in CHF patients. miR-182-5p combined with BDNF can assist the diagnosis of CHF and predict a poor prognosis.
Collapse
Affiliation(s)
- Fang Fang
- Department of Cardiovascular Medicine, Xianning Central Hospital, No. 228 Jingui Road, Xian'an District, Xianning City, 437000, Hubei Province, China.
| | - Xiaonan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shenyang Medical College, Xianning, 110000, Liaoning Province, China
| | - Bin Li
- Department of Cardiovascular Medicine, Xianning Central Hospital, No. 228 Jingui Road, Xian'an District, Xianning City, 437000, Hubei Province, China
| | - Shouyi Gan
- Department of Cardiovascular Medicine, Xianning Central Hospital, No. 228 Jingui Road, Xian'an District, Xianning City, 437000, Hubei Province, China
| |
Collapse
|
20
|
Gryshkova V, Lushbough I, Palmer J, Burrier R, Delaunois A, Donley E, Valentin JP. microRNAs signatures as potential biomarkers of structural cardiotoxicity in human-induced pluripotent stem-cell derived cardiomyocytes. Arch Toxicol 2022; 96:2033-2047. [PMID: 35488128 DOI: 10.1007/s00204-022-03280-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
Identification of early biomarkers of heart injury and drug-induced cardiotoxicity is important to eliminate harmful drug candidates early in preclinical development and to prevent severe drug effects. The main objective of this study was to investigate the expression of microRNAs (miRNAs) in human-induced pluripotent stem cell cardiomyocytes (hiPSC-CM) in response to a broad range of cardiotoxic drugs. Next generation sequencing was applied to hiPSC-CM treated for 72 h with 40 drugs falling into the categories of functional (i.e., ion channel blockers), structural (changes in cardiomyocytes structure), and general (causing both functional and structural) cardiotoxicants as well as non-cardiotoxic drugs. The largest changes in miRNAs expression were observed after treatments with structural or general cardiotoxicants. The number of deregulated miRNAs was the highest for idarubicin, mitoxantrone, and bortezomib treatments. RT-qPCR validation confirmed upregulation of several miRNAs across multiple treatments at therapeutically relevant concentrations: hsa-miR-187-3p, hsa-miR-146b-5p, hsa-miR-182-5p (anthracyclines); hsa-miR-365a-5p, hsa-miR-185-3p, hsa-miR-184, hsa-miR-182-5p (kinase inhibitors); hsa-miR-182-5p, hsa-miR-126-3p and hsa-miR-96-5p (common some anthracyclines, kinase inhibitors and bortezomib). Further investigations showed that an upregulation of hsa-miR-187-3p and hsa-miR-182-5p could serve as a potential biomarker of structural cardiotoxicity and/or an additional endpoint to characterize cardiac injury in vitro.
Collapse
Affiliation(s)
| | - Isabel Lushbough
- UCB Biopharma SRL, Braine L'Alleud, Belgium
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | |
Collapse
|
21
|
Ding J, Jiang H, Su B, Wang S, Chen X, Tan Y, Shen L, Wang J, Shi M, Lin H, Zhang Z. DNMT1/miR-130a/ZEB1 Regulatory Pathway Affects the Inflammatory Response in Lipopolysaccharide-Induced Sepsis. DNA Cell Biol 2022; 41:479-486. [PMID: 35486848 DOI: 10.1089/dna.2021.1060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Sepsis is a global health care issue that affects millions of people. DNA methyltransferase I (DNMT1)-mediated DNA methylation is involved in a number of human diseases by affecting many types of cellular progression events. However, the role and underlying molecular mechanism of DNMT1 in development of sepsis remain largely unknown. Lipopolysaccharide (LPS) induced lung fibrosis in the sepsis mouse model, and DNMT1 was upregulated in lung tissues of a sepsis mouse model compared with lung tissues from control mice. Then, this study demonstrated that LPS induced the production of interleukin (IL)-7 and tumor necrosis factor (TNF)-α and promoted DNMT1 expression in primary type II alveolar epithelial cells (AECII cells). Knockdown of DNMT1 inhibited IL-7 and TNF-α secretion in AECII cells exposed to LPS. Further study demonstrated that DNMT1 repressed the expression of miR-130a in AECII cells with or without LPS exposure. Next, this study demonstrated that miR-130a inhibited ZEB1 expression in AECII cells exposed to LPS. Ultimately, this study revealed the role of the DNMT1/miR-130a/ZEB1 regulatory pathway in AECII cells exposed to LPS. Overall, our data revealed that LPS induced the secretion of inflammatory factors by modulating the DNMT1/miR-130a/ZEB1 regulatory pathway in AECII cells, thus providing a novel theoretical basis that might be beneficial for establishment of diagnostic and therapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Jurong Ding
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongbin Jiang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bo Su
- Department of Central Laboratory, and Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shanmei Wang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaolan Chen
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanlin Tan
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Shen
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingjing Wang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Minxing Shi
- Department of Respirology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haixu Lin
- Department of Central Laboratory, and Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhemin Zhang
- Department of Respirology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Gong CW, Yuan MM, Qiu BQ, Wang LJ, Zou HX, Hu T, Lai SQ, Liu JC. Identification and Validation of Ferroptosis-Related Biomarkers in Septic Cardiomyopathy via Bioinformatics Analysis. Front Genet 2022; 13:827559. [PMID: 35495160 PMCID: PMC9043284 DOI: 10.3389/fgene.2022.827559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
Septic cardiomyopathy (SCM) is a cardiac dysfunction caused by severe sepsis and septic shock that increases the risk of heart failure and death and its molecular mechanism remains unclear. Ferroptosis, a novel form of programmed cell death, has been reported to be present in the heart tissue of patients with sepsis, which demonstrated that ferroptosis may be a potential mechanism of myocardial injury in SCM. Therefore, we explored the role of ferroptosis-related genes (FRGs) in SCM and aimed to identify pivotal ferroptosis-related targets in SCM and potential therapeutic targets involved in the pathological process of SCM. To explore the regulatory mechanisms of ferroptosis in SCM, we identified differentially expressed genes (DEGs) in SCM and FRGs by bioinformatics analysis, and further identified hub genes. And the crucial microRNAs (miRNAs)-FRGs regulatory network was subsequently constructed. Finally, several candidate drugs associated with the hub genes were predicted, and Real-time quantitative reverse Transcription PCR (qRT-PCR) and western blotting analysis were performed to confirm the abnormal expression of hub genes. In this study, we identified several FRGs that may be involved in the pathogenesis of SCM, which helps us further clarify the role of ferroptosis in SCM and deeply understand the molecular mechanisms and potential therapeutic targets of SCM.
