1
|
Vinhais da Silva AV, Chesseron S, Benouna O, Rollin J, Roger S, Bourguignon T, Chadet S, Ivanes F. P2 purinergic receptors at the heart of pathological left ventricular remodeling following acute myocardial infarction. Am J Physiol Heart Circ Physiol 2025; 328:H550-H564. [PMID: 39884315 DOI: 10.1152/ajpheart.00599.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/17/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Pathological left ventricular remodeling is a complex process following an acute myocardial infarction, leading to architectural disorganization of the cardiac tissue. This phenomenon is characterized by sterile inflammation and the exaggerated development of fibrotic tissue, which is noncontractile and poorly conductive, responsible for organ dysfunction and heart failure. At present, specific therapies are lacking for both prevention and treatment of this condition, and no biomarkers are currently validated to identify at-risk patients. Physiopathological understanding of this process is limited, probably due to the combination of the multicellular responses involved that are initially necessary for tissue healing but may be detrimental in the longer term. Current research focuses on understanding and modulating the inflammatory response, a key aspect of the tissue healing process. Inflammation is triggered by the release of inflammatory mediators from cardiomyocytes undergoing cell death in the context of ischemia-reperfusion injury. Among them, extracellular ATP is a strong mediator of inflammation through the activation of P2 purinergic receptors, regulating the behavior of all the cellular actors of the postmyocardial infarction response and impacting organ function and recovery. Rather than considering each cellular protagonist independently, this review provides an integrated overview of the inflammatory and tissue response to myocardial infarction by members of the P2 receptor family. Finally, it explores the possibility of reducing pathological left ventricular remodeling through the modulation of these receptors and their associated signaling pathways.
Collapse
Affiliation(s)
- Ana Valéria Vinhais da Silva
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Simon Chesseron
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Oumnia Benouna
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Jérôme Rollin
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
- Service d'Hématologie-Hémostase, CHU de Tours, Tours, France
| | - Sébastien Roger
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Thierry Bourguignon
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
- Service de Chirurgie Cardiaque, CHU de Tours, Tours, France
| | - Stéphanie Chadet
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
| | - Fabrice Ivanes
- Inserm UMR1327 ISCHEMIA Membrane Signalling and Inflammation in reperfusion injuries, Université de Tours, Tours, France
- Service de Cardiologie, CHU de Tours, Tours, France
| |
Collapse
|
2
|
LoBue A, Li Z, Heuser SK, Li J, Leo F, Vornholz L, Dunaway LS, Suvorava T, Isakson BE, Cortese-Krott MM. Generation and characterization of a conditional eNOS knock out mouse model for cell-specific reactivation of eNOS in gain-of-function studies. Nitric Oxide 2024; 153:106-113. [PMID: 39419166 DOI: 10.1016/j.niox.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) in the vessel wall regulates blood pressure and cardiovascular hemodynamics. In this study, we generated conditional eNOS knock out (KO) mice characterized by a duplicated/inverted exon 2 flanked with two pairs of loxP regions (eNOSinv/inv); a Cre-recombinase activity induces cell-specific reactivation of eNOS, as a result of a flipping of the inverted exon 2 (eNOSfl). This work aimed to test the efficiency of the Cre-mediated cell-specific recombination and the resulting eNOS expression/function. As proof of concept, we crossed eNOSinv/inv mice with DeleterCrepos (DelCrepos) mice, expressing Cre recombinase in all cells. We generated heterozygous eNOSfl/inv or homozygous eNOSfl/fl mice, and eNOSinv/inv littermate mice. We found that both eNOSfl/fl and eNOSfl/inv mice express eNOS and the overall expression level depends on the number of mutated alleles, while eNOSinv/inv mice did not show any eNOS expression. Vascular endothelial function was restored in eNOSfl/fl and eNOSfl/inv mice, as determined by ACh-dependent vasodilation of aortic rings. Cre-dependent reactivation of eNOS in eNOSfl/fl and eNOSfl/inv mice rescued eNOSinv/inv (phenotypically global eNOS KO) mice from hypertension. These findings demonstrate that eNOS expression is restored in eNOSfl/fl mice at comparable physiological levels of WT mice, and its functional activity is independent on the number of the reactivated alleles. Therefore, eNOSinv/inv mice are a useful model for studying the effects of conditional reactivation of eNOS and gene dosage effects in specific cells for gain-of-function studies.
Collapse
Affiliation(s)
- Anthea LoBue
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Zhixin Li
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Sophia K Heuser
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Junjie Li
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Francesca Leo
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Lukas Vornholz
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Tatsiana Suvorava
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Group, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden; CARID, Cardiovascular Research Institute Düsseldorf, Germany.
| |
Collapse
|
3
|
Prajapati AK, Shah G. Exploring in vivo and in vitro models for heart failure with biomarker insights: a review. Egypt Heart J 2024; 76:141. [PMID: 39432214 PMCID: PMC11493927 DOI: 10.1186/s43044-024-00568-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a condition characterized by the heart's inability to meet the body's demands, resulting in various complications. Two primary types of HF exist, namely HF with preserved left ventricular ejection fraction (LVEF) and HF reduced with LVEF. The progression of HF involves compensatory mechanisms such as cardiac hypertrophy, fibrosis, and alterations in gene expression. Pressure overload and volume overload are common etiologies of HF, with pressure overload often stemming from conditions like hypertension, leading to left ventricular hypertrophy and fibrosis. In contrast, volume overload can arise from chronic valvular regurgitant disease, also inducing left ventricular hypertrophy. MAIN BODY In vitro cell culture techniques serve as vital tools in studying HF pathophysiology, allowing researchers to investigate cellular responses and potential therapeutic targets. Additionally, biomarkers, measurable biological characteristics, play a crucial role in diagnosing and predicting HF. Some notable biomarkers include adrenomedullin, B-type natriuretic peptide, copeptin, galectin-3, interleukin-6, matrix metalloproteinases (MMPs), midregional pro-atrial natriuretic peptide, myostatin, procollagen type I C-terminal propeptide, procollagen type III N-terminal propeptide and tissue inhibitors of metalloproteinases (TIMPs). These biomarkers aid in HF diagnosis, assessing its severity, and monitoring treatment response, contributing to a deeper understanding of the disease and potentially leading to improved management strategies and outcomes. CONCLUSIONS This review provides comprehensive insights into various in vivo models of HF, commonly utilized cell lines in HF research, and pivotal biomarkers with diagnostic relevance for HF. By synthesizing this information, researchers gain valuable resources to further explore HF pathogenesis, identify novel therapeutic targets, and enhance diagnostic and prognostic approaches.
Collapse
Affiliation(s)
- Anil Kumar Prajapati
- Pharmacology Department, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
- Research Scholar, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Gaurang Shah
- Pharmacology Department, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India.
| |
Collapse
|
4
|
Wei YR, Hou YL, Yin YJ, Li Z, Liu Y, Han NX, Wang ZX, Liu L, Wang XQ, Hao YJ, Ma K, Gu JJ, Jia ZH. Tongxinluo Activates PI3K/AKT Signaling Pathway to Inhibit Endothelial Mesenchymal Transition and Attenuate Myocardial Fibrosis after Ischemia-Reperfusion in Mice. Chin J Integr Med 2024; 30:608-615. [PMID: 38386252 DOI: 10.1007/s11655-024-3652-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 02/23/2024]
Abstract
OBJECTIVE To investigate the potential role of Tongxinluo (TXL) in attenuating myocardial fibrosis after myocardial ischemia-reperfusion injury (MIRI) in mice. METHODS A MIRI mouse model was established by left anterior descending coronary artery ligation for 45 min. According to a random number table, 66 mice were randomly divided into 6 groups (n=11 per group): the sham group, the model group, the LY-294002 group, the TXL group, the TXL+LY-294002 group and the benazepril (BNPL) group. The day after modeling, TXL and BNPL were administered by gavage. Intraperitoneal injection of LY-294002 was performed twice a week for 4 consecutive weeks. Echocardiography was used to measure cardiac function in mice. Masson staining was used to evaluate the degree of myocardial fibrosis in mice. Qualitative and quantitative analysis of endothelial mesenchymal transition (EndMT) after MIRI was performed by immunohistochemistry, immunofluorescence staining and flow cytometry, respectively. The protein expressions of platelet endothelial cell adhesion molecule-1 (CD31), α-smoth muscle actin (α-SMA), phosphatidylinositol-3-kinase (PI3K) and phospho protein kinase B (p-AKT) were assessed using Western blot. RESULTS TXL improved cardiac function in MIRI mice, reduced the degree of myocardial fibrosis, increased the expression of CD31 and inhibited the expression of α-SMA, thus inhibited the occurrence of EndMT (P<0.05 or P<0.01). TXL significantly increased the protein expressions of PI3K and p-AKT (P<0.05 or P<0.01). There was no significant difference between TXL and BNPL group (P>0.05). In addition, the use of the PI3K/AKT pathway-specific inhibitor LY-294002 to block this pathway and combination with TXL intervention, eliminated the protective effect of TXL, further supporting the protective effect of TXL. CONCLUSION TXL activated the PI3K/AKT signaling pathway to inhibit EndMT and attenuated myocardial fibrosis after MIRI in mice.
Collapse
Affiliation(s)
- Ya-Ru Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Yun-Long Hou
- Shijiazhuang Yiling Pharmaceutical New Drug Evaluation Center, Shijiazhuang, 050035, China
| | - Yu-Jie Yin
- Hebei Institute of Integrated Traditional and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Department of Cardiology, Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ning-Xin Han
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zi-Xuan Wang
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Lu Liu
- Hebei Institute of Integrated Traditional and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Department of Cardiology, Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Xiao-Qi Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Yuan-Jie Hao
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, China
| | - Kun Ma
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Jiao-Jiao Gu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Zhen-Hua Jia
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050090, China.
- Hebei Institute of Integrated Traditional and Western Medicine, Shijiazhuang, 050035, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
- Department of Cardiology, Hebei Yiling Hospital, Shijiazhuang, 050091, China.
| |
Collapse
|
5
|
Pan D, Xu L, Chen P, Miao L, Tian Y, Shi D, Guo M. Panax Quinquefolium Saponins enhances angiogenesis in rats with diabetes and myocardial infarction. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117252. [PMID: 37777023 DOI: 10.1016/j.jep.2023.117252] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/17/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xi Yang Shen (Panax quinquefolium L.) was originally recorded in "Ben Cao Cong Xin" edited by Wu Yiluo during the Qing Dynasty. Panax Quinquefolium Saponins (PQS) is the main component derived from Panax quinquefolium L, and has been wildly used in the treatment of coronary heart disease. AIM OF THE STUDY This study aims to explore the potential role and underlying mechanisms of PQS in promoting angiogenesis in rats with diabetes and myocardial infarction. MATERIALS AND METHODS Echocardiograms were used to assess cardiac function, while the heart weight to tibia length ratio was calculated to determine cardiac hypertrophy. Hematoxylin and eosin, periodic acid-Schiff and Masson's trichrome staining were used to examine cardiac morphology, myocyte diameter, and myocardial fibrosis. Immunofluorescence staining was employed to evaluate arteriolar density. The transcriptomes were analyzed and bioinformatic analyses were conducted to predict the potential angiogenesis-promoting mechanism of PQS. In addition, RT-PCR and western blotting was utilized to examine the expression of genes and proteins influenced by PQS. RESULTS PQS improved blood glucose, ameliorated cardiac function, reduced cardiac hypertrophy, and enhanced myocardial morphology in diabetic rats with myocardial infarction. PQS was also found to decrease myocyte diameter, curtail myocardial fibrosis, and increase arteriolar density. However, the effects of PQS were abolished following the deletion of protein kinase C δ (PKCδ). Molecular docking predicted strong interactions between the major blood components of PQS and PKCδ. Transcriptomic and bioinformatic analyses indicated that PQS may bolster angiogenesis by activating the VEGF/PI3K-Akt/eNOS pathway in rats with diabetes and myocardial infarction. Finally, the study demonstrated that PQS could inhibit the expression of PKCδ and stimulate the activation of the VEGF/PI3K-Akt/eNOS pathway. CONCLUSIONS PQS improves blood glucose, enhances cardiac function, mitigates cardiac damage, and boosts arteriolar density. The angiogenic impact of PQS appears to be, at least partially, due to its modulation of the PKCδ-mediated VEGF/PI3K-Akt/eNOS signaling pathway in rats with diabetes and myocardial infarction.
