51
|
Li C, Ying S, Wu X, Zhu T, Zhou Q, Zhang Y, Liu Y, Zhu R, Hu H. CTRP1 Aggravates Cardiac Fibrosis by Regulating The NOX2/P38 Pathway in Macrophages. CELL JOURNAL 2022; 24:732-740. [PMID: 36527345 PMCID: PMC9790075 DOI: 10.22074/cellj.2022.557327.1043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE C1q/TNF-related proteins 1 (CTRP1) is a recently identified adiponectin associated with obesity-linked disorders and adverse cardiovascular events. The effect of CTRP1 on cardiac fibrosis has not yet been fully elucidated; thus, we aimed to explore this association. MATERIALS AND METHODS In this experimental study, a mouse model of cardiac fibrosis was established by administering isoproterenol (ISO) (subcutaneously injecting 10 mg/kg/day for 3 days and then 5 mg/kg/day for 11 days). Mice were also injected with recombinant CTRP1 protein (200 μg/kg) 14 days after the final ISO administration. Adult mouse fibroblasts were isolated and stimulated with transforming growth factor (TGF) β1, followed by treatment with recombinant CTRP1. Primary bone marrow-derived macrophages were isolated from C57BL/6J mice and treated with recombinant CTRP1 as well. RESULTS CTRP1 level was increased in mouse plasma and heart tissue 2 weeks after ISO injection. Our findings indicated that recombinant CTRP1 injection aggravated ISO-induced cardiac fibrosis and dysfunction. However, recombinant CTRP1 did not alter TGFβ1-induced fibroblast proliferation and activation or collagen transcription. Recombinant CTRP1 exacerbated ISO-induced macrophage infiltration and inflammatory response. We determined that macrophages treated with recombinant CTRP1 showed increased pro-inflammatory cytokine release. Fibroblasts co-cultured with macrophages treated with recombinant CTRP1 showed increased proliferation and collagen transcription. We also found that CTRP1 upregulated the NADPH oxidase 2 (NOX2)/p38 pathway in macrophages. When we inhibited p38 signaling, the pro-inflammatory effect of CTRP1 on macrophages was counteracted. Fibroblasts co-cultured with macrophages treated with a p38 inhibitor also showed limited proliferation and collagen transcription. CONCLUSION Cardiac fibrosis was aggravated with the activation of the NOX2/p38 pathway in macrophages after CTRP1 treatment.
Collapse
Affiliation(s)
- Chenyu Li
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - Shaozhen Ying
- Department of Cardiology, Jiangxi provincial People’s Hospital, Affiliated to Nanchang University, Nanchang, Jiangxi, China
| | - Xiaolin Wu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - Tongjian Zhu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - Qing Zhou
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - Yue Zhang
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - Yongsheng Liu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - Rui Zhu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China
| | - He Hu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei,
P.R. China,Department of CardiologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and
ScienceXiangyangHubeiP.R. China
| |
Collapse
|
52
|
Liu J, Zhang R, Wang D, Lin Y, Bai C, Nie N, Gao S, Zhang Q, Chang H, Ren C. Elucidating the role of circNFIB in myocardial fibrosis alleviation by endogenous sulfur dioxide. BMC Cardiovasc Disord 2022; 22:492. [PMID: 36404310 PMCID: PMC9677687 DOI: 10.1186/s12872-022-02909-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/21/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND To investigate the role of circNFIB in the alleviation of myocardial fibrosis by endogenous sulfur dioxide (SO2). METHODS We stimulated cultured neonatal rat cardiac fibroblasts with transforming growth factor-β1 (TGF-β1) and developed an in vitro myocardial fibrosis model. Lentivirus vectors containing aspartate aminotransferase 1 (AAT1) cDNA were used to overexpress AAT1, and siRNA was used to silence circNFIB. The SO2, collagen, circNFIB, Wnt/β-catenin, and p38 MAPK pathways were examined in each group. RESULTS In the in vitro TGF-β1-induced myocardial fibrosis model, endogenous SO2/AAT1 expression was significantly decreased, and collagen levels in the cell supernatant and type I and III collagen expression, as well as α-SMA expression, were all significantly increased. TGF-β1 also significantly reduced circNFIB expression. AAT1 overexpression significantly reduced myocardial fibrosis while significantly increasing circNFIB expression. Endogenous SO2 alleviated myocardial fibrosis after circNFIB expression was blocked. We discovered that circNFIB plays an important role in the alleviation of myocardial fibrosis by endogenous SO2 by inhibiting the Wnt/β-catenin and p38 MAPK pathways. CONCLUSION Endogenous SO2 promotes circNFIB expression, which inhibits the Wnt/β-catenin and p38 MAPK signaling pathways, consequently alleviating myocardial fibrosis.