Collapse
Affiliation(s)
- Cheng-Wu Gong
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ming-Ming Yuan
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bai-Quan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li-Jun Wang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hua-Xi Zou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tie Hu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Song-Qing Lai
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Ji-Chun Liu, ; Song-Qing Lai,
| | - Ji-Chun Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Ji-Chun Liu, ; Song-Qing Lai,
| |
Collapse
|
23
|
Zhang T, Wang LL, Guan J, Zhou Y, Cheng P, Zou L. MicroRNA-125a/b-5p promotes malignant behavior in multiple myeloma cells and xenograft tumor growth by targeting DIS3. Kaohsiung J Med Sci 2022; 38:574-584. [PMID: 35394705 DOI: 10.1002/kjm2.12534] [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: 09/26/2021] [Revised: 12/17/2021] [Accepted: 01/26/2022] [Indexed: 11/11/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy with a high prevalence and is characterized by the clonal expansion of malignant plasma cells. As a new tumor suppressor, defective in sister chromatid joining (DIS3) was reported to be a gene closely related to MM. This study elucidated the biological functions and underlying mechanisms of DIS3 in MM. DIS3 mRNA and protein levels were detected using RT-qPCR and western blotting, respectively. Methyl thiazolyl tetrazolium assays, flow cytometry analyses, Transwell assays, and wound healing assays were performed to detect the proliferation, apoptosis, invasion, and migration of MM cells. The binding relationship between miR-125a/b-5p and DIS3 was verified using luciferase reporter assays and RNA pulldown assays. Xenograft tumor models were established in nude mice to investigate the effects of miR-125a/b-5p and DIS3 on tumor growth in vivo. DIS3 levels were downregulated in MM cells, and DIS3 upregulation inhibited the malignant behaviors of MM cells. Mechanistically, miR-125a/b-5p directly targeted the 3' untranslated region of DIS3. The expression of miR-125a/b-5p was upregulated in MM cells, miR-125a/b-5p knockdown inhibited the malignant behaviors of MM cells, and the inhibitory effect was reversed by DIS3 downregulation. The results of in vivo experiments indicated that miR-125a/b-5p promoted tumor growth by downregulating DIS3. Overall, miR-125a/b-5p promotes MM cellular processes and xenograft tumor growth by targeting DIS3.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Lan-Lan Wang
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Jun Guan
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Ying Zhou
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Ping Cheng
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Liang Zou
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| |
Collapse
|
24
|
Identification and Characterization of the miRNAs and Cytokines in Response to Leishmania infantum Infection with Different Response to Treatment. Acta Parasitol 2022; 67:403-410. [PMID: 34622398 DOI: 10.1007/s11686-021-00474-5] [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: 03/25/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Due to the complexity of cytokine and microRNA function in progression and/or suppression of an infection, in this study, we examined miR-3473f, miR-2128, miR-6994-5p, miR-7093-3p, miR-5128, miR-574-5p, miR-7235, IL-2, IL-4, IL-5, IL-10 and IL-13 in patients with VL caused by Leishmania infantum in an in vivo study. METHODS Sampling was carried out from patient with leishmaniasis and with different responses to treatment during March 2016-January 2020. DNA was extracted and purified using QIAamp Kit. The L. infantum were cultured in DMEM medium and protein content was determined by the Micro BCA Protein Assay Kit. Cytokines were evaluated using a MILLIPLEX MAP Mouse Cytokine/Chemokine Panel I kit. The relative expression of miRNAs was measured in duplicate using automatic thermocycler ABI Prism 7500 sequence detection system (Applied Bio-systems) using the TaqMan MicroRNA Assay kit. RESULTS The real-time PCR assay revealed that miR-2128, miR-6994-5p, miR-7093-3p, miR-5128, miR-574-5p and miR-7235 were down-regulated and miR-3473 were up-regulated in patients with semi-resistance and resistance parasite strain (P < 0.05). In the current work, cytokine patterns in patients who were slow-to-clear or unable-to-clear L. infantum infection during drug treatment were seen to have decreased protective Th1 cytokines (IL-2, IL-12, TNF-α, and IFN-ɤ, P < 0.001) and increased Th2 cytokines (IL-5, IL-10, and IL-13, P < 0.001). No association was seen with IL-4 in patients with different treatment outcomes. CONCLUSION Overall, the results of a recent study have shown that cytokines and microRNAs can play a key role in response to treatment, and more comprehensive studies are needed to support this hypothesis.
Collapse
|
25
|
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: 3.7] [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
|
26
|
Mirna M, Schmutzler L, Topf A, Boxhammer E, Sipos B, Hoppe UC, Lichtenauer M. Treatment with Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) Does Not Affect Outcome in Patients with Acute Myocarditis or Myopericarditis. J Cardiovasc Dev Dis 2022; 9:jcdd9020032. [PMID: 35200686 PMCID: PMC8880264 DOI: 10.3390/jcdd9020032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Previous animal studies reported an association of non-steroidal anti-inflammatory drugs (NSAIDs) with adverse outcomes in acute myocarditis, which is why these drugs are currently not recommended in affected patients. In this retrospective case-control study, we sought to investigate the effects of NSAID treatment in patients with acute myocarditis and myopericarditis to complement the available evidence. Method: A total of 114 patients with acute myocarditis were retrospectively enrolled. Demographical, clinical and laboratory data were extracted from hospital records. Patients who received NSAIDs (n = 39, 34.2%) were compared to controls. Follow-up on all-cause mortality was acquired for two years. Propensity score matching was additionally conducted to account for covariate imbalances between groups. Results: Treatment with NSAIDs was neither associated with a worse outcome (p = 0.115) nor with significant differences in left ventricular systolic function (p = 0.228) or in-hospital complications (p = 0.507). Conclusion: Treatment with NSAIDs was not associated with adverse outcomes in our study cohort. Together with the findings of previous studies, our results indicate that these drugs could be safely administered in patients with myocarditis and myopericarditis.
