1
|
Shityakov S, Nagai M, Ergün S, Braunger BM, Förster CY. The Protective Effects of Neurotrophins and MicroRNA in Diabetic Retinopathy, Nephropathy and Heart Failure via Regulating Endothelial Function. Biomolecules 2022; 12:biom12081113. [PMID: 36009007 PMCID: PMC9405668 DOI: 10.3390/biom12081113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a common disease affecting more than 537 million adults worldwide. The microvascular complications that occur during the course of the disease are widespread and affect a variety of organ systems in the body. Diabetic retinopathy is one of the most common long-term complications, which include, amongst others, endothelial dysfunction, and thus, alterations in the blood-retinal barrier (BRB). This particularly restrictive physiological barrier is important for maintaining the neuroretina as a privileged site in the body by controlling the inflow and outflow of fluid, nutrients, metabolic end products, ions, and proteins. In addition, people with diabetic retinopathy (DR) have been shown to be at increased risk for systemic vascular complications, including subclinical and clinical stroke, coronary heart disease, heart failure, and nephropathy. DR is, therefore, considered an independent predictor of heart failure. In the present review, the effects of diabetes on the retina, heart, and kidneys are described. In addition, a putative common microRNA signature in diabetic retinopathy, nephropathy, and heart failure is discussed, which may be used in the future as a biomarker to better monitor disease progression. Finally, the use of miRNA, targeted neurotrophin delivery, and nanoparticles as novel therapeutic strategies is highlighted.
Collapse
Affiliation(s)
- Sergey Shityakov
- Division of Chemoinformatics, Infochemistry Scientific Center, Lomonosova Street 9, 191002 Saint-Petersburg, Russia
| | - Michiaki Nagai
- Department of Cardiology, Hiroshima City Asa Hospital, 2-1-1 Kabeminami, Aaskita-ku, Hiroshima 731-0293, Japan
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
| | - Barbara M. Braunger
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
- Correspondence: (B.M.B.); (C.Y.F.)
| | - Carola Y. Förster
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Würzburg University, 97080 Würzburg, Germany
- Correspondence: (B.M.B.); (C.Y.F.)
| |
Collapse
|
2
|
Lin KH, Ng SC, Paul CR, Chen HC, Zeng RY, Liu JS, Padma VV, Huang CY, Kuo WW. MicroRNA-210 repression facilitates advanced glycation end-product (AGE)-induced cardiac mitochondrial dysfunction and apoptosis via JNK activation. J Cell Biochem 2021; 122:1873-1885. [PMID: 34545968 DOI: 10.1002/jcb.30146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022]
Abstract
Hyperglycemia results in the formation of reactive oxygen species which in turn causes advanced glycation end products (AGEs) formation, leading to diabetic cardiomyopathy. Our previous study showed that AGE-induced reactive oxygen species-dependent apoptosis is mediated via protein kinase C delta (PKCδ)-enhanced mitochondrial damage in cardiomyocytes. By using microRNA (miRNA) database, miRNA-210 was predicted to target c-Jun N-terminal kinase (JNK), which were previously identified as downstream of PKCδ in regulating mitochondrial function. Therefore, we hypothesized that miR-210 mediates PKCδ-dependent upregulation of JNK to cause cardiac mitochondrial damage and apoptosis following AGE exposure. AGE-exposed cells showed activated cardiac JNK, PKCδ, and apoptosis, which were reversed by treatment with a JNK inhibitor and PKCδ-KD (deficient kinase). Cardiac miR-210 and mitochondrial function were downregulated following AGE exposure. Furthermore, JNK was upregulated and involved in AGE-induced mitochondrial damage. Interestingly, luciferase activity of the miR-210 mimic plus JNK WT-3'-untranslated region overexpressed group was significantly lower than that of miR-210 mimic plus JNK MT-3'UTR group, indicating that JNK is a target of miR-210. Moreover, JNK activation induced by AGEs was reduced by treatment with the miR-210 mimic and reversed by treatment with the miR-210 inhibitor, indicating the regulatory function of miR-210 in JNK activation following AGE exposure. Additionally, JNK-dependent mitochondrial dysfunction and apoptosis were reversed following treatment with the miR-210 mimic, while the miR-210 inhibitor showed no effect on JNK-induced mitochondrial dysfunction and apoptosis in AGE-exposed cardiac cells. Taken together, our study showed that PKCδ-enhanced JNK-dependent mitochondrial damage is mediated through the reduction of miR-210 in cardiomyocytes following AGE exposure.
Collapse
Affiliation(s)
- Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung, Taiwan, ROC.,Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Shang-Chuan Ng
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan, ROC
| | - Catherine R Paul
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan, ROC
| | - Hong-Chen Chen
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, ROC.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ren-You Zeng
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan, ROC
| | - Jian-Sheng Liu
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,Department of Thoracic, China Medical University Beigang Hospital, Yunlin, Taiwan, ROC
| | - Viswanadha V Padma
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Chih-Yang Huang
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan, ROC.,Department of Biotechnology, Translational Research Laboratory, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu, India.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC.,Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan, ROC.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
3
|
Zhou R, Wang L, Zhao G, Chen D, Song X, Momtazi-Borojeni AA, Yuan H. Circulating exosomal microRNAs as emerging non-invasive clinical biomarkers in heart failure: Mega bio-roles of a nano bio-particle. IUBMB Life 2020; 72:2546-2562. [PMID: 33053610 DOI: 10.1002/iub.2396] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/22/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022]
Abstract
Exosomes are nano-sized extracellular vesicles containing a cell-specific biologically active cargo of proteins and genetic materials. Exosomes are constitutively released from almost all cell-types and affect neighboring or distant cells through a complex intercellular exchange of the genetic information and/or regulation of certain gene expressions that change the function and behavior of recipient cells. Those released into body fluids are the major mediators of intercellular communications. The success of the biological functions of exosomes is highly mediated by the effective transfer of microRNAs (miRs). Exosomes secreted by a damaged or diseased heart can exhibit alterations in the miRs' profile that may reflect the cellular origin and (patho)physiological state, as a "signature" or "fingerprint" of the donor cell. It has been shown that the transportation of cardiac-specific miRs in exosomes can be rapidly detected and measured, holding great potential as biomarkers in heart diseases. Currently, the search for new biomarkers of heart diseases remains a large and increasing enterprise. Notably, circulating exosomal miRs (Exo-miRs) have successfully gained huge interests for their diagnostic and prognostic potentials. The present review highlights circulating Exo-miRs explored for diagnosis/prognosis and outcome prediction in patients with heart failure (HF). To this end, we explain the feasibility of exosomes as clinical biomarkers, discuss the priority of circulating Exo-miRs over non-exosomal ones as a biomarker, and then outline reported circulating Exo-miRs having the biomarker function in HF patients, together with their mechanism of action. In conclusion, circulating Exo-miRs represent emerging diagnostic (Exo-miR-92b-5p, Exo-miR-146a, Exo-miR-181c, and Exo-miR-495) and prognostic (Exo-miR-192, Exo-miR-194, Exo-miR-34a, Exo-miR-425, Exo-miR-744) biomarkers for HF.