Collapse
Affiliation(s)
- Deng Pan
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Lin Xu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Pengfei Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Miao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Tian
- Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ming Guo
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Wang Y, Liu Y, Li X, Yao L, Mbadhi M, Chen S, Lv Y, Bao X, Chen L, Chen S, Zhang J, Wu Y, Lv J, Shi L, Tang J. Vagus nerve stimulation-induced stromal cell-derived factor-l alpha participates in angiogenesis and repair of infarcted hearts. ESC Heart Fail 2023; 10:3311-3329. [PMID: 37641543 PMCID: PMC10682864 DOI: 10.1002/ehf2.14475] [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: 12/23/2022] [Revised: 06/02/2023] [Accepted: 07/02/2023] [Indexed: 08/31/2023] Open
Abstract
AIMS We aim to explore the role and mechanism of vagus nerve stimulation (VNS) in coronary endothelial cells and angiogenesis in infarcted hearts. METHODS AND RESULTS Seven days after rat myocardial infarction (MI) was prepared by ligation of the left anterior descending coronary artery, the left cervical vagus nerve was treated with electrical stimulation 1 h after intraperitoneal administration of the α7-nicotinic acetylcholine inhibitor mecamylamine or the mAChR inhibitor atropine or 3 days after local injection of Ad-shSDF-1α into the infarcted heart. Cardiac tissue acetylcholine (ACh) and serum ACh, tumour necrosis factor α (TNF-α), interleukin 1β (IL-1β) and interleukin 6 (IL-6) levels were detected by ELISA to determine whether VNS was successful. An inflammatory injury model in human coronary artery endothelial cells (HCAECs) was established by lipopolysaccharide and identified by evaluating TNF-α, IL-1β and IL-6 levels and tube formation. Immunohistochemistry staining was performed to evaluate CD31-positive vessel density and stromal cell-derived factor-l alpha (SDF-1α) expression in the MI heart in vivo and the expression and distribution of SDF-1α, C-X-C motif chemokine receptor 4 and CXCR7 in HCAECs in vitro. Western blotting was used to detect the levels of SDF-1α, V-akt murine thymoma viral oncogene homolog (AKT), phosphorylated AKT (pAKT), specificity protein 1 (Sp1) and phosphorylation of Sp1 in HCAECs. Left ventricular performance, including left ventricular systolic pressure, left ventricular end-diastolic pressure and rate of the rise and fall of ventricular pressure, should be evaluated 28 days after VNS treatment. VNS was successfully established for MI therapy with decreases in serum TNF-α, IL-1β and IL-6 levels and increases in cardiac tissue and serum ACh levels, leading to increased SDF-1α expression in coronary endothelial cells of MI hearts, triggering angiogenesis of MI hearts with increased CD31-positive vessel density, which was abolished by the m/nAChR inhibitors mecamylamine and atropine or knockdown of SDF-1α by shRNA. ACh promoted SDF-1α expression and its distribution along with the branch of the formed tube in HCAECs, resulting in an increase in the number of tubes formed in HCAECs. ACh increased the levels of pAKT and phosphorylation of Sp1 in HCAECs, resulting in inducing SDF-1α expression, and the specific effects could be abolished by mecamylamine, atropine, the PI3K/AKT blocker wortmannin or the Sp1 blocker mithramycin. Functionally, VNS improved left ventricular performance, which could be abolished by Ad-shSDF-1α. CONCLUSIONS VNS promoted angiogenesis to repair the infarcted heart by inducing SDF-1α expression and redistribution along new branches during angiogenesis, which was associated with the m/nAChR-AKT-Sp1 signalling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Department of Pathology, Renmin HospitalHubei University of MedicineShiyanPR China
| | - Yun Liu
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Xing‐yuan Li
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Lu‐yuan Yao
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Department of Anesthesiology, Institute of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanPR China
| | - MagdaleenaNaemi Mbadhi
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Shao‐Juan Chen
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Department of Stomatology, Taihe HospitalHubei University of MedicineShiyanPR China
| | - Yan‐xia Lv
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
| | - Xin Bao
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Experimental Medical Center, Guoyao‐Dong Feng HospitalHubei University of MedicineShiyanPR China
| | - Long Chen
- Experimental Medical Center, Guoyao‐Dong Feng HospitalHubei University of MedicineShiyanPR China
| | - Shi‐You Chen
- Department of SurgeryUniversity of MissouriColumbiaMissouriUSA
| | - Jing‐xuan Zhang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| | - Yan Wu
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| | - Jing Lv
- Department of Anesthesiology, Institute of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanPR China
| | - Liu‐liu Shi
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| | - Jun‐ming Tang
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei Key Laboratory of Embryonic Stem Cell ResearchHubei University of MedicineShiyanPR China
- Institute of Basic Medical Sciences, Institute of BiomedicineHubei University of MedicineShiyanPR China
| |
Collapse
|
7
|
Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H, Peng C. Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res 2023; 197:106953. [PMID: 37804925 DOI: 10.1016/j.phrs.2023.106953] [Citation(s) in RCA: 5] [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: 06/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic multimorbidity (CMM) is an increasingly significant global public health concern. It encompasses the coexistence of multiple cardiometabolic diseases, including hypertension, stroke, heart disease, atherosclerosis, and T2DM. A crucial component to the development of CMM is the disruption of endothelial homeostasis. Therefore, therapies targeting endothelial cells through multi-targeted and multi-pathway approaches hold promise for preventing and treatment of CMM. Curcumin, a widely used dietary supplement derived from the golden spice Carcuma longa, has demonstrated remarkable potential in treatment of CMM through its interaction with endothelial cells. Numerous studies have identified various molecular targets of curcumin (such as NF-κB/PI3K/AKT, MAPK/NF-κB/IL-1β, HO-1, NOs, VEGF, ICAM-1 and ROS). These findings highlight the efficacy of curcumin as a therapeutic agent against CMM through the regulation of endothelial function. It is worth noting that there is a close relationship between the progression of CMM and endothelial damage, characterized by oxidative stress, inflammation, abnormal NO bioavailability and cell adhesion. This paper provides a comprehensive review of curcumin, including its availability, pharmacokinetics, pharmaceutics, and therapeutic application in treatment of CMM, as well as the challenges and future prospects for its clinical translation. In summary, curcumin shows promise as a potential treatment option for CMM, particularly due to its ability to target endothelial cells. It represents a novel and natural lead compound that may offer significant therapeutic benefits in the management of CMM.
Collapse
Affiliation(s)
- Fei Tang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dong Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
8
|
Martín-Bórnez M, Falcón D, Morrugares R, Siegfried G, Khatib AM, Rosado JA, Galeano-Otero I, Smani T. New Insights into the Reparative Angiogenesis after Myocardial Infarction. Int J Mol Sci 2023; 24:12298. [PMID: 37569674 PMCID: PMC10418963 DOI: 10.3390/ijms241512298] [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: 07/03/2023] [Revised: 07/23/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Myocardial infarction (MI) causes massive loss of cardiac myocytes and injury to the coronary microcirculation, overwhelming the limited capacity of cardiac regeneration. Cardiac repair after MI is finely organized by complex series of procedures involving a robust angiogenic response that begins in the peri-infarcted border area of the infarcted heart, concluding with fibroblast proliferation and scar formation. Efficient neovascularization after MI limits hypertrophied myocytes and scar extent by the reduction in collagen deposition and sustains the improvement in cardiac function. Compelling evidence from animal models and classical in vitro angiogenic approaches demonstrate that a plethora of well-orchestrated signaling pathways involving Notch, Wnt, PI3K, and the modulation of intracellular Ca2+ concentration through ion channels, regulate angiogenesis from existing endothelial cells (ECs) and endothelial progenitor cells (EPCs) in the infarcted heart. Moreover, cardiac repair after MI involves cell-to-cell communication by paracrine/autocrine signals, mainly through the delivery of extracellular vesicles hosting pro-angiogenic proteins and non-coding RNAs, as microRNAs (miRNAs). This review highlights some general insights into signaling pathways activated under MI, focusing on the role of Ca2+ influx, Notch activated pathway, and miRNAs in EC activation and angiogenesis after MI.
Collapse
Affiliation(s)
- Marta Martín-Bórnez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Débora Falcón
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Rosario Morrugares
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
- Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, 14071 Córdoba, Spain
| | - Geraldine Siegfried
- RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France (A.-M.K.)
| | - Abdel-Majid Khatib
- RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France (A.-M.K.)
| | - Juan A. Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003 Caceres, Spain;
| | - Isabel Galeano-Otero
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| |
Collapse
|
9
|
Early Development of Cardiac Fibrosis in Young Old-Father Offspring. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8770136. [PMID: 36193084 PMCID: PMC9526616 DOI: 10.1155/2022/8770136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
Cardiac aging is characterized by progressive fibrosis. Epidemiological studies have found that advanced paternal age is associated with an increased risk of heart failure in the next generation. This study is aimed at evaluating the effect of paternal age, in the young male rat progeny, on cardiac phenotype under circulatory stress conditions. Offspring rats were obtained by mating old males (24 months old) with young females (two months old) and by mating young males (two months old) with the same young females. Hypertension was induced in old father offspring (OFO) rats and young old father (YFO) offspring rats using L-NAME (N(ω)-nitro-L-arginine methyl ester). The OFO L-NAME rats showed a high blood pressure phenotype associated with substantial cardiac hypertrophy and an exacerbation of cardiac fibrosis compared to the YFO L-NAME rats. Histological analysis of heart tissue showed an expansion of the extracellular matrix, with fibroblasts displaying markers of epicardial origin (Tcf21, Tbx18, and Wt1) in the OFO group. Moreover, western blot and protein phosphorylation antibody array identified the TGF-β2 receptor pathway as preferentially activated in aged hearts as well as in OFO cardiac tissue treated with L-NAME. In addition, old father offspring rats (OFO+OFO L-NAME) had increased cardiac DNA methylation. In young hypertensive progeny, advanced paternal age at conception may be a risk factor for early progression towards cardiac fibrosis. An intergenerational transmission may be behind the paternal age-related cardiac remodeling in the young offspring.
Collapse
|
10
|
Zhang YG, Liu XX, Zong JC, Zhang YTJ, Dong R, Wang N, Ma ZH, Li L, Wang SL, Mu YL, Wang SS, Liu ZM, Han LW. Investigation Driven by Network Pharmacology on Potential Components and Mechanism of DGS, a Natural Vasoprotective Combination, for the Phytotherapy of Coronary Artery Disease. Molecules 2022; 27:molecules27134075. [PMID: 35807320 PMCID: PMC9268537 DOI: 10.3390/molecules27134075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/14/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Phytotherapy offers obvious advantages in the intervention of Coronary Artery Disease (CAD), but it is difficult to clarify the working mechanisms of the medicinal materials it uses. DGS is a natural vasoprotective combination that was screened out in our previous research, yet its potential components and mechanisms are unknown. Therefore, in this study, HPLC-MS and network pharmacology were employed to identify the active components and key signaling pathways of DGS. Transgenic zebrafish and HUVECs cell assays were used to evaluate the effectiveness of DGS. A total of 37 potentially active compounds were identified that interacted with 112 potential targets of CAD. Furthermore, PI3K-Akt, MAPK, relaxin, VEGF, and other signal pathways were determined to be the most promising DGS-mediated pathways. NO kit, ELISA, and Western blot results showed that DGS significantly promoted NO and VEGFA secretion via the upregulation of VEGFR2 expression and the phosphorylation of Akt, Erk1/2, and eNOS to cause angiogenesis and vasodilation. The result of dynamics molecular docking indicated that Salvianolic acid C may be a key active component of DGS in the treatment of CAD. In conclusion, this study has shed light on the network molecular mechanism of DGS for the intervention of CAD using a network pharmacology-driven strategy for the first time to aid in the intervention of CAD.
Collapse
Affiliation(s)
- You-Gang Zhang
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
| | - Xia-Xia Liu
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030000, China
| | - Jian-Cheng Zong
- Chenland Research Institute, Irvine, CA 92697, USA; (J.-C.Z.); (L.L.); (S.-L.W.)
| | - Yang-Teng-Jiao Zhang
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
| | - Rong Dong
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
| | - Na Wang
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030000, China
| | - Zhi-Hui Ma
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, China
| | - Li Li
- Chenland Research Institute, Irvine, CA 92697, USA; (J.-C.Z.); (L.L.); (S.-L.W.)
| | - Shang-Long Wang
- Chenland Research Institute, Irvine, CA 92697, USA; (J.-C.Z.); (L.L.); (S.-L.W.)
| | - Yan-Ling Mu
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
| | - Song-Song Wang
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
| | - Zi-Min Liu
- Chenland Nutritionals Inc., Irvine, CA 92697, USA
- Correspondence: (Z.-M.L.); (L.-W.H.)
| | - Li-Wen Han
- School of Pharmacy and Pharmaceutical Science, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China; (Y.-G.Z.); (X.-X.L.); (Y.-T.-J.Z.); (R.D.); (N.W.); (Z.-H.M.); (Y.-L.M.); (S.-S.W.)
