1
|
Ochoa Bernal MA, Song Y, Joshi N, Burns GW, Paul EN, Vegter E, Hrbek S, Sempere LF, Fazleabas AT. The Regulation of MicroRNA-21 by Interleukin-6 and Its Role in the Development of Fibrosis in Endometriotic Lesions. Int J Mol Sci 2024; 25:8994. [PMID: 39201680 PMCID: PMC11354763 DOI: 10.3390/ijms25168994] [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: 06/25/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Endometriosis is one of the most common causes of chronic pelvic pain and infertility that affects 10% of women of reproductive age. It is currently defined as the presence of endometrial epithelial and stromal cells at ectopic sites; however, advances in endometriosis research have some authors believing that endometriosis should be re-defined as "a fibrotic condition in which endometrial stroma and epithelium can be identified". microRNAs (miRNAs) are regulatory molecules that potentially play a role in endometriotic lesion development. There is evidence that suggests that miRNAs, including microRNA-21 (miR-21), participate in fibrotic processes in different organs, including the heart, kidney, liver and lungs. The objective of this study was to understand the role of miR-21 and the mechanisms that can contribute to the development of fibrosis by determining how IL-6 regulates miR-21 expression and how this miRNA regulates the transforming growth factor beta (TGF-β) signaling pathway to promote fibrosis. We investigated the expression of miR-21 in the baboon and mouse model of endometriosis and its correlation with fibrosis. We demonstrated that inflammation and fibrosis are present at a very early stage of endometriosis and that the inflammatory environment in the peritoneal cavity, which includes interleukin 6 (IL-6), can regulate the expression of miR-21 in vitro and in vivo.
Collapse
Affiliation(s)
- Maria Ariadna Ochoa Bernal
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Yong Song
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| | - Niraj Joshi
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| | - Gregory W. Burns
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| | - Emmanuel N. Paul
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| | - Erin Vegter
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| | - Samantha Hrbek
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| | - Lorenzo F. Sempere
- Precision Health Program and Department of Radiology Michigan State University, East Lansing, MI 48824, USA;
| | - Asgerally T. Fazleabas
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (M.A.O.B.); (Y.S.); (N.J.); (G.W.B.); (E.N.P.); (E.V.); (S.H.)
| |
Collapse
|
2
|
Fan X, Feng K, Liu Y, Yang L, Zhao Y, Tian L, Tang Y, Wang X. miR-135a Regulates Atrial Fibrillation by Targeting Smad3. Cardiovasc Ther 2023; 2023:8811996. [PMID: 37187923 PMCID: PMC10181910 DOI: 10.1155/2023/8811996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/12/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Background Atrial fibrillation (AF) is the most common arrhythmia in clinical. Atrial fibrosis is a hallmark feature of atrial structural remodeling in AF, which is regulated by the TGF-β1/Smad3 pathway. Recent studies have implicated that miRNAs are involved in the process of AF. However, the regulatory mechanisms of miRNAs remain largely unknown. This study is aimed at investigating the function and regulatory network of miR-135a in AF. Methods In vivo, the plasma was collected from patients with AF and non-AF subjects. Adult SD rats were induced by acetylcholine (ACh) (66 μg/ml)-CaCl2 (10 mg/ml) to establish an AF rat model. In vitro, atrial fibroblasts (AFs), isolated from adult SD rats, were treated with high-frequency electrical stimulation (HES) (12 h) and hypoxia (24 h) to mimic the AF and atrial fibrosis, respectively. miR-135a expression was detected through quantitative real-time polymerase chain reaction (qRT-PCR). The association between miR-135a and Smad3 was speculated by the TargetScan database and confirmed by the luciferase reporter assay. Fibrosis-related genes, Smad3, and TRPM7 were all assessed. Results The expression of miR-135a was markedly decreased in the plasma of AF patients and AF rats, which was consistent with that in HES-treated and hypoxia-treated AFs. Smad3 was identified as a target of miR-135a. the downregulation of miR-135a was associated with the enhancement of Smad3/TRPM7 expressions in AFs. Additionally, the knockdown of Smad3 significantly reduced the expression of TRPM7 and further inhibited atrial fibrosis. Conclusions Our study demonstrates that miR-135a regulates AF via Smad3/TRPM7, which is a potential therapeutic target for AF.
Collapse
Affiliation(s)
- Xueting Fan
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmacy, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Kai Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yonghui Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Leixi Yang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yizhuo Zhao
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmacy, Ningbo First Hospital, Ningbo Hospital of Zhejiang University, Ningbo 315010, China
| | - Liping Tian
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaozhi Wang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
3
|
Samra M, Srivastava K. Non-coding RNA and their potential role in cardiovascular diseases. Gene 2023; 851:147011. [DOI: 10.1016/j.gene.2022.147011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 10/21/2022] [Indexed: 11/27/2022]
|
4
|
miR-96-5p regulates myocardial infarction-induced cardiac fibrosis via Smad7/Smad3 pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1874-1888. [PMID: 36789690 PMCID: PMC10157616 DOI: 10.3724/abbs.2022175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Fibrotic remodelling contributes to heart failure in myocardial infarction. MicroRNAs (miRNAs) play a crucial role in myocardial fibrosis. However, current antifibrotic therapeutic strategies using miRNAs are far from effective. In this study, we aim to investigate the effect of miR-96-5p on cardiac fibrosis. Our work reveals a significant upregulation of miR-96-5p level in the ventricular tissues of myocardial infarction mice, as well as in neonatal rat cardiac fibroblasts stimulated with TGF-β or Ang II as shown by qPCR assay. In myocardial infarction mice, miR-96-5p knockdown using antagomir alleviates the aggravated cardiac fibrosis and exacerbated myocardial function caused by myocardial infarction surgery as shown by the echocardiography and Masson's staining analysis. In contrast, immunofluorescence staining results reveal that miR-96-5p overexpression in neonatal rat cardiac fibroblasts contributes to an increase in the expressions of fibrosis-associated genes and promotes the proliferation and differentiation of cardiac fibroblasts. Conversely, miR-96-5p downregulation using inhibitor presents adverse consequences. Furthermore, Smad7 expression is downregulated in fibrotic cardiac tissues, and the Smad7 gene is identified as a direct target of miR-96-5p by dual luciferase assay. Indeed, Smad7 knockdown weakens the anti-fibrotic effect of the miR-96-5p inhibitor on cardiac fibroblasts. Moreover, Smad3 phosphorylation is elevated in fibrotic cardiac tissues, and interestingly, the Smad3 inhibitor suppresses the profibrotic effect of the miR-96-5p mimic. Taken together, our findings demonstrate that the Smad7/Smad3 signaling pathway mediates the profibrotic effect of miR-96-5p in cardiac fibrosis.
Collapse
|
5
|
Molecular Approaches and Echocardiographic Deformation Imaging in Detecting Myocardial Fibrosis. Int J Mol Sci 2022; 23:ijms231810944. [PMID: 36142856 PMCID: PMC9501415 DOI: 10.3390/ijms231810944] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/11/2022] [Accepted: 09/16/2022] [Indexed: 12/25/2022] Open
Abstract
The pathological remodeling of myocardial tissue is the main cause of heart diseases. Several processes are involved in the onset of heart failure, and the comprehension of the mechanisms underlying the pathological phenotype deserves special attention to find novel procedures to identify the site of injury and develop novel strategies, as well as molecular druggable pathways, to counteract the high degree of morbidity associated with it. Myocardial fibrosis (MF) is recognized as a critical trigger for disruption of heart functionality due to the excessive accumulation of extracellular matrix proteins, in response to an injury. Its diagnosis remains focalized on invasive techniques, such as endomyocardial biopsy (EMB), or may be noninvasively detected by cardiac magnetic resonance imaging (CMRI). The detection of MF by non-canonical markers remains a challenge in clinical practice. During the last two decades, two-dimensional (2D) speckle tracking echocardiography (STE) has emerged as a new non-invasive imaging modality, able to detect myocardial tissue abnormalities without specifying the causes of the underlying histopathological changes. In this review, we highlighted the clinical utility of 2D-STE deformation imaging for tissue characterization, and its main technical limitations and criticisms. Moreover, we focalized on the importance of coupling 2D-STE examination with the molecular approaches in the clinical decision-making processes, in particular when the 2D-STE does not reflect myocardial dysfunction directly. We also attempted to examine the roles of epigenetic markers of MF and hypothesized microRNA-based mechanisms aiming to understand how they match with the clinical utility of echocardiographic deformation imaging for tissue characterization and MF assessment.
Collapse
|
6
|
Mahtal N, Lenoir O, Tinel C, Anglicheau D, Tharaux PL. MicroRNAs in kidney injury and disease. Nat Rev Nephrol 2022; 18:643-662. [PMID: 35974169 DOI: 10.1038/s41581-022-00608-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by degrading or repressing the translation of their target messenger RNAs. As miRNAs are critical regulators of cellular homeostasis, their dysregulation is a crucial component of cell and organ injury. A substantial body of evidence indicates that miRNAs are involved in the pathophysiology of acute kidney injury (AKI), chronic kidney disease and allograft damage. Different subsets of miRNAs are dysregulated during AKI, chronic kidney disease and allograft rejection, which could reflect differences in the physiopathology of these conditions. miRNAs that have been investigated in AKI include miR-21, which has an anti-apoptotic role, and miR-214 and miR-668, which regulate mitochondrial dynamics. Various miRNAs are downregulated in diabetic kidney disease, including the miR-30 family and miR-146a, which protect against inflammation and fibrosis. Other miRNAs such as miR-193 and miR-92a induce podocyte dedifferentiation in glomerulonephritis. In transplantation, miRNAs have been implicated in allograft rejection and injury. Further work is needed to identify and validate miRNAs as biomarkers of graft function and of kidney disease development and progression. Use of combinations of miRNAs together with other molecular markers could potentially improve diagnostic or predictive power and facilitate clinical translation. In addition, targeting specific miRNAs at different stages of disease could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Nassim Mahtal
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France
| | - Olivia Lenoir
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| | - Claire Tinel
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Dany Anglicheau
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| |
Collapse
|
7
|
Dong Y, Peng N, Dong L, Tan S, Zhang X. Non-coding RNAs: Important participants in cardiac fibrosis. Front Cardiovasc Med 2022; 9:937995. [PMID: 35966549 PMCID: PMC9365961 DOI: 10.3389/fcvm.2022.937995] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
Cardiac remodeling is a pathophysiological process activated by diverse cardiac stress, which impairs cardiac function and leads to adverse clinical outcome. This remodeling partly attributes to cardiac fibrosis, which is a result of differentiation of cardiac fibroblasts to myofibroblasts and the production of excessive extracellular matrix within the myocardium. Non-coding RNAs mainly include microRNAs and long non-coding RNAs. These non-coding RNAs have been proved to have a profound impact on biological behaviors of various cardiac cell types and play a pivotal role in the development of cardiac fibrosis. This review aims to summarize the role of microRNAs and long non-coding RNAs in cardiac fibrosis associated with pressure overload, ischemia, diabetes mellitus, aging, atrial fibrillation and heart transplantation, meanwhile shed light on the diagnostic and therapeutic potential of non-coding RNAs for cardiac fibrosis.