Collapse
|
27
|
Li R, Chen X, Li X, Huang G, Lu C, Wen Z, Chen Z, Lai Y. A four-miRNA signature in serum as a biomarker for bladder cancer diagnosis. Am J Transl Res 2022; 14:4606-4616. [PMID: 35958461 PMCID: PMC9360833 DOI: pmid/35958461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Urinary bladder cancer (BCa) is globally the 10th most frequent cancer. As a novel diagnostic tool, miRNA in serum screening is non-invasive. This project aimed to determine particular serum miRNAs as novel biomarkers for diagnosing urinary BCa. METHODS We designed a three-phase study with 122 healthy controls (HCs) and 132 BCa patients. The 30 miRNAs' expressions in serum from HCs and BCa patients were detected during the screening phase. The miRNAs with the most dysregulation were tested in the training (HCs vs. BCa, 30 each) and validation (80 HCs vs. 82 BCa) phase further. The diagnostic ability of these candidate miRNAs was estimated by the receiver operating characteristic (ROC) curves as well as the area under the ROC curve (AUC). The miRNAs' target genes and their annotations to functions were predicted utilizing bioinformatic assays. RESULTS Six serum miRNAs (miR-124-3p, miR-182-5p, miR-1-3p, miR-196a-5p, miR-23b-3p and miR-34a-5p) had significantly different expression between BCa patients and HCs in the training and validation phase. The four-microRNA panel improved the diagnostic value, with AUC =0.985. The result of bioinformatic analysis showed that these miRNAs' target genes in the panel may be related to the MAPK signaling pathway in bladder cancer. CONCLUSIONS Our study identified a four-miRNA panel that is a non-invasive new biomarker for diagnosing BCa.
Collapse
Affiliation(s)
- Rongkang Li
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical UniversityHefei 230032, Anhui, China
| | - Xuan Chen
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Xinji Li
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Guocheng Huang
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Chong Lu
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical UniversityHefei 230032, Anhui, China
| | - Zhenyu Wen
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Zebo Chen
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
| | - Yongqing Lai
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical UniversityShenzhen 518036, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical UniversityHefei 230032, Anhui, China
| |
Collapse
|
28
|
Arghiani N, Nissan T, Matin MM. Role of microRNAs in COVID-19 with implications for therapeutics. Biomed Pharmacother 2021; 144:112247. [PMID: 34601190 PMCID: PMC8463393 DOI: 10.1016/j.biopha.2021.112247] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 02/09/2023] Open
Abstract
COVID-19 is a pneumonia-like disease with highly transmittable and pathogenic properties caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which infects both animals and humans. Although many efforts are currently underway to test possible therapies, there is no specific FDA approved drug against SARS-CoV-2 yet. miRNA-directed gene regulation controls the majority of biological processes. In addition, the development and progression of several human diseases are associated with dysregulation of miRNAs. In this regard, it has been shown that changes in miRNAs are linked to severity of COVID-19 especially in patients with respiratory diseases, diabetes, heart failure or kidney problems. Therefore, targeting these small noncoding-RNAs could potentially alleviate complications from COVID-19. Here, we will review the roles and importance of host and RNA virus encoded miRNAs in COVID-19 pathogenicity and immune response. Then, we focus on potential miRNA therapeutics in the patients who are at increased risk for severe disease.
Collapse
Affiliation(s)
- Nahid Arghiani
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom
| | - Tracy Nissan
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran.
| |
Collapse
|
29
|
Zhang L, Tan W, Song X, Wang S, Tang L, Chen Y, Yu H, Jiang P, Liu J. Methylprednisolone Attenuates Lipopolysaccharide-Induced Sepsis by Modulating the Small Nucleolar RNA Host Gene 5/Copine 1 Pathway. DNA Cell Biol 2021; 40:1396-1406. [PMID: 34767734 DOI: 10.1089/dna.2021.0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sepsis has become a major public health problem worldwide. Methylprednisolone sodium succinate (MP) is a commonly used drug to prevent inflammation. However, the role and underlying mechanism of MP in sepsis remain vague. MP inhibited the lipopolysaccharide (LPS)-induced production of tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-17 and suppressed cell growth in alveolar type II epithelial cells (ATII cells). Small nucleolar RNA host gene 5 (SNHG5) expression was inhibited by LPS and restored by MP. Upregulation of SNHG5 inhibited the cellular role of LPS in ATII cells, and further, downregulation of SNHG5 inhibited the cellular role of MP in ATII cells under LPS conditions. SNHG5 elevated the expression of Copine 1 (CPNE1) by enhancing the mRNA stability of CPNE1. Increasing CPNE1 expression restored the silenced SNHG5-induced inhibitor role of MP in ATII cells under LPS conditions. Finally, MP attenuated lung injury and TNF-α and IL-17 secretion in an LPS-induced sepsis mouse model. Overall, this study investigated the mechanism underlying the effect of MP treatment in sepsis and, for the first time, revealed the important role of the SNHG5/CPNE1 pathway in the development and treatment of sepsis and the potential to serve as a diagnostic and therapeutic target for sepsis.
Collapse
Affiliation(s)
- Li Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Tan
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xinmiao Song
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shanmei Wang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Tang
- Department of Central Laboratory, and Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hanqing Yu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinming Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Zhang L, Li WP. New Cu(II) coordination polymer inhibits cervical cancer development by regulating BRCA1 protein via miR-9-5p. J Inorg Biochem 2021; 226:111655. [PMID: 34740040 DOI: 10.1016/j.jinorgbio.2021.111655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/23/2021] [Accepted: 10/23/2021] [Indexed: 01/22/2023]
Abstract
A novel Cu(II)-based coordination polymer [chemical composition, {[CuL(CH3CO2)](H2O)(DMF)}n (1, DMF = N,N-dimethylformamide) was successfully prepared via Cu(NO3)2·3H2O reaction with HL ligand in DMF and H2O mixture by using a hetero-donor ligand 4-(bis(4-(4H-1,2,4-triazol-4-yl)phenyl)amino)benzoic acid (HL) featuring carboxylic acid and triazole groups. Reverse transcription-polymerase chain reaction (RT-PCR) was adopted to determine miR-9-5p expression in cervical cancer cells after compound treatment. Bioinformatic analysis and luciferase reporter assay were utilized to confirm miR-9-5p and BRCA1 interaction to discover the potential goal of miR-9-5p in cervical cancer cells. Cell Counting Kit-8 (CCK-8) and reactive oxygen species (ROS) detection kits were adopted to examine cancer cell proliferation and ROS accumulation after compound treatment.