- Correspondence: (Z.-M.L.); (L.-W.H.)
| |
Collapse
|
11
|
Musicante M, Kim HH, Chen Y, Liao F, Bhattacharya SK, Lu L, Sun Y. Regulation of endothelial nitric oxide synthase in cardiac remodeling. Int J Cardiol 2022; 364:96-101. [PMID: 35654172 DOI: 10.1016/j.ijcard.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/25/2022] [Accepted: 05/06/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Our previous study demonstrated that endothelial nitric oxide synthase (eNOS) gene serves as a candidate for modifiers of hypertrophic cardiomyopathy (HCM), which alters severity of HCM phenotypes. Herein, we sought to further elucidate the role of eNOS on cardiac myocyte hypertrophy and fibrosis, the major phenotypes of HCM. METHODS Male eNOS-deficient mice (eNOS-/-) and wild type control mice (eNOS+/+, C57B1/6 J) were used in this study. Myocyte size was analyzed in hematoxylin/eosin stained sections using an image analyzing system. Cardiac β-myosin heavy chain (β-MHC) and α-skeletal actin (α-SKA) levels, markers of myocyte hypertrophy were evaluated by Western blot. Cardiac collagen volume fraction (CVF) was examined in picrosirius red stained section using an image analyzing system. Cardiac expression of tissue inhibitor of metalloproteinase 1 (TIMP-1) and transforming growth factor beta 1 (TGF-β1), markers of fibrosis, were determined by Western blot. RESULTS Compared to eNOS+/+ mice, we found that; 1) myocyte size was significantly increased in eNOS-/- mice; 2) cardiac expression of β-MHC was markedly elevated, while α-SKA levels remained unchanged in eNOS-/- mice; 3) cardiac total and interstitial CVF levels were significantly higher in eNOS-/- mice; and 4) cardiac TIMP-1 levels were significantly greater in eNOS-/- mice, however, cardiac TGF-β1 was not differently expressed between the two groups. CONCLUSION The current study revealed that eNOS plays a beneficial role in cardiac remodeling, preventing the heart from development of myocyte hypertrophy and cardiac fibrosis. These findings support our previous report that eNOS may modify the severity of HCM phenotypes.
Collapse
Affiliation(s)
- Meryl Musicante
- University of Tennessee Health Science Center, United States of America
| | - Hannah H Kim
- University of Tennessee Health Science Center, United States of America
| | - Yuanjian Chen
- Division of Cardiovascular Diseases, Department of Medicine(,) University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine(,) University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America.
| | - Yao Sun
- Division of Cardiovascular Diseases, Department of Medicine(,) University of Tennessee Health Science Center, Memphis, TN, United States of America.
| |
Collapse
|
12
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 316] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
13
|
Regional Diversities in Fibrogenesis Weighed as a Key Determinant for Atrial Arrhythmogenesis. Biomedicines 2021; 9:biomedicines9121900. [PMID: 34944715 PMCID: PMC8698388 DOI: 10.3390/biomedicines9121900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 11/18/2022] Open
Abstract
Atrial fibrosis plays a key role in atrial myopathy, resulting in the genesis of atrial fibrillation (AF). The abnormal distribution of fibrotic tissue, electrical coupling, paracrine interactions, and biomechanical–electrical interactions have all been suggested as causes of fibrosis-related arrhythmogenesis. Moreover, the regional difference in fibrogenesis, specifically the left atrium (LA) exhibiting a higher arrhythmogenesis and level of fibrosis than the right atrium (RA) in AF, is a key contributor to atrial arrhythmogenesis. LA fibroblasts have greater profibrotic cellular activities than RA fibroblasts, but knowledge about the regional diversity of atrial regional fibrogenesis remains limited. This article provides a comprehensive review of research findings on the association between fibrogenesis and arrhythmogenesis from laboratory to clinical evidence and updates the current understanding of the potential mechanism underlying the difference in fibrogenesis between the LA and RA.
Collapse
|
14
|
Akita K, Isoda K, Ohtomo F, Isobe S, Niida T, Sato-Okabayashi Y, Sano M, Shimada K, Iwakura Y, Minamino T. Blocking of interleukin-1 suppresses angiotensin II-induced renal injury. Clin Sci (Lond) 2021; 135:2035-2048. [PMID: 34402864 DOI: 10.1042/cs20201406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 11/17/2022]
Abstract
Clinical hypertension (HT) is associated with renal inflammation and elevated circulating levels of proinflammatory cytokines. Interleukin (IL)-1 receptor antagonist (IL-1Ra) is one of the most important anti-inflammatory cytokines and plays a crucial role in inflammation. Inhibition of IL-1 may contribute to modulation of the Angiotensin II (Ang II)-induced HT response. The present study aimed to elucidate the effects of IL-1Ra and anti-IL-1β antibody (01BSUR) on Ang II-induced renal injury. To determine the contribution of IL-1Ra to Ang II-induced renal inflammation, male wildtype (WT) and IL-1Ra-deficient (IL-1Ra-/-) mice were infused with Ang II (1000 ng/kg/min) using subcutaneous osmotic pump for 14 days. We checked renal function, histological change, and several mRNA expressions 14 days after infusion. Fourteen days after infusion, systolic blood pressure (197 ± 5 vs 169 ± 9 mmHg, P<0.05) in IL-1Ra-/- mice significantly increased compared with WT mice. Furthermore, on day 14 of Ang II infusion, plasma IL-6 was 5.9-fold higher in IL-1Ra-/- versus WT mice (P<0.001); renal preproendothelin-1 mRNA expression was also significantly higher in IL-1Ra-/- mice (P<0.05). In addition, renal histology revealed greater damage in IL-1Ra-/- mice compared with WT mice 14 days after infusion. Finally, we administrated 01BSUR to both IL-1Ra-/- and WT mice, and 01BSUR treatment decreased Ang II-induced HT and renal damage (glomerular injury and fibrosis of the tubulointerstitial area) in both IL-1Ra-/- and WT mice compared with IgG2a treatment. Inhibition of IL-1 decreased Ang II-induced HT and renal damage in both IL-1Ra-/- and WT mice, suggesting suppression of IL-1 may provide an additional strategy to protect against renal damage in hypertensive patients.
Collapse
Affiliation(s)
- Koji Akita
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Kikuo Isoda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Cardiology, Juntendo University Nerima Hospital, Nerimaku, Tokyo, Japan
| | - Fumie Ohtomo
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Sarasa Isobe
- Division of Cardiology, Keio University, Shinanomachi, Tokyo, Japan
| | - Tomiharu Niida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yayoi Sato-Okabayashi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Motoaki Sano
- Division of Cardiology, Keio University, Shinanomachi, Tokyo, Japan
| | - Kazunori Shimada
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
15
|
Zhu Q, Zhang Y, Tang J, Tang N, He Y, Chen X, Gao S, Xu Y, Liu Z. Ultrasound-Targeted Microbubble Destruction Accelerates Angiogenesis and Ameliorates Left Ventricular Dysfunction after Myocardial Infarction in Mice. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2692-2701. [PMID: 34130882 DOI: 10.1016/j.ultrasmedbio.2021.04.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/25/2021] [Accepted: 04/25/2021] [Indexed: 06/12/2023]
Abstract
Failure of coronary recanalization within 12 h or no flow in the myocardium after percutaneous coronary intervention is associated with high mortality from myocardial infarction, and insufficient angiogenesis in the border zone results in the expansion of infarct area. In this study, we examined the effects of ultrasound-targeted microbubble destruction (UTMD) on angiogenesis and left ventricular dysfunction in a mouse model of myocardial infarction. Fifty-four mice with MI were treated with no UTMD, ultrasound (US) alone or UTMD four times (days 1, 3, 5 and 7), and another 18 mice underwent sham operation and therapy. Therapeutic US was generated with a linear transducer connected to a commercial diagnostic US system (VINNO70). UTMD was performed with the VINNO70 at a peak negative pressure of 0.8 MPa and lipid microbubbles. Transthoracic echocardiography was performed on the first and seventh days. The results indicated that UTMD decreased the infarct size ratio from 78.1 ± 5.3% (untreated) to 43.3 ± 6.4%, accelerated angiogenesis and ameliorated left ventricular dysfunction. The ejection fraction increased from 25.05 ± 8.52% (untreated) to 42.83 ± 9.44% (UTMD). Compared with that in other groups, expression of vascular endothelial growth factor and endothelial nitric oxide synthase and release of nitric oxide were significantly upregulated after UTMD treatment, indicating angiogenesis. Therefore, UTMD is a potential physical approach in the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jiawei Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Najiao Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ying He
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoqin Chen
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shunji Gao
- Department of Ultrasound, General Hospital of Central Theater Command, Wuhan, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
16
|
Role of PI3K/Akt signaling pathway in cardiac fibrosis. Mol Cell Biochem 2021; 476:4045-4059. [PMID: 34244974 DOI: 10.1007/s11010-021-04219-w] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/29/2021] [Indexed: 12/26/2022]
Abstract
Heart failure (HF) is considered as a severe health problem worldwide, while cardiac fibrosis is one of the main driving factors for the progress of HF. Cardiac fibrosis was characterized by changes in cardiomyocytes, cardiac fibroblasts, ratio of collagen (COL) I/III, and the excessive production and deposition of extracellular matrix (ECM), thus forming a scar tissue, which leads to pathological process of cardiac structural changes and systolic as well as diastolic dysfunction. Cardiac fibrosis is a common pathological change of many advanced cardiovascular diseases including ischemic heart disease, hypertension, and HF. Accumulated studies have proven that phosphoinositol-3 kinase (PI3K)/Akt signaling pathway is involved in regulating the occurrence, progression and pathological formation of cardiac fibrosis via regulating cell survival, apoptosis, growth, cardiac contractility and even the transcription of related genes through a series of molecules including mammalian target of rapamycin (mTOR), glycogen synthase kinase 3 (GSK-3), forkhead box proteins O1/3 (FoxO1/3), and nitric oxide synthase (NOS). Thus, the review focuses on the role of PI3K/Akt signaling pathway in the cardiac fibrosis. The information reviewed here should be significant in understanding the role of PI3K/Akt in cardiac fibrosis and contribute to the design of further studies related to PI3K/Akt and the cardiac fibrotic response, as well as sought to shed light on a potential treatment for cardiac fibrosis.
Collapse
|
17
|
Wang Y, Wang M, Samuel CS, Widdop RE. Preclinical rodent models of cardiac fibrosis. Br J Pharmacol 2021; 179:882-899. [PMID: 33973236 DOI: 10.1111/bph.15450] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 11/30/2022] Open
Abstract
Cardiac fibrosis (scarring), characterised by an increased deposition of extracellular matrix (ECM) proteins, is a hallmark of most types of cardiovascular disease and plays an essential role in heart failure progression. Inhibition of cardiac fibrosis could improve outcomes in patients with cardiovascular diseases and particularly heart failure. However, pharmacological treatment of the ECM build-up is still lacking. In this context, preclinical models of heart disease are important tools for understanding the complex pathogenesis involved in the development of cardiac fibrosis which in turn could identify new therapeutic targets and the facilitation of antifibrotic drug discovery. Many preclinical models have been used to study cardiac fibrosis and each model provides mechanistic insights into the many factors that contribute to cardiac fibrosis. This review discusses the most frequently used rodent models of cardiac fibrosis and also provides context for the use of particular models of heart failure.
Collapse
Affiliation(s)
- Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
18
|
Stewart L, Turner NA. Channelling the Force to Reprogram the Matrix: Mechanosensitive Ion Channels in Cardiac Fibroblasts. Cells 2021; 10:990. [PMID: 33922466 PMCID: PMC8145896 DOI: 10.3390/cells10050990] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibroblasts (CF) play a pivotal role in preserving myocardial function and integrity of the heart tissue after injury, but also contribute to future susceptibility to heart failure. CF sense changes to the cardiac environment through chemical and mechanical cues that trigger changes in cellular function. In recent years, mechanosensitive ion channels have been implicated as key modulators of a range of CF functions that are important to fibrotic cardiac remodelling, including cell proliferation, myofibroblast differentiation, extracellular matrix turnover and paracrine signalling. To date, seven mechanosensitive ion channels are known to be functional in CF: the cation non-selective channels TRPC6, TRPM7, TRPV1, TRPV4 and Piezo1, and the potassium-selective channels TREK-1 and KATP. This review will outline current knowledge of these mechanosensitive ion channels in CF, discuss evidence of the mechanosensitivity of each channel, and detail the role that each channel plays in cardiac remodelling. By better understanding the role of mechanosensitive ion channels in CF, it is hoped that therapies may be developed for reducing pathological cardiac remodelling.