Collapse
|
8
|
Ciccarelli M, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Heymans S, Hooghiemstra A, Leeuwis A, Hermkens D, Tocchetti CG, van der Velden J, Zacchigna S, Thum T. Reciprocal organ interactions during heart failure: a position paper from the ESC Working Group on Myocardial Function. Cardiovasc Res 2021; 117:2416-2433. [PMID: 33483724 PMCID: PMC8562335 DOI: 10.1093/cvr/cvab009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/20/2021] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Heart failure-either with reduced or preserved ejection fraction (HFrEF/HFpEF)-is a clinical syndrome of multifactorial and gender-dependent aetiology, indicating the insufficiency of the heart to pump blood adequately to maintain blood flow to meet the body's needs. Typical symptoms commonly include shortness of breath, excessive fatigue with impaired exercise capacity, and peripheral oedema, thereby alluding to the fact that heart failure is a syndrome that affects multiple organ systems. Patients suffering from progressed heart failure have a very limited life expectancy, lower than that of numerous cancer types. In this position paper, we provide an overview regarding interactions between the heart and other organ systems, the clinical evidence, underlying mechanisms, potential available or yet-to-establish animal models to study such interactions and finally discuss potential new drug interventions to be developed in the future. Our working group suggests that more experimental research is required to understand the individual molecular mechanisms underlying heart failure and reinforces the urgency for tailored therapeutic interventions that target not only the heart but also other related affected organ systems to effectively treat heart failure as a clinical syndrome that affects and involves multiple organs.
Collapse
Affiliation(s)
- Michele Ciccarelli
- University of Salerno, Department of Medicine, Surgery and Dentistry, Via S. Allende 1, 84081, Baronissi(Salerno), Italy
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2DZ, UK
| | - Inês Falcao-Pires
- Department of Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Mauro Giacca
- King’s College London, Molecular Medicine Laboratory, 125 Caldharbour Lane, London WC2R2LS, United Kingdom
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34129 Trieste, Italy
| | - Nazha Hamdani
- Department of Clinical Pharmacology and Molecular Cardiology, Institute of Physiology, Ruhr University Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Universitätsstraße 150, D-44801 Bochum, Germany
| | - Stéphane Heymans
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, Bus 911, 3000 Leuven, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
- ICIN-Netherlands Heart Institute, Holland Heart House, Moreelsepark 1, 3511 EP Utrecht, the Netherlands
| | - Astrid Hooghiemstra
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081HZ, Amsterdam, The Netherlands
- Department of Medical Humanities, Amsterdam Public Health Research Institute, Amsterdam UMC, Location VUmc, De Boelelaan 1089a, 1081HV, Amsterdam, The Netherlands
| | - Annebet Leeuwis
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081HZ, Amsterdam, The Netherlands
| | - Dorien Hermkens
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, the Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1118, 1081HZ Amsterdam, the Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume, 447, 34129 Trieste, Italy
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano, 99, 34149 Trieste, Italy
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Nicolai-Fuchs-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
9
|
Abstract
Objective Arteriovenous fistulae (AVF) placed for hemodialysis have high flow rates that can stimulate left ventricular (LV) hypertrophy. LV hypertrophy generally portends poor cardiac outcomes, yet clinical studies point to superior cardiac-specific outcomes for patients with AVF when compared with other dialysis modalities. We hypothesize that AVF induce physiologic cardiac hypertrophy with cardioprotective features. Methods We treated 9- to 11-week-old C57Bl/6 male and female mice with sham laparotomy or an aortocaval fistula via a 25G needle. Cardiac chamber size and function were assessed with serial echocardiography, and cardiac computed tomography angiography. Hearts were harvested at 5 weeks postoperatively, and the collagen content was assessed with Masson's trichrome. Bulk messenger RNA sequencing was performed from LV of sham and AVF mice at 10 days. Differentially expressed genes were analyzed using Ingenuity Pathway Analysis (Qiagen) to identify affected pathways and predict downstream biological effects. Results Mice with AVF had similar body weight and wet lung mass, but increased cardiac mass compared with sham-operated mice. AVF increased cardiac output while preserving LV systolic and diastolic function, as well as indices of right heart function; all four cardiac chambers were enlarged, with a slight decrement in the relative LV wall thickness. Histology showed preserved collagen density within each of the four chambers without areas of fibrosis. RNA sequencing captured 19 384 genes, of which 857 were significantly differentially expressed, including transcripts from extracellular matrix-related genes, ion channels, metabolism, and cardiac fetal genes. The top upstream regulatory molecules predicted include activation of angiogenic (Vegf, Akt1), procardiomyocyte survival (Hgf, Foxm1, Erbb2, Lin9, Areg), and inflammation-related (CSF2, Tgfb1, TNF, Ifng, Ccr2, IL6) genes, as well as the inactivation of cardiomyocyte antiproliferative factors (Cdkn1a, FoxO3, α-catenin). The predicted downstream effects include a decrease in heart damage, and increased arrhythmia, angiogenesis, and cardiogenesis. There were no significant sex-dependent differences in the AVF-stimulated cardiac adaptation. Conclusions AVF stimulate adaptive cardiac hypertrophy in wild-type mice without heart failure or pathologic fibrosis. Transcriptional correlates suggest AVF-induced cardiac remodeling has some cardioprotective, although also arrhythmogenic features. (JVS–Vascular Science 2021;2:110-28.) Clinical Relevance Arteriovenous fistulae (AVF) are commonly used as access for hemodialysis in patients with end-stage renal disease. AVF induce a high-output state that is associated with long-term structural cardiac remodeling, including left ventricle hypertrophy, but this element has uncertain clinical significance. Although left ventricle hypertrophy has traditionally been associated with an increased risk of cardiovascular disease, clinical studies have suggested that cardiac-specific outcomes of patients with end-stage renal disease were better with AVF compared with other dialysis modalities. This study uses a mouse model of AVF to study the structural, functional, and molecular correlates of AVF-induced cardiac remodeling. It finds that AVF causes an adaptive cardiac hypertrophy without functional decline or fibrosis. Transcriptional correlates suggest an electrical remodeling and the upregulation of proangiogenic, procardiogenic, and prosurvival factors, implying that AVF-induced cardiac hypertrophy is potentially cardioprotective, but also arrhythmogenic.
Collapse
|
10
|
Novák J, Macháčková T, Krejčí J, Bienertová-Vašků J, Slabý O. MicroRNAs as theranostic markers in cardiac allograft transplantation: from murine models to clinical practice. Theranostics 2021; 11:6058-6073. [PMID: 33897899 PMCID: PMC8058726 DOI: 10.7150/thno.56327] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Congestive heart failure affects about 23 million people worldwide, and cardiac allograft transplantation remains one of the last options for patients with terminal refractory heart failure. Besides the infectious or oncological complications, the prognosis of patients after heart transplantation is affected by acute cellular or antibody-mediated rejection and allograft vasculopathy development. Current monitoring of both conditions requires the performance of invasive procedures (endomyocardial biopsy sampling and coronary angiography or optical coherence tomography, respectively) that are costly, time-demanding, and non-comfortable for the patient. Within this narrative review, we focus on the potential pathophysiological and clinical roles of microRNAs (miRNAs, miRs) in the field of cardiac allograft transplantation. Firstly, we provide a general introduction about the status of cardiac allograft function monitoring and the discovery of miRNAs as post-transcriptional regulators of gene expression and clinically relevant biomarkers found in the extracellular fluid. After this general introduction, information from animal and human studies are summarized to underline the importance of miRNAs both in the pathophysiology of the rejection process, the possibility of its modulation by altering miRNAs levels, and last but not least, about the use of miRNAs in the clinical practice to diagnose or predict the rejection occurrence.
Collapse
Affiliation(s)
- Jan Novák
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5-A18, 625 00, Brno, Czech Republic
- Second Department of Internal Medicine, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Kamenice 5-A35, 625 00, Brno, Czech Republic
| | - Táňa Macháčková
- Central European Institute of Technology, Masaryk University, Kamenice 5-A35, 625 00, Brno, Czech Republic
| | - Jan Krejčí
- Department of Cardiovascular Diseases, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691, Brno, Czech Republic
| | - Julie Bienertová-Vašků
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5-A18, 625 00, Brno, Czech Republic
- RECETOX, Faculty of Sciences, Masaryk University, Kamenice 5-A29, 625 00, Brno, Czech Republic
| | - Ondřej Slabý
- Central European Institute of Technology, Masaryk University, Kamenice 5-A35, 625 00, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| |
Collapse
|
11
|
Yang Z, Han F, Liao T, Zheng H, Luo Z, Ma M, He J, Li L, Ye Y, Zhang R, Huang Z, Zhang Y, Sun Q. Artemisinin Attenuates Transplant Rejection by Inhibiting Multiple Lymphocytes and Prolongs Cardiac Allograft Survival. Front Immunol 2021; 12:634368. [PMID: 33717174 PMCID: PMC7943449 DOI: 10.3389/fimmu.2021.634368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/07/2021] [Indexed: 01/06/2023] Open
Abstract
Immunological rejection is an important factor resulting in allograft dysfunction, and more valid therapeutic methods need to be explored to improve allograft outcomes. Many researches have indicated that artemisinin and its derivative exhibits immunosuppressive functions, apart from serving as a traditional anti-malarial drug. In this assay, we further explored the therapeutic effects of artemisinin for transplant rejection in a rat cardiac transplantation model. We found that it markedly attenuated allograft rejection and histological injury and significantly prolonged the survival of allograft. Upon further exploring the mechanism, we demonstrated that artemisinin not only attenuated T cell-mediated rejection (TCMR) by reducing effector T cell infiltration and inflammatory cytokine secretion and increasing regulatory T cell infiltration and immunoregulatory cytokine levels, but also attenuated antibody-mediated rejection (ABMR) through inhibition of B cells activation and antibody production. Furthermore, artemisinin also reduced macrophage infiltration in allografts, which was determined to be important for TCMR and ABMR. Moreover, we demonstrated that artemisinin significantly inhibited the function of pure T cells, B cells, and macrophages in vitro. All in all, this study provide evidence that artemisinin significantly attenuates TCMR and ABMR by targeting multiple effectors. Therefore, this agent might have potential for use in clinical settings to protect against transplant rejection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhengyu Huang
- Research Institute of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yannan Zhang
- Research Institute of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiquan Sun
- Research Institute of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Liu B, Wang B, Zhang X, Lock R, Nash T, Vunjak-Novakovic G. Cell type-specific microRNA therapies for myocardial infarction. Sci Transl Med 2021; 13:eabd0914. [PMID: 33568517 PMCID: PMC8848299 DOI: 10.1126/scitranslmed.abd0914] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
Current interventions fail to recover injured myocardium after infarction and prompt the need for development of cardioprotective strategies. Of increasing interest is the therapeutic use of microRNAs to control gene expression through specific targeting of mRNAs. In this Review, we discuss current microRNA-based therapeutic strategies, describing the outcomes and limitations of key microRNAs with a focus on target cell types and molecular pathways. Last, we offer a perspective on the outlook of microRNA therapies for myocardial infarction, highlighting the outstanding challenges and emerging strategies.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bryan Wang
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor Nash
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| |
Collapse
|
13
|
Schimmel K, Stojanović SD, Huang CK, Jung M, Meyer MH, Xiao K, Grote-Levi L, Bär C, Pfanne A, Mitzka S, Just A, Geffers R, Bock K, Kenneweg F, Kleemiß F, Falk CS, Fiedler J, Thum T. Combined high-throughput library screening and next generation RNA sequencing uncover microRNAs controlling human cardiac fibroblast biology. J Mol Cell Cardiol 2021; 150:91-100. [PMID: 33127387 DOI: 10.1016/j.yjmcc.2020.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Myocardial fibrosis is a hallmark of the failing heart, contributing to the most common causes of deaths worldwide. Several microRNAs (miRNAs, miRs) controlling cardiac fibrosis were identified in recent years; however, a more global approach to identify miRNAs involved in fibrosis is missing. METHODS AND RESULTS Functional miRNA mimic library screens were applied in human cardiac fibroblasts (HCFs) to identify annotated miRNAs inducing proliferation. In parallel, miRNA deep sequencing was performed after subjecting HCFs to proliferating and resting stimuli, additionally enabling discovery of novel miRNAs. In-depth in vitro analysis confirmed the pro-fibrotic nature of selected, highly conserved miRNAs miR-20a-5p and miR-132-3p. To determine downstream cellular pathways and their role in the fibrotic response, targets of the annotated miRNA candidates were modulated by synthetic siRNA. We here provide evidence that repression of autophagy and detoxification of reactive oxygen species by miR-20a-5p and miR-132-3p explain some of their pro-fibrotic nature on a mechanistic level. CONCLUSION We here identified both miR-20a-5p and miR-132-3p as crucial regulators of fibrotic pathways in an in vitro model of human cardiac fibroblast biology.