Collapse
Affiliation(s)
- Jia Liu
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ranran Zhang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dahai Wang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi Lin
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Bai
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nana Nie
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shan Gao
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiuye Zhang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Chang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chongmin Ren
- grid.412521.10000 0004 1769 1119Department of orthopedic oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
53
|
Antar SA, Saleh MA, Al-Karmalawy AA. Investigating the possible mechanisms of pirfenidone to be targeted as a promising anti-inflammatory, anti-fibrotic, anti-oxidant, anti-apoptotic, anti-tumor, and/or anti-SARS-CoV-2. Life Sci 2022; 309:121048. [PMID: 36209833 PMCID: PMC9536875 DOI: 10.1016/j.lfs.2022.121048] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/23/2022] [Accepted: 10/02/2022] [Indexed: 01/10/2023]
Abstract
Pirfenidone (PFD) is a non-peptide synthetic chemical that inhibits the production of transforming growth factor-beta 1 (TGF-β1), tumor necrosis factor-alpha (TNF-α), platelet-derived growth factor (PDGF), Interleukin 1 beta (IL-1β), and collagen 1 (COL1A1), all of which have been linked to the prevention or removal of excessive scar tissue deposition in many organs. PFD has been demonstrated to decrease apoptosis, downregulate angiotensin-converting enzyme (ACE) receptor expression, reduce inflammation through many routes, and alleviate oxidative stress in pneumocytes and other cells while protecting them from COVID-19 invasion and cytokine storm. Based on the mechanism of action of PFD and the known pathophysiology of COVID-19, it was recommended to treat COVID-19 patients. The use of PFD as a treatment for a range of disorders is currently being studied, with an emphasis on outcomes related to reduced inflammation and fibrogenesis. As a result, rather than exploring the molecule's chemical characteristics, this review focuses on innovative PFD efficacy data. Briefly, herein we tried to investigate, discuss, and illustrate the possible mechanisms of actions for PFD to be targeted as a promising anti-inflammatory, anti-fibrotic, anti-oxidant, anti-apoptotic, anti-tumor, and/or anti-SARS-CoV-2 candidate.
Collapse
Affiliation(s)
- Samar A Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.
| | - Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, the United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| |
Collapse
|
54
|
Wang X, Guo Y, Gao Y, Ren C, Huang Z, Liu B, Li X, Chang L, Shen K, Ding H, Zhang H, Tian Z, Hacker M, Zhang S, Wang Y, Li J, Li X, Huo L. Feasibility of 68Ga-Labeled Fibroblast Activation Protein Inhibitor PET/CT in Light-Chain Cardiac Amyloidosis. JACC Cardiovasc Imaging 2022; 15:1960-1970. [PMID: 36357138 DOI: 10.1016/j.jcmg.2022.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/23/2022] [Accepted: 06/16/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Systemic amyloid light chain (AL) amyloidosis is the most common type of amyloidosis, leading to cardiomyocyte necrosis and interstitial fibrosis. Gallium-68-labeled fibroblast activation protein inhibitor 04 (68Ga-FAPI-04) has recently been introduced for imaging fibroblast activation in cardiac diseases. To date, cardiac fibroblast and cardiac amyloidosis (CA) phenotype activities have not been mapped. OBJECTIVES The aim of this study was to evaluate the feasibility of 68Ga-FAPI-04 positron emission tomography (PET)/computed tomography (CT) in assessing AL CA. METHODS Thirty consecutive patients (mean age: 59.1 ± 7.7 years; 20 men, 10 women) with biopsy-proven AL amyloidosis were enrolled prospectively (including 27 with AL CA and 3 without AL CA). All patients underwent 68Ga-FAPI-04 PET/CT (107.4 ± 26.5 MBq). Global standardized uptake values and left ventricular (LV) molecular volume were calculated in correlation to echocardiography (n = 30), cardiac magnetic resonance (n = 18), and clinical biomarkers. Subsequently, the patients were categorized as having patchy (PET-patchy), extensive (PET-extensive), and negative (PET-negative) patterns. RESULTS Of all patients, 80% (24 of 30) showed increased LV uptake (PET-patchy [n = 4] vs PET-extensive [n = 20]), whereas 6 patients did not show visible myocardial uptake. Standardized uptake value ratio and LV molecular volume were significantly higher in the PET-extensive than the PET-patchy group (2.79 mL ± 1.22 mL vs 1.53 mL ± 0.66 mL [P = 0.045] and 198.3 mL ± 59.97 mL vs 127.8 mL ± 25.82 [P = 0.005], respectively). Additionally, 68Ga-FAPI-04 uptake was significantly correlated with clinical biomarkers (Mayo stage and N-terminal pro-brain natriuretic peptide), interventricular septal thickness, left ventricular ejection fraction (LVEF), LV end-systolic volume, extracellular volume, and LV global strain (P < 0.05). CONCLUSIONS 68Ga-FAPI-04 PET/CT is feasible in detecting myocardial fibroblast activation in patients with AL CA in correlation with myocardial remodeling. It might provide complementary information on cardiac molecular characterization and staging of disease.
Collapse
Affiliation(s)
- Xuezhu Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yubo Guo
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yajuan Gao
- Department of Hematology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Ren
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | - Bowei Liu
- Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Xiao Li
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Long Chang
- Department of Hematology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaini Shen
- Department of Hematology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyan Ding
- Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Zhuang Tian
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Shuyang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yining Wang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Li
- Department of Hematology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
55
|
Liu H, Fan P, Jin F, Huang G, Guo X, Xu F. Dynamic and static biomechanical traits of cardiac fibrosis. Front Bioeng Biotechnol 2022; 10:1042030. [PMID: 36394025 PMCID: PMC9659743 DOI: 10.3389/fbioe.2022.1042030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/20/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiac fibrosis is a common pathology in cardiovascular diseases which are reported as the leading cause of death globally. In recent decades, accumulating evidence has shown that the biomechanical traits of fibrosis play important roles in cardiac fibrosis initiation, progression and treatment. In this review, we summarize the four main distinct biomechanical traits (i.e., stretch, fluid shear stress, ECM microarchitecture, and ECM stiffness) and categorize them into two different types (i.e., static and dynamic), mainly consulting the unique characteristic of the heart. Moreover, we also provide a comprehensive overview of the effect of different biomechanical traits on cardiac fibrosis, their transduction mechanisms, and in-vitro engineered models targeting biomechanical traits that will aid the identification and prediction of mechano-based therapeutic targets to ameliorate cardiac fibrosis.