Collapse
Affiliation(s)
- Lin Zhang
- Luohe Medical College, Luohe, Henan, China.
| | | |
Collapse
|
31
|
Chen W, Gao G, Yan M, Yu M, Shi K, Yang P. Long noncoding RNA MAPKAPK5-AS1 promoted lipopolysaccharide-induced inflammatory damage in the myocardium by sponging microRNA-124-3p/E2F3. Mol Med 2021; 27:131. [PMID: 34666672 PMCID: PMC8524853 DOI: 10.1186/s10020-021-00385-1] [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] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Myocardial dysfunction caused by sepsis (SIMD) leads to high mortality in critically ill patients. We investigated the function and mechanism of long non-coding RNA MAPKAPK5-AS1 (lncRNA MAPKAPK-AS1) on lipopolysaccharide (LPS)-induced inflammation response in vivo and in vitro. METHOD Male SD rats were utilized for in vivo experiments. Rat cardiomyocytes (H9C2) were employed for in vitro experiments. Western blotting was employed to measure protein expression, and RT-PCR was performed to measure mRNA expression of inflammation factors. TUNEL and flow cytometry were carried out to evulate cell apoptosis. RESULT The results showed that the expression of MAPKAPK5-AS1 was increased, while the expression of miR-124-3p was decreased in the inflammatory damage induced by LPS in vivo and in vitro. Knockdown of MAPKAPK5-AS1 reduced LPS-induced cell apoptosis and inflammation response, while overexpression of miR-124-3p weakened the effects of MAPKAPK5-AS1 knockdown on LPS-induced cell apoptosis and inflammation response. Moreover, miR-124-3p was identified as a downstream miRNA of MAPKAPK5-AS1, and E2F3 was a target of miR-214-3p. MAPKAPK5-AS1 knockdown increased the expression of miR-124-3p, while miR-124-3p overexpression reduced the expression of MAPKAPK5-AS1. In addition, miR-124-3p was found to downregulate E2F3 expression in H9C2 cells. CONCLUSION MAPKAPK5-AS1/miR-124-3p/E2F3 axis regulates LPS-related H9C2 cell apoptosis and inflammatory response.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Guangyuan Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Mengjie Yan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Ming Yu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Kaiyao Shi
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China.
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
32
|
Gao T, Zou M, Shen T, Duan S. Dysfunction of miR-802 in tumors. J Clin Lab Anal 2021; 35:e23989. [PMID: 34558723 PMCID: PMC8605121 DOI: 10.1002/jcla.23989] [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: 06/01/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
Recent studies have shown that miR-802 is abnormally expressed in many tumors. miR-802 is expressed at low levels in tissues and cells of gastric cancer, colorectal cancer, breast cancer, cervical cancer, epithelial ovarian cancer, tongue squamous cell carcinoma, oral squamous cell carcinoma, esophageal squamous cell carcinoma, laryngeal squamous cell carcinoma, and melanoma. In contrast, miR-802 is overexpressed in hepatocellular carcinoma, bladder urothelial cancer, osteosarcoma, and cholesteatoma tissue cells. It should be noted that the results of studies on the expression of miR-802 in pancreatic cancer, prostate cancer, and lung cancer are inconsistent. Current studies have found that miR-802 can target and regulate genes in different tumors, and affect the regulation of the Wnt signaling pathway, EMT signaling pathway, PI3K/AKT signaling pathway, ERK signaling pathway, and Hedgehog signaling pathway. At the same time, miR-802 is regulated by the endogenous competition of four ceRNAs, including circDONSON, IGFL2-AS1, MIR155HG, and MIR4435-2HG. This article reviews the abnormal expression of miR-802 in a variety of tumors, expounds the mechanism by which miR-802 affects tumor progression by regulating different target genes, and elaborates the network of miR-802-related ceRNAs. We also summarized the limitations of miR-802 research and looked forward to the potential application of miR-802 in the diagnosis and prognosis of tumors.
Collapse
Affiliation(s)
- Tong Gao
- Medical Genetics Center, Ningbo University School of Medicine, Ningbo, China
| | - Mengsha Zou
- Medical Genetics Center, Ningbo University School of Medicine, Ningbo, China
| | - Tiancheng Shen
- Medical Genetics Center, Ningbo University School of Medicine, Ningbo, China
| | - Shiwei Duan
- Medical Genetics Center, Ningbo University School of Medicine, Ningbo, China.,School of Medicine, Zhejiang University City College, Hangzhou, China
| |
Collapse
|
33
|
Yu Y, Ou-Yang WX, Zhang H, Jiang T, Tang L, Tan YF, Luo HY, Xiao ZH, Li SJ. MiR-125b enhances autophagic flux to improve septic cardiomyopathy via targeting STAT3/HMGB1. Exp Cell Res 2021; 409:112842. [PMID: 34563514 DOI: 10.1016/j.yexcr.2021.112842] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 01/05/2023]
Abstract
We explore the role of miR-125b in septic cardiomyopathy, focusing on miR-125b/STAT3/HMGB1 axis. CLP mouse model and LPS-stimulated primary rat cardiomyocytes (CMs) and H9C2 cell were used as in vivo and in vitro models of septic cardiomyopathy, respectively. qRT-PCR and western blot were performed to measure expression levels of miR-125b, STAT3, HMGB1, and autophagy-related proteins. MTT assay was employed to examine LPS toxicity. Dual luciferase activity assay and CHIP were performed to validate interactions of miR-125b/STAT3 and STAT3/HMGB1 promoter. Immunostaining was used to assess the level of autophagic flux. ROS level was measured by fluorescence assay. Heart functions were examined via intracoronary Doppler ultrasound. miR-125b was diminished while STAT3 and HMGB1 were elevated in the heart tissue following CLP surgery and in LPS-treated H9C2 cells. LPS treatment up-regulated ROS generation and suppressed autophagic flux. Overexpression of miR-125b mimics or knockdown of STAT3 or HMGB1 alleviated LPS-induced hindrance of autophagic flux and ROS production. miR-125b directly targeted STAT3 mRNA and STAT3 bound with HMGB1 promoter. Overexpression of miR-125b mitigated myocardial dysfunction induced by CLP in vivo. Hyperactivation of STAT3/HMGB1 caused by reduced miR-125b contributes to ROS generation and the hindrance of autophagic flux during septic cardiomyopathy, leading to myocardial dysfunction.