Collapse
Affiliation(s)
- Runfa Zhou
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Leiyan Wang
- Clinical Skill Training Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Gang Zhao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Dan Chen
- Department of Cardiology Electrocardiogram Room, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoning Song
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Amir A Momtazi-Borojeni
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
4
|
MicroRNA-21 prevents excessive inflammation and cardiac dysfunction after myocardial infarction through targeting KBTBD7. Cell Death Dis 2018; 9:769. [PMID: 29991775 PMCID: PMC6039462 DOI: 10.1038/s41419-018-0805-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 11/18/2022]
Abstract
The excessive inflammation triggered by damage-associated molecular patterns (DAMPs) after myocardial infarction (MI) is responsible for the development of cardiac dysfunction and adverse remodeling, while the mechanisms by which inflammation is fine tuned remain to be fully elucidated. MicroRNA-21 (miR-21) has been shown to function in cardiovascular diseases, while its role in inflammatory responses and cardiac function post MI in mice remains unknown. Here, we found that miR-21 expression was markedly increased in border and infarct areas of cardiac tissues during the early inflammatory phase of MI model established by ligating the left-anterior descending coronary artery. MiR-21 knockout mice had decreased survival rates, worse cardiac dysfunction, and increased infarct and scar areas after MI compared with WT mice. MiR-21 knockout mice showed significantly higher levels of inflammatory cytokines including IL-1β, IL-6, and TNF-α in cardiac tissues, as well as infiltration of CD11b+ monocytes/macrophages with higher expression level of inflammatory cytokines. MI induced the great release of high mobility group protein B1 (HMGB1) and heat shock protein 60 (HSP60) in cardiac tissue. MiR-21 deficiency significantly promoted the inflammatory cytokine production triggered by DAMPs in macrophages, whereas, miR-21 overexpression markedly inhibited the inflammatory cytokine production. Mechanistically, miR-21 deficiency enhanced p38 and NF-κB signaling activation in cardiac tissue post MI and macrophages treated with DAMPs. MiR-21 was found to directly target kelch repeat and BTB (POZ) domain containing 7 (KBTBD7), which promoted DAMP-triggered inflammatory responses in macrophages. Furthermore, KBTBD7 interacted with MKK3/6 and promoted their activation, which in turn enhanced the activation of downstream p38 and NF-κB signaling induced by DAMPs. Therefore, our findings demonstrate that miR-21 attenuates inflammation, cardiac dysfunction, and maladaptive remodeling post MI through targeting KBTBD7 and inhibiting p38 and NF-κB signaling activation, suggesting that miR-21 may function as a novel potential therapeutic target for MI.
Collapse
|
5
|
Abstract
Epidemiological and experimental observations tend to prove that environment, lifestyle or nutritional challenges influence heart functions together with genetic factors. Furthermore, when occurring during sensitive windows of heart development, these environmental challenges can induce an 'altered programming' of heart development and shape the future heart disease risk. In the etiology of heart diseases driven by environmental challenges, epigenetics has been highlighted as an underlying mechanism, constituting a bridge between environment and heart health. In particular, micro-RNAs which are involved in each step of heart development and functions seem to play a crucial role in the unfavorable programming of heart diseases. This review describes the latest advances in micro-RNA research in heart diseases driven by early exposure to challenges and discusses the use of micro-RNAs as potential targets in the reversal of the pathophysiology.
Collapse
|
6
|
Abstract
microRNAs (miRs) are targets for genomic aberrations and emerging treatments against cancer. It has been demonstrated that targeting miR-569 may potentially benefit patients with ovarian or breast cancer. However, the exact roles of miR-569 remain unclear in human lung cancer cells. Using the reverse transcription-quantitative polymerase chain reaction (RT-qPCR), it was demonstrated that miR-569 expression was consistently decreased in lung cancer cells. As well as cell proliferation and migration inhibition, apoptosis and cell arrest at the G1 phase were induced following reversion of miR-569 expression in lung cancer cells. The present study demonstrated that miR-569 was able to downregulate FOS and high mobility group A2 mRNA and protein expression using RT-qPCR and western blot analysis. The observed role of miRNA-569 in lung cancer cells in the present study suggested that it may be a novel and promising therapeutic target, and a novel biomarker for detecting lung cancer.
Collapse
Affiliation(s)
- Yi Ping Zheng
- Department of Geriatrics, The First Affiliated Hospital of Dalian Medical College, Dalian, Liaoning 116000, P.R. China
| | - Linxia Wu
- Department of Geriatrics, The First Affiliated Hospital of Dalian Medical College, Dalian, Liaoning 116000, P.R. China
| | - Jie Gao
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical College, Dalian, Liaoning 116000, P.R. China
| | - Yanfu Wang
- Department of Geriatrics, The First Affiliated Hospital of Dalian Medical College, Dalian, Liaoning 116000, P.R. China
| |
Collapse
|
7
|
Qipshidze Kelm N, Piell KM, Wang E, Cole MP. MicroRNAs as predictive biomarkers for myocardial injury in aged mice following myocardial infarction. J Cell Physiol 2018; 233:5214-5221. [PMID: 29150941 DOI: 10.1002/jcp.26283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 11/13/2017] [Indexed: 12/23/2022]
Abstract
The occurrence of myocardial infarction (MI) increases appreciably with age. In the Framingham Heart Study, the incidence of MI more than doubles for men and increases more than five-fold in women (ages 55-64 years compared to 85-94 years). MicroRNAs (miRNAs) quantitatively regulate their target's expression post-transcriptionally by either silencing action through binding at the 3'UTR domains or degrading the messages at their coding regions. In either case, these regulations affect the cardiac transcriptional output and cardiac function. Among the known cardiac associated miRNA, miRNA-1, miRNA-133a, and miRNA-34a have been shown to induce adverse structural remodeling to impair cardiac contractile function. In the present study, an in vivo model of MI in young (3 month) and old (22 month) mice is used to investigate the possible role whereby these three miRNAs exert negative effects on heart function following MI. Herein we demonstrate that in older mouse heart, all three microRNAs show increased levels of expression, while miRNA-1 shows a further increase in old mouse heart following MI, which corresponds to left ventricular (LV) wall thinning. These structural changes in cardiac tissue may causes downstream LV dilation and subsequent LV dysfunction. Results presented here suggest that significantly elevated levels of miRNA-1 in post-MI old heart could be predictive of cardiac injury in older mice as the high risk biomarker for MI in older individuals.