Collapse
Affiliation(s)
- Katharina Schimmel
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Stevan D Stojanović
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Cheng-Kai Huang
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Martin H Meyer
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Lea Grote-Levi
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Angelika Pfanne
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Saskia Mitzka
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Research Group Genome Analytics, Braunschweig, Germany
| | - Katharina Bock
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Felix Kleemiß
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Christine S Falk
- Transplant Immunology, Integrated Research and Treatment Centre Transplantation, Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
14
|
Zhao H, Yang H, Geng C, Chen Y, Pang J, Shu T, Zhao M, Tang Y, Li Z, Li B, Hou C, Song X, Wu A, Guo X, Chen S, Liu B, Yan C, Wang J. Role of IgE-FcεR1 in Pathological Cardiac Remodeling and Dysfunction. Circulation 2020; 143:1014-1030. [PMID: 33305586 DOI: 10.1161/circulationaha.120.047852] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Immunoglobulin E (IgE) belongs to a class of immunoglobulins involved in immune response to specific allergens. However, the roles of IgE and IgE receptor (FcεR1) in pathological cardiac remodeling and heart failure are unknown. METHODS Serum IgE levels and cardiac FcεR1 expression were assessed in diseased hearts from human and mouse. The role of FcεR1 signaling in pathological cardiac remodeling was explored in vivo by FcεR1 genetic depletion, anti-IgE antibodies, and bone marrow transplantation. The roles of the IgE-FcεR1 pathway were further evaluated in vitro in primary cultured rat cardiomyocytes and cardiac fibroblasts (CFs). RNA sequencing and bioinformatic analyses were used to identify biochemical changes and signaling pathways that are regulated by IgE/FcεR1. RESULTS Serum IgE levels were significantly elevated in patients with heart failure as well as in 2 mouse cardiac disease models induced by chronic pressure overload via transverse aortic constriction and chronic angiotensin II infusion. Interestingly, FcεR1 expression levels were also significantly upregulated in failing hearts from human and mouse. Blockade of the IgE-FcεR1 pathway by FcεR1 knockout alleviated transverse aortic constriction- or angiotensin II-induced pathological cardiac remodeling or dysfunction. Anti-IgE antibodies (including the clinical drug omalizumab) also significantly alleviated angiotensin II-induced cardiac remodeling. Bone marrow transplantation experiments indicated that IgE-induced cardiac remodeling was mediated through non-bone marrow-derived cells. FcεR1 was found to be expressed in both cardiomyocytes and CFs. In cultured rat cardiomyocytes, IgE-induced cardiomyocyte hypertrophy and hypertrophic marker expression were abolished by depleting FcεR1. In cultured rat CFs, IgE-induced CF activation and matrix protein production were also blocked by FcεR1 deficiency. RNA sequencing and signaling pathway analyses revealed that transforming growth factor-β may be a critical mediator, and blocking transforming growth factor-β indeed alleviated IgE-induced cardiomyocyte hypertrophy and cardiac fibroblast activation in vitro. CONCLUSIONS Our findings suggest that IgE induction plays a causative role in pathological cardiac remodeling, at least partially via the activation of IgE-FcεR1 signaling in cardiomyocytes and CFs. Therapeutic strategies targeting the IgE-FcεR1 axis may be effective for managing IgE-mediated cardiac remodeling.
Collapse
Affiliation(s)
- Hongmei Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Hongqin Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Chi Geng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Yang Chen
- Department of Pharmacology, School of Basic Medical Sciences, Inner Mongolia Medical University, Huhhot, China (Y.C.)
| | - Junling Pang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Ting Shu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Meijun Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Yaqin Tang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Zhiwei Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Baicun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Cuiliu Hou
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Xiaomin Song
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| | - Aoxue Wu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing (A.W., X.G.)
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing (A.W., X.G.)
| | - Si Chen
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, NY (S.C., B.L., C.Y.)
| | - Bin Liu
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, NY (S.C., B.L., C.Y.)
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, NY (S.C., B.L., C.Y.)
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing (H.Z., H.Y., C.G., J.P., T.S., M.Z., Y.T., Z.L., B.L., C.H., X.S., J.W.)
| |
Collapse
|
15
|
Florio MC, Magenta A, Beji S, Lakatta EG, Capogrossi MC. Aging, MicroRNAs, and Heart Failure. Curr Probl Cardiol 2020; 45:100406. [PMID: 30704792 PMCID: PMC10544917 DOI: 10.1016/j.cpcardiol.2018.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 12/23/2018] [Indexed: 12/12/2022]
Abstract
Aging is a major risk factor for heart failure, one of the leading causes of death in Western society. The mechanisms that underlie the different forms of heart failure have been elucidated only in part and the role of noncoding RNAs is still poorly characterized. Specifically, microRNAs (miRNAs), a class of small noncoding RNAs that can modulate gene expression at the posttranscriptional level in all cells, including myocardial and vascular cells, have been shown to play a role in heart failure with reduced ejection fraction. In contrast, miRNAs role in heart failure with preserved ejection fraction, the predominant form of heart failure in the elderly, is still unknown. In this review, we will focus on age-dependent miRNAs in heart failure and on some other conditions that are prevalent in the elderly and are frequently associated with heart failure with preserved ejection fraction.
Collapse
|
16
|
Ding Y, Wang Y, Zhang W, Jia Q, Wang X, Li Y, Lv S, Zhang J. Roles of Biomarkers in Myocardial Fibrosis. Aging Dis 2020; 11:1157-1174. [PMID: 33014530 PMCID: PMC7505259 DOI: 10.14336/ad.2020.0604] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Myocardial fibrosis is observed in various cardiovascular diseases and plays a key role in the impairment of cardiac function. Endomyocardial biopsy, as the gold standard for the diagnosis of myocardial fibrosis, has limitations in terms of clinical application. Therefore, biomarkers have been recommended for noninvasive assessment of myocardial fibrosis. This review discusses the role of biomarkers in myocardial fibrosis from the perspective of collagen.
Collapse
Affiliation(s)
- Yuejia Ding
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yuan Wang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Wanqin Zhang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qiujin Jia
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoling Wang
- 3Qian'an Hospital of Traditional Chinese Medicine, Qian'an 064400, China
| | - Yanyang Li
- 4Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shichao Lv
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.,2Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300000, China
| | - Junping Zhang
- 1First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| |
Collapse
|
17
|
Function analysis of differentially expressed microRNAs in TGF-β1-induced cardiac fibroblasts differentiation. Biosci Rep 2020; 39:BSR20182048. [PMID: 31527065 PMCID: PMC6822545 DOI: 10.1042/bsr20182048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 08/20/2019] [Accepted: 09/13/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Cardiac fibroblasts differentiation plays a critical role in cardiac remodeling and failure, but the underlying molecular mechanisms are still poorly understood. MicroRNAs (miRNAs) had been identified as important regulators during cell differentiation. The aim of the present study was to screen the miRNAs involved in regulation of cardiac fibroblasts differentiation. METHODS The differentiation of rat cardiac fibroblasts into myofibroblasts was induced by transforming growth factor-β1 (TGF-β1). Small RNA sequencing was then applied to detect the differentially expressed miRNAs. RESULTS A total of 450 known miRNAs were detected, and 127 putative novel miRNAs were predicted by miRDeep2 analysis. DEGseq analysis and qRT-PCR confirmed that 24 known miRNAs were differentially expressed in TGF-β1-induced cardiac fibroblasts, including three up-regulated miRNAs and 21 down-regulated miRNAs. After miRNAs target genes prediction by miRanda algorithm, pathway analysis showed that these potential target genes were involved in Calcium signaling pathway, Type II diabetes mellitus, and Glutamatergic synapse pathway, etc. Meanwhile, seven putative miRNAs were also detected differentially expressed during TGF-β1-induced cardiac fibroblasts differentiation. CONCLUSIONS These differentially expressed miRNAs might play critical roles in cardiac fibroblasts differentiation. Altered expression of miRNAs may yield new insights into the underlying mechanisms of cardiac fibrosis and provide novel mechanism-based therapeutic strategies for cardiac fibrosis.
Collapse
|
18
|
Liu X, Zhang Y, Jiang P, Cai J, Fu Q, Li X, Li Z. Ultrasonic cardiogram and MiRNA-21 analysis of cardiac dysfunction in patients with cardiac arrest following cardiopulmonary resuscitation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 190:105284. [PMID: 32018074 DOI: 10.1016/j.cmpb.2019.105284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/08/2019] [Accepted: 12/12/2019] [Indexed: 06/10/2023]
Abstract
PURPOSE To explore correlations between the serum level of miRNA-21 expression and cardiac dysfunction severity after cardiopulmonary resuscitation (CPR) using ultrasonic cardiogram. METHODS Thirty-nine patients with cardiopulmonary arrest receiving successful CPR and forty-one healthy participants were recruited in the study. Reverse Transcription-Polymerase Chain Reaction (RT-PCR) and immunochemiluminometric assays was used to examine the serum miRNA-21 level and the concentration of cardiac troponins T and I, respectively. Indices of Electrocardiogram (ECG) and cardiac dysfunction measured by ultrasound of patients in the case group were used to assess cardiac function after CPR. Furthermore, the correlation between the serum level of miRNA-21 expression and severity of cardiac dysfunction was analyzed by Spearman correlation analysis. RESULTS As compared to the control group, the serum level of miRNA-21 expression, as well as cardiac troponin T and I levels in the case group were significantly higher (p = 0.000). The miRNA-21 expression level in the patients at IV grade of cardiac function were substantially higher than patients at III grade (p = 0.015). There was no significant difference in level of cardiac troponins T and I between patients at III grade and patients at IV grade (p > 0.05). Further, Spearman correlation analysis revealed that the level of miRNA-21 expression was negatively correlated with cardiac function index in the ultrasound imaging: E peak, E/A value, LVEF and LVEDD (r = 0.617, 0.535, 0.612, 0.573, P = 0.012, 0.009, 0.008, 0.011), but was positively correlated with the level of cardiac troponins T and I (r = 0.546,0.582, P = 0.006,0.007) and the severity of cardiac dysfunction (r = 0.859, p < 0.05). CONCLUSION The level of miRNA-21 is higher after CPR is closely related to the severity of cardiac dysfunction that is measured by ultrasound, suggesting that it may serve as a potential biomarker.