Collapse
Affiliation(s)
- Ying Yu
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Wen-Xian Ou-Yang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hui Zhang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Tao Jiang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Lian Tang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Yan-Fang Tan
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hai-Yan Luo
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Zheng-Hui Xiao
- Emergence Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Shuang-Jie Li
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China.
| |
Collapse
|
34
|
Akseh S, Nemati M, Zamani-Gharehchamani E, Rezaie Nezhad Zamani A, Jodati A, Pezeshkian M, Nouri M, Gholizadeh D, Safaie N, Faridvand Y. Amnion membrane proteins attenuate LPS-induced inflammation and apoptosis by inhibiting TLR4/NF-κB pathway and repressing MicroRNA-155 in rat H9c2 cells. Immunopharmacol Immunotoxicol 2021; 43:487-494. [PMID: 34227443 DOI: 10.1080/08923973.2021.1945086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Amnion membrane (AM) has been popular for the treatment of inflammatory disorders due to its cell repairing properties. This current study aims to find the underlying mechanisms of amnion membrane proteins (AMPs) against the pro-inflammatory miRNA, miR-155, miR-146, and anti-apoptotic microRNA, miR-21, in LPS-treated H9c2 cells. METHODS Cell viability and apoptosis were determined by MTT assay and annexin V/PI staining. The production of the cytokines, TNF-α and IL-6 were evaluated by using qPCR and Enzyme-linked immunosorbent assay (ELISA), respectively. In addition, the expression of miRNAs was quantified by qPCR, and also the protein level of TLR4 and NF-kβ was determined with western blotting. RESULTS We found that AMPs ameliorated LPS-induced reduction of cell viability and augment apoptosis in H9c2 cells. AMPs efficiently inhibited cytokine expression (IL-6 and TNF-α) and activity of TLR4/NF-κB pathway in LPS-treated H9c2 cells. Correspondingly, in parallel with the suppression of pro-inflammatory cytokines and apoptosis, AMPs mitigated pro-inflammatory miRNA, miR-155 expression, while, the expression of miR-155 was found to be increased in LPS-treated H9c2 cells. Also, AMPs activated miR-146 expression in H9c2 cells under LPS treatment. Additionally, the elevated expression of miR-21 provoked by LPS was further enhanced by AMPs. CONCLUSIONS In conclusion, AMPs could alleviate LPS-induced cardiomyocytes cells injury via up-regulation of miR-21, miR-146, and suppression of TLR4/NF-κB pathway, which plays a key role in the down-regulation of LPS-mediated miR-155 and inflammatory cytokine expression.
Collapse
Affiliation(s)
- Saeideh Akseh
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Molecular Genetics, Islamic Azad University, Ahar, Iran
| | - Maryam Nemati
- Department of Genetic, Islamic Azad University, Tabriz, Iran
| | | | | | - Ahmadreza Jodati
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Pezeshkian
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Gholizadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Safaie
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Faridvand
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Song W, Zhang T, Yang N, Zhang T, Wen R, Liu C. Inhibition of micro RNA miR-122-5p prevents lipopolysaccharide-induced myocardial injury by inhibiting oxidative stress, inflammation and apoptosis via targeting GIT1. Bioengineered 2021; 12:1902-1915. [PMID: 34002676 PMCID: PMC8806731 DOI: 10.1080/21655979.2021.1926201] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Myocardial injury resulting from sepsis is the leading cause of death worldwide. Micro RNA miR-122-5p is involved in various physiological and pathological processes and is highly expressed in the heart of septic rats. However, its function in sepsis-caused myocardial injury remains elusive. Herein, a rat model of septic myocardial injury was established by intraperitoneal injection of lipopolysaccharide (LPS), and cardiomyocyte H9c2 was exposed to LPS to induce sepsis-related inflammatory injury in vitro. Inhibition of miR-122-5p suppressed LPS-triggered myocardial injury evidenced by decreased heart weight index (HWI), reduced inflammatory cell infiltration and cell rupture, and reduced cardiac marker enzymes cTnI and LDH. MiR-122-5p inhibition inhibited ROS production and enhanced the activities of antioxidant enzymes CAT, SOD and GSH-px in LPS-treated rats and H9c2 cells. MiR-122-5p inhibition reduced the production of pro-inflammatory cytokines TNF-α, IL-6 and IL-1β, and inhibited cell apoptosis along with decreased cleaved-caspase 3 induced by LPS. Moreover, increased GIT1 expression was found following miR-122-5p inhibition. We further verified GIT1 as a target of miR-122-5p, and silencing GIT1 partially reversed the benefits of miR-122-5p loss in LPS-injured H9c2 cells. The HO-1 and NQO-1 expression and Nrf-2 activation were enhanced by miR-122-5p inhibition, which was reversed by GIT1 depletion, indicating the involvement of Nrf-2/HO-1 signaling in regulating miR-122-5p/GIT1-mediated cardioprotection. Taken together, our data suggest that inhibition of miR-122-5p may mitigate sepsis-triggered myocardial injury through inhibiting inflammation, oxidative stress and apoptosis via targeting GIT1, which provides a possible therapeutic target for sepsis.
Collapse
Affiliation(s)
- Wenliang Song
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Tiening Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Tao Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chunfeng Liu
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
36
|
Wang W, Yang N, Wen R, Liu CF, Zhang TN. Long Noncoding RNA: Regulatory Mechanisms and Therapeutic Potential in Sepsis. Front Cell Infect Microbiol 2021; 11:563126. [PMID: 34055659 PMCID: PMC8149942 DOI: 10.3389/fcimb.2021.563126] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/28/2021] [Indexed: 12/17/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection and is characterized by a hyperinflammatory state accompanied by immunosuppression. Long noncoding RNAs (lncRNAs) are noncoding RNAs longer than 200 nucleotides and have important roles in mediating various biological processes. Recently, lncRNAs were found to exert both promotive and inhibitory immune functions in sepsis, thus participating in sepsis regulation. Additionally, several studies have revealed that lncRNAs are involved in sepsis-induced organ dysfunctions, including cardiovascular dysfunction, acute lung injury, and acute kidney injury. Considering the lack of effective biomarkers for early identification and specific treatment for sepsis, lncRNAs may be promising biomarkers and even targets for sepsis therapies. This review systematically highlights the recent advances regarding the roles of lncRNAs in sepsis and sheds light on their use as potential biomarkers and treatment targets for sepsis.
Collapse
Affiliation(s)
| | | | | | - Chun-Feng Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
37
|
Yan C, Xu Z, Huang W. Cellular Senescence Affects Cardiac Regeneration and Repair in Ischemic Heart Disease. Aging Dis 2021; 12:552-569. [PMID: 33815882 PMCID: PMC7990367 DOI: 10.14336/ad.2020.0811] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/11/2020] [Indexed: 01/10/2023] Open
Abstract
Ischemic heart disease (IHD) is defined as a syndrome of ischemic cardiomyopathy. Myogenesis and angiogenesis in the ischemic myocardium are important for cardiomyocyte (CM) survival, improving cardiac function and decreasing the progression of heart failure after IHD. Cellular senescence is a state of permanent irreversible cell cycle arrest caused by stress that results in a decline in cellular functions, such as proliferation, migration, homing, and differentiation. In addition, senescent cells produce the senescence-associated secretory phenotype (SASP), which affects the tissue microenvironment and surrounding cells by secreting proinflammatory cytokines, chemokines, growth factors, and extracellular matrix degradation proteins. The accumulation of cardiovascular-related senescent cells, including vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs), CMs and progenitor cells, is an important risk factor of cardiovascular diseases, such as vascular aging, atherosclerotic plaque formation, myocardial infarction (MI) and ventricular remodeling. This review summarizes the processes of angiogenesis, myogenesis and cellular senescence after IHD. In addition, this review focuses on the relationship between cellular senescence and cardiovascular disease and the mechanism of cellular senescence. Finally, we discuss a potential therapeutic strategy for MI targeting senescent cells.