Collapse
Affiliation(s)
- Natia Qipshidze Kelm
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, Kentucky.,Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Kellianne M Piell
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, Kentucky.,Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Eugenia Wang
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, Kentucky.,Gheens Center on Aging, University of Louisville School of Medicine, Louisville, Kentucky
| | - Marsha P Cole
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, Kentucky.,Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky.,Gheens Center on Aging, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
8
|
Lu Y, Wu F. A new miRNA regulator, miR-672, reduces cardiac hypertrophy by inhibiting JUN expression. Gene 2018; 648:21-30. [PMID: 29339068 DOI: 10.1016/j.gene.2018.01.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 12/17/2017] [Accepted: 01/11/2018] [Indexed: 01/04/2023]
Abstract
Cardiac hypertrophy is one of the initial symptoms of many heart diseases. We found that miR-672-5p may participate in the regulation of heart disease development in mouse, but the association between miR-672-5p and cardiac hypertrophy remains unclear. In the present study, we found that the abundance of miR-672-5p decreased in hypertrophic cardiomyocytes induced by phenylephrine, angiotensin II (Ang II) and insulin-like growth factor 1. Putative target genes of miR-672-5p were identified using four pipelines, miRWalk, miRanda, RNA22 and Targetscan, and a total of 834 genes were predicted by all four pipelines. Among these target genes, 98 were associated with the development of heart disease. PPI networks showed that the Jun proto-oncogene product (JUN), a subunit of the AP-1 transcription factor, had the highest node degree, and it was defined as the hub gene of the PPI networks. Luciferase assays showed that miR-672-5p bound to the 3' UTR of the JUN gene and decreased luciferase activity, indicating that JUN is a target of miR-672-5p. Finally, we found that increasing the abundance of miR-672-5p in cardiomyocytes controlled the relative cell area in Ang II-stimulated hypertrophic cardiomyocytes. Correspondingly, the abundance of JUN, a target of miR-672-5p, was decreased in hypertrophic cardiomyocytes on both mRNA and protein levels, implying that miR-672-5p had suppressive effects on cardiac hypertrophy through regulating the expression of Jun in cardiomyocytes.
Collapse
Affiliation(s)
- Yili Lu
- Department of Pediatrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Fangli Wu
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
9
|
Beg F, Wang R, Saeed Z, Devaraj S, Masoor K, Nakshatri H. Inflammation-associated microRNA changes in circulating exosomes of heart failure patients. BMC Res Notes 2017; 10:751. [PMID: 29258606 PMCID: PMC5735935 DOI: 10.1186/s13104-017-3090-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 12/13/2017] [Indexed: 12/21/2022] Open
Abstract
Objective MiR-486 and miR-146a are cardiomyocyte-enriched microRNAs that control cell survival and self-regulation of inflammation. These microRNAs are released into circulation and are detected in plasma or in circulating exosomes. Little is known whether heart failure affects their release into circulation, which this study investigated. Results Total and exosome-specific microRNAs in plasma of 40 heart failure patients and 20 controls were prepared using the miRVana Kit. We measured exosomal and total plasma microRNAs separately because exosomes serve as cargos that transfer biological materials and alter signaling in distant organs, whereas microRNAs in plasma indicate the level of tissue damage and are mostly derived from dead cells. qRT-PCR was used to quantify miR-486, miR-146a, and miR-16. Heart failure did not significantly affect plasma miR-486/miR-16 and miR-146a/miR-16 ratio, although miR-146a/miR-16 showed a trend of elevated expression (2.3 ± 0.79, p = 0.27). By contrast, circulating exosomal miR-146a/miR-16 ratio was higher in heart failure patients (2.46 ± 0.51, p = 0.05). miR-146a is induced in response to inflammation as a part of inflammation attenuation circuitry. Indeed, Tnfα and Gm-csf increased miR-146a but not miR-486 in the cardiomyocyte cell line H9C2. These results, if confirmed in a larger study, may help to develop circulating exosomal miR-146a as a biomarker of heart failure.
Collapse
Affiliation(s)
- Faheemullah Beg
- Department of Internal Medicine, IU School of Medicine, Indianapolis, IN, USA
| | - Ruizhong Wang
- Department of Surgery, IU School of Medicine, C218C, 980 West Walnut St., Indianapolis, IN, 46202, USA
| | - Zeb Saeed
- Department of Internal Medicine, IU School of Medicine, Indianapolis, IN, USA
| | - Srikant Devaraj
- Center for Business and Economic Research, Ball State University, Muncie, IN, USA
| | - Kamalesh Masoor
- Department of Cardiology, Richard L Roudebush VAMC, Indianapolis, IN, USA
| | - Harikrishna Nakshatri
- Department of Surgery, IU School of Medicine, C218C, 980 West Walnut St., Indianapolis, IN, 46202, USA. .,Department of Biochemistry and Molecular Biology, IU School of Medicine, C218C, 980 West Walnut St., Indianapolis, IN, 46202, USA. .,Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
10
|
Quast C, Alter C, Ding Z, Borg N, Schrader J. Adenosine Formed by CD73 on T Cells Inhibits Cardiac Inflammation and Fibrosis and Preserves Contractile Function in Transverse Aortic Constriction-Induced Heart Failure. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003346. [PMID: 28404626 DOI: 10.1161/circheartfailure.116.003346] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Structural damage during heart failure development leads to increased infiltration of leukocytes. Because purinergic signaling on immune cells may impact on the inflammatory response, we evaluated the role of ecto-5'-nucleotidase (CD73) on the development of heart failure after transverse aortic constriction (TAC) using global and T-cell-specific CD73-/- mice. METHODS AND RESULTS Leukocytes infiltrating the failing heart were analyzed by a multistep enzymatic procedure over a period of 16 weeks using fluorescence-activated cell sorting. TAC significantly enhanced the infiltration of leukocytes, especially T cells. The fraction of CD73 expressing cells increased over time exclusively on cytotoxic T cells, T-helper cells, and regulatory T cells. Cardiac function significantly declined in T-cell-specific CD4-Cre+/-CD73flox/flox mice identical to that observed in global CD73 mutants and was associated with enhanced fibrosis (collagen, laminin, vimentin, periostin). Expression analysis by quantitative reverse transcription polymerase chain reaction of extracellular purine degrading enzymes and P1 and P2 receptors on T cells isolated from the injured heart revealed profound upregulation of the enzymatic machinery for hydrolysis of extracellular adenosine triphosphate and nicotinamide adenine dinucleotide, both pathways converging in the formation of AMP and adenosine via CD73. Among the P1 receptors, only the A2a receptor was significantly upregulated after TAC. T cells isolated from TAC-treated hearts show enhanced production of proinflammatory cytokines (interleukin-3, interleukin-6, interleukin-13, interleukin-17, macrophage inflammatory proteins-1α, and macrophage inflammatory proteins-1β) when CD73 was lacking. CONCLUSIONS Our data provide first evidence that CD73 on T cells plays an important anti-inflammatory role in TAC-induced heart failure, which is associated with antifibrotic activity and reduced production of proinflammatory cytokines most likely by activation of the adenosine A2a receptor.
Collapse
Affiliation(s)
- Christine Quast
- From the Division of Cardiology, Pulmonary Diseases and Vascular Medicine (C.Q.) and Department of Molecular Cardiology (C.Q., C.A., Z.D., N.B., J.S.), University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | - Christina Alter
- From the Division of Cardiology, Pulmonary Diseases and Vascular Medicine (C.Q.) and Department of Molecular Cardiology (C.Q., C.A., Z.D., N.B., J.S.), University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | - Zhaoping Ding
- From the Division of Cardiology, Pulmonary Diseases and Vascular Medicine (C.Q.) and Department of Molecular Cardiology (C.Q., C.A., Z.D., N.B., J.S.), University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | - Nadine Borg
- From the Division of Cardiology, Pulmonary Diseases and Vascular Medicine (C.Q.) and Department of Molecular Cardiology (C.Q., C.A., Z.D., N.B., J.S.), University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | - Jürgen Schrader
- From the Division of Cardiology, Pulmonary Diseases and Vascular Medicine (C.Q.) and Department of Molecular Cardiology (C.Q., C.A., Z.D., N.B., J.S.), University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany.