Collapse
Affiliation(s)
- Xing Liu
- Department of Emergency. Shenzhen Longhua District Center Hospital, Shenzhen, Guangdong 518110, PR China
| | - Yongguang Zhang
- Department of Medicine, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510000, PR China
| | - Peng Jiang
- Department of Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518101, PR China
| | - Jiachen Cai
- Department of Medicine, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510000, PR China
| | - Qiuhong Fu
- Department of Emergency. Shenzhen Longhua District Center Hospital, Shenzhen, Guangdong 518110, PR China
| | - Xiaolei Li
- Department of Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518101, PR China
| | - Zhou Li
- Department of Medicine, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510000, PR China.
| |
Collapse
|
19
|
An Overview of Non-coding RNAs and Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:3-45. [PMID: 32285403 DOI: 10.1007/978-981-15-1671-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease management and timely diagnosis remain a major dilemma. Delineating molecular mechanisms of cardiovascular diseases is opening horizon in the field of molecular medicines and in the development of early diagnostic markers. Non-coding RNAs are the highly functional and vibrant nucleic acids and are known to be involved in the regulation of endothelial cells, vascular and smooth muscles cells, cardiac metabolism, ischemia, inflammation and many processes in cardiovascular system. This chapter is comprehensively focusing on the overview of the non-coding RNAs including their discovery, generation, classification and functional regulation. In addition, overview regarding different non-coding RNAs as long non-coding, siRNAs and miRNAs involvement in the cardiovascular diseases is also addressed. Detailed functional analysis of this vast group of highly regulatory molecules will be promising for shaping future drug discoveries.
Collapse
|
20
|
MicroRNA-142-5p is Up-regulated on Allogeneic Immune Responses and Up-regulates MHC Class II Expression in Human Umbilical Vein Endothelial Cells. Transplant Proc 2020; 53:408-416. [PMID: 32616346 DOI: 10.1016/j.transproceed.2020.05.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE MicroRNA could be biomarker and therapeutic target for rejection. The aim of this study was to investigate the role of miR-142-5p in allogeneic immune responses using in vitro and in vivo models. MATERIALS AND METHODS Primary and immortalized human umbilical vein endothelial cells (HUVECs) were cultured with unrelated blood mononuclear cells to induce allogeneic immune responses. Syngeneic and allogeneic skin graft was performed in mice. Flow cytometry, quantitative reverse transcription-polymerase chain reaction, and Western blotting was performed to understand the underlying mechanisms. RESULTS miR-142-5p was up-regulated in primary HUVEC and a HUVEC line when allogeneic immune responses were elicited. miR-142-5p was also up-regulated in the murine allogeneic skin graft. Overexpression of miR-142-5p in HUVEC increased the expression of HLA-ABC and HLA-DR additively to allogeneic immune responses, suggesting a possible increase in alloantigen presentation. Inhibition of miR-142-5p reduced the expression of HLA-DR. ZEB1, a putative target gene of miR-142-5p, was down-regulated in HUVEC on allogeneic immune response as well as in murine allogeneic skin graft. CONCLUSION These results suggest that the up-regulation of miR-142-5p on allogeneic immune response might facilitate endothelial activation to exacerbate rejection.
Collapse
|
21
|
Li J, Wei L, Han Z, Chen Z, Zhang Q. Long non-coding RNA X-inactive specific transcript silencing ameliorates primary graft dysfunction following lung transplantation through microRNA-21-dependent mechanism. EBioMedicine 2020; 52:102600. [PMID: 31981974 PMCID: PMC6976928 DOI: 10.1016/j.ebiom.2019.102600] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/25/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Background Primary graft dysfunction (PGD) is a known acute lung injury (ALI) and a major cause of fatality post-lung transplantation. Though some long non-coding RNAs (lncRNAs) have been studied in ALI through regulation of microRNAs (miRNAs), their effects on PGD remain undefined. The present study aims to explore the underlying mechanism of lncRNA X-inactive specific transcript (XIST) in PGD after lung transplantation. Methods Initially, the expression of miR-21, IL-12A and XIST was determined by RT-qPCR and western blot analysis. The dual luciferase reporter assay, RNA pull-down and RIP assay were performed to identify the targeting relationship between miR-21 and IL-12A and the binding relationship between miR-21 and XIST. Loss- and gain-of-function investigations were conducted in rats treated with prolonged cold ischemia and polymorphonuclear neutrophils (PMNs). Findings miR-21 was decreased, whilst XIST and IL-12A were increased in the bronchoalveolar lavage fluid of PGD patients after lung transplantation. Enhanced miR-21 expression in rats and PMNs resulted in downregulated expression of pro-inflammatory factors and chemokines, and enhanced the apoptosis of PMNs. XIST was found to upregulate IL-12A expression in a miR-21-dependent manner. Additionally, XIST silencing enhanced the apoptosis of PMNs and inhibited the neutrophil extracellular trap (NET) formation through upregulation of miR-21 but downregulation of IL-12A in vivo. Interpretation In summary, lncRNA XIST upregulates IL-12A by binding to miR-21, thereby inducing NET formation and accelerating PGD after lung transplantation. This suggests that inhibition of XIST and NET may be beneficial for the treatment of PGD.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, PR China
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, PR China.
| | - Zhijun Han
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, PR China
| | - Zhong Chen
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, PR China
| | - Quan Zhang
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450003, PR China
| |
Collapse
|
22
|
Chiasson V, Takano APC, Guleria RS, Gupta S. Deficiency of MicroRNA miR-1954 Promotes Cardiac Remodeling and Fibrosis. J Am Heart Assoc 2019; 8:e012880. [PMID: 31640463 PMCID: PMC6898847 DOI: 10.1161/jaha.119.012880] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/18/2019] [Indexed: 12/17/2022]
Abstract
Background Cardiac fibrosis occurs because of disruption of the extracellular matrix network leading to myocardial dysfunction. Angiotensin II (AngII) has been implicated in the development of cardiac fibrosis. Recently, microRNAs have been identified as an attractive target for therapeutic intervention in cardiac pathologies; however, the underlying mechanism of microRNAs in cardiac fibrosis remains unclear. Next-generation sequencing analysis identified a novel characterized microRNA, miR-1954, that was significantly reduced in AngII-infused mice. The finding led us to hypothesize that deficiency of miR-1954 triggers cardiac fibrosis. Methods and Results A transgenic mouse was created using α-MHC (α-myosin heavy chain) promoter and was challenged with AngII infusion. AngII induced cardiac hypertrophy and remodeling. The in vivo overexpression of miR-1954 showed significant reduction in cardiac mass and blood pressure in AngII-infused mice. Further analysis showed significant reduction in cardiac fibrotic genes, hypertrophy marker genes, and an inflammatory gene and restoration of a calcium-regulated gene (Atp2a2 [ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2]; also known as SERCA2), but no changes were observed in apoptotic genes. THBS1 (thrombospondin 1) is indicated as a target gene for miR-1954. Conclusions Our findings provide evidence, for the first time, that miR-1954 plays a critical role in cardiac fibrosis by targeting THBS1. We conclude that promoting the level of miR-1954 would be a promising strategy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
| | | | - Rakeshwar S. Guleria
- Department of Medical PhysiologyTexas A&M UniversityTempleTX
- VISN 17 Center of Excellence on Returning War VeteransWacoTX
| | - Sudhiranjan Gupta
- Department of BiologyBaylor UniversityWacoTX
- Department of Medical PhysiologyTexas A&M UniversityTempleTX
- VISN 17 Center of Excellence on Returning War VeteransWacoTX
| |
Collapse
|
23
|
Chou CH, Hung CS, Liao CW, Wei LH, Chen CW, Shun CT, Wen WF, Wan CH, Wu XM, Chang YY, Wu VC, Wu KD, Lin YH. IL-6 trans-signalling contributes to aldosterone-induced cardiac fibrosis. Cardiovasc Res 2019; 114:690-702. [PMID: 29360942 DOI: 10.1093/cvr/cvy013] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 01/18/2018] [Indexed: 12/13/2022] Open
Abstract
Aims An excess of aldosterone results in cardiac remodelling and fibrosis. Interleukin-6 (IL-6) is a key mediator in the fibrotic process; however, the effect of aldosterone on the expression of IL-6 remains unclear. We investigated whether aldosterone induces the expression of IL-6 and thereby contributes to the fibrotic process. Methods and results In this clinical study, we prospectively enrolled 25 patients with primary aldosteronism (PA) and 26 patients with essential hypertension (EH). The PA patients had higher plasma IL-6 levels, left ventricular mass index, degree of myocardial fibrosis, and more impaired diastolic function than the EH patients. In addition, plasma IL-6 levels were positively correlated with 24-h urinary aldosterone and echocardiographic parameters. In cell studies, we investigated the possible molecular mechanism how aldosterone-induced IL-6 secretion and the further effects of collagen production. Aldosterone significantly induced IL-6 protein and mRNA production in human umbilical vein endothelial cells. Intracellular signalling occurred through the mineralocorticoid receptor/PI3K/Akt/NF-kB pathway. In cardiac fibroblasts, IL-6 trans-signalling played a critical role in aldosterone-induced IL-6-enhanced fibrosis-related factor expression. To further investigate the role of IL-6 trans-signalling in aldosterone-induced cardiac fibrosis, we measured the severity of myocardial fibrosis in aldosterone infusion mice models including an IL-6 chemical inhibitor and Sgp130 Knockin Transgenic Mice. Mice receiving recombinant soluble gp130 and Sgp130 Knockin Transgenic Mice prevented myocardial fibrosis and cardiac hypertrophy by aldosterone infusion. Conclusions IL-6 trans-signalling contributes to aldosterone-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and National Taiwan University College of Medicine, 100, Taipei, Taiwan
| | - Chi-Sheng Hung
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Telehealth Center, National Taiwan University Hospital, 100, Taipei, Taiwan
| | - Che-Wei Liao
- Department of Internal Medicine, National Taiwan University Hospital 300, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Lin-Hung Wei
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and National Taiwan University College of Medicine, 100, Taipei, Taiwan
| | - Ching-Way Chen
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, 640, Douliu, Taiwan
| | - Chia-Tung Shun
- Department and Graduate Institute of Forensic Medicine, College of Medicine, National Taiwan University, 100, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital, 100, Taipei, Taiwan
| | - Wen-Fen Wen
- Department of Pathology, National Taiwan University Hospital, 100, Taipei, Taiwan
| | - Cho-Hua Wan
- Taiwan Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Xue-Ming Wu
- Department of Internal Medicine, Taoyuan General Hospital, 300, Taoyuan, Taiwan
| | - Yi-Yao Chang
- Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, 220, New Taipei City, Taiwan
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kwan-Dun Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Hung Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | |
Collapse
|
24
|
Mohammadzadeh N, Lunde IG, Andenæs K, Strand ME, Aronsen JM, Skrbic B, Marstein HS, Bandlien C, Nygård S, Gorham J, Sjaastad I, Chakravarti S, Christensen G, Engebretsen KVT, Tønnessen T. The extracellular matrix proteoglycan lumican improves survival and counteracts cardiac dilatation and failure in mice subjected to pressure overload. Sci Rep 2019; 9:9206. [PMID: 31235849 PMCID: PMC6591256 DOI: 10.1038/s41598-019-45651-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Left ventricular (LV) dilatation is a key step in transition to heart failure (HF) in response to pressure overload. Cardiac extracellular matrix (ECM) contains fibrillar collagens and proteoglycans, important for maintaining tissue integrity. Alterations in collagen production and cross-linking are associated with cardiac LV dilatation and HF. Lumican (LUM) is a collagen binding proteoglycan with increased expression in hearts of patients and mice with HF, however, its role in cardiac function remains poorly understood. To examine the role of LUM in pressure overload induced cardiac remodeling, we subjected LUM knock-out (LUMKO) mice to aortic banding (AB) and treated cultured cardiac fibroblasts (CFB) with LUM. LUMKO mice exhibited increased mortality 1-14 days post-AB. Echocardiography revealed increased LV dilatation, altered hypertrophic remodeling and exacerbated contractile dysfunction in surviving LUMKO 1-10w post-AB. LUMKO hearts showed reduced collagen expression and cross-linking post-AB. Transcriptional profiling of LUMKO hearts by RNA sequencing revealed 714 differentially expressed transcripts, with enrichment of cardiotoxicity, ECM and inflammatory pathways. CFB treated with LUM showed increased mRNAs for markers of myofibroblast differentiation, proliferation and expression of ECM molecules important for fibrosis, including collagens and collagen cross-linking enzyme lysyl oxidase. In conclusion, we report the novel finding that lack of LUM attenuates collagen cross-linking in the pressure-overloaded heart, leading to increased mortality, dilatation and contractile dysfunction in mice.