Collapse
Affiliation(s)
- Han Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of China, Zhengzhou, China
| | - Pengbei Fan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of China, Zhengzhou, China
| | - Fanli Jin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of China, Zhengzhou, China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Xiaogang Guo
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
56
|
Tao Y, Gao C, Qian D, Cao D, Han L, Yang L. Regulatory mechanism of fibrosis-related genes in patients with heart failure. Front Genet 2022; 13:1032572. [DOI: 10.3389/fgene.2022.1032572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Heart failure (HF) is a complex clinical syndrome characterized by the inability to match cardiac output with metabolic needs. Research on regulatory mechanism of fibrosis-related genes in patients with HF is very limited. In order to understand the mechanism of fibrosis in the development and progression of HF, fibrosis -related hub genes in HF are screened and verified.Methods: RNA sequencing data was obtained from the Gene Expression Omnibus (GEO) cohorts to identify differentially expressed genes (DEGs). Thereafter, fibrosis-related genes were obtained from the GSEA database and that associated with HF were screened out. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis was carried out to analyze the biological function of fibrosis-related DEGs. The protein-protein interaction (PPI) network of hub genes was constructed via the STRING database. Moreover, the diagnostic value of hub genes for HF was confirmed using ROC curves and expression analysis. Finally, quantitative real time PCR was used to detect the expression levels of mRNAs.Results: A total of 3, 469 DEGs were identified closely related to HF, and 1, 187 fibrosis-related DEGs were obtained and analyzed for GO and KEGG enrichment. The enrichment results of fibrosis-related DEGs were consistent with that of DEGs. A total of 10 hub genes (PPARG, KRAS, JUN, IL10, TLR4, STAT3, CXCL8, CCL2, IL6, IL1β) were selected via the PPI network. Receiver operating characteristic curve analysis was estimated in the test cohort, and 6 genes (PPARG, KRAS, JUN, IL10, TLR4, STAT3) with AUC more than 0.7 were identified as diagnosis genes. Moreover, miRNA-mRNA and TF-mRNA regulatory networks were constructed. Finally, quantitative real time PCR revealed these 6 genes may be used as the potential diagnostic biomarkers of HF.Conclusion: In this study, 10 fibrosis-related hub genes in the HF were identified and 6 of them were demonstrated as potential diagnostic biomarkers for HF.
Collapse
|
57
|
Al-Kuraishy HM, Al-Gareeb AI, Alkhuriji AF, Al-Megrin WAI, Elekhnawy E, Negm WA, De Waard M, Batiha GES. Investigation of the impact of rosuvastatin and telmisartan in doxorubicin-induced acute cardiotoxicity. Biomed Pharmacother 2022; 154:113673. [PMID: 36942604 DOI: 10.1016/j.biopha.2022.113673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 12/06/2022] Open
Abstract
Cardiac injury is the main dose-limiting factor for doxorubicin (Dox) use as an anticancer agent. The cardiotoxicity of Dox is linked to a number of complex mechanisms, including oxidative stress, mitochondrial damage, intracellular calcium dysregulation, and apoptosis/necrosis. This study investigates several aspects of Dox-induced cardiotoxicity. We investigated the effects of pre-treatment with rosuvastatin and telmisartan, which were used in different doses alone or combination, on the acute cardiotoxicity induced by Dox. The results of this study showed that Dox induced significant pathological changes in the cardiomyocytes. Adverse effects were observed on several biomarkers related to cardiac damage like cardiac troponin I (cTnI) and lactate dehydrogenase (LDH), oxidative stress like malondialdehyde (MDA), an inflammatory process like interleukin-17 (IL-17) with important histopathological changes. We illusterate the cardio-protective contribution of the two pharmacological agents against the acute cardiotoxic effects of Dox. This is manifested by the significant improvement in the biomarker levels and the associated histological damage. This study points out the beneficial use of both rosuvastatin and telmisartan alone or in combination as a clinical option for decreasing the acute toxicity of Dox on cardiomyocytes.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-mustansiriyiah University, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-mustansiriyiah University, Iraq.
| | - Afrah Fahad Alkhuriji
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Wafa Abdullah I Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120 Saint-Egrève, France; L'institut du Thorax, INSERM, CNRS, UNIV NANTES, F-44007 Nantes, France; LabEx Ion Channels, Science & Therapeutics, Université de Nice Sophia-Antipolis, F-06560 Valbonne, France.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AL Beheira, Egypt.