| |
Collapse
|
11
|
Wang D, Zhai G, Ji Y, Jing H. microRNA-10a Targets T-box 5 to Inhibit the Development of Cardiac Hypertrophy. Int Heart J 2017; 58:100-106. [PMID: 28100873 DOI: 10.1536/ihj.16-020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The mechanism of cardiac hypertrophy involving microRNAs (miRNAs) is attracting increasing attention. Our study aimed to investigate the role of miR-10a in cardiac hypertrophy development and the underlying regulatory mechanism.Transverse abdominal aortic constriction (TAAC) surgery was performed to establish a cardiac hypertrophy rat model, and angiotensin II (AngII) was used to induce cardiac hypertrophy in cultured neonatal rat cardiomyocytes. Expression of T-box 5 (TBX5) and miR-10a was altered by cell transfection of siRNA or miRNA mimic/inhibitor. Leucine incorporation assay, histological and cytological examination, quantitative real-time PCR (qRT-PCR), and Western blot were performed to detect the effects of miR-10a and TBX5 on cardiac hypertrophy. Dual-luciferase reporter assay was conducted to verify the regulation of TBX5 by miR-10a.miR-10a was down-regulated, and TBX5 was up-regulated in the rat model and AngII-stimulated cardiomyocytes. miR-10a inhibited TBX5 expression by directly targeting the binding site in Tbx5 3'UTR. Overexpression of miR-10a in AngII-treated cardiomyocytes decreased relative cell area, and significantly reduced the mRNA levels of natriuretic peptide A (Nppa), myosin heavy chain 7 cardiac muscle beta (Myh7), and leucine incorporation (P < 0.01 or P < 0.001). Knockdown of Tbx5 had similar effects on AngII-induced cardiomyocytes.Our findings indicate that miR-10a may inhibit cardiac hypertrophy via targeting Tbx5. Thus, miR-10a provides promising therapeutic strategies for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dan Wang
- Fifth Department of Cardiology, Zhengzhou Central Hospital
| | | | | | | |
Collapse
|
12
|
Mishra PK, Ying W, Nandi SS, Bandyopadhyay GK, Patel KK, Mahata SK. Diabetic Cardiomyopathy: An Immunometabolic Perspective. Front Endocrinol (Lausanne) 2017; 8:72. [PMID: 28439258 PMCID: PMC5384479 DOI: 10.3389/fendo.2017.00072] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022] Open
Abstract
The heart possesses a remarkable inherent capability to adapt itself to a wide array of genetic and extrinsic factors to maintain contractile function. Failure to sustain its compensatory responses results in cardiac dysfunction, leading to cardiomyopathy. Diabetic cardiomyopathy (DCM) is characterized by left ventricular hypertrophy and reduced diastolic function, with or without concurrent systolic dysfunction in the absence of hypertension and coronary artery disease. Changes in substrate metabolism, oxidative stress, endoplasmic reticulum stress, formation of extracellular matrix proteins, and advanced glycation end products constitute the early stage in DCM. These early events are followed by steatosis (accumulation of lipid droplets) in cardiomyocytes, which is followed by apoptosis, changes in immune responses with a consequent increase in fibrosis, remodeling of cardiomyocytes, and the resultant decrease in cardiac function. The heart is an omnivore, metabolically flexible, and consumes the highest amount of ATP in the body. Altered myocardial substrate and energy metabolism initiate the development of DCM. Diabetic hearts shift away from the utilization of glucose, rely almost completely on fatty acids (FAs) as the energy source, and become metabolically inflexible. Oxidation of FAs is metabolically inefficient as it consumes more energy. In addition to metabolic inflexibility and energy inefficiency, the diabetic heart suffers from impaired calcium handling with consequent alteration of relaxation-contraction dynamics leading to diastolic and systolic dysfunction. Sarcoplasmic reticulum (SR) plays a key role in excitation-contraction coupling as Ca2+ is transported into the SR by the SERCA2a (sarcoplasmic/endoplasmic reticulum calcium-ATPase 2a) during cardiac relaxation. Diabetic cardiomyocytes display decreased SERCA2a activity and leaky Ca2+ release channel resulting in reduced SR calcium load. The diabetic heart also suffers from marked downregulation of novel cardioprotective microRNAs (miRNAs) discovered recently. Since immune responses and substrate energy metabolism are critically altered in diabetes, the present review will focus on immunometabolism and miRNAs.
Collapse
Affiliation(s)
- Paras K. Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, USA
- *Correspondence: Paras K. Mishra, ; Sushil K. Mahata,
| | - Wei Ying
- Department of Medicine, Metabolic Physiology and Ultrastructural Biology Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Shyam Sundar Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gautam K. Bandyopadhyay
- Department of Medicine, Metabolic Physiology and Ultrastructural Biology Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Kaushik K. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushil K. Mahata
- Department of Medicine, Metabolic Physiology and Ultrastructural Biology Laboratory, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, Metabolic Physiology and Ultrastructural Biology Laboratory, VA San Diego Healthcare System, San Diego, CA, USA
- *Correspondence: Paras K. Mishra, ; Sushil K. Mahata,
| |
Collapse
|
13
|
Morishima M, Iwata E, Nakada C, Tsukamoto Y, Takanari H, Miyamoto S, Moriyama M, Ono K. Atrial Fibrillation-Mediated Upregulation of miR-30d Regulates Myocardial Electrical Remodeling of the G-Protein-Gated K(+) Channel, IK.ACh. Circ J 2016; 80:1346-55. [PMID: 27180889 DOI: 10.1253/circj.cj-15-1276] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) begets AF in part due to atrial remodeling, the molecular mechanisms of which have not been completely elucidated. This study was conducted to identify microRNA(s) responsible for electrical remodeling in AF. METHODS AND RESULTS The expression profiles of 1205 microRNAs, in cardiomyocytes from patients with persistent AF and from age-, gender-, and cardiac function-matched control patients with normal sinus rhythm, were examined by use of a microRNA microarray platform. Thirty-nine microRNAs differentially expressed in AF patients' atria were identified, including miR-30d, as a candidate responsible for ion channel remodeling by in silico analysis. MiR-30d was significantly upregulated in cardiomyocytes from AF patients, whereas the mRNA and protein levels ofCACNA1C/Cav1.2 andKCNJ3/Kir3.1, postulated targets of miR-30d, were markedly reduced.KCNJ3/Kir3.1 expression was downregulated by transfection of the miR-30 precursor, concomitant with a reduction of the acetylcholine-sensitive inward-rectifier K(+)current (IK.ACh).KCNJ3/Kir3.1 (but notCACNA1C/Cav1.2) expression was enhanced by the knockdown of miR-30d. The Ca(2+)ionophore, A23187, induced a dose-dependent upregulation of miR-30d, followed by the suppression ofKCNJ3mRNA expression. Blockade of protein kinase C signaling blunted the [Ca(2+)]i-dependent downregulation of Kir3.1 via miR-30d. CONCLUSIONS The downward remodeling ofIK.AChis attributed, at least in part, to deranged Ca(2+)handling, leading to the upregulation of miR-30d in human AF, revealing a novel post-transcriptional regulation ofIK.ACh. (Circ J 2016; 80: 1346-1355).