Collapse
Affiliation(s)
- Naiyereh Mohammadzadeh
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Center for Molecular Medicine Norway, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Kine Andenæs
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Mari E Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Biljana Skrbic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Henriette S Marstein
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Caroline Bandlien
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Ståle Nygård
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Shukti Chakravarti
- Department of Medicine, Johns Hopkins University, Baltimore, PhD, USA
- Department of Ophthalmology and Pathology, NYU Langone Health, Alexandria Life Sciences Center, West Tower, New York, NY, NY10011, USA
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Kristin V T Engebretsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Department of Surgery, Vestre Viken Hospital, Drammen, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.
- KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
25
|
Micheu MM, Scarlatescu AI, Scafa-Udriste A, Dorobantu M. The Winding Road of Cardiac Regeneration-Stem Cell Omics in the Spotlight. Cells 2018; 7:255. [PMID: 30544622 PMCID: PMC6315576 DOI: 10.3390/cells7120255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/18/2022] Open
Abstract
Despite significant progress in treating ischemic cardiac disease and succeeding heart failure, there is still an unmet need to develop effective therapeutic strategies given the persistent high-mortality rate. Advances in stem cell biology hold great promise for regenerative medicine, particularly for cardiac regeneration. Various cell types have been used both in preclinical and clinical studies to repair the injured heart, either directly or indirectly. Transplanted cells may act in an autocrine and/or paracrine manner to improve the myocyte survival and migration of remote and/or resident stem cells to the site of injury. Still, the molecular mechanisms regulating cardiac protection and repair are poorly understood. Stem cell fate is directed by multifaceted interactions between genetic, epigenetic, transcriptional, and post-transcriptional mechanisms. Decoding stem cells' "panomic" data would provide a comprehensive picture of the underlying mechanisms, resulting in patient-tailored therapy. This review offers a critical analysis of omics data in relation to stem cell survival and differentiation. Additionally, the emerging role of stem cell-derived exosomes as "cell-free" therapy is debated. Last but not least, we discuss the challenges to retrieve and analyze the huge amount of publicly available omics data.
Collapse
Affiliation(s)
- Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alina Ioana Scarlatescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| |
Collapse
|
26
|
Gao Q, Xu L, Yang Q, Guan TJ. MicroRNA-21 contributes to high glucose-induced fibrosis in peritoneal mesothelial cells in rat models by activation of the Ras-MAPK signaling pathway via Sprouty-1. J Cell Physiol 2018; 234:5915-5925. [PMID: 30515805 DOI: 10.1002/jcp.26941] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/13/2018] [Indexed: 12/26/2022]
Abstract
Peritoneal fibrosis remains to be one of the most severe causes of failure in continuous peritoneal dialysis. The current study cultured peritoneal mesothelial cells in high glucose to stimulate the environment of peritoneal fibrosis model in rats, and investigate whether microRNA-21 (miR-21) targeting Sprouty-1 affects high glucose-induced fibrosis in peritoneal mesothelial cells via the rennin angiotensin system (Ras)-mitogen-activated protein kinase (MAPK) signaling pathway, as well as potential mechanisms. Peritoneal tissues in fibrosis rats were collected to extract peritoneal mesothelial cells, which, after in vitro culture, were transfected with a series of mimic or inhibitor of miR-21, or the small interfering RNA (siRNA) against Sprouty-1. Reverse-transcription quantitative polymerase chain reaction and western blot analyses were performed to determine the levels of related genes or proteins. MTT assay and flow cytometry were conducted to observe the cell viability and cell apoptosis of peritoneal mesothelial cells. Dual-luciferase reporter gene assay revealed that Sprouty-1 is the target gene of miR-21. Peritoneal fibrosis manifested with elevated miR-21, extracellular-signal-regulated kinase (ERK), c-Jun NH2-terminal protein kinase (JNK), RAS and p38MAPK but reduced Sprouty-1. Cells transfected with miR-21 mimic exhibited decreased Sprouty-1 expressions, but increased levels of ERK, JNK, RAS, and p38MAPK. As for cellular process, miR-21 mimic or siRNA against Sprouty-1 exposure reduced cell viability, which resulted in more cells arrested at the G1 stage, and induced apoptosis. In contrast, miR-21 inhibitor exposure was observed to have induced effects on peritoneal mesothelial cells. These key findings provide evidence that miR-21 inhibits Sprouty-1 to promote the progression of fibrosis in peritoneal mesothelial cells by activating the Ras-MAPK signaling pathway.
Collapse
Affiliation(s)
- Qing Gao
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Lin Xu
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Qian Yang
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Tian-Jun Guan
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
27
|
Syed M, Ball JP, Mathis KW, Hall ME, Ryan MJ, Rothenberg ME, Yanes Cardozo LL, Romero DG. MicroRNA-21 ablation exacerbates aldosterone-mediated cardiac injury, remodeling, and dysfunction. Am J Physiol Endocrinol Metab 2018; 315:E1154-E1167. [PMID: 30153065 PMCID: PMC6336952 DOI: 10.1152/ajpendo.00155.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Primary aldosteronism is characterized by excess aldosterone secretion by the adrenal gland independent of the renin-angiotensin system and accounts for ~10% of hypertensive patients. Excess aldosterone causes cardiac hypertrophy, fibrosis, inflammation, and hypertension. The molecular mechanisms that trigger the onset and progression of aldosterone-mediated cardiac injury remain incompletely understood. MicroRNAs (miRNAs) are endogenous, small, noncoding RNAs that have been implicated in multiple cardiac pathologies; however, their regulation and role in aldosterone-mediated cardiac injury and dysfunction remains mostly unknown. We previously reported that microRNA-21 (miR-21) is the most upregulated miRNA by excess aldosterone in the left ventricle in a rat experimental model of primary aldosteronism. To elucidate the role of miR-21 in aldosterone-mediated cardiac injury and dysfunction, miR-21 knockout mice and their wild-type littermates were treated with aldosterone infusion and salt in the drinking water for 2 or 8 wk. miR-21 genetic ablation exacerbated aldosterone/salt-mediated cardiac hypertrophy and cardiomyocyte cross-sectional area. Furthermore, miR-21 genetic ablation increased the cardiac expression of fibrosis and inflammation markers and fetal gene program. miR-21 genetic ablation increased aldosterone/salt-mediated cardiac dysfunction but did not affect aldosterone/salt-mediated hypertension. miR-21 target gene Sprouty 2 may be implicated in the cardiac effects of miR-21 genetic ablation. Our study shows that miR-21 genetic ablation exacerbates aldosterone/salt-mediated cardiac hypertrophy, injury, and dysfunction blood pressure independently. These results suggest that miR-21 plays a protective role in the cardiac pathology triggered by excess aldosterone. Furthermore, miR-21 supplementation may be a novel therapeutic approach to abolish or mitigate excess aldosterone-mediated cardiovascular deleterious effects in primary aldosteronism.