Collapse
Affiliation(s)
- Chi Yan
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi, China.
- Department of Cardiology, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Guangxi, China.
| | - Zhimeng Xu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China.
| | - Weiqiang Huang
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi, China.
- Department of Cardiology, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Guangxi, China.
| |
Collapse
|
38
|
Anti-Interleukin-16 Neutralizing Antibody Treatment Alleviates Sepsis-Induced Cardiac Injury and Dysfunction via the Nuclear Factor Erythroid-2 Related Factor 2 Pathway in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6616422. [PMID: 33628366 PMCID: PMC7896865 DOI: 10.1155/2021/6616422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 11/17/2022]
Abstract
Several interleukin (IL) members have been reported to participate in sepsis. In this study, the effects of IL-16 on sepsis-induced cardiac injury and dysfunction were examined, and the related mechanisms were detected. IL-16 expression in septic mice was first measured, and the results showed that both cardiac and serum IL-16 expression levels were increased in mice with sepsis induced by LPS or cecal ligation and puncture (CLP) compared with control mice. Then, IL-16 was neutralized, and the effects on lipopolysaccharide- (LPS-) induced cardiac injury were detected. The results showed that an anti-IL-16 neutralizing antibody (nAb) significantly reduced mortality and increased serum lactate dehydrogenase (LDH), creatine kinase myocardial bound (CK-MB), and cardiac troponin T (cTnT) levels while improving cardiac function in mice with LPS-induced sepsis. Neutralization of IL-16 also increased the activation of antioxidant pathways and the expression of antioxidant factors in septic mice while decreasing the activation of prooxidant pathways and the expression of prooxidants. Treatment with the anti-IL-16 nAb increased mitochondrial apoptosis-inducing factor (AIF) expression, decreased nuclear AIF and cleaved poly-ADP-ribose polymerase (PARP) expression, and decreased TUNEL-positive cell percentages in LPS-treated mice. Additionally, treatment with CPUY192018, the nuclear factor erythroid-2 related factor 2 (Nrf2) pathway, significantly increased mortality and reversed the above effects in mice treated with LPS and the anti-IL-16 nAb. Our results showed that the anti-IL-16 nAb regulates oxidative stress through the Nrf2 pathway and participates in the regulation of cardiac injury in septic mice. Neutralization of IL-16 may be a beneficial strategy for the prevention of cardiac injury and dysfunction in sepsis patients.
Collapse
|
39
|
miR-362-3p Targets Orosomucoid 1 to Promote Cell Proliferation, Restrain Cell Apoptosis and Thereby Mitigate Hypoxia/Reoxygenation-Induced Cardiomyocytes Injury. Cardiovasc Toxicol 2021; 21:387-398. [PMID: 33459949 DOI: 10.1007/s12012-020-09631-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022]
Abstract
This study aimed to investigate the mechanism of how miR-362-3p/orosomucoid 1 (ORM1) involved in hypoxia/reoxygenation (H/R)-induced cardiomyocytes injury. Based on data obtained from Gene Expression Omnibus (GEO) database, we revealed that ORM1 was highly expressed and positively correlated with the expression of inflammatory factors (MAPK1, MAPK3, IL1B and CASP9). miR-362-3p was identified as an upstream regulatory miRNA of ORM1 and negatively modulated the mRNA and protein expression levels of ORM1 in H/R-injured cardiomyocytes. Moreover, we found that miR-362-3p was downregulated in cardiomyocytes injured by H/R. The promoting influence of miR-362-3p mimic on the proliferation and the inhibitory effect of miR-362-3p mimic on the apoptosis of H/R-stimulated cardiomyocytes were eliminated by overexpression of ORM1. Furthermore, miR-362-3p affected the expression of MAPK1, MAPK3, IL1B and CASP9 in H/R-injured cardiomyocytes through targeting ORM1. Our outcomes illustrated that miR-362-3p exhibited a protective influence on H/R-induced cardiomyocytes through targeting ORM1.
Collapse
|
40
|
Sun J, Sun X, Chen J, Liao X, He Y, Wang J, Chen R, Hu S, Qiu C. microRNA-27b shuttled by mesenchymal stem cell-derived exosomes prevents sepsis by targeting JMJD3 and downregulating NF-κB signaling pathway. Stem Cell Res Ther 2021; 12:14. [PMID: 33413595 PMCID: PMC7791667 DOI: 10.1186/s13287-020-02068-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background Exosomal microRNAs (miRs) derived from mesenchymal stem cells (MSCs) have been shown to play roles in the pathophysiological processes of sepsis. Moreover, miR-27b is highly enriched in MSC-derived exosomes. Herein, we aimed to investigate the potential role and downstream molecular mechanism of exosomal miR-27b in sepsis. Methods Inflammation was induced in bone marrow-derived macrophages (BMDMs) by lipopolysaccharide (LPS), and mice were made septic by cecal ligation and puncture (CLP). The expression pattern of miR-27b in MSC-derived exosomes was characterized using RT-qPCR, and its downstream gene was predicted by in silico analysis. The binding affinity between miR-27b, Jumonji D3 (JMJD3), or nuclear factor κB (NF-κB) was characterized to identify the underlying mechanism. We induced miR-27b overexpression or downregulation, along with silencing of JMJD3 or NF-κB to examine their effects on sepsis. The production of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 was detected by ELISA. Results miR-27b was highly expressed in MSC-derived exosomes. Mechanistic investigations showed that miR-27b targeted JMJD3. miR-27b decreased expression of pro-inflammatory genes by inhibiting the recruitment of JMJD3 and NF-κB at gene promoter region. Through this, MSC-derived exosomal miR-27b diminished production of pro-inflammatory cytokines in LPS-treated BMDMs and septic mice, which could be rescued by upregulation of JMJD3 and NF-κB. Besides, in vitro findings were reproduced by in vivo findings. Conclusion These data demonstrated that exosomal miR-27b derived from MSCs inhibited the development of sepsis by downregulating JMJD3 and inactivating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jia Sun
- ShenZhen Beike Biotechnology Research Institute, No. 59, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China.,Intervention and Cell Therapy Center, Shenzhen Hospital of Peking University, Shenzhen, 518057, People's Republic of China
| | - Xuan Sun
- Hematology Department, Shenzhen People's Hospital, Shenzhen, 518020, People's Republic of China
| | - Junhui Chen
- Intervention and Cell Therapy Center, Shenzhen Hospital of Peking University, Shenzhen, 518057, People's Republic of China
| | - Xin Liao
- ShenZhen Beike Biotechnology Research Institute, No. 59, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China
| | - Yixuan He
- ShenZhen Beike Biotechnology Research Institute, No. 59, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China
| | - Jinsong Wang
- ShenZhen Beike Biotechnology Research Institute, No. 59, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China
| | - Rui Chen
- ShenZhen Beike Biotechnology Research Institute, No. 59, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China
| | - Sean Hu
- ShenZhen Beike Biotechnology Research Institute, No. 59, Gaoxin South 9th Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China. .,Clinical Medical Research Center, Shenzhen People's Hospital, Shenzhen, 518020, People's Republic of China.