Collapse
Affiliation(s)
- Masaki Morishima
- Department of Pathophysiology, Oita University School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Cardiac hypertrophy is a primary pathological change associated with cardiovascular diseases. Dysregulated microRNAs are frequent in cardiovascular diseases and contribute to cardiac hypertrophy by regulating a series of targeted genes. In this study, a rat model of cardiac hypertrophy was created by transverse abdominal aortic constriction, and cardiomyocyte hypertrophy in cultured neonatal rat cardiomyocytes was induced using angiotensin II (AngII) to investigate the role of miR-101 in myocardial hypertrophy. We demonstrated that miR-101 was downregulated in both the transverse abdominal aortic constriction rat model and hypertrophic cardiac myocytes. The overexpression of miR-101 in neonatal rat cardiomyocytes, which was accompanied by a reduced Rab1a level, inhibits 3 cardinal features of cardiomyocyte hypertrophy: fetal gene expression, protein synthesis, and cell enlargement. Conversely, the downregulation of miR-101 reverses these effects. Furthermore, the luciferase reporter system demonstrated that Rab1a is a target gene of miR-101, and the ectopic expression of Rab1a can reverse the cardiomyocyte hypertrophy inhibitory activity of miR-101. Taken together, our findings identify miR-101 as an important regulator in cardiac hypertrophy and implicate the potential application of miR-101 in the therapy of cardiac hypertrophy.
Collapse
|
15
|
Li N, Si B, Ju JF, Zhu M, You F, Wang D, Ren J, Ning YS, Zhang FQ, Dong K, Huang J, Yu WQ, Wang TJ, Qiao B. Nicotine Induces Cardiomyocyte Hypertrophy Through TRPC3-Mediated Ca 2+/NFAT Signalling Pathway. Can J Cardiol 2015; 32:1260.e1-1260.e10. [PMID: 26952156 DOI: 10.1016/j.cjca.2015.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/11/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Nicotine is thought to be an important risk factor for the development of cardiovascular diseases. However, the effects of nicotine on cardiomyocyte hypertrophy are poorly understood. The present study was designed to explore the role of nicotine in cardiomyocyte hypertrophy and its underlying mechanism. METHODS We used primary cardiomyocytes isolated from Wistar rats to examine the effects of nicotine on intracellular Ca2+ mobilization and hypertrophy determined by immunofluorescence, quantitative polymerase chain reaction, and western blot analysis. A luciferase reporter assay was used to examine the activity of NFAT signalling. RESULTS We found that nicotine caused cardiomyocyte hypertrophy, which was accompanied by increased intracellular Ca2+. Nicotine-enhanced intracellular Ca2+ concentration ([Ca2+]i) was significantly abolished by store-operated Ca2+ entry (SOCE) and TRPC inhibitors. Knockdown of TRPC3 significantly decreased nicotine-induced SOCE and hypertrophy. Moreover, calcineurin-nuclear factor of activated T cells (NFAT) is involved in TRPC3-mediated Ca2+ signalling and cardiomyocyte hypertrophy. Notably, upregulation of TRPC3 by nicotine requires TRPC3-mediated Ca2+ influx and calcineurin-NFAT signalling activation. CONCLUSIONS Our findings demonstrate that the prohypertrophic effect of nicotine on cardiomyocytes is dependent on enhanced TRPC3 expression through a calcium-dependent regulatory loop, which could become a potential target for prevention and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Na Li
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Biao Si
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Ji-Feng Ju
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Meng Zhu
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Feng You
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Dong Wang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Jie Ren
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Yan-Song Ning
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Feng-Quan Zhang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Kai Dong
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Jing Huang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Wen-Qian Yu
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Tong-Jian Wang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China.
| | - Bin Qiao
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China.
| |
Collapse
|
16
|
Palomer X, Capdevila-Busquets E, Botteri G, Davidson MM, Rodríguez C, Martínez-González J, Vidal F, Barroso E, Chan TO, Feldman AM, Vázquez-Carrera M. miR-146a targets Fos expression in human cardiac cells. Dis Model Mech 2015; 8:1081-91. [PMID: 26112171 PMCID: PMC4582106 DOI: 10.1242/dmm.020768] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/12/2015] [Indexed: 01/22/2023] Open
Abstract
miR-146a is a microRNA whose transcript levels are induced in the heart upon activation of NF-κB, a transcription factor induced by pro-inflammatory molecules (such as TNF-α) that is strongly related to the pathogenesis of cardiac disorders. The main goal of this study consisted of studying new roles of miR-146a in cardiac pathological processes caused by the pro-inflammatory cytokine TNF-α. Our results demonstrate that miR-146a transcript levels were sharply increased in cardiac ventricular tissue of transgenic mice with specific overexpression of TNF-α in the heart, and also in a cardiomyocyte cell line of human origin (AC16) exposed to TNF-α. Among all the in silico predicted miR-146a target genes, Fos mRNA and protein levels notably decreased after TNF-α treatment or miR-146a overexpression. These changes correlated with a diminution in the DNA-binding activity of AP-1, the Fos-containing transcription factor complex. Interestingly, AP-1 inhibition was accompanied by a reduction in matrix metalloproteinase (MMP)-9 mRNA levels in human cardiac cells. The specific regulation of this MMP by miR-146a was further confirmed at the secretion and enzymatic activity levels, as well as after anti-miR-mediated miR-146a inhibition. The results reported here demonstrate that Fos is a direct target of miR-146a activity and that downregulation of the Fos–AP-1 pathway by miR-146a has the capacity to inhibit MMP-9 activity. Given that MMP-9 is an AP-1 target gene involved in cardiac remodeling, myocardial dysfunction and progression of heart failure, these findings suggest that miR-146a might be a new and promising therapeutic tool for treating cardiac disorders associated with enhanced inflammation in the heart. Summary: These findings demonstrate that Fos is a direct target of miR-146a activity and that downregulation of the Fos–AP-1 pathway by miR-146a can inhibit MMP-9 activity.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology and Therapeutic Chemistry, IBUB (Institut de Biomedicina de la Universitat de Barcelona) and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy, University of Barcelona, Diagonal 643, Barcelona E-08028, Spain
| | - Eva Capdevila-Busquets
- Department of Pharmacology and Therapeutic Chemistry, IBUB (Institut de Biomedicina de la Universitat de Barcelona) and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy, University of Barcelona, Diagonal 643, Barcelona E-08028, Spain
| | - Gaia Botteri
- Department of Pharmacology and Therapeutic Chemistry, IBUB (Institut de Biomedicina de la Universitat de Barcelona) and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy, University of Barcelona, Diagonal 643, Barcelona E-08028, Spain
| | - Mercy M Davidson
- Department of Radiation Oncology, Columbia University, P&S 11-451, 630 West 168th Street, New York, NY 10032, USA
| | - Cristina Rodríguez
- Centro de Investigación Cardiovascular, CSIC-ICCC, IIB-Sant Pau, Avda. Sant Antoni Maria Claret 167, Barcelona 08025, Spain
| | - José Martínez-González
- Centro de Investigación Cardiovascular, CSIC-ICCC, IIB-Sant Pau, Avda. Sant Antoni Maria Claret 167, Barcelona 08025, Spain
| | - Francisco Vidal
- Unitat de Diagnòstic i Teràpia Molecular, Banc de Sang i Teixits, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Emma Barroso
- Department of Pharmacology and Therapeutic Chemistry, IBUB (Institut de Biomedicina de la Universitat de Barcelona) and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy, University of Barcelona, Diagonal 643, Barcelona E-08028, Spain