Collapse
Affiliation(s)
- Maryam Syed
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jana P Ball
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Keisa W Mathis
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- G.V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center , Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
28
|
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, UK
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, UK.,Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, 31-008 Anny 12, Krakow, Poland
| |
Collapse
|
29
|
Wang J, Duan L, Gao Y, Zhou S, Liu Y, Wei S, An S, Liu J, Tian L, Wang S. Angiotensin II receptor blocker valsartan ameliorates cardiac fibrosis partly by inhibiting miR-21 expression in diabetic nephropathy mice. Mol Cell Endocrinol 2018; 472:149-158. [PMID: 29233785 DOI: 10.1016/j.mce.2017.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/29/2017] [Accepted: 12/08/2017] [Indexed: 12/21/2022]
Abstract
Cardiac fibrosis with diabetic nephropathy (DN) is one of major diabetic complications. miR-21 and MMP-9 were closely associated with fibrosis diseases. Angiotensin II receptor blockers (ARB) have cardioprotective effects. However, it remains unclear whether miR-21 was involved in the mechanism of cardiac fibrosis with DN by target MMP-9 and ARB ameliorates cardiac fibrosis partly by inhibiting miR-21 expression. In this study, In Situ Hybridization(ISH), RT-PCR, cell transfection, western blotting and laser confocal telescope were used, respectively. ISH showed that miR-21, concentrated in cytoplasmic foci in the proximity of the nucleus, was mainly localized in cardiac fibroblasts and at relatively low levels in cardiomyocytes within cardiac tissue with DN. RT-PCR showed that miR-21 expression was significantly enhanced in cardiac tissue with DN, accompanied by the increase of col-IV, FN, CVF, PVCA, LVMI, HWI and NT-pro-BNP (p < 0.05). Bioinformatics analysis and Luciferase reporter gene assays showed that MMP-9 was a validated target of miR-21. Furthermore, cell transfection experiments showed that miR-21 overexpression directly decreased MMP-9 expression. Interestingly, miR-21 levels in cardiac tissue was positively correlated with ACR (r = -0.870, P = 0.003), whereas, uncorrelated with SBP, HbA1C and T-Cho (p > 0.05). More importantly, ARB can significantly decrease miR-21 expression in cardiac tissue, cardiac fibroblasts and serum. Overall, our results suggested that miR-21 may contribute to the pathogenesis of cardiac fibrosis with DN by target MMP-9, and that miR-21 may be a new possible therapeutic target for ARB in cardiac fibrosis with DN.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Endocrinology, Gansu Provincial People's Hospital, Gansu Key Laboratory of Endocrine and Metabolism, 204 Donggang West Road, Lanzhou, 730000, PR China.
| | - Lijun Duan
- Department of Gynecology and Obstetrics, Gansu Provincial People's Hospital, 204 Donggang West Road, Lanzhou 730000, PR China.
| | - Yanbin Gao
- Metabolic Disease Center, School of Traditional Chinese Medical, Capital Medical University, Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, 100069, PR China
| | - Shuhong Zhou
- Department of Rheumatology and Immunology, Gansu Provincial People's Hospital, 204 Donggang West Road, Lanzhou, 730000, PR China
| | - Yongming Liu
- Department of Geriatric Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, PR China
| | - Suhong Wei
- Department of Endocrinology, Gansu Provincial People's Hospital, Gansu Key Laboratory of Endocrine and Metabolism, 204 Donggang West Road, Lanzhou, 730000, PR China
| | - Siqin An
- Department of Endocrinology, Gansu Provincial People's Hospital, Gansu Key Laboratory of Endocrine and Metabolism, 204 Donggang West Road, Lanzhou, 730000, PR China
| | - Jing Liu
- Department of Endocrinology, Gansu Provincial People's Hospital, Gansu Key Laboratory of Endocrine and Metabolism, 204 Donggang West Road, Lanzhou, 730000, PR China
| | - Liming Tian
- Department of Endocrinology, Gansu Provincial People's Hospital, Gansu Key Laboratory of Endocrine and Metabolism, 204 Donggang West Road, Lanzhou, 730000, PR China
| | - Shaocheng Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Hospital, Tianjin, 300070, PR China
| |
Collapse
|
30
|
Lu X, Wu J, Ma M, Wu X, Wen J, Yu J. An integrated deep sequencing analysis of microRNAs in transplanted corneas. Mol Immunol 2018; 101:429-439. [DOI: 10.1016/j.molimm.2018.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/28/2018] [Accepted: 06/09/2018] [Indexed: 10/28/2022]
|
31
|
Guzik TJ, Cosentino F. Epigenetics and Immunometabolism in Diabetes and Aging. Antioxid Redox Signal 2018; 29:257-274. [PMID: 28891325 PMCID: PMC6012980 DOI: 10.1089/ars.2017.7299] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE A strong relationship between hyperglycemia, impaired insulin pathway, and cardiovascular disease in type 2 diabetes (T2D) is linked to oxidative stress and inflammation. Immunometabolic pathways link these pathogenic processes and pose important potential therapeutic targets. Recent Advances: The link between immunity and metabolism is bidirectional and includes the role of inflammation in the pathogenesis of metabolic disorders such as T2D, obesity, metabolic syndrome, and hypertension and the role of metabolic factors in regulation of immune cell functions. Low-grade inflammation, oxidative stress, balance between superoxide and nitric oxide, and the infiltration of macrophages, T cells, and B cells in insulin-sensitive tissues lead to metabolic impairment and accelerated aging. CRITICAL ISSUES Inflammatory infiltrate and altered immune cell phenotype precede development of metabolic disorders. Inflammatory changes are tightly linked to alterations in metabolic status and energy expenditure and are controlled by epigenetic mechanisms. FUTURE DIRECTIONS A better comprehension of these mechanistic insights is of utmost importance to identify novel molecular targets. In this study, we describe a complex scenario of epigenetic changes and immunometabolism linking to diabetes and aging-associated vascular disease. Antioxid. Redox Signal. 29, 257-274.
Collapse
Affiliation(s)
- Tomasz J. Guzik
- BHF Centre for Research Excellence, Institute of Cardiovascular and Medical Research (ICAMS), University of Glasgow, Glasgow, United Kingdom
- Department of Internal and Agricultural Medicine, Laboratory of Translational Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Affiliation(s)
- Tina Lucas
- From the Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Germany (T.L., A.B., S.D.)
- German Center of Cardiovascular Research, Partner Site Rhein-Main, Frankfurt, Germany (T.L., A.B., S.D.)
| | - Angelika Bonauer
- From the Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Germany (T.L., A.B., S.D.)
- German Center of Cardiovascular Research, Partner Site Rhein-Main, Frankfurt, Germany (T.L., A.B., S.D.)
| | - Stefanie Dimmeler
- From the Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Germany (T.L., A.B., S.D.)
- German Center of Cardiovascular Research, Partner Site Rhein-Main, Frankfurt, Germany (T.L., A.B., S.D.)
| |
Collapse
|
33
|
Di Francesco A, Fedrigo M, Santovito D, Natarelli L, Castellani C, De Pascale F, Toscano G, Fraiese A, Feltrin G, Benazzi E, Nocco A, Thiene G, Valente M, Valle G, Schober A, Gerosa G, Angelini A. MicroRNA signatures in cardiac biopsies and detection of allograft rejection. J Heart Lung Transplant 2018; 37:1329-1340. [PMID: 30174164 DOI: 10.1016/j.healun.2018.06.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/28/2018] [Accepted: 06/20/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Identification of heart transplant (HTx) rejection currently relies on immunohistology and immunohistochemistry. We aimed to identify specific sets of microRNAs (miRNAs) to characterize acute cellular rejection (ACR), antibody-mediated rejection (pAMR), and mixed rejection (MR) in monitoring formalin-fixed paraffin-embedded (FFPE) endomyocardial biopsies (EMBs) in HTx patients. METHODS In this study we selected 33 adult HTx patients. For each, we chose the first positive EMB for study of each type of rejection. The next-generation sequencing (NGS) IonProton technique and reverse transcript quantitative polymerase chain reaction (RT-qPCR) analysis were performed on FFPE EMBs. Using logistic regression analysis we created unique miRNA signatures as predictive models of each rejection. In situ PCR was carried out on the same EMBs. RESULTS We obtained >2,257 mature miRNAs from all the EMBs. The 3 types of rejection showed a different miRNA profile for each group. The logistic regression model formed by miRNAs 208a, 126-5p, and 135a-5p identified MR; that formed by miRNAs 27b-3p, 29b-3p, and 199a-3p identified ACR; and that formed by miRNAs 208a, 29b-3p, 135a-5p, and 144-3p identified pAMR. The expression of miRNAs on tissue, through in situ PCR, showed different expressions of the same miRNA in different rejections. miRNA 126-5p was expressed in endothelial cells in ACR but in cardiomyocytes in pAMR. In ACR, miRNA 29b-3p was significantly overexpressed and detected in fibroblasts, whereas in pAMR it was underexpressed and detected only in cardiomyocytes. CONCLUSIONS miRNA profiling on FFPE EMBs differentiates the 3 types of rejection. Localization of expression of miRNAs on tissue showed different expression of the same miRNA for different cells, suggesting different roles of the same miRNA in different rejections.
Collapse
Affiliation(s)
- Andrea Di Francesco
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Marny Fedrigo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy.
| | - Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximillians University, Munich, Germany
| | - Lucia Natarelli
- Institute for Cardiovascular Prevention, Ludwig-Maximillians University, Munich, Germany
| | - Chiara Castellani
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | | | - Giuseppe Toscano
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Angela Fraiese
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Giuseppe Feltrin
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Elena Benazzi
- Organ and Tissue Transplantation Immunology, Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Angela Nocco
- Organ and Tissue Transplantation Immunology, Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Gaetano Thiene
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Marialuisa Valente
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Giorgio Valle
- CRIBI Biotechnology Centre, University of Padova, Padova, Italy
| | - Andreas Schober
- Institute for Cardiovascular Prevention, Ludwig-Maximillians University, Munich, Germany
| | - Gino Gerosa
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Annalisa Angelini
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| |
Collapse
|
34
|
Bao S, Zheng J, Li N, Huang C, Chen M, Cheng Q, Yu K, Chen S, Zhu M, Shi G. Serum MicroRNA Levels as a Noninvasive Diagnostic Biomarker for the Early Diagnosis of Hepatitis B Virus-Related Liver Fibrosis. Gut Liver 2018; 11:860-869. [PMID: 28750488 PMCID: PMC5669603 DOI: 10.5009/gnl16560] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/06/2017] [Accepted: 02/22/2017] [Indexed: 12/12/2022] Open
Abstract
Background/Aims To investigate the role of selected serum microRNA (miRNA) levels as potential noninvasive biomarkers for differentiating S0-S2 (early fibrosis) from S3-S4 (late fibrosis) in patients with a chronic hepatitis B virus (HBV) infection. Methods One hundred twenty-three treatment-naive patients with a chronic HBV infection who underwent a liver biopsy were enrolled in this study. The levels of selected miRNAs were measured using a real-time quantitative polymerase chain reaction assay. A logistic regression analysis was performed to assess factors associated with fibrosis progression. Receiver operating characteristic (ROC) curve and discriminant analyses validated these the ability of these predicted variables to discriminate S0-S2 from S3-S4. Results Serum miR-29, miR-143, miR-223, miR-21, and miR-374 levels were significantly downregulated as fibrosis progressed from S0-S2 to S3-S4 (p<0.05), but not miR-16. The multivariate logistic regression analysis identified a panel of three miRNAs and platelets that were associated with a high diagnostic accuracy in discriminating S0-S2 from S3-S4, with an area under the curve of 0.936. Conclusions The levels of the studied miRNAs, with the exception of miR-16, varied with fibrosis progression. A panel was identified that was capable of discriminating S0-S2 from S3-S4, indicating that serum miRNA levels could serve as a potential noninvasive biomarker of fibrosis progression.