| | - Chen Qiu
- Respiratory and Critical Care Medicine Department, Shenzhen People's Hospital, No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020, Guangdong Province, People's Republic of China.
| |
Collapse
|
41
|
Topf A, Mirna M, Ohnewein B, Jirak P, Kopp K, Fejzic D, Haslinger M, Motloch LJ, Hoppe UC, Berezin A, Lichtenauer M. The Diagnostic and Therapeutic Value of Multimarker Analysis in Heart Failure. An Approach to Biomarker-Targeted Therapy. Front Cardiovasc Med 2020; 7:579567. [PMID: 33344515 PMCID: PMC7746655 DOI: 10.3389/fcvm.2020.579567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Heart failure is a pathophysiological state, which is still associated with high morbidity and mortality despite established therapies. Diverse well-known biomarkers fail to assess the variety of individual pathophysiology in the context of heart failure. Methods: An analysis of prospective, multimarker-specific therapeutic approaches to heart failure based on studies in current literature was performed. A total of 159 screened publications in the field of biomarkers in heart failure were hand-selected and found to be eligible for this study by a team of experts. Results: Established biomarkers of the inflammatory axis, matrix remodeling, fibrosis and oxidative stress axis, as well as potential therapeutic interventions were investigated. Interaction with end organs, such as cardio-hepatic, cardio-renal and cardio-gastrointestinal interactions show the complexity of the syndrome and could be of further therapeutic value. MicroRNAs are involved in a wide variety of physiologic and pathophysiologic processes in heart failure and could be useful in diagnostic as well as therapeutic setting. Conclusion: Based on our analysis by a biomarker-driven approach in heart failure therapy, patients could be treated more specifically in long term with a consideration of different aspects of heart failure. New studies evaluating a multimarker – based therapeutic approach could lead in a decrease in the morbidity and mortality of heart failure patients.
Collapse
Affiliation(s)
- Albert Topf
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Moritz Mirna
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Bernhard Ohnewein
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Peter Jirak
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Kristen Kopp
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Dzeneta Fejzic
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Michael Haslinger
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Lukas J Motloch
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Uta C Hoppe
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Alexander Berezin
- Internal Medicine Department, State Medical University, Zaporozhye, Ukraine
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
42
|
Jirak P, Wernly B, Lichtenauer M, Franz M, Knost T, Abusamrah T, Kelm M, Bimpong-Buta NY, Jung C. Next-generation sequencing analysis of circulating micro-RNA expression in response to parabolic flight as a spaceflight analogue. NPJ Microgravity 2020; 6:31. [PMID: 33298968 PMCID: PMC7606465 DOI: 10.1038/s41526-020-00121-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 09/11/2020] [Indexed: 01/15/2023] Open
Abstract
Understanding physiologic reactions to weightlessness is an indispensable requirement for safe human space missions. This study aims to analyse changes in the expression of circulating miRNAs following exposure to gravitational changes. Eight healthy volunteers (age: 24.5 years, male: 4, female: 4) were included. Each subject underwent 31 short-term phases of weightlessness and hypergravity induced by parabolic flight as a spaceflight analogue. At baseline, 1 and 24 h after parabolic flight, venous blood was withdrawn. Analysis of circulating miRNAs in serum was conducted by means of next generation sequencing. In total, 213 miRNAs were robustly detected (TPM > 5) by small RNA sequencing in all 24 samples. Four miRNAs evidenced a significant change in expression after adjusting for multiple testing. Only miR-223-3p showed a consistent significant decrease 24 h after parabolic flight compared to baseline values and values at 1 h after parabolic flight. miR-941 and miR-24-3p showed a significant decrease 24 h after parabolic flight compared to 1 h after parabolic flight but not to baseline values. miR-486-5p showed a significant increase 24 h after parabolic flight compared to 1 h after parabolic flight but not to baseline values. A target network analysis identified genes of the p53 signaling pathway and the cell cycle highly enriched among the targets of the four microRNAs. Our findings suggest cellular adaption to gravitational changes at the post-transcriptional level. Based on our results, we suggest a change in cell cycle regulation as potential explanation for adaptational changes observed in space missions.
Collapse
Affiliation(s)
- Peter Jirak
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Marcus Franz
- Department of Internal Medicine I, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Thorben Knost
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Thaer Abusamrah
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Nana-Yaw Bimpong-Buta
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Duesseldorf, Germany.
| |
Collapse
|
43
|
Nrp-1 Mediated Plasmatic Ago2 Binding miR-21a-3p Internalization: A Novel Mechanism for miR-21a-3p Accumulation in Renal Tubular Epithelial Cells during Sepsis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2370253. [PMID: 32923478 PMCID: PMC7453242 DOI: 10.1155/2020/2370253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022]
Abstract
The mechanism underlying sepsis-associated acute kidney injury (SAKI), which is an independent risk factor for sepsis-associated death, is unclear. A previous study indicates that during sepsis miR-21a-3p accumulates in renal tubular epithelial cells (TECs) as the mediator of inflammation and mediates TEC malfunction by manipulating its metabolism. However, the specific mechanism responsible for the accumulation of miR-21a-3p in TECs during sepsis is unrevealed. In this study, a cecal ligation and puncture- (CLP-) induced sepsis rat model and rat TEC line were used to elucidate the mechanism. Firstly, miR-21a-3p and Ago2 levels were found out to increase in both plasma and TECs during sepsis, and the increase of intracellular Ago2 and miR-21a-3p could be mitigated when Ago2 was either inactivated or downregulated in septic plasma. Moreover, membrane Nrp-1 expression of TECs was increased significantly during sepsis and Nrp-1 knockdown also mitigated the rise of both the intracellular Ago2 and miR-21a-3p levels in TECs incubated with septic plasma. Furthermore, it was revealed that Ago2 can be internalized by TECs mediated with Nrp-1 and this process had no effect on the intracellular content of miR-21a-3p. Both Ago2 and miR-21a-3p could bind to TECs derived Nrp-1 directly. Finally, it was determined that miR-21a-3p was internalized by TECs via Nrp-1 and Ago2 facilitated this process. Taken together, it can be concluded from our results that Ago2 binding miR-21a-3p from septic plasma can be actively internalized by TECs via Nrp-1 mediated cell internalization, and this mechanism is crucial for the rise of intracellular miR-21a-3p content of TECs during sepsis. These findings will improve our understanding of the mechanisms underlying SAKI and aid in developing novel therapeutic strategies.