| | - Tung O Chan
- Department of Medicine, The Center for Translational Medicine, Jefferson Medical College, 1025 Walnut Street, Philadelphia, PA 19107, USA
| | - Arthur M Feldman
- Departments of Medicine and Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N, Broad Street, Philadelphia, PA 19140, USA
| | - Manuel Vázquez-Carrera
- Department of Pharmacology and Therapeutic Chemistry, IBUB (Institut de Biomedicina de la Universitat de Barcelona) and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy, University of Barcelona, Diagonal 643, Barcelona E-08028, Spain
| |
Collapse
|
17
|
van den Hoogen P, van den Akker F, Deddens JC, Sluijter JPG. Heart Failure in Chronic Myocarditis: A Role for microRNAs? Curr Genomics 2015; 16:88-94. [PMID: 26085807 PMCID: PMC4467309 DOI: 10.2174/1389202916999150120153344] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/20/2014] [Accepted: 01/06/2015] [Indexed: 12/22/2022] Open
Abstract
Myocarditis is an inflammatory disease of the heart, which can persist over a long time. During this time, known as the chronic phase of myocarditis, ongoing inflammation damages the cardiomyocytes. The loss of cardiac cells culminates in the development of dilated cardiomyopathy, often followed by non-ischemic heart failure due to diminished cardiac function. During the course of the disease, expression levels of non-coding small RNAs, called microRNAs (miRNAs), change. Although mainly studied in the acute setting, some of these changes in expression level appear to persist in the chronic phase. In addition to being a much-needed diagnostic tool, these miRNA could provide new treatment options. miRNA-based intervention strategies already showed promising results in the treatment of ischemic cardiovascular diseases in preclinical animal models. By implementing more knowledge on the role of miRNAs in the progression towards heart failure, this can potentially be used in the development of miRNA-based therapeutic interventions in the treatment of myocarditis and thereby preventing the progression towards heart failure. The first part of this review will focus on the natural course of myocarditis and the progression towards heart failure. Secondly, we will discuss the current knowledge on alterations of miRNA expression patterns, and suggest some possible future interventions.
Collapse
Affiliation(s)
- P van den Hoogen
- Department of Cardiology, University Medical Center Utrecht, the Netherlands; ; ICIN - Netherlands Heart Institute, Utrecht, The Netherlands
| | - F van den Akker
- Department of Cardiology, University Medical Center Utrecht, the Netherlands
| | - J C Deddens
- Department of Cardiology, University Medical Center Utrecht, the Netherlands
| | - J P G Sluijter
- Department of Cardiology, University Medical Center Utrecht, the Netherlands; ; ICIN - Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
18
|
Avlas O, Bragg A, Fuks A, Nicholson JD, Farkash A, Porat E, Aravot D, Levy-Drummer RS, Cohen C, Shainberg A, Arad M, Hochhauser E. TLR4 Expression Is Associated with Left Ventricular Dysfunction in Patients Undergoing Coronary Artery Bypass Surgery. PLoS One 2015; 10:e0120175. [PMID: 26030867 PMCID: PMC4451004 DOI: 10.1371/journal.pone.0120175] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 01/28/2015] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Toll-like receptor 4 (TLR4) is an innate immune receptor expressed in immune cells and the heart. Activation of the immune system following myocardial ischemia causes the release of proinflammatory mediators that may negatively influence heart function. AIM The aim of this study is to determine whether TLR4 is activated in peripheral monocytes and heart tissue taken from patients with varying degrees of myocardial dysfunction caused by coronary artery diseases and scheduled for coronary artery bypass graft (CABG) surgery before 12 months following operation. METHODS AND RESULTS Patients (n = 44) undergoing CABG surgery having left ventricular ejection fraction ≤ 45% ('reduced EF', n = 20) were compared to patients with preserved EF >45% ('preserved EF' group, n = 24). 'Reduced EF' patients exhibited increased TLR4 expression in monocytes (2.78±0.49 vs. 1.76±0.07 rMFI, p = 0.03). Plasma levels of C-reactive protein, microRNA miR-320a, brain natriuretic peptide (pro BNP) and NADPH oxidase (NOX4) were also significantly different between the 'preserved EF' and 'reduced EF'groups. Elevated TLR4 gene expression levels in the right auricle correlated with those of EF (p<0.008), NOX4 (p<0.008) and miR320, (p<0.04). In contrast, no differences were observed in peripheral monocyte TLR2 expression. After CABG surgery, monocyte TLR4 expression decreased in all patients, reaching statistical significance in the 'reduced EF' group. CONCLUSION TLR4 is activated in peripheral monocytes and heart tissue obtained from patients with ischemic heart disease and reduced left ventricular function. Coronary revascularization decreases TLR4 expression. We therefore propose that TLR4 plays a pathogenic role and may serve as an additional marker of ischemic myocardial dysfunction.
Collapse
Affiliation(s)
- Orna Avlas
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Arieh Bragg
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Avi Fuks
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - James D. Nicholson
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Ariel Farkash
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Eyal Porat
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Dan Aravot
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Rachel S. Levy-Drummer
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Cyrille Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Asher Shainberg
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Michael Arad
- Heart Failure Service and Leviev Heart Institute, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Edith Hochhauser
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
- * E-mail:
| |
Collapse
|
19
|
MCP-1-induced protein attenuates post-infarct cardiac remodeling and dysfunction through mitigating NF-κB activation and suppressing inflammation-associated microRNA expression. Basic Res Cardiol 2015; 110:26. [PMID: 25840774 DOI: 10.1007/s00395-015-0483-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/21/2022]
Abstract
MCP-1-induced protein (MCPIP, also known as ZC3H12A) has recently been uncovered to act as a negative regulator of inflammation. Expression of MCPIP was elevated in the ventricular myocardium of patients with ischemic heart failure. However, the role of MCPIP in the development of post-infarct cardiac inflammation and remodeling is unknown. The objective of the present study was to investigate whether MCPIP exerts an inhibitory effect on the cardiac inflammatory response and adverse remodeling after myocardial infarction (MI). Mice with cardiomyocyte-specific expression of MCPIP and their wild-type littermates (FVB/N) were subjected to permanent ligation of left coronary artery. The levels of MCPIP were significantly increased in the ischemic myocardium and sustained for 4 weeks after MI. Acute infarct size was comparable between groups. However, constitutive overexpression of MCPIP in the murine heart resulted in improved survival rate, decreased cardiac hypertrophy, less of fibrosis and scar formation, and better cardiac performance at 28 days after MI, along with a markedly reduced monocytic cell infiltration, less cytokine expression, decreased caspase-3/7 activities and apoptotic cell death compared to the wild-type hearts. Cardiomyocyte-specific expression of MCPIP also attenuated activation of cardiac NF-κB signaling and expression of inflammation-associated microRNAs (miR-126, -146a, -155, and -199a) when compared with the post-infarct wild-type hearts. In vitro, MCPIP expression suppressed hypoxia-induced NF-κB-luciferase activity in cardiomyocytes. In conclusion, MCPIP expression in the ischemic myocardium protects against adverse cardiac remodeling and dysfunction following MI by modulation of local myocardial inflammation, possibly through mitigating NF-κB signaling and suppressing inflammation-associated microRNA expression.