Collapse
Affiliation(s)
- Suxia Bao
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianming Zheng
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ning Li
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Chong Huang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Mingquan Chen
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Cheng
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Kangkang Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Shengshen Chen
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengqi Zhu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Guangfeng Shi
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Goldberg L, Tirosh-Wagner T, Vardi A, Abbas H, Pillar N, Shomron N, Nevo-Caspi Y, Paret G. Circulating MicroRNAs: a Potential Biomarker for Cardiac Damage, Inflammatory Response, and Left Ventricular Function Recovery in Pediatric Viral Myocarditis. J Cardiovasc Transl Res 2018; 11:319-328. [PMID: 29916103 DOI: 10.1007/s12265-018-9814-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/12/2018] [Indexed: 01/02/2023]
Abstract
Viral myocarditis (VM) can be a life-threatening event resulting in cardiac failure, chronic cardiomyopathy, and death. VM typically includes three phases, i.e., acute, subacute, and resolution/chronic. We prospectively investigated cardiac- and inflammatory-associated plasma-circulating miRNA levels in eight pediatric patients with VM during the three stages of the disease. The level of cardiac-associated miR-208a was significantly elevated during the acute phase compared with the subacute and resolution/chronic phases. The level of cardiac- and inflammatory-associated miR-21 was significantly elevated during the acute phase compared to the resolution/chronic phase. Moreover, cardiac-associated miR-208b levels during the subacute phase correlated with systolic left ventricular function recovery as measured during the resolution/chronic phase. The findings of our study demonstrate an association between cardiac damage and the inflammatory response and the expression of miR-208a and miR-21 during the pathological progression of myocarditis. We also found that miR-208b levels exhibit a prognostic significance for left ventricular functional recovery.
Collapse
Affiliation(s)
- Lior Goldberg
- Department of Pediatric Critical Care Medicine, Safra Children's Hospital, Sheba Medical Center, 5262100, Tel Hashomer, Israel. .,Faculty of Medicine, Tel-Aviv University, 6997801, Tel-Aviv, Israel.
| | - Tal Tirosh-Wagner
- Department of Pediatric Critical Care Medicine, Safra Children's Hospital, Sheba Medical Center, 5262100, Tel Hashomer, Israel.,Faculty of Medicine, Tel-Aviv University, 6997801, Tel-Aviv, Israel
| | - Amir Vardi
- Department of Pediatric Critical Care Medicine, Safra Children's Hospital, Sheba Medical Center, 5262100, Tel Hashomer, Israel
| | - Haya Abbas
- Department of Pediatric Critical Care Medicine, Safra Children's Hospital, Sheba Medical Center, 5262100, Tel Hashomer, Israel
| | - Nir Pillar
- Faculty of Medicine, Tel-Aviv University, 6997801, Tel-Aviv, Israel
| | - Noam Shomron
- Faculty of Medicine, Tel-Aviv University, 6997801, Tel-Aviv, Israel
| | - Yael Nevo-Caspi
- Department of Pediatric Critical Care Medicine, Safra Children's Hospital, Sheba Medical Center, 5262100, Tel Hashomer, Israel
| | - Gideon Paret
- Department of Pediatric Critical Care Medicine, Safra Children's Hospital, Sheba Medical Center, 5262100, Tel Hashomer, Israel.,Faculty of Medicine, Tel-Aviv University, 6997801, Tel-Aviv, Israel
| |
Collapse
|
36
|
Wang W, Tang S, Li H, Liu R, Su Y, Shen L, Sun M, Ning B. MicroRNA-21a-5p promotes fibrosis in spinal fibroblasts after mechanical trauma. Exp Cell Res 2018; 370:24-30. [PMID: 29883711 DOI: 10.1016/j.yexcr.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/03/2018] [Accepted: 06/04/2018] [Indexed: 02/06/2023]
Abstract
Traumatic spinal cord injury (SCI) causes permanent disability to at least 180,000 people per year worldwide. Early regulation of spinal fibroblast proliferation may inhibit fibrotic scar formation, allowing the creation of a favorable environment for neuronal regeneration and thereby enhancing recovery from traumatic SCIs. In this study, we aimed to identify the role of microRNA-21a-5p (miR-21a-5p) in regulating spinal fibroblasts after mechanical trauma and to investigate the dysregulation of miR-21a-5p in the pathological process of spinal SCI. We investigated the differential expression of microRNAs in primary spinal fibroblasts after mechanical trauma and found that the expression of miR-21a-5p was higher in spinal fibroblasts after scratch damage (SD). In addition, mouse spinal fibroblasts were transfected with miR-21a-5p mimics/inhibitor, and the role of miR-21a-5p in spinal fibrogenic activation was analyzed. These experiments demonstrated that miR-21a-5p overexpression promoted fibrogenic activity in spinal fibroblasts after mechanical trauma, as well as enhancing proliferation and attenuating apoptosis in spinal fibroblasts. Finally, the potential role of miR-21a-5p in regulating the Smad signaling pathway was examined. MiR-21a-5p activated the Smad signaling pathway by enhancing Smad2/3 phosphorylation. These results suggest that miR-21a-5p promotes spinal fibrosis after mechanical trauma. Based on these findings, we propose a close relationship between miR-21a-5p and spinal fibrosis, providing a new potential therapeutic target for SCI.
Collapse
Affiliation(s)
- Wenzhao Wang
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Shi Tang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Hongfei Li
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Ronghan Liu
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yanlin Su
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Lin Shen
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Mingjie Sun
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Bin Ning
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
37
|
Ottaviani L, Sansonetti M, da Costa Martins PA. Myocardial cell-to-cell communication via microRNAs. Noncoding RNA Res 2018; 3:144-153. [PMID: 30175287 PMCID: PMC6114265 DOI: 10.1016/j.ncrna.2018.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/30/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Lara Ottaviani
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Marida Sansonetti
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
38
|
Keating BJ, Pereira AC, Snyder M, Piening BD. Applying genomics in heart transplantation. Transpl Int 2018; 31:278-290. [PMID: 29363220 PMCID: PMC5990370 DOI: 10.1111/tri.13119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/18/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022]
Abstract
While advances in patient care and immunosuppressive pharmacotherapies have increased the lifespan of heart allograft recipients, there are still significant comorbidities post-transplantation and 5-year survival rates are still significant, at approximately 70%. The last decade has seen massive strides in genomics and other omics fields, including transcriptomics, with many of these advances now starting to impact heart transplant clinical care. This review summarizes a number of the key advances in genomics which are relevant for heart transplant outcomes, and we highlight the translational potential that such knowledge may bring to patient care within the next decade.
Collapse
Affiliation(s)
- Brendan J. Keating
- Division of Transplantation, Department of Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Michael Snyder
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
39
|
miR-21 suppression prevents cardiac alterations induced by d-galactose and doxorubicin. J Mol Cell Cardiol 2018; 115:130-141. [PMID: 29329959 DOI: 10.1016/j.yjmcc.2018.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/01/2018] [Accepted: 01/08/2018] [Indexed: 01/02/2023]
Abstract
d-galactose (d-gal)-induced cardiac alterations and Doxorubicin (Dox)-induced cardiomyocyte senescence are commonly used models to study cardiac aging. Accumulating evidence has suggested that microRNAs (miRNAs, miRs) are critically involved in the regulation of cellular and organismal aging and age-related diseases. However, little has been revealed about the roles of miRNAs in cardiac alterations induced by d-gal and Dox. In this study, we used miRNA arrays to investigate the dysregulated miRNAs in heart samples from 15month-old versus 2month-old male C57BL/6 mice and further validated them in d-gal-induced pseudo-aging mouse model and Dox-induced cardiomyocyte senescence in vitro model. We confirmed a significant increase of miR-21 in all these models by quantitative reverse transcription polymerase chain reactions. We further demonstrated that miR-21 was able to promote Dox-induced cardiomyocyte senescence whereas suppression of miR-21 could prevent that, as determined by percentage of β-gal-positive cells and gene markers of aging. Phosphatase and tensin homolog (PTEN) was identified as a target gene of miR-21, mediating its effect in increasing cardiomyocyte senescence. Finally, we found that miR-21 knockout mice were resistant to d-gal-induced alterations in aging-markers and cardiac function. Collectively, this study provides direct evidence that inhibition of miR-21 is protective against d-gal-induced cardiac alterations and Dox-induced cardiomyocyte senescence via targeting PTEN. Inhibition of miR-21 might be a novel strategy to combat cardiac aging.
Collapse
|
40
|
Nong Q, Li S, Wu Y, Liu D. LncRNA COL1A2-AS1 inhibits the scar fibroblasts proliferation via regulating miR-21/Smad7 pathway. Biochem Biophys Res Commun 2017; 495:319-324. [PMID: 29117538 DOI: 10.1016/j.bbrc.2017.11.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/04/2017] [Indexed: 01/28/2023]
Abstract
lncRNA COL1A2-AS1 (COL1A2 antisense RNA 1), a lncRNA overexpressed in hypertrophic scar, has been demonstrated to be involved in the hypertrophic scar formation. However, the mechanisms of lncRNA COL1A2-AS1 inhibiting the scar fibroblasts proliferation remains not well understood. In this study, we demonstrated that lncRNA COL1A2-AS1 was upregulated in hypertrophic scar tissue and fibroblasts, and suppressed fibroblasts proliferation by promoting Smad7 expression. Furthermore, we found that miR-21 was involved in lncRNA COL1A2-AS1-induced expression of Smad7, by which COL1A2-AS1 acted as endogenous sponge to adsorb miR-21 and in turn regulated Smad7 and a cascade of molecular to play a protective role in hypertrophic scar. In addition, overexpression of miR-21 attenuated COL1A2-AS1-mediated proliferation suppression of hypertrophic scar fibroblasts. In conclusion, our study demonstrated that COL1A2-AS1/miR-21/Smad pathway plays an important role in inhibiting hypertrophic scar formation, and suggested this novel pathway may be a new target for hypertrophic scar treatment.
Collapse
Affiliation(s)
- Qingwen Nong
- Burns & Plastic Surgery, The First Affiliate Hospital of Guangxi Medical University, Nanning 530021, China
| | - Shuntang Li
- Burns & Plastic Surgery, The First Affiliate Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yajun Wu
- Burns & Plastic Surgery, The First Affiliate Hospital of Guangxi Medical University, Nanning 530021, China
| | - Daen Liu
- Burns & Plastic Surgery, The First Affiliate Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
41
|
Abstract
AbstractMyocardial fibrosis is observed in many cardiovascular diseases including hypertension, heart failure and cardiomyopathy. Myocardial fibrosis has been proved to be reversible and treatable only under timely intervention, which makes early detection and assessment of fibrosis crucial. Aside from tissue biopsy as the gold standard for the diagnosis of myocardial fibrosis, circulating biomarkers have been adopted as noninvasive assessment of this lesion. Dysregulated collagen deposition is thought to be the major cause of myocardial fibrosis. Collagens, procollagens, TGF-β, TIMP, galectin-3, and microRNAs are thought to be indicators of myocardial fibrosis. In this review, we summarize the molecules that are frequently used as biomarkers in diagnosis of cardiac fibrosis. Mechanisms of fibrosis that they take part in are also introduced.