Collapse
Affiliation(s)
| | - Neil A. Turner
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, UK;
| |
Collapse
|
19
|
Yun W, Qian L, Yuan R, Xu H. Periplocymarin protects against myocardial fibrosis induced by β-adrenergic activation in mice. Biomed Pharmacother 2021; 139:111562. [PMID: 33839492 DOI: 10.1016/j.biopha.2021.111562] [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: 12/27/2020] [Revised: 03/24/2021] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
Periplocymarin is an effective component of Periplocae Cortex, which was wildly used as an ingredient in Traditional Chinese Medicine. Our group previously reported that periplocymarin exerted cardiotonic role via promoting calcium influx. However, its exact role in the pathogenesis of myocardial fibrosis has not been elucidated yet. The present study was aimed at determining the potential effect and underlying mechanism of periplocymarin in isoproterenol (ISO)-induced myocardial fibrosis. C57BL/6 mice were subcutaneously injected with ISO (5 mg/kg/day) or saline for 1 week. The early-to-atrial wave ratio (E/A ratio) measured by echocardiography revealed that ISO-induced heart stiffness was remarkably reversed by administration of periplocymarin (5 mg/kg/day). Masson trichrome staining exhibited that treatment of periplocymarin reduced the excessive deposition of extracellular matrix (ECM). Further investigations employing real-time PCR and western blot demonstrated that periplocymarin suppressed the expression of fibrosis related genes (Col1a1, Col3a1, Acta2 and Tgfb1) and proteins (Collagen I, Collagen III, α-SMA and TGF-β1) induced by ISO. Metabolomics analysis demonstrated that periplocymarin ameliorated the disorders triggered by ISO and many of the differential metabolic substances were involved in amino acid, glucose and lipid metabolism. Further analysis using network pharmacology revealed that three key genes, namely NOS2, NOS3 and Ptgs2, may be the potential targets of periplocymarin and responsible for the disorders. Validation using heart tissues showed that the mRNA expression of NOS3 was decreased while Ptgs2 was increased upon ISO treatment, which were reversed by periplocymarin. Moreover, the expression of COX-2 (Ptgs2 encoded protein) was consistent with the aspect of Ptgs2 mRNA, while eNOS (NOS3 encoded protein) expression was unchanged. In vitro studies exhibited that periplocymarin exerts anti-fibrotic function via regulating at least eNOS and COX-2 in cardiomyocyte. Taken together, periplocymarin protects against myocardial fibrosis induced by β-adrenergic activation, the potential mechanism was that periplocymarin targeted on, at least eNOS and COX-2, to improve the metabolic processes of cardiomyocyte and thus attenuated the myocardial fibrosis. Our study highlighted that periplocymarin is a potential therapeutic agent for the prevention of myocardial fibrosis.
Collapse
Affiliation(s)
- Weijing Yun
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Lei Qian
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Ruqiang Yuan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Hu Xu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
20
|
Angiotensin II-Induced Cardiovascular Fibrosis Is Attenuated by NO-Sensitive Guanylyl Cyclase1. Cells 2020; 9:cells9112436. [PMID: 33171621 PMCID: PMC7695185 DOI: 10.3390/cells9112436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
In the NO/cGMP signaling cascade, relevant in the cardiovascular system, two NO-sensitive guanylyl cyclase (NO-GC) isoforms are responsible for NO-dependent cGMP generation. Here, the impact of the major NO-GC isoform, NO-GC1, on fibrosis development in the cardiovascular system was studied in NO-GC1-deficient mice treated with AngiotensinII (AngII), known to induce vascular and cardiac remodeling. Morphometric analysis of NO-GC1 KO’s aortae demonstrated an enhanced increase of perivascular area after AngII treatment accompanied by a higher aortic collagen1 mRNA content. Increased perivascular fibrosis also occurred in cardiac vessels of AngII-treated NO-GC1 KO mice. In line, AngII-induced interstitial fibrosis was 32% more pronounced in NO-GC1 KO than in WT myocardia associated with a higher cardiac Col1 and other fibrotic marker protein content. In sum, increased perivascular and cardiac interstitial fibrosis together with the enhanced collagen1 mRNA content in AngII-treated NO-GC1-deficient mice represent an exciting manifestation of antifibrotic properties of cGMP formed by NO-GC1, a finding with great pharmaco-therapeutic implications.
Collapse
|
21
|
Caporarello N, Meridew JA, Aravamudhan A, Jones DL, Austin SA, Pham TX, Haak AJ, Moo Choi K, Tan Q, Haresi A, Huang SK, Katusic ZS, Tschumperlin DJ, Ligresti G. Vascular dysfunction in aged mice contributes to persistent lung fibrosis. Aging Cell 2020; 19:e13196. [PMID: 32691484 PMCID: PMC7431829 DOI: 10.1111/acel.13196] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/28/2020] [Accepted: 06/21/2020] [Indexed: 12/23/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease thought to result from impaired lung repair following injury and is strongly associated with aging. While vascular alterations have been associated with IPF previously, the contribution of lung vasculature during injury resolution and fibrosis is not well understood. To compare the role of endothelial cells (ECs) in resolving and non-resolving models of lung fibrosis, we applied bleomycin intratracheally to young and aged mice. We found that injury in aged mice elicited capillary rarefaction, while injury in young mice resulted in increased capillary density. ECs from the lungs of injured aged mice relative to young mice demonstrated elevated pro-fibrotic and reduced vascular homeostasis gene expression. Among the latter, Nos3 (encoding the enzyme endothelial nitric oxide synthase, eNOS) was transiently upregulated in lung ECs from young but not aged mice following injury. Young mice deficient in eNOS recapitulated the non-resolving lung fibrosis observed in aged animals following injury, suggesting that eNOS directly participates in lung fibrosis resolution. Activation of the NO receptor soluble guanylate cyclase in human lung fibroblasts reduced TGFβ-induced pro-fibrotic gene and protein expression. Additionally, loss of eNOS in human lung ECs reduced the suppression of TGFβ-induced lung fibroblast activation in 2D and 3D co-cultures. Altogether, our results demonstrate that persistent lung fibrosis in aged mice is accompanied by capillary rarefaction, loss of EC identity, and impaired eNOS expression. Targeting vascular function may thus be critical to promote lung repair and fibrosis resolution in aging and IPF.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey A Meridew
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Aja Aravamudhan
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Dakota L Jones
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Susan A Austin
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Tho X Pham
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Andrew J Haak
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kyoung Moo Choi
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Qi Tan
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Adil Haresi
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zvonimir S Katusic
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Masoud AG, Lin J, Azad AK, Farhan MA, Fischer C, Zhu LF, Zhang H, Sis B, Kassiri Z, Moore RB, Kim D, Anderson CC, Vederas JC, Adam BA, Oudit GY, Murray AG. Apelin directs endothelial cell differentiation and vascular repair following immune-mediated injury. J Clin Invest 2020; 130:94-107. [PMID: 31738185 DOI: 10.1172/jci128469] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/18/2019] [Indexed: 01/06/2023] Open
Abstract
Sustained, indolent immune injury of the vasculature of a heart transplant limits long-term graft and recipient survival. This injury is mitigated by a poorly characterized, maladaptive repair response. Vascular endothelial cells respond to proangiogenic cues in the embryo by differentiation to specialized phenotypes, associated with expression of apelin. In the adult, the role of developmental proangiogenic cues in repair of the established vasculature is largely unknown. We found that human and minor histocompatibility-mismatched donor mouse heart allografts with alloimmune-mediated vasculopathy upregulated expression of apelin in arteries and myocardial microvessels. In vivo, loss of donor heart expression of apelin facilitated graft immune cell infiltration, blunted vascular repair, and worsened occlusive vasculopathy in mice. In vitro, an apelin receptor agonist analog elicited endothelial nitric oxide synthase activation to promote endothelial monolayer wound repair and reduce immune cell adhesion. Thus, apelin acted as an autocrine growth cue to sustain vascular repair and mitigate the effects of immune injury. Treatment with an apelin receptor agonist after vasculopathy was established markedly reduced progression of arterial occlusion in mice. Together, these initial data identify proangiogenic apelin as a key mediator of coronary vascular repair and a pharmacotherapeutic target for immune-mediated injury of the coronary vasculature.
Collapse
Affiliation(s)
| | - Jiaxin Lin
- Department of Surgery.,Department of Medical Microbiology and Immunology, and
| | | | | | - Conrad Fischer
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Hao Zhang
- Department of Medicine.,Mazankowski Heart Institute, Edmonton, Alberta, Canada
| | - Banu Sis
- Department of Laboratory Medicine and Pathology and
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Colin C Anderson
- Department of Surgery.,Department of Medical Microbiology and Immunology, and
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Gavin Y Oudit
- Department of Medicine.,Mazankowski Heart Institute, Edmonton, Alberta, Canada
| | | |
Collapse
|
23
|
Kinins and nitric oxide in patients with chronic chagas disease and systemic arterial hypertension. Cardiovasc Pathol 2020; 49:107257. [PMID: 32674046 DOI: 10.1016/j.carpath.2020.107257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/31/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chronic Chagas disease (CCHD) associated with Systemic Arterial Hypertension (SAH) is frequently found in areas where the disease is endemic. The pathogenesis of patients with both pathologies (CCHD-SAH) is unsettled. Nitric Oxide (NO) and Kinins are important players in the myocardial inflammation process in experimental CCHD. No previous study has addressed this question in patients with CCHD, particularly in those with CCHD-SAH. Accordingly, this study was undertaken in an attempt to contribute to the understanding of the pathogenesis of patients with CCHD-SAH. METHODS Thirty-seven patients with a positive serology for Chagas disease were enrolled; 15 had CCHD alone, 22 had CCHD-SAH (abnormal ECG/Doppler echocardiogram plus a systolic blood pressure > 140 mmHg or diastolic blood pressure > 90 mmHg on admission), and 11 had SAH alone. Thirty healthy individuals matched by age and sex served as controls. Plasma High-molecular (Hkg) and low-molecular weight (LKg) kininogens, plasma kallikrein levels (Pkal and Tcal), Kininase II, and plasma NO were measured. RESULTS HKg and LKg were lower in CCHD-SAH patients in comparison with other groups (P < .0001). Pkal and Tcal were higher in CCHD-SAH patients in comparison with the other groups (P< .0001). Kininase II levels were similar in SAH, CCHD, and CCHD-SAH patients, but lower in comparison with controls (P< .0001). NO levels were similar in CCHD and CCHD-SAH patients, but higher in comparison with SAH patients and controls (P > .0001). CONCLUSION Such findings suggest increased Kinin and NO activity in patients with CCHD-SAH, thus contributing to the understanding of the pathogenesis of this condition.
Collapse
|
24
|
Therapeutic potential of miR-21 regulation by human peripheral blood derived-small extracellular vesicles in myocardial infarction. Clin Sci (Lond) 2020; 134:985-999. [DOI: 10.1042/cs20191077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 01/15/2023]
Abstract
Abstract
Small extracellular vesicles (sEVs) as natural membranous vesicles are on the frontiers of nanomedical research, due to their ability to deliver therapeutic molecules such as microRNAs (miRNAs). The miRNA-21 (miR-21) is thought to be involved in the initiation and development of myocardial infarction (MI). Here, we examined whether miR-21 regulation using human peripheral blood-derived sEVs (PB-sEVs) could serve as a potential therapeutic strategy for MI. First, we examined miR-21 levels in hypoxic conditions and validated the ability of PB-sEVs to serve as a potential delivery system for miRNAs. Further, bioinformatics analysis and luciferase assay were performed to identify target genes of miR-21 mechanistically. Among numerous target pathways, we focused on nitrogen metabolism, which remains relatively unexplored compared with other possible miR-21-mediated pathways; hence, we aimed to determine novel target genes of miR-21 related to nitrogen metabolism. In hypoxic conditions, the expression of miR-21 was significantly up-regulated and correlated with nitric oxide synthase 3 (NOS3) levels, which in turn influences cardiac function. The down-regulation of miR-21 expression by PB-sEVs loaded with anti-miR-21 significantly improved survival rates, consistent with the augmentation of cardiac function. However, the up-regulation of miR-21 expression by PB-sEVs loaded with miR-21 reversed these effects. Mechanistically, miR-21 targeted and down-regulated the mRNA and protein expression of striatin (STRN), which could regulate NOS3 expression. In conclusion, we identified a novel therapeutic strategy to improve cardiac function by regulating the expression of miR-21 with PB-sEVs as an miR-21 or anti-miR-21 delivery vehicle and confirmed the miR-21-associated nitrogen metabolic disorders in MI.
Collapse
|
25
|
Gao Z, Gao Q, Lv X. MicroRNA-668-3p Protects Against Oxygen-Glucose Deprivation in a Rat H9c2 Cardiomyocyte Model of Ischemia-Reperfusion Injury by Targeting the Stromal Cell-Derived Factor-1 (SDF-1)/CXCR4 Signaling Pathway. Med Sci Monit 2020; 26:e919601. [PMID: 31997826 PMCID: PMC7003666 DOI: 10.12659/msm.919601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background Ischemia-reperfusion injury (IRI) results from the restoration of blood supply to ischemic organs, including the heart. Expression of microRNA-668-3p (miR-668-3p) is known to protect the kidney from IRI. This study aimed to investigate the role of miR-668-3p in oxygen-glucose deprivation (OGD) in a rat H9c2 cardiomyocyte model of IRI. Material/Methods Rat H9c2 cardiomyocytes were cultured in glucose-free Dulbecco’s modified Eagle’s medium (DMEM) under anaerobic conditions, followed by oxygenation, to create the OGD model of IRI. The luciferase reporter assay evaluated the interaction between stromal cell-derived factor-1 (SDF-1), or CXC motif chemokine 12 (CXCL12), and miR-668-3p. Protein and mRNA levels of SDF-1, CXCR4, Bcl2, Bax, cleaved caspase-3, endothelial nitric oxide synthase (eNOS), and phosphorylated endothelial nitric oxide synthase (p-eNOS) were analyzed by Western blot and quantitative reverse transcription-polymerase chain reaction (RT-qPCR), and apoptosis were assessed by flow cytometry. Enzyme-linked immunosorbent assay (ELISA) measured reactive oxygen species (ROS), including malondialdehyde (MDA), nitric oxide (NO), p-eNOS, and the inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and monocyte chemoattractant protein-1 (MCP-1) in H9c2 cell supernatants. Results In the OGD rat H9c2 cardiomyocyte model of IRI, miR-668-3p levels were reduced. Overexpression of miR-668-3p inhibited SDF-1, CXCR4, the expression of inflammatory cytokines, markers of oxidative stress, and p-eNOS. The overexpression of SDF-1 reversed these findings. Overexpression of SDF-1 promoted cell apoptosis, which was reduced by miR-668-3p. Conclusions In the OGD rat H9c2 cardiomyocyte model of IRI, miR-668-3p suppressed mediators of inflammation and oxidative stress and enhanced cell viability through the SDF-1/CXCR4 signaling pathway.