Collapse
|
44
|
Sachinidis A. Cardiotoxicity and Heart Failure: Lessons from Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes and Anticancer Drugs. Cells 2020; 9:cells9041001. [PMID: 32316481 PMCID: PMC7226145 DOI: 10.3390/cells9041001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) are discussed as disease modeling for optimization and adaptation of therapy to each individual. However, the fundamental question is still under debate whether stem-cell-based disease modeling and drug discovery are applicable for recapitulating pathological processes under in vivo conditions. Drug treatment and exposure to different chemicals and environmental factors can initiate diseases due to toxicity effects in humans. It is well documented that drug-induced cardiotoxicity accelerates the development of heart failure (HF). Until now, investigations on the understanding of mechanisms involved in HF by anticancer drugs are hindered by limitations of the available cellular models which are relevant for human physiology and by the fact that the clinical manifestation of HF often occurs several years after its initiation. Recently, we identified similar genomic biomarkers as observed by HF after short treatment of hiPSCs-derived cardiomyocytes (hiPSC-CMs) with different antitumor drugs such as anthracyclines and etoposide (ETP). Moreover, we identified common cardiotoxic biological processes and signal transduction pathways which are discussed as being crucial for the survival and function of cardiomyocytes and, therefore, for the development of HF. In the present review, I discuss the applicability of the in vitro cardiotoxicity test systems as modeling for discovering preventive mechanisms/targets against cardiotoxicity and, therefore, for novel HF therapeutic concepts.
Collapse
Affiliation(s)
- Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| |
Collapse
|
45
|
Wang J, Li X, Liu Y, Peng C, Zhu H, Tu G, Yu X, Li Z. CircHIPK3 Promotes Pyroptosis in Acinar Cells Through Regulation of the miR-193a-5p/GSDMD Axis. Front Med (Lausanne) 2020; 7:88. [PMID: 32318575 PMCID: PMC7154086 DOI: 10.3389/fmed.2020.00088] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Acute pancreatitis (AP), especially severe acute pancreatitis (SAP), is an extremely dangerous illness with a high mortality rate. Pyroptotic cells release their cellular contents and inflammatory factors, aggravating the inflammatory response. Pyroptosis may be the main mode of acinar cell death during AP. The circular RNA circHIPK3 is expressed in pancreatic tissue and is associated with inflammatory response. In this study, we focused on the role and underlying mechanism of circHIPK3 in AP. We found that the expression of circHIPK3 was significantly elevated in serum of patients with AP and in caerulein-stimulated AR42J cells and was associated with caspase-1 and caspase-11 activation. circHIPK3 silencing ameliorated caerulein-induced cell damage and reduced the release of inflammatory factors IL-1β, IL-6, IL-8, and TNF-α and inhibited the activation of caspase-1 and caspase-11. In addition, circHIPK3 bound to miR-193a-5p and negatively regulated its expression. Inhibition of miR-193a-5p increased the release of IL-1β, IL-6, IL-8, and TNF-α and activated caspase-1 and caspase-11, thereby counteracting the effect of circHIPK3 silencing on caerulein-induced cell damage. Furthermore, we identified GSDMD as a target gene of miR-193a-5p, which is the key gene for pyroptosis. Interfering with the expression of GSDMD can increase cell viability, reduce the secretion of inflammatory cytokines, and suppress the activation of cleaved caspase-1 and caspase-11. Silencing GSDMD reversed the effects of miR-193a-5p inhibitors on caerulein-induced damage. In conclusion, circHIPK3 promotes pyroptosis in acinar cells through regulation of the miR-193a-5p/GSDMD axis, which eventually aggravates AP disease.
Collapse
Affiliation(s)
- Jiale Wang
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xia Li
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yunfei Liu
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Peng
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Zhu
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Guangping Tu
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhiqiang Li
- Department of Hepatobiliary and Pancreatic Surgery II, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
46
|
Tikhomirov R, Reilly-O’Donnell B, Catapano F, Faggian G, Gorelik J, Martelli F, Emanueli C. Exosomes: From Potential Culprits to New Therapeutic Promise in the Setting of Cardiac Fibrosis. Cells 2020; 9:E592. [PMID: 32131460 PMCID: PMC7140485 DOI: 10.3390/cells9030592] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022] Open
Abstract
Fibrosis is a significant global health problem associated with many inflammatory and degenerative diseases affecting multiple organs, individually or simultaneously. Fibrosis develops when extracellular matrix (ECM) remodeling becomes excessive or uncontrolled and is associated with nearly all forms of heart disease. Cardiac fibroblasts and myofibroblasts are the main effectors of ECM deposition and scar formation. The heart is a complex multicellular organ, where the various resident cell types communicate between themselves and with cells of the blood and immune systems. Exosomes, which are small extracellular vesicles, (EVs), contribute to cell-to-cell communication and their pathophysiological relevance and therapeutic potential is emerging. Here, we will critically review the role of endogenous exosomes as possible fibrosis mediators and discuss the possibility of using stem cell-derived and/or engineered exosomes as anti-fibrotic agents.
Collapse
Affiliation(s)
- Roman Tikhomirov
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (R.T.); (B.R.-O.); (F.C.); (J.G.)
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular Science, The University of Verona, Policlinico G., B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy; (G.F.); (F.M.)
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese Milano, Italy
| | - Benedict Reilly-O’Donnell
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (R.T.); (B.R.-O.); (F.C.); (J.G.)
| | - Francesco Catapano
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (R.T.); (B.R.-O.); (F.C.); (J.G.)
| | - Giuseppe Faggian
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular Science, The University of Verona, Policlinico G., B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy; (G.F.); (F.M.)
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (R.T.); (B.R.-O.); (F.C.); (J.G.)
| | - Fabio Martelli
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular Science, The University of Verona, Policlinico G., B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy; (G.F.); (F.M.)
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (R.T.); (B.R.-O.); (F.C.); (J.G.)
| |
Collapse
|