Collapse
Affiliation(s)
- Zhe An
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun130033, China
| | - Guang Yang
- Department of Molecular Biology, College of Basic Medical Science, Jilin University, Changchun130061, China
| | - Haikuo Zheng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun130033, China
| | - Wei Nie
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun130033, China
| | - Guohui Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun130033, China
| |
Collapse
|
42
|
Shah P, Bristow MR, Port JD. MicroRNAs in Heart Failure, Cardiac Transplantation, and Myocardial Recovery: Biomarkers with Therapeutic Potential. Curr Heart Fail Rep 2017; 14:454-464. [DOI: 10.1007/s11897-017-0362-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
43
|
Sabia C, Picascia A, Grimaldi V, Amarelli C, Maiello C, Napoli C. The epigenetic promise to improve prognosis of heart failure and heart transplantation. Transplant Rev (Orlando) 2017; 31:249-256. [PMID: 28882368 DOI: 10.1016/j.trre.2017.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/03/2017] [Accepted: 08/07/2017] [Indexed: 12/14/2022]
Abstract
Heart transplantation is still the only possible life-saving treatment for end-stage heart failure, the critical epilogue of several cardiac diseases. Epigenetic mechanisms are being intensively investigated because they could contribute to establishing innovative diagnostic and predictive biomarkers, as well as ground-breaking therapies both for heart failure and heart transplantation rejection. DNA methylation and histone modifications can modulate the innate and adaptive immune response by acting on the expression of immune-related genes that, in turn, are crucial determinants of transplantation outcome. Epigenetic drugs acting on methylation and histone-modification pathways may modulate Treg activity by acting as immunosuppressive agents. Moreover, the identification of non-invasive and reliable epigenetic biomarkers for the prediction of allograft rejection and for monitoring immunosuppressive therapies represents an attractive perspective in the management of transplanted patients. MiRNAs seem to fit particularly well to this purpose because they are differently expressed in patients at high and low risk of rejection and are detectable in biological fluids besides biopsies. Although increasing evidence supports the involvement of epigenetic tags in heart failure and transplantation, further short and long-term clinical studies are needed to translate the possible available findings into clinical setting.
Collapse
Affiliation(s)
- Chiara Sabia
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria, Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Università degli Studi della Campania "L. Vanvitelli", Italy.
| | - Antonietta Picascia
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria, Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Università degli Studi della Campania "L. Vanvitelli", Italy
| | - Vincenzo Grimaldi
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria, Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Università degli Studi della Campania "L. Vanvitelli", Italy; Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Cristiano Amarelli
- Department of Cardiovascular Surgery and Transplants, Monaldi Hospital, Azienda dei Colli, Naples, Italy
| | - Ciro Maiello
- Department of Cardiovascular Surgery and Transplants, Monaldi Hospital, Azienda dei Colli, Naples, Italy
| | - Claudio Napoli
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria, Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Università degli Studi della Campania "L. Vanvitelli", Italy; SDN Foundation, Institute of Diagnostic and Nuclear Development, IRCCS, Via Gianturco 113, 80143 Naples, Italy
| |
Collapse
|
44
|
Shen Y, Xie X, Li Z, Huang Y, Ma L, Shen X, Liu Y, Zhao Y. Interleukin-17-induced expression of monocyte chemoattractant protein-1 in cardiac myocytes requires nuclear factor κB through the phosphorylation of p65. Microbiol Immunol 2017; 61:280-286. [PMID: 28593659 DOI: 10.1111/1348-0421.12495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/26/2017] [Accepted: 05/26/2017] [Indexed: 01/24/2023]
Affiliation(s)
- Yan Shen
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Xin Xie
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Zhuolun Li
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Yan Huang
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Li Ma
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Xinhe Shen
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Yanyue Liu
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| | - Yuxia Zhao
- Department of Clinical Laboratory; The First Affiliated Hospital of Zhengzhou University; No. 1 JianShe Road Zhengzhou 450052 China
| |
Collapse
|
45
|
Li X, Chen H, Wang S, Dai J, Yan L, Wang J, Sun Y. Tacrolimus induces fibroblasts apoptosis and reduces epidural fibrosis by regulating miR-429 and its target of RhoE. Biochem Biophys Res Commun 2017; 490:1197-1204. [PMID: 28669722 DOI: 10.1016/j.bbrc.2017.06.181] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
Tacrolimus (FK506) has been demonstrated to reduce epidural fibrosis. However, the detailed mechanism of action has not been elucidated. Aberrant miR-429 is involved in many diseases. The aim of this study was to describe the exact mechanism of FK506 induced apoptosis in fibroblasts and the prevention of epidural fibrosis. FK506 induced fibroblast apoptosis was evaluated using CCK-8 assays, flow cytometry, and western blotting. The expression of miR-429 in fibroblasts treated with FK506 was determined by RT-qPCR. Additionally, luciferase activity assays were used to determine the target relationship between miR-429 and RhoE. Flow cytometry and western blot analysis were used to determine the effects of FK506 and miR-429 on fibroblast apoptosis. The effects of FK506 and RhoE on fibroblast apoptosis were determined by CCK-8 assay, flow cytometry, and western blotting. We also evaluate the effects of FK506 and miR-429 on epidural fibrosis in rats by using histological analysis and TUNEL-staining. The results revealed FK506 induces fibroblast apoptosis and significantly downregulates miR-429 expression in fibroblasts. Additionally, miR-429 downregulation caused the apoptosis of fibroblasts. The luciferase activity assay confirmed that RhoE is a direct target of miR-429 and RhoE promotes fibroblast apoptosis. The rat model demonstrated miR-429 inhibition promotes fibroblast apoptosis and epidural fibrosis, which is consistent with the results of FK506 treatment. Our study demonstrates that FK506 induces fibroblast apoptosis and reduces epidural fibrosis by regulating miR-429 expression and its target of RhoE.
Collapse
Affiliation(s)
- Xiaolei Li
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Hui Chen
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Shuguang Wang
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Lianqi Yan
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China.
| | - Yu Sun
- Department of Orthopedics and Orthopedic Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China.
| |
Collapse
|
46
|
Zhou R, Wang C, Wen C, Wang D. miR-21 promotes collagen production in keloid via Smad7. Burns 2017; 43:555-561. [DOI: 10.1016/j.burns.2016.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/11/2016] [Accepted: 09/13/2016] [Indexed: 02/06/2023]
|
47
|
Dueñas A, Aranega AE, Franco D. More than Just a Simple Cardiac Envelope; Cellular Contributions of the Epicardium. Front Cell Dev Biol 2017; 5:44. [PMID: 28507986 PMCID: PMC5410615 DOI: 10.3389/fcell.2017.00044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022] Open
Abstract
The adult pumping heart is formed by distinct tissue layers. From inside to outside, the heart is composed by an internal endothelial layer, dubbed the endocardium, a thick myocardial component which supports the pumping capacity of the heart and exteriorly covered by a thin mesothelial layer named the epicardium. Cardiac insults such as coronary artery obstruction lead to ischemia and thus to an irreversible damage of the myocardial layer, provoking in many cases heart failure and death. Thus, searching for new pathways to regenerate the myocardium is an urgent biomedical need. Interestingly, the capacity of heart regeneration is present in other species, ranging from fishes to neonatal mammals. In this context, several lines of evidences demonstrated a key regulatory role for the epicardial layer. In this manuscript, we provide a state-of-the-art review on the developmental process leading to the formation of the epicardium, the distinct pathways controlling epicardial precursor cell specification and determination and current evidences on the regenerative potential of the epicardium to heal the injured heart.
Collapse
Affiliation(s)
- Angel Dueñas
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| | - Amelia E Aranega
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| | - Diego Franco
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| |
Collapse
|
48
|
Bartman CM, Oyama Y, Brodsky K, Khailova L, Walker L, Koeppen M, Eckle T. Intense light-elicited upregulation of miR-21 facilitates glycolysis and cardioprotection through Per2-dependent mechanisms. PLoS One 2017; 12:e0176243. [PMID: 28448534 PMCID: PMC5407766 DOI: 10.1371/journal.pone.0176243] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
A wide search for ischemic preconditioning (IPC) mechanisms of cardioprotection identified the light elicited circadian rhythm protein Period 2 (Per2) to be cardioprotective. Studies on cardiac metabolism found a key role for light elicited Per2 in mediating metabolic dependence on carbohydrate metabolism. To profile Per2 mediated pathways following IPC of the mouse heart, we performed a genome array and identified 352 abundantly expressed and well-characterized Per2 dependent micro RNAs. One prominent result of our in silico analysis for cardiac Per2 dependent micro RNAs revealed a selective role for miR-21 in the regulation of hypoxia and metabolic pathways. Based on this Per2 dependency, we subsequently found a diurnal expression pattern for miR-21 with higher miR-21 expression levels at Zeitgeber time (ZT) 15 compared to ZT3. Gain or loss of function studies for miR-21 using miRNA mimics or miRNA inhibitors and a Seahorse Bioanalyzer uncovered a critical role of miR-21 for cellular glycolysis, glycolytic capacity, and glycolytic reserve. Exposing mice to intense light, a strategy to induce Per2, led to a robust induction of cardiac miR-21 tissue levels and decreased infarct sizes, which was abolished in miR-21-/- mice. Similarly, first translational studies in humans using intense blue light exposure for 5 days in healthy volunteers resulted in increased plasma miR-21 levels which was associated with increased phosphofructokinase activity, the rate-limiting enzyme in glycolysis. Together, we identified miR-21 as cardioprotective downstream target of Per2 and suggest intense light therapy as a potential strategy to enhance miR-21 activity and subsequent carbohydrate metabolism in humans.
Collapse
Affiliation(s)
- Colleen Marie Bartman
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
- Department of Cell and Developmental Biology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
| | - Yoshimasa Oyama
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
- Department of Anesthesiology and Intensive Care Medicine, Oita University Faculty of Medicine, Oita, Japan
| | - Kelley Brodsky
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
- Division of Cardiology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
| | - Ludmila Khailova
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
| | - Lori Walker
- Division of Cardiology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
| | - Michael Koeppen
- Department of Anesthesiology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
- Department of Cell and Developmental Biology, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
- Division of Cardiology, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
49
|
Gupta SK, Thum T. MiR-142-3p is a paracrine mediator between T cells and endothelium during allograft rejection. Cardiovasc Res 2017; 113:431-433. [PMID: 28339725 DOI: 10.1093/cvr/cvx031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Affiliation(s)
- Shashi K Gupta
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuber Str.1, 30625 Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuber Str.1, 30625 Hannover, Germany.,Excellence Cluster REBIRTH, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.,National Heart and Lung Institute, Imperial College, Sydney St, Chelsea, London SW3 6NP, UK
| |
Collapse
|
50
|
MiR-21 is required for anti-tumor immune response in mice: an implication for its bi-directional roles. Oncogene 2017; 36:4212-4223. [PMID: 28346427 DOI: 10.1038/onc.2017.62] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/29/2016] [Accepted: 02/08/2017] [Indexed: 12/17/2022]
Abstract
Here we show that miR-21, a microRNA known for its oncogenic activity, is also essential for mediating immune responses against tumor. Knockout of miR-21 in mice slowed the proliferation of both CD4+ and CD8+ cells, reduced their cytokine production and accelerated the grafted tumor growth. Further investigations indicated that miR-21 could activate CD4+ and CD8+ T cells via the PTEN/Akt pathway in response to stimulations. Taken together, these data suggest the key functions of miR-21 in mediating anti-tumor immune response and thereby uncover a bi-directional role of this traditionally known 'oncomiR' in tumorigenesis. Our study may provide new insights for the design of cancer therapies targeting microRNAs, with an emphasis on the dynamic and possibly unexpected role of these molecules.
Collapse
|