Collapse
Affiliation(s)
- Zhan Gao
- Department of Cardiovascular Surgery, Children's Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Qiang Gao
- Department of Cardiovascular Surgery, Children's Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Xiaodong Lv
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland)
| |
Collapse
|
26
|
Yu B, Sladojevic N, Blair JE, Liao JK. Targeting Rho-associated coiled-coil forming protein kinase (ROCK) in cardiovascular fibrosis and stiffening. Expert Opin Ther Targets 2020; 24:47-62. [PMID: 31906742 PMCID: PMC7662835 DOI: 10.1080/14728222.2020.1712593] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 01/04/2020] [Indexed: 02/07/2023]
Abstract
Introduction: Pathological cardiac fibrosis, through excessive extracellular matrix protein deposition from fibroblasts and pro-fibrotic immune responses and vascular stiffening is associated with most forms of cardiovascular disease. Pathological cardiac fibrosis and stiffening can lead to heart failure and arrythmias and vascular stiffening may lead to hypertension. ROCK, a serine/threonine kinase downstream of the Rho-family of GTPases, may regulate many pro-fibrotic and pro-stiffening signaling pathways in numerous cell types.Areas covered: This article outlines the molecular mechanisms by which ROCK in fibroblasts, T helper cells, endothelial cells, vascular smooth muscle cells, and macrophages mediate fibrosis and stiffening. We speculate on how ROCK could be targeted to inhibit cardiovascular fibrosis and stiffening.Expert opinion: Critical gaps in knowledge must be addressed if ROCK inhibitors are to be used in the clinic. Numerous studies indicate that each ROCK isoform may play differential roles in regulating fibrosis and may have opposing roles in specific tissues. Future work needs to highlight the isoform- and tissue-specific contributions of ROCK in fibrosis, and how isoform-specific ROCK inhibitors in murine models and in clinical trials affect the pathophysiology of cardiac fibrosis and stiffening. This could progress knowledge regarding new treatments for heart failure, arrythmias and hypertension and the repair processes after myocardial infarction.
Collapse
Affiliation(s)
- Brian Yu
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nikola Sladojevic
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - John E Blair
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - James K Liao
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
27
|
Miguel V, Lamas S. Redox distress in organ fibrosis: The role of noncoding RNAs. OXIDATIVE STRESS 2020:779-820. [DOI: 10.1016/b978-0-12-818606-0.00037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
28
|
Loss of secretin results in systemic and pulmonary hypertension with cardiopulmonary pathologies in mice. Sci Rep 2019; 9:14211. [PMID: 31578376 PMCID: PMC6775067 DOI: 10.1038/s41598-019-50634-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
More than 1 billion people globally are suffering from hypertension, which is a long-term incurable medical condition that can further lead to dangerous complications and death if left untreated. In earlier studies, the brain-gut peptide secretin (SCT) was found to be able to control blood pressure by its cardiovascular and pulmonary effects. For example, serum SCT in patients with congestive heart failure was one-third of the normal level. These observations strongly suggest that SCT has a causal role in blood pressure control, and in this report, we used constitutive SCT knockout (SCT−/−) mice and control C57BL/6N mice to investigate differences in the morphology, function, underlying mechanisms and response to SCT treatment. We found that SCT−/− mice suffer from systemic and pulmonary hypertension with increased fibrosis in the lungs and heart. Small airway remodelling and pulmonary inflammation were also found in SCT−/− mice. Serum NO and VEGF levels were reduced and plasma aldosterone levels were increased in SCT−/− mice. Elevated cardiac aldosterone and decreased VEGF in the lungs were observed in the SCT−/− mice. More interestingly, SCT replacement in SCT−/− mice could prevent the development of heart and lung pathologies compared to the untreated group. Taken together, we comprehensively demonstrated the critical role of SCT in the cardiovascular and pulmonary systems and provide new insight into the potential role of SCT in the pathological development of cardiopulmonary and cardiovascular diseases.
Collapse
|
29
|
Magdaleno F, Blajszczak CC, Charles-Niño CL, Guadrón-Llanos AM, Vázquez-Álvarez AO, Miranda-Díaz AG, Nieto N, Islas-Carbajal MC, Rincón-Sánchez AR. Aminoguanidine reduces diabetes-associated cardiac fibrosis. Exp Ther Med 2019; 18:3125-3138. [PMID: 31572553 DOI: 10.3892/etm.2019.7921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
Aminoguanidine (AG) inhibits advanced glycation end products (AGEs) and advanced oxidation protein products (AOPP) accumulated as a result of excessive oxidative stress in diabetes. However, the molecular mechanism by which AG reduces AGE-associated damage in diabetes is not well understood. Thus, we investigated whether AG supplementation mitigates oxidative-associated cardiac fibrosis in rats with type 2 diabetes mellitus (T2DM). Forty-five male Wistar rats were divided into three groups: Control, T2DM and T2DM+AG. Rats were fed with a high-fat, high-carbohydrate diet (HFCD) for 2 weeks and rendered diabetic using low-dose streptozotocin (STZ) (20 mg/kg), and one group was treated with AG (20 mg/kg) up to 25 weeks. In vitro experiments were performed in primary rat myofibroblasts to confirm the antioxidant and antifibrotic effects of AG and to determine if blocking the receptor for AGEs (RAGE) prevents the fibrogenic response in myofibroblasts. Diabetic rats exhibited an increase in cardiac fibrosis resulting from HFCD and STZ injections. By contrast, AG treatment significantly reduced cardiac fibrosis, α-smooth muscle actin (αSMA) and oxidative-associated Nox4 and Nos2 mRNA expression. In vitro challenge of myofibroblasts with AG under T2DM conditions reduced intra- and extracellular collagen type I expression and Pdgfb, Tgfβ1 and Col1a1 mRNAs, albeit with similar expression of Tnfα and Il6 mRNAs. This was accompanied by reduced phosphorylation of ERK1/2 and SMAD2/3 but not of AKT1/2/3 and STAT pathways. RAGE blockade further attenuated collagen type I expression in AG-treated myofibroblasts. Thus, AG reduces oxidative stress-associated cardiac fibrosis by reducing pERK1/2, pSMAD2/3 and collagen type I expression via AGE/RAGE signaling in T2DM.
Collapse
Affiliation(s)
- Fernando Magdaleno
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico.,Department of Pathology, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | | | - Claudia Lisette Charles-Niño
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico.,Department of Microbiology and Pathology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Alma Marlene Guadrón-Llanos
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Alan Omar Vázquez-Álvarez
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Alejandra Guillermina Miranda-Díaz
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Natalia Nieto
- Department of Pathology, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - María Cristina Islas-Carbajal
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Ana Rosa Rincón-Sánchez
- Institute of Molecular Biology and Gene Therapy, Department of Molecular Biology and Genomics, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| |
Collapse
|
30
|
Saliba Y, Jebara V, Hajal J, Maroun R, Chacar S, Smayra V, Abramowitz J, Birnbaumer L, Farès N. Transient Receptor Potential Canonical 3 and Nuclear Factor of Activated T Cells C3 Signaling Pathway Critically Regulates Myocardial Fibrosis. Antioxid Redox Signal 2019; 30:1851-1879. [PMID: 30318928 PMCID: PMC6486676 DOI: 10.1089/ars.2018.7545] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Cardiac fibroblasts (CFs) are emerging as major contributors to myocardial fibrosis (MF), a final common pathway of many etiologies of heart disease. Here, we studied the functional relevance of transient receptor potential canonical 3 (TRPC3) channels and nuclear factor of activated T cells c3 (NFATc3) signaling in rodent and human ventricular CFs, and whether their modulation would limit MF. RESULTS A positive feedback loop between TRPC3 and NFATc3 drove a rat ventricular CF fibrotic phenotype. In these cells, polyphenols (extract of grape pomace polyphenol [P.E.]) decreased basal and angiotensin II-mediated Ca2+ entries through a direct modulation of TRPC3 channels and subsequently NFATc3 signaling, abrogating myofibroblast differentiation, fibrosis and inflammation, as well as an oxidative stress-associated phenotype. N(ω)-nitro-l-arginine methyl ester (l-NAME) hypertensive rats developed coronary perivascular, sub-epicardial, and interstitial fibrosis with induction of embryonic epicardial progenitor transcription factors in activated CFs. P.E. treatment reduced ventricular CF activation by modulating the TRPC3-NFATc3 pathway, and it ameliorated echocardiographic parameters, cardiac stress markers, and MF in l-NAME hypertensive rats independently of blood pressure regulation. Further, genetic deletion (TRPC3-/-) and pharmacological channel blockade with N-[4-[3,5-Bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-4-methyl-benzenesulfonamide (Pyr10) blunted ventricular CF activation and MF in l-NAME hypertensive mice. Finally, TRPC3 was present in human ventricular CFs and upregulated in MF, whereas pharmacological modulation of TRPC3-NFATc3 decreased proliferation and collagen secretion. Innovation and Conclusion: We demonstrate that TRPC3-NFATc3 signaling is modulated by P.E. and critically regulates ventricular CF phenotype and MF. These findings strongly argue for P.E., through TRPC3 targeting, as potential and interesting therapeutics for MF management.
Collapse
Affiliation(s)
- Youakim Saliba
- Laboratoire de Recherche en Physiologie et Physiopathologie, Pôle Technologie Santé, Faculté de Médecine, Université Saint Joseph, Beirut, Lebanon
| | - Victor Jebara
- Faculté de Médecine, Université Saint Joseph, Beirut, Lebanon
| | - Joelle Hajal
- Laboratoire de Recherche en Physiologie et Physiopathologie, Pôle Technologie Santé, Faculté de Médecine, Université Saint Joseph, Beirut, Lebanon
| | - Richard Maroun
- Unité de Recherche Technologie et Valorisation Alimentaire, Centre d'Analyses et de Recherche, Faculté des Sciences, Université Saint-Joseph, Beirut, Lebanon
| | - Stéphanie Chacar
- Laboratoire de Recherche en Physiologie et Physiopathologie, Pôle Technologie Santé, Faculté de Médecine, Université Saint Joseph, Beirut, Lebanon
- Unité de Recherche Technologie et Valorisation Alimentaire, Centre d'Analyses et de Recherche, Faculté des Sciences, Université Saint-Joseph, Beirut, Lebanon
| | - Viviane Smayra
- Faculté de Médecine, Université Saint Joseph, Beirut, Lebanon
| | - Joel Abramowitz
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina
- Institute of Biomedical Research (BIOMED), School of Medicine, Catholic University of Argentina, Buenos Aires, Argentina
| | - Nassim Farès
- Laboratoire de Recherche en Physiologie et Physiopathologie, Pôle Technologie Santé, Faculté de Médecine, Université Saint Joseph, Beirut, Lebanon
- Address correspondence to: Prof. Nassim Farès, Laboratoire de Physiologie et Physiopathologie, Pôle Technologie Santé, Faculté de Médecine, Université Saint Joseph, Damascus Street, BP 11-5076 - Riad El Solh, Beirut 1107 2180, Lebanon
| |
Collapse
|
31
|
Jackson EK, Mi E, Ritov VB, Gillespie DG. Extracellular Ubiquitin(1-76) and Ubiquitin(1-74) Regulate Cardiac Fibroblast Proliferation. Hypertension 2019; 72:909-917. [PMID: 30354710 DOI: 10.1161/hypertensionaha.118.11666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SDF-1α (stromal cell-derived factor-1α) is a CXCR4-receptor agonist and DPP4 (dipeptidyl peptidase 4) substrate. SDF-1α, particularly when combined with sitagliptin to block the metabolism of SDF-1α by DPP4, stimulates proliferation of cardiac fibroblasts via the CXCR4 receptor; this effect is greater in cells from spontaneously hypertensive rats versus Wistar-Kyoto normotensive rats. Emerging evidence indicates that ubiquitin(1-76) exists in plasma and is a potent CXCR4-receptor agonist. Therefore, we hypothesized that ubiquitin(1-76), similar to SDF-1α, should increase proliferation of cardiac fibroblasts. Contrary to our working hypothesis, ubiquitin(1-76) did not stimulate cardiac fibroblast proliferation, yet unexpectedly antagonized the proproliferative effects of SDF-1α combined with sitagliptin. In this regard, ubiquitin(1-76) was more potent in spontaneously hypertensive versus Wistar-Kyoto cells. In the presence of 6bk (selective inhibitor of insulin-degrading enzyme [IDE]; an enzyme known to convert ubiquitin(1-76) to ubiquitin(1-74)), ubiquitin(1-76) no longer antagonized the proproliferative effects of SDF-1α/sitagliptin. Ubiquitin(1-74) also antagonized the proproliferative effects of SDF-1α/sitagliptin, and this effect of ubiquitin(1-74) was not blocked by 6bk and was >10-fold more potent compared with ubiquitin(1-76). Neither ubiquitin(1-76) nor ubiquitin(1-74) inhibited the proproliferative effects of the non-CXCR4 receptor agonist neuropeptide Y (activates Y1 receptors). Cardiac fibroblasts expressed IDE mRNA, protein, and activity and converted ubiquitin(1-76) to ubiquitin(1-74). Spontaneously hypertensive fibroblasts expressed greater IDE activity. Extracellular ubiquitin(1-76) blocks the proproliferative effects of SDF-1α/sitagliptin via its conversion by IDE to ubiquitin(1-74), a potent CXCR4 antagonist. Thus, IDE inhibitors, particularly when combined with DPP4 inhibitors or hypertension, could increase the risk of cardiac fibrosis.
Collapse
Affiliation(s)
- Edwin K Jackson
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| | - Eric Mi
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| | - Vladimir B Ritov
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| | - Delbert G Gillespie
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| |
Collapse
|
32
|
Zhu ZD, Ye JM, Fu XM, Wang XC, Ye JY, Wu XR, Hua P, Liao YQ, Xuan W, Duan JL, Li WY, Fu H, Xia ZH, Zhang X. DDAH2 alleviates myocardial fibrosis in diabetic cardiomyopathy through activation of the DDAH/ADMA/NOS/NO pathway in rats. Int J Mol Med 2018; 43:749-760. [PMID: 30569164 PMCID: PMC6317674 DOI: 10.3892/ijmm.2018.4034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/27/2018] [Indexed: 01/02/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a form of idiopathic heart disease, with signs including hypertrophy of myocardial cells, hypertension‑independent fibrosis and coronary artery disease. Considering the involvement of dimethylarginine dimethylaminohydrolase 2 (DDAH2) in diabetes, it was hypothesized that DDAH2 may be beneficial to cardiac function and myocardial fibrosis during the progression of DCM with involvement of the DDAH/asymmetric NG, NGdimethyl‑L‑arginine (ADMA)/nitric oxide synthase (NOS)/nitric oxide (NO) signaling pathway. Following establishment of diabetic rat models, diabetes‑related blood biochemical indices and cardiac function were measured in diabetic rats treated with lentivirus expressing DDAH2, short hairpin RNA against DDAH2, or L‑NNA (inhibitor of NOS) to identify the roles of DDAH2 in DCM. The functional roles of DDAH2 in DCM were further determined through detection of the levels of collagen I, matrix metalloproteinase 2 (MMP2) and tissue inhibitor of metalloproteinase 2 (TIMP2). The H9C2 myocardial cell line was selected for in vitro experiments. The effects of DDAH2 on the migration of myocardial cells under high glucose conditions were also examined. To further investigate the underlying regulatory mechanism of DDAH2 in DCM, the contents of ADMA and NO, and the activities of DDAH and NOS were observed. The DCM model rats treated with DDAH2 exhibited reduced left ventricular end‑diastolic pressure, and decreased blood glucose, total cholesterol, triglyceride, fasting blood glucose, and fasting insulin levels, but exhibited increased left ventricular systolic pressure and maximum rate of left ventricular pressure rise/fall levels in myocardial tissues. Myocardial cells under high glucose conditions treated with DDAH2 showed reductions in collagen I, MMP2 and TIMP2, indicating that DDAH2 reduced cell migration. Decreased levels of ADMA and NO but increased levels of DDAH and NOS were observed following treatment with DDAH2, indicating that the DDAH/ADMA/NOS/NO pathway was activated. These results reveal that the overexpression of DDAH2 attenuates myocardial fibrosis and protects against DCM through activation of the DDAH/ADMA/NOS/NO pathway in DCM rats. These results indicate that DDAH2 is a potential therapeutic candidate for the treatment of DCM.
Collapse
Affiliation(s)
- Zhen-Dong Zhu
- Yunnan Research Center for Geriatric Diseases, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Ji-Ming Ye
- Department of Pharmacy, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Xue-Mei Fu
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Xue-Chang Wang
- Department of Pharmacy, the Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, Kunming, Yunnan 650011, P.R. China
| | - Ji-Yun Ye
- Pathogenic Organisms Department of Experimental Center, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Xin-Ran Wu
- Center Laboratory, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, Kunming, Yunnan 650011, P.R. China
| | - Peng Hua
- Department of Pharmacy, the Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, Kunming, Yunnan 650011, P.R. China
| | - Yu-Qiong Liao
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Wei Xuan
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Jin-Lan Duan
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Wei-Yuan Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Hui Fu
- Clinic Laboratory, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Zhong-Hua Xia
- Clinical Medical College of Dali University, Dali, Yunnan 671003, P.R. China
| | - Xuan Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
33
|
Chung CC, Lin YK, Chen YC, Kao YH, Yeh YH, Chen YJ. Factor Xa inhibition by rivaroxaban regulates fibrogenesis in human atrial fibroblasts with modulation of nitric oxide synthesis and calcium homeostasis. J Mol Cell Cardiol 2018; 123:128-138. [PMID: 30213724 DOI: 10.1016/j.yjmcc.2018.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/19/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Rivaroxaban, a widely used factor Xa inhibitor in reducing stroke in atrial fibrillation (AF) patients has multiple biological effects with activation of protease-activated receptor (PAR) signaling. Atrial fibrosis plays a critical role in the pathophysiology of AF. In this study, we evaluated whether rivaroxaban regulates atrial fibroblast activity and its underlying mechanisms. METHODS AND RESULTS Migration, proliferation analyses, nitric oxide (NO) production assay, calcium fluorescence imaging, and western blots were conducted in human atrial fibroblasts with or without rivaroxaban (100 nmol/L or 300 nmol/L) and co-administration of L-NAME (L-NG-nitro arginine methyl ester, 100 μmol/L), EGTA (Ethylene glycol tetra-acetic acid, 1 mmol/L), thrombin (0.5 U/mL), PAR1 agonist peptide (TFLLR-NH2, 100 μmol/L), PAR1 inhibitor (SCH79797, 0.5 μmol/L) and PAR2 inhibitor (GB83, 10 μmol/L). Atrial fibrosis was examined in isoproterenol (100 mg/kg, subcutaneous injection)-treated rats with or without rivaroxaban (10 mg/kg/day orally for 14 consecutive days). Rivaroxaban reduced the migration, pro-collagen type I production, and proliferation of atrial fibroblasts. Rivaroxaban decreased phosphorylated endothelial NO synthase (eNOS) (Thr 495, an inhibitory phosphorylated site of eNOS), and calcium (Ca2+) entry, and increased NO production. Moreover, L-NAME blocked the effects of rivaroxaban on fibroblast collagen and NO production. In the presence of EGTA, the migratory capability was similarly decreased in atrial fibroblasts with and without treatment with rivaroxaban (100 nmol/L), which suggests that rivaroxaban decreases migratory capability of atrial fibroblasts by inhibiting Ca2+ entry. Additionally, rivaroxaban significantly attenuated the effects of thrombin, and TFLLR-NH2 on migratory, proliferative, and pro-collagen type I production capability in atrial fibroblasts. SCH79797 or GB83 decreased pro-collagen type I production, migration, and proliferation capability in fibroblasts, but combined SCH79797 or GB83 with and without rivaroxaban had similar fibroblast activity. Moreover, rivaroxaban significantly decreased atrial fibrosis in isoproterenol-treated rats. CONCLUSIONS Rivaroxaban (100-300 nmol/L) regulates atrial fibroblast activity and atrial fibrosis by increasing NO production and decreasing Ca2+ entry through inhibition of PAR signaling.
Collapse
Affiliation(s)
- Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang Gung Memorial Hospital, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Jen Chen
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
34
|
Kazakov A, Hall RA, Werner C, Meier T, Trouvain A, Rodionycheva S, Nickel A, Lammert F, Maack C, Böhm M, Laufs U. Raf kinase inhibitor protein mediates myocardial fibrosis under conditions of enhanced myocardial oxidative stress. Basic Res Cardiol 2018; 113:42. [PMID: 30191336 PMCID: PMC6133069 DOI: 10.1007/s00395-018-0700-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/15/2018] [Indexed: 12/21/2022]
Abstract
Fibrosis is a hallmark of maladaptive cardiac remodelling. Here we report that genome-wide quantitative trait locus (QTL) analyses in recombinant inbred mouse lines of C57BL/6 J and DBA2/J strains identified Raf Kinase Inhibitor Protein (RKIP) as genetic marker of fibrosis progression. C57BL/6 N-RKIP−/− mice demonstrated diminished fibrosis induced by transverse aortic constriction (TAC) or CCl4 (carbon tetrachloride) treatment compared with wild-type controls. TAC-induced expression of collagen Iα2 mRNA, Ki67+ fibroblasts and marker of oxidative stress 8-hydroxyguanosine (8-dOHG)+ fibroblasts as well as the number of fibrocytes in the peripheral blood and bone marrow were markedly reduced in C57BL/6 N-RKIP−/− mice. RKIP-deficient cardiac fibroblasts demonstrated decreased migration and fibronectin production. This was accompanied by a two-fold increase of the nuclear accumulation of nuclear factor erythroid 2-related factor 2 (Nrf2), the main transcriptional activator of antioxidative proteins, and reduced expression of its inactivators. To test the importance of oxidative stress for this signaling, C57BL/6 J mice were studied. C57BL/6 J, but not the C57BL/6 N-strain, is protected from TAC-induced oxidative stress due to mutation of the nicotinamide nucleotide transhydrogenase gene (Nnt). After TAC surgery, the hearts of Nnt-deficient C57BL/6 J-RKIP−/− mice revealed diminished oxidative stress, increased left ventricular (LV) fibrosis and collagen Iα2 as well as enhanced basal nuclear expression of Nrf2. In human LV myocardium from both non-failing and failing hearts, RKIP-protein correlated negatively with the nuclear accumulation of Nrf2. In summary, under conditions of Nnt-dependent enhanced myocardial oxidative stress induced by TAC, RKIP plays a maladaptive role for fibrotic myocardial remodeling by suppressing the Nrf2-related beneficial effects.
Collapse
Affiliation(s)
- Andrey Kazakov
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany.
| | - Rabea A Hall
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie, Diabetologie und Ernährungsmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 77, 66421, Homburg, Germany
| | - Christian Werner
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - Timo Meier
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - André Trouvain
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - Svetlana Rodionycheva
- Klinik für Thorax- und Herz-Gefäßchirurgie, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 57, 66421, Homburg, Germany
| | - Alexander Nickel
- Deutsches Zentrum für Herzinsuffizienz, Universitätsklinikum Würzburg, am Schwarzenberg 15, A15, 97078, Würzburg, Germany
| | - Frank Lammert
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie, Diabetologie und Ernährungsmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 77, 66421, Homburg, Germany
| | - Christoph Maack
- Deutsches Zentrum für Herzinsuffizienz, Universitätsklinikum Würzburg, am Schwarzenberg 15, A15, 97078, Würzburg, Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstrasse 20, 04103, Leipzig, Germany
| |
Collapse
|
35
|
Kazakov A, Laufs U. Healthy and Unhealthy Cardiac Progenitor Cells Modify the Pathogenesis of Myocardial Diseases. Circ Res 2018; 119:10-2. [PMID: 27340265 DOI: 10.1161/circresaha.116.309012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Andrey Kazakov
- From the Klinik für Innere Medizin III Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes Homburg, Saarland, Germany
| | - Ulrich Laufs
- From the Klinik für Innere Medizin III Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes Homburg, Saarland, Germany
| |
Collapse
|
36
|
Zhao Y, Liu C, Liu J, Kong Q, Mao Y, Cheng H, Li N, Zhang X, Li C, Li Y, Liu L, Ding Z. HSPA12B promotes functional recovery after ischaemic stroke through an eNOS-dependent mechanism. J Cell Mol Med 2018; 22:2252-2262. [PMID: 29411514 PMCID: PMC5867065 DOI: 10.1111/jcmm.13507] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/23/2017] [Indexed: 02/01/2023] Open
Abstract
Stroke is the leading cause of disability worldwide. HSPA12B, a heat-shock protein recently identified expression specifically in endothelial cells, is able to promote angiogenesis. Here, we have investigated its effects on functional recovery at chronic phase of ischaemic stroke. Ischaemic stroke was induced by 60 min. of middle cerebral artery occlusion in transgenic mice with overexpression of HSPA12B (HSPA12B Tg) and wild-type littermates (WT). HSPA12B Tg mice demonstrated a significant higher survival rate than WT mice within 28 days post-stroke. Significant improved neurological functions, increased spontaneous locomotor activity and decreased anxiety were detected inHSPA12B Tg mice compared with WT controls within 21 days post-stroke. Stroke-induced hippocampal degeneration was attenuated in HSPA12B Tg mice examined at day 28 post-stroke. Interestingly, HSPA12B Tg mice showed enhanced peri-infarct angiogenesis (examined 28 days post-stroke) and hippocampal neurogenesis (examined 7 days post-stroke), respectively, compared to WT mice. The stroke-induced eNOS phosphorylation and TGF-β1 expression were augmented in HSPA12B Tg mice. However, administration with eNOS inhibitor L-NAME diminished the HSPA12B-induced protection in neurological functional recovery and mice survival post-stroke. The data suggest that HSPA12B promoted functional recovery and survival after stroke in an eNOS-dependent mechanism. Targeting HSPA12B expression may have a therapeutic potential for the stroke-evoked functional disability and mortality.
Collapse
Affiliation(s)
- Yanlin Zhao
- Department of GeriatricsJiangsu Provincial Key Laboratory of GeriatricsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Chang Liu
- Departments of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Jiali Liu
- Department of GeriatricsJiangsu Provincial Key Laboratory of GeriatricsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Qiuyue Kong
- Departments of AnesthesiologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Yu Mao
- Department of GeriatricsJiangsu Provincial Key Laboratory of GeriatricsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Hao Cheng
- Departments of AnesthesiologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Nan Li
- Departments of AnesthesiologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Xioajin Zhang
- Department of GeriatricsJiangsu Provincial Key Laboratory of GeriatricsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Chuanfu Li
- Departments of SurgeryEast Tennessee State UniversityJohnson CityTNUSA
| | - Yuehua Li
- Department of PathophysiologyNanjing Medical UniversityNanjingChina
- Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingChina
| | - Li Liu
- Department of GeriatricsJiangsu Provincial Key Laboratory of GeriatricsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjingChina
| | - Zhengnian Ding
- Departments of AnesthesiologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| |
Collapse
|
37
|
Florijn BW, Bijkerk R, van der Veer EP, van Zonneveld AJ. Gender and cardiovascular disease: are sex-biased microRNA networks a driving force behind heart failure with preserved ejection fraction in women? Cardiovasc Res 2017; 114:210-225. [DOI: 10.1093/cvr/cvx223] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/23/2017] [Indexed: 01/08/2023] Open
Abstract
AbstractCardiovascular disease (CVD) is the primary cause of death among men and women worldwide. Nevertheless, our comprehension of how CVD progresses in women and elicits clinical outcomes is lacking, leading CVD to be under-diagnosed and under-treated in women. A clear example of this differential presentation of CVD pathophysiologies in females is the strikingly higher prevalence of heart failure with preserved ejection fraction (HFpEF). Women with a history of pre-eclampsia or those who present with co-morbidities such as obesity, hypertension, and diabetes mellitus are at increased risk of developing HFpEF. Long understood to be a critical CVD risk factor, our understanding of how gender differentially affects the development of CVD has been greatly expanded by extensive genomic and transcriptomic studies. These studies uncovered a pivotal role for differential microRNA (miRNA) expression in response to systemic inflammation, where their co-ordinated expression forms a post-transcriptional regulatory network that instigates microcirculation defects. Importantly, the potential sex-biased expression of the given miRNAs may explain sex-specific cardiovascular pathophysiologies in women, such as HFpEF. Sex-biased miRNAs are regulated by oestrogen (E2) in their transcription and processing or are expressed from loci on the X-chromosome due to incomplete X-chromosome inactivation. Interestingly, while E2-induced miRNAs predominantly appear to serve protective functions, it could be argued that many X-linked miRNAs have been found to challenge microvascular and myocardial integrity. Therefore, menopausal E2 deficiency, resulting in protective miRNA loss, and the augmentation of X-linked miRNA expression, may well contribute to the molecular mechanisms that underlie the female-specific cardiovascular aetiology in HFpEF.
Collapse
Affiliation(s)
- Barend W Florijn
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Roel Bijkerk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| |
Collapse
|
38
|
Jackson EK, Zhang Y, Gillespie DD, Zhu X, Cheng D, Jackson TC. SDF-1α (Stromal Cell-Derived Factor 1α) Induces Cardiac Fibroblasts, Renal Microvascular Smooth Muscle Cells, and Glomerular Mesangial Cells to Proliferate, Cause Hypertrophy, and Produce Collagen. J Am Heart Assoc 2017; 6:JAHA.117.007253. [PMID: 29114002 PMCID: PMC5721794 DOI: 10.1161/jaha.117.007253] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Activated cardiac fibroblasts (CFs), preglomerular vascular smooth muscle cells (PGVSMCs), and glomerular mesangial cells (GMCs) proliferate, cause hypertrophy, and produce collagen; in this way, activated CFs contribute to cardiac fibrosis, and activated PGVSMCs and GMCs promote renal fibrosis. In heart and kidney diseases, SDF-1α (stromal cell-derived factor 1α; endogenous CXCR4 [C-X-C motif chemokine receptor 4] receptor agonist) levels are often elevated; therefore, it is important to know whether and how the SDF-1α/CXCR4 axis activates CFs, PGVSMCs, or GMCs. METHODS AND RESULTS Here we investigated whether SDF-1α activates CFs, PGVSMCs, and GMCs to proliferate, hypertrophy, or produce collagen. DPP4 (dipeptidyl peptidase 4) inactivates SDF-1α and previous experiments show that growth-promoting peptides have greater effects in cells from genetically-hypertensive animals. Therefore, we performed experiments in the absence and presence of sitagliptin (DPP4 inhibitor) and in cells from normotensive Wistar-Kyoto rats and spontaneously hypertensive rats. Our studies show (1) that spontaneously hypertensive and Wistar-Kyoto rat CFs, PGVSMCs, and GMCs express CXCR4 receptors and DPP4 activity; (2) that chronic treatment with physiologically relevant concentrations of SDF-1α causes concentration-dependent increases in the proliferation (cell number) and hypertrophy (3H-leucine incorporation) of and collagen production (3H-proline incorporation) by CFs, PGVSMCs, and GMCs; (3) that sitagliptin augments these effects of SDF-1α; (4) that interactions between SDF-1α and sitagliptin are greater in spontaneously hypertensive rat cells; (5) that CXCR4 antagonism (AMD3100) blocks all effects of SDF-1α; and (6) that SDF-1α/CXCR4 signal transduction likely involves the RACK1 (receptor for activated C kinase 1)/Gβγ/PLC (phospholipase C)/PKC (protein kinase C) signaling complex. CONCLUSIONS The SDF-1α/CXCR4 axis drives proliferation and hypertrophy of and collagen production by CFs, PGVSMCs, and GMCs, particularly in cells from genetically hypertensive animals and when DPP4 is inhibited.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Yumeng Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Delbert D Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xiao Zhu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Travis C Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
39
|
Xu B, Bobek G, Makris A, Hennessy A. Antihypertensive methyldopa, labetalol, hydralazine, and clonidine reversed tumour necrosis factor-α inhibited endothelial nitric oxide synthase expression in endothelial-trophoblast cellular networks. Clin Exp Pharmacol Physiol 2017; 44:421-427. [PMID: 27998008 DOI: 10.1111/1440-1681.12712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 12/08/2016] [Accepted: 12/11/2016] [Indexed: 11/29/2022]
Abstract
Medications used to control hypertension in pregnancy also improve trophoblast and endothelial cellular interaction in vitro. Tumour necrosis factor-α (TNF-α) inhibits trophoblast and endothelial cellular interactions and simultaneously decreases endothelial nitric oxide synthase (eNOS) expression. This study investigated whether antihypertensive medications improved these cellular interactions by modulating eNOS and inducible nitric oxide synthase (iNOS) expression. Human uterine myometrial microvascular endothelial cells (UtMVECs) were pre-incubated with (or without) low dose TNF-α (0.5 ng/mL) or TNF-α plus soluble fms-like tyrosine kinase-1 (sFlt-1) (100 ng/mL). The endothelial cells were cultured on Matrigel. After endothelial cellular networks appeared, trophoblast derived HTR-8/SVneo cells were co-cultured in the presence of clinically relevant doses of methyldopa, labetalol, hydralazine or clonidine for 24 hours. Cells were retrieved from the Matrigel to extract mRNA and eNOS and iNOS expression were examined by quantitative PCR. Methyldopa, labetalol, hydralazine and clonidine reversed the inhibitory effect of TNF-α on eNOS mRNA expression. After pre-incubating endothelial cells with TNF-α and sFlt-1, all the medications except methyldopa lost their effect on eNOS mRNA expression. In the absence of TNF-α, antihypertensive medications did not change eNOS expression. The mRNA expression of iNOS was not affected by TNF-α or any medications. This study shows that selected antihypertensive medications used in the treatment of hypertension in pregnancy increase eNOS expression in vitro when induced by the inflammatory TNF-α. The anti-angiogenic molecule sFlt-1 may antagonise the potential benefit of these medications by interfering with the NOS pathway.
Collapse
Affiliation(s)
- Bei Xu
- School of Medicine, Western Sydney University, Sydney, NSW, Australia.,Vascular Immunology Research Laboratory, The Heart Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Gabriele Bobek
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Angela Makris
- School of Medicine, Western Sydney University, Sydney, NSW, Australia.,Vascular Immunology Research Laboratory, The Heart Research Institute, University of Sydney, Sydney, NSW, Australia.,Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
| | - Annemarie Hennessy
- School of Medicine, Western Sydney University, Sydney, NSW, Australia.,Vascular Immunology Research Laboratory, The Heart Research Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
40
|
Polonchuk L, Chabria M, Badi L, Hoflack JC, Figtree G, Davies MJ, Gentile C. Cardiac spheroids as promising in vitro models to study the human heart microenvironment. Sci Rep 2017; 7:7005. [PMID: 28765558 PMCID: PMC5539326 DOI: 10.1038/s41598-017-06385-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional in vitro cell systems are a promising alternative to animals to study cardiac biology and disease. We have generated three-dimensional in vitro models of the human heart ("cardiac spheroids", CSs) by co-culturing human primary or iPSC-derived cardiomyocytes, endothelial cells and fibroblasts at ratios approximating those present in vivo. The cellular organisation, extracellular matrix and microvascular network mimic human heart tissue. These spheroids have been employed to investigate the dose-limiting cardiotoxicity of the common anti-cancer drug doxorubicin. Viability/cytotoxicity assays indicate dose-dependent cytotoxic effects, which are inhibited by the nitric oxide synthase (NOS) inhibitor L-NIO, and genetic inhibition of endothelial NOS, implicating peroxynitrous acid as a key damaging agent. These data indicate that CSs mimic important features of human heart morphology, biochemistry and pharmacology in vitro, offering a promising alternative to animals and standard cell cultures with regard to mechanistic insights and prediction of toxic effects in human heart tissue.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Mamta Chabria
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Laura Badi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Jean-Christophe Hoflack
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Gemma Figtree
- Sydney Medical School, University of Sydney, Sydney, 2000, Australia
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Carmine Gentile
- Sydney Medical School, University of Sydney, Sydney, 2000, Australia.
- Heart Research Institute, Newtown, 2041, Australia.
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
41
|
Angiotensin II dependent cardiac remodeling in the eel Anguilla anguilla involves the NOS/NO system. Nitric Oxide 2017; 65:50-59. [DOI: 10.1016/j.niox.2017.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/08/2017] [Accepted: 02/13/2017] [Indexed: 11/19/2022]
|
42
|
Lin RJ, Su ZZ, Liang SM, Chen YY, Shu XR, Nie RQ, Wang JF, Xie SL. Role of Circulating Fibrocytes in Cardiac Fibrosis. Chin Med J (Engl) 2017; 129:326-31. [PMID: 26831236 PMCID: PMC4799578 DOI: 10.4103/0366-6999.174503] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE It is revealed that circulating fibrocytes are elevated in patients/animals with cardiac fibrosis, and this review aims to provide an introduction to circulating fibrocytes and their role in cardiac fibrosis. DATA SOURCES This review is based on the data from 1994 to present obtained from PubMed. The search terms were "circulating fibrocytes " and "cardiac fibrosis ". STUDY SELECTION Articles and critical reviews, which are related to circulating fibrocytes and cardiac fibrosis, were selected. RESULTS Circulating fibrocytes, which are derived from hematopoietic stem cells, represent a subset of peripheral blood mononuclear cells exhibiting mixed morphological and molecular characteristics of hematopoietic and mesenchymal cells (CD34+/CD45+/collagen I+). They can produce extracellular matrix and many cytokines. It is shown that circulating fibrocytes participate in many fibrotic diseases, including cardiac fibrosis. Evidence accumulated in recent years shows that aging individuals and patients with hypertension, heart failure, coronary heart disease, and atrial fibrillation have more circulating fibrocytes in peripheral blood and/or heart tissue, and this elevation of circulating fibrocytes is correlated with the degree of fibrosis in the hearts. CONCLUSIONS Circulating fibrocytes are effector cells in cardiac fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuang-Lun Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong 510120, China
| |
Collapse
|
43
|
Wang Q, Zhang Y, Li D, Zhang Y, Tang B, Li G, Yang Y, Yang D. Transgenic overexpression of transient receptor potential vanilloid subtype 1 attenuates isoproterenol-induced myocardial fibrosis in mice. Int J Mol Med 2016; 38:601-9. [PMID: 27314441 DOI: 10.3892/ijmm.2016.2648] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 06/08/2016] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential vanilloid subtype 1 (TRPV1) is a non-selective cation channel with high permeability to Ca2+. Intracellular Ca2+ signaling is an essential regulator of endothelial nitric oxide (NO) synthase (eNOS) that plays a beneficial role in myocardial fibrosis. The aim of the present study was to determine the role of TRPV1 in isoproterenol-induced myocardial fibrosis. Transgenic mice overexpressing TRPV1 were generated on a C57BL/6J genetic background. An animal model of myocardial fibrosis was created by subcutaneously injecting the mice with isoproterenol. We found that the wild-type mice exhibited a significant increase in heart/body weight ratio, left ventricle/body weight ratio, left ventricular end-diastolic pressure (LVEDP), the cardiac fibrotic lesion area and collagen content, as well as a marked decrease in eNOS phosphorylation and NO/cyclic guanosine monophosphate (cGMP) levels at 2 weeks after the administration of isoproterenol (all p<0.01). However, these changes were significantly attenuated in the TRPV1 transgenic mice (p<0.05 or p<0.01). Moreover, the beneficial effects on myocardial fibrosis exerted by the overexpression of TRPV1 were attenuated by the administration of the eNOS inhibitor, Nω-nitro-L-arginine methyl ester (L-NAME) (all p<0.05). Similar anti-fibrotic effects were observed in in vitro experiments with primary cultured cardiac fibroblasts. The findings of our study suggest that TRPV1 overexpression attenuates isoproterenol‑induced myocardial fibrosis.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - Yunrong Zhang
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - De Li
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - Yan Zhang
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - Bing Tang
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - Gang Li
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - Yongjian Yang
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| | - Dachun Yang
- Department of Cardiology, Chengdu Military General Hospital, Jinniu, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
44
|
Cui L, Wang Y, Yu R, Li B, Xie S, Gao Y, Wang X, Zhu M. Jia-Shen decoction-medicated serum inhibits angiotensin-II induced cardiac fibroblast proliferation via the TGF-β1/Smad signaling pathway. Mol Med Rep 2016; 14:1610-6. [PMID: 27315199 PMCID: PMC4940101 DOI: 10.3892/mmr.2016.5405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 05/27/2016] [Indexed: 11/05/2022] Open
Abstract
Jia-Shen decoction (JSD) is a traditional Chinese medicine, which is used widely to treat chronic heart failure. However, the underlying mechanism remains to be elucidated. The present study aimed to investigate the mechanism underlying the effects of JSD on cardiac fibroblast (CF) proliferation and differentiation. The CFs were obtained from the hearts of neonatal (1‑3‑day old) Sprague‑Dawley rats and treated with JSD-medicated serum (JSDS) with or without angiotensin II (Ang II). Cell proliferation was assessed using Cell Counting Kit‑8 reagent. In addition, the mRNA expression levels of transforming growth factor‑β1 (TGF‑β1) and phosphorylated small mothers against decapentaplegic (p‑Smad)2/3 and their protein expression levels were analyzed. CF proliferation was significantly increased in the Ang II‑treated group, compared with the control group (P<0.05). The expression levels of collagen, α‑smooth muscle actin, TGF‑β1 and p‑Smad2/3 were also increased in the Ang II‑treated group (P<0.05). Following JSDS treatment, the increased levels of collagen and cell proliferation were inhibited, and the increased expression levels of p‑Smad2 and p‑Smad3 were also inhibited (P<0.05). These data suggested that JSDS may inhibit CF proliferation via attenuating the TGF‑β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Lin Cui
- Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Youping Wang
- Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Rui Yu
- Department of Internal Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Bin Li
- Division of Cardiology, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Shiyang Xie
- Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Yuan Gao
- Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Xiaoxiao Wang
- Central Laboratory, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Mingjun Zhu
- Division of Cardiology, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
45
|
Jose Corbalan J, Vatner DE, Vatner SF. Myocardial apoptosis in heart disease: does the emperor have clothes? Basic Res Cardiol 2016; 111:31. [PMID: 27043720 DOI: 10.1007/s00395-016-0549-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/24/2016] [Indexed: 01/06/2023]
Abstract
Since the discovery of a novel mechanism of cell death that differs from traditional necrosis, i.e., apoptosis, there have been numerous studies concluding that increased apoptosis augments myocardial infarction and heart failure and that limiting apoptosis protects the heart. Importantly, the vast majority of cells in the heart are non-myocytes with only roughly 30 % myocytes, yet almost the entire field studying apoptosis in the heart has disregarded non-myocyte apoptosis, e.g., only 4.7 % of 423 studies on myocardial apoptosis in the past 3 years quantified non-myocyte apoptosis. Accordingly, we reviewed the history of apoptosis in the heart focusing first on myocyte apoptosis, followed by the history of non-myocyte apoptosis in myocardial infarction and heart failure. Apoptosis of several of the major non-myocyte cell types in the heart (cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, macrophages and leukocytes) may actually be responsible for affecting the severity of myocardial infarction and heart failure. In summary, even though it is now known that the majority of apoptosis in the heart occurs in non-myocytes, very little work has been done to elucidate the mechanisms by which non-myocyte apoptosis might be responsible for the adverse effects of apoptosis in myocardial infarction and heart failure. The goal of this review is to provide an impetus for future work in this field on non-myocyte apoptosis that will be required for a better understanding of the role of apoptosis in the heart.
Collapse
Affiliation(s)
- J Jose Corbalan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
46
|
C-kit(+) resident cardiac stem cells improve left ventricular fibrosis in pressure overload. Stem Cell Res 2015; 15:700-711. [PMID: 26587804 DOI: 10.1016/j.scr.2015.10.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/16/2015] [Accepted: 10/26/2015] [Indexed: 11/23/2022] Open
Abstract
To investigate the effect of resident cardiac stem cells (RCSC) on myocardial remodeling, c-kit(+) RCSC were isolated from hearts of C57Bl/6-Tg (ACTb-EGFP)1Osb/J mice expressing green fluorescent protein and expanded in vitro. C57/Bl6N wildtype mice were subjected to transverse aortic constriction (TAC, 360 μm) or sham-operation. 5 × 10(5) c-kit(+) RCSC or c-kit(-) cardiac cells or cell buffer were infused intravenously 24 h post-surgery (n = 11-24 per group). Hypoxia-inducible factor-1α-mRNA in left ventricles of TAC mice was enhanced 24 h after transplantation. 35 days post-TAC, the density of c-kit(+) RCSC in the myocardium was increased by two-fold. Infusion of c-kit(+) resident cardiac stem cells post-TAC markedly reduced myocardial fibrosis and the expression of collagen Iα2 and connective tissue growth factor. Infusion of c-kit(-) cardiac cells did not ameliorate cardiac fibrosis. In parallel, expression of pro-angiogenic mediators (FGFb, IL-4, IL-6, TGFß, leptin) and the density of CD31(+) and CD31(+) GFP(+) endothelial cells were increased. Transplantation reduced brain- and atrial natriuretic peptides and the cardiomyocyte cross-sectional area. Infusion of c-kit(+) resident cardiac stem reduced the rate of apoptosis and oxidative stress in cardiomyocytes and in non-cardiomyocyte cells.
Collapse
|
47
|
Campbell KA, Li X, Biendarra SM, Terzic A, Nelson TJ. Nos3-/- iPSCs model concordant signatures of in utero cardiac pathogenesis. J Mol Cell Cardiol 2015; 87:228-36. [PMID: 26344701 DOI: 10.1016/j.yjmcc.2015.08.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Through genome-wide transcriptional comparisons, this study interrogates the capacity of in vitro differentiation of induced pluripotent stem cells (iPSCs) to accurately model pathogenic signatures of developmental cardiac defects. METHODS AND RESULTS Herein, we studied the molecular etiology of cardiac defects in Nos3(-/-) mice via transcriptional analysis of stage-matched embryonic tissues and iPSC-derived cells. In vitro comparisons of differentiated cells were calibrated to in utero benchmarks of health and disease. Integrated systems biology analysis of WT and Nos3(-/-) transcriptional profiles revealed 50% concordant expression patterns between in utero embryonic tissues and ex vivo iPSC-derived cells. In particular, up-regulation of glucose metabolism (p-value=3.95×10(-12)) and down-regulation of fatty acid metabolism (p-value=6.71×10(-12)) highlight a bioenergetic signature of early Nos3 deficiency during cardiogenesis that can be recapitulated in iPSC-derived differentiated cells. CONCLUSIONS The in vitro concordance of early Nos3(-/-) disease signatures supports the utility of iPSCs as a cellular model of developmental heart defects. Moreover, this study supports the use of iPSCs as a platform to pinpoint initial stages of congenital cardiac pathogenesis.
Collapse
Affiliation(s)
- Katherine A Campbell
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Regenerative Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of General Internal Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Xing Li
- Department of Health Sciences Research, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Biomedical Statistics and Informatics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Sherri M Biendarra
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Terzic
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Regenerative Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Cardiovascular Diseases, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Department of Medical Genetics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy J Nelson
- Department of Molecular Pharmacology and Experimental Therapeutics, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Regenerative Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Cardiovascular Diseases, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of General Internal Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Center for Transplantation and Clinical Regeneration, 200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA; Division of Pediatric Cardiology,200 First Street SW, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
48
|
Bai X, Qi Z, Song G, Zhao X, Zhao H, Meng X, Liu C, Bing W, Bi Y. Effects of Monocyte Chemotactic Protein-1 and Nuclear Factor of Kappa B Pathway in Rejection of Cardiac Allograft in Rat. Transplant Proc 2015; 47:2010-6. [DOI: 10.1016/j.transproceed.2015.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/26/2015] [Accepted: 05/14/2015] [Indexed: 01/04/2023]
|
49
|
Yamamoto T, Shimano M, Inden Y, Takefuji M, Yanagisawa S, Yoshida N, Tsuji Y, Hirai M, Murohara T. Alogliptin, a dipeptidyl peptidase-4 inhibitor, regulates the atrial arrhythmogenic substrate in rabbits. Heart Rhythm 2015; 12:1362-9. [DOI: 10.1016/j.hrthm.2015.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 12/27/2022]
|
50
|
Puhl SL, Müller A, Wagner M, Devaux Y, Böhm M, Wagner DR, Maack C. Exercise attenuates inflammation and limits scar thinning after myocardial infarction in mice. Am J Physiol Heart Circ Physiol 2015; 309:H345-59. [PMID: 26001415 DOI: 10.1152/ajpheart.00683.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 05/10/2015] [Indexed: 12/16/2022]
Abstract
Although exercise mediates beneficial effects in patients after myocardial infarction (MI), the underlying mechanisms as well as the question of whether an early start of exercise after MI is safe or even beneficial are incompletely resolved. The present study analyzed the effects of exercise before and reinitiated early after MI on cardiac remodeling and function. Male C57BL/6N mice were housed sedentary or with the opportunity to voluntarily exercise for 6 wk before MI induction (ligation of the left anterior descending coronary artery) or sham operation. After a 5-day exercise-free phase after MI, mice were allowed to reexercise for another 4 wk. Exercise before MI induced adaptive hypertrophy with moderate increases in heart weight, cardiomyocyte diameter, and left ventricular (LV) end-diastolic volume, but without fibrosis. In sedentary mice, MI induced eccentric LV hypertrophy with massive fibrosis but maintained systolic LV function. While in exercised mice gross LV end-diastolic volumes and systolic function did not differ from sedentary mice after MI, LV collagen content and thinning of the infarcted area were reduced. This was associated with ameliorated activation of inflammation, mediated by TNF-α, IL-1β, and IL-6, as well as reduced activation of matrix metalloproteinase 9. In contrast, no differences in the activation patterns of various MAPKs or adenosine receptor expressions were observed 5 wk after MI in sedentary or exercised mice. In conclusion, continuous exercise training before and with an early reonset after MI ameliorates adverse LV remodeling by attenuating inflammation, fibrosis, and scar thinning. Therefore, an early reonset of exercise after MI can be encouraged.
Collapse
Affiliation(s)
- Sarah-Lena Puhl
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; and
| | - Andreas Müller
- Klinik für Interventionelle Radiologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Michael Wagner
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; and
| | - Yvan Devaux
- Laboratory of Cardiovascular Research, Centre de Recherche Public-Santé, Luxembourg; and
| | - Michael Böhm
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; and
| | - Daniel R Wagner
- Division of Cardiology, Centre Hospitalier Luxembourg, Luxembourg
| | - Christoph Maack
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; and
| |
Collapse
|