Collapse
|
20
|
Huynh K, Van Tassell B, Chow SL. Predicting therapeutic response in patients with heart failure: the story of C-reactive protein. Expert Rev Cardiovasc Ther 2015; 13:153-61. [PMID: 25578159 DOI: 10.1586/14779072.2015.1000307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Heart failure continues to be a major public health burden in the USA. With markedly high rates of morbidity and mortality upon diagnosis, effective treatment and prognosis are critical in the management of chronic heart failure. Growing evidence now supports the hypothesis that inflammation plays a key role in the progression and worsening of heart failure. Of the various inflammatory mediators identified, C-reactive protein, an acute phase inflammatory marker, has been associated with poor prognosis in patients with heart failure. Several interventional studies have been investigated to explore C-reactive protein modulation and potential treatment options and health outcomes; however, further studies are warranted before C-reactive protein-targeted therapy may be recommended in the management of heart failure.
Collapse
Affiliation(s)
- Kitty Huynh
- Western University of Health Sciences, Pharmacy Practice, 309 E. Second Street, Pomona, 91766, USA
| | | | | |
Collapse
|
21
|
Differential expression of dicer, miRNAs, and inflammatory markers in diabetic Ins2+/- Akita hearts. Cell Biochem Biophys 2014; 68:25-35. [PMID: 23797610 DOI: 10.1007/s12013-013-9679-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetic cardiomyopathy is a leading cause of morbidity and mortality, and Insulin2 mutant (Ins2+/-) Akita is a genetic mice model of diabetes relevant to humans. Dicer, miRNAs, and inflammatory cytokines are associated with heart failure. However, the differential expression of miRNAs, dicer, and inflammatory molecules are not clear in diabetic cardiomyopathy of Akita. We measured the levels of miRNAs, dicer, pro-inflammatory tumor necrosis factor alpha (TNFα), and anti-inflammatory interleukin 10 (IL-10) in C57BL/6J (WT) and Akita hearts. The results revealed increased heart to body weight ratio and robust expression of brain natriuretic peptide (BNP: a hypertrophy marker) suggesting cardiac hypertrophy in Akita. The multiplex RT-PCR, qPCR, and immunoblotting showed up regulation of dicer, whereas miRNA array elicited spread down regulation of miRNAs in Akita including dramatic down regulation of let-7a, miR-130, miR-142-3p, miR-148, miR-338, miR-345-3p, miR-384-3p, miR-433, miR-450, miR-451, miR-455, miR-494, miR-499, miR-500, miR-542-3p, miR-744, and miR-872. Conversely, miR-295 is induced in Akita. Cardiac TNFα is upregulated at mRNA (RT-PCR and qPCR), protein (immunoblotting), and cellular (immunohistochemistry and confocal microscopy) levels, and is robust in hypertrophic cardiomyocytes suggesting direct association of TNFα with hypertrophy. Contrary to TNFα, cardiac IL-10 is downregulated in Akita. In conclusion, induction of dicer and TNFα, and attenuation of IL-10 and majority of miRNA are associated with cardiomyopathy in Akita and could be used for putative therapeutic target for heart failure in diabetics.
Collapse
|
22
|
Dong Z, Wu P, Li Y, Shen Y, Xin P, Li S, Wang Z, Dai X, Zhu W, Wei M. Myocardial infarction worsens glomerular injury and microalbuminuria in rats with pre-existing renal impairment accompanied by the activation of ER stress and inflammation. Mol Biol Rep 2014; 41:7911-21. [PMID: 25173645 DOI: 10.1007/s11033-014-3685-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/20/2014] [Indexed: 01/01/2023]
Abstract
Deterioration of renal function occurs after chronic heart failure in approximately one-third of patients, particularly in those with pre-existing renal impairment such as diabetic nephropathy. Impaired renal function in these patients is always associated with a worse prognosis. However, the mechanisms underlying such deterioration of renal function are still largely unknown. In three separate protocols, we compared 1) sham operation (Ctr, n = 10) with surgically induced myocardial infarction (MI, n = 10); 2) unilateral nephrectomy (UNX, n = 10) with UNX + MI (n = 10); and 3) STZ-induced type 1 diabetes (DB, n = 10) with DB + MI (n = 10). The differences between combined injury models (UNX + MI, DB + MI) and simple MI were also examined. Renal remodeling, function, ER stress (CHOP and GRP78) and inflammation (infiltration of inflammatory cells, NF-κB p65) were evaluated 12 weeks after MI. In common SD rats, MI activated less glomerular ER stress and inflammation, resulting in a minor change of glomerular remodeling and microalbuminuria. However, MI significantly increased the glomerular expression of GRP78 and CHOP in UNX and DB rats. In addition, it also promoted the infiltration of CD4+ T cells, particularly inflammatory cytokine (IFN-γ, IL-17, IL-4)-producing CD4+ T cells, and the expression of NF-κB p65 in the glomeruli. By contrast, significant glomerular fibrosis, glomerulosclerosis, podocyte injury and microalbuminuria were found in rats with UNX + MI and DB + MI. MI significantly increased chronic glomerular injury and microalbuminuria at 12 weeks in rats with pre-existing renal impairment, i.e., UNX and DB, but not common SD rats. These changes were accompanied by increased glomerular ER stress and immune-associated inflammation.
Collapse
Affiliation(s)
- Zhifeng Dong
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, NO 600, Street Yishan, Shanghai, 200233, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Cardiac hypertrophy and fibrosis are two closely related adaptive response mechanisms of the myocardium to mechanical, metabolic, and genetic stress that finally contribute to the development of heart failure (HF). This relation is based on a dynamic interplay between many cell types including cardiomyocytes and fibroblasts during disease progression. Both cell types secrete a variety of growth factors, cytokines, and hormones that influence hypertrophic cardiomyocyte growth and fibrotic fibroblast activation in a paracrine and autocrine manner. It has become evident that, aside proteinous signals, microRNAs (miRNAs) and possible other RNA species such as long non-coding RNAs are potential players in such a cell-to-cell communication. By directly acting as paracrine signals or by modulating downstream intercellular signalling mediators, miRNAs can act as moderators of the intercellular crosstalk. These small regulators can potentially be secreted in a 'mircrine' fashion, so that miRNAs can be assumed as the message itself. This review will summarize the recent findings about the paracrine crosstalk between cardiac fibroblasts and cardiomyocytes and addresses how miRNAs may be involved in this interplay. It also highlights therapeutic strategies targeting factors of pathological communication for the treatment of HF.
Collapse
Affiliation(s)
- Janika Viereck
- Institute of Molecular and Translational Therapeutic Strategies , IFB-Tx, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover D-30625, Germany
| | | | | | | |
Collapse
|
24
|
Abstract
Cardiovascular diseases in children comprise a large public health problem. The major goals of paediatric cardiologists and paediatric cardiovascular researchers are to identify the cause(s) of these diseases to improve treatment and preventive protocols. Recent studies show the involvement of microRNAs (miRs) in different aspects of heart development, function, and disease. Therefore, miR-based research in paediatric cardiovascular disorders is crucial for a better understanding of the underlying pathogenesis of the disease, and unravelling novel, efficient, preventive, and therapeutic means. The ultimate goal of such research is to secure normal cardiac development and hence decrease disabilities, improve clinical outcomes, and decrease the morbidity and mortality among children. This review focuses on the role of miRs in different paediatric cardiovascular conditions in an effort to encourage miR-based research in paediatric cardiovascular disorders.