| |
Collapse
|
58
|
Cardiac fibrosis in oncologic therapies. CURRENT OPINION IN PHYSIOLOGY 2022; 29. [DOI: 10.1016/j.cophys.2022.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
59
|
Zhu F, Li P, Sheng Y. Treatment of myocardial interstitial fibrosis in pathological myocardial hypertrophy. Front Pharmacol 2022; 13:1004181. [PMID: 36249793 PMCID: PMC9561344 DOI: 10.3389/fphar.2022.1004181] [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: 07/27/2022] [Accepted: 09/14/2022] [Indexed: 01/09/2023] Open
Abstract
Pathological myocardial hypertrophy can be caused by a variety of diseases, mainly accompanied by myocardial interstitial fibrosis (MIF), which is a diffuse and patchy process, appearing as a combination of interstitial micro-scars and perivascular collagen fiber deposition. Different stimuli may trigger MIF without cell death by activating a variety of fibrotic signaling pathways in mesenchymal cells. This manuscript summarizes the current knowledge about the mechanism and harmful outcomes of MIF in pathological myocardial hypertrophy, discusses the circulating and imaging biomarkers that can be used to identify this lesion, and reviews the currently available and potential future treatments that allow the individualized management of patients with pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Fuyu Zhu
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Peng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Yanhui Sheng, ; Peng Li,
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China,Department of Cardiology, Jiangsu Province Hospital, Nanjing, China,*Correspondence: Yanhui Sheng, ; Peng Li,
| |
Collapse
|
60
|
Wu J, Liu T, Shi S, Fan Z, Hiram R, Xiong F, Cui B, Su X, Chang R, Zhang W, Yan M, Tang Y, Huang H, Wu G, Huang C. Dapagliflozin reduces the vulnerability of rats with pulmonary arterial hypertension-induced right heart failure to ventricular arrhythmia by restoring calcium handling. Cardiovasc Diabetol 2022; 21:197. [PMID: 36171554 PMCID: PMC9516842 DOI: 10.1186/s12933-022-01614-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/01/2022] [Indexed: 12/20/2022] Open
Abstract
Background Malignant ventricular arrhythmia (VA) is a major contributor to sudden cardiac death (SCD) in patients with pulmonary arterial hypertension (PAH)-induced right heart failure (RHF). Recently, dapagliflozin (DAPA), a sodium/glucose cotransporter-2 inhibitor (SGLT2i), has been found to exhibit cardioprotective effects in patients with left ventricular systolic dysfunction. In this study, we examined the effects of DAPA on VA vulnerability in a rat model of PAH-induced RHF. Methods Rats randomly received monocrotaline (MCT, 60 mg/kg) or vehicle via a single intraperitoneal injection. A day later, MCT-injected rats were randomly treated with placebo, low-dose DAPA (1 mg/kg/day), or high-dose (3 mg/kg/day) DAPA orally for 35 days. Echocardiographic analysis, haemodynamic experiments, and histological assessments were subsequently performed to confirm the presence of PAH-induced RHF. Right ventricle (RV) expression of calcium (Ca2+) handling proteins were detected via Western blotting. RV expression of connexin 43 (Cx43) was determined via immunohistochemical staining. An optical mapping study was performed to assess the electrophysiological characteristics in isolated hearts. Cellular Ca2+ imaging from RV cardiomyocytes (RVCMs) was recorded using Fura-2 AM or Fluo-4 AM. Results High-dose DAPA treatment attenuated RV structural remodelling, improved RV function, alleviated Cx43 remodelling, increased the conduction velocity, restored the expression of key Ca2+ handling proteins, increased the threshold for Ca2+ and action potential duration (APD) alternans, decreased susceptibility to spatially discordant APD alternans and spontaneous Ca2+ events, promoted cellular Ca2+ handling, and reduced VA vulnerability in PAH-induced RHF rats. Low-dose DAPA treatment also showed antiarrhythmic effects in hearts with PAH-induced RHF, although with a lower level of efficacy. Conclusion DAPA administration reduced VA vulnerability in rats with PAH-induced RHF by improving RVCM Ca2+ handling. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01614-5.
Collapse
Affiliation(s)
- Jinchun Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Department of Cardiology, Qinghai Provincial People's Hospital, No.2 Gong He Road, Xining, 810007, People's Republic of China
| | - Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Zhixing