Collapse
|
25
|
Gupta MK, Halley C, Duan ZH, Lappe J, Viterna J, Jana S, Augoff K, Mohan ML, Vasudevan NT, Na J, Sossey-Alaoui K, Liu X, Liu CG, Tang WHW, Naga Prasad SV. miRNA-548c: a specific signature in circulating PBMCs from dilated cardiomyopathy patients. J Mol Cell Cardiol 2013; 62:131-41. [PMID: 23735785 DOI: 10.1016/j.yjmcc.2013.05.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/29/2013] [Accepted: 05/19/2013] [Indexed: 12/19/2022]
Abstract
High fidelity genome-wide expression analysis has strengthened the idea that microRNA (miRNA) signatures in peripheral blood mononuclear cells (PBMCs) can be potentially used to predict the pathology when anatomical samples are inaccessible like the heart. PBMCs from 48 non-failing controls and 44 patients with relatively stable chronic heart failure (ejection fraction of ≤ 40%) associated with dilated cardiomyopathy (DCM) were used for miRNA analysis. Genome-wide miRNA-microarray on PBMCs from chronic heart failure patients identified miRNA signature uniquely characterized by the downregulation of miRNA-548 family members. We have also independently validated downregulation of miRNA-548 family members (miRNA-548c & 548i) using real time-PCR in a large cohort of independent patient samples. Independent in silico Ingenuity Pathway Analysis (IPA) of miRNA-548 targets shows unique enrichment of signaling molecules and pathways associated with cardiovascular disease and hypertrophy. Consistent with specificity of miRNA changes with pathology, PBMCs from breast cancer patients showed no alterations in miRNA-548c expression compared to healthy controls. These studies suggest that miRNA-548 family signature in PBMCs can therefore be used to detect early heart failure. Our studies show that cognate networking of predicted miRNA-548 targets in heart failure can be used as a powerful ancillary tool to predict the ongoing pathology.
Collapse
Affiliation(s)
- Manveen K Gupta
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Despite our cognizance that diabetes can enhance the chances of heart failure, causes multiorgan failure,and contributes to morbidity and mortality, it is rapidly increasing menace worldwide. Less attention has been paid to alert prediabetics through determining the comprehensive predictors of diabetic cardiomyopathy (DCM) and ameliorating DCM using novel approaches. DCM is recognized as asymptomatic progressing structural and functional remodeling in the heart of diabetics, in the absence of coronary atherosclerosis and hypertension. The three major stages of DCM are: (1) early stage, where cellular and metabolic changes occur without obvious systolic dysfunction; (2) middle stage, which is characterized by increased apoptosis, a slight increase in left ventricular size, and diastolic dysfunction and where ejection fraction (EF) is <50%; and (3) late stage, which is characterized by alteration in microvasculature compliance, an increase in left ventricular size, and a decrease in cardiac performance leading to heart failure. Recent investigations have revealed that DCM is multifactorial in nature and cellular, molecular, and metabolic perturbations predisposed and contributed to DCM. Differential expression of microRNA (miRNA), signaling molecules involved in glucose metabolism, hyperlipidemia, advanced glycogen end products, cardiac extracellular matrix remodeling, and alteration in survival and differentiation of resident cardiac stem cells are manifested in DCM. A sedentary lifestyle and high fat diet causes obesity and this leads to type 2 diabetes and DCM. However, exercise training improves insulin sensitivity, contractility of cardiomyocytes, and cardiac performance in type 2 diabetes. These findings provide new clues to diagnose and mitigate DCM. This review embodies developments in the field of DCM with the aim of elucidating the future perspectives of predictors and prevention of DCM.
Collapse
Affiliation(s)
| | | | - Paras K Mishra
- Correspondence: Paras Kumar Mishra, Department of Physiology and Biophysics, School of Medicine, 500 S Preston Street, HSC-A Room 1216, University of Louisville, Louisville, KY 40202, USA, Tel +1 502 852 3627, Fax +1 502 852 6239, Email
| |
Collapse
|
27
|
Corsten MF, Papageorgiou A, Verhesen W, Carai P, Lindow M, Obad S, Summer G, Coort SLM, Hazebroek M, van Leeuwen R, Gijbels MJJ, Wijnands E, Biessen EAL, De Winther MPJ, Stassen FRM, Carmeliet P, Kauppinen S, Schroen B, Heymans S. MicroRNA profiling identifies microRNA-155 as an adverse mediator of cardiac injury and dysfunction during acute viral myocarditis. Circ Res 2012; 111:415-25. [PMID: 22715471 DOI: 10.1161/circresaha.112.267443] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Viral myocarditis results from an adverse immune response to cardiotropic viruses, which causes irreversible myocyte destruction and heart failure in previously healthy people. The involvement of microRNAs and their usefulness as therapeutic targets in this process are unknown. OBJECTIVE To identify microRNAs involved in viral myocarditis pathogenesis and susceptibility. METHODS AND RESULTS Cardiac microRNAs were profiled in both human myocarditis and in Coxsackievirus B3-injected mice, comparing myocarditis-susceptible with nonsusceptible mouse strains longitudinally. MicroRNA responses diverged depending on the susceptibility to myocarditis after viral infection in mice. MicroRNA-155, -146b, and -21 were consistently and strongly upregulated during acute myocarditis in both humans and susceptible mice. We found that microRNA-155 expression during myocarditis was localized primarily in infiltrating macrophages and T lymphocytes. Inhibition of microRNA-155 by a systemically delivered LNA-anti-miR attenuated cardiac infiltration by monocyte-macrophages, decreased T lymphocyte activation, and reduced myocardial damage during acute myocarditis in mice. These changes were accompanied by the derepression of the direct microRNA-155 target PU.1 in cardiac inflammatory cells. Beyond the acute phase, microRNA-155 inhibition reduced mortality and improved cardiac function during 7 weeks of follow-up. CONCLUSIONS Our data show that cardiac microRNA dysregulation is a characteristic of both human and mouse viral myocarditis. The inflammatory microRNA-155 is upregulated during acute myocarditis, contributes to the adverse inflammatory response to viral infection of the heart, and is a potential therapeutic target for viral myocarditis.
Collapse
Affiliation(s)
- Maarten F Corsten
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Current World Literature. Curr Opin Cardiol 2012; 27:318-26. [DOI: 10.1097/hco.0b013e328352dfaf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Mann DL. The emerging role of small non-coding RNAs in the failing heart: big hopes for small molecules. Cardiovasc Drugs Ther 2012; 25:149. [PMID: 21573764 DOI: 10.1007/s10557-011-6292-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Douglas L Mann
- Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
30
|
Remme WJ. Cardiovascular Drugs and Therapy Celebrates its 25th Year of Publication with a New Section: Education in Cardiovascular Therapy. Cardiovasc Drugs Ther 2011; 25:109-10. [DOI: 10.1007/s10557-011-6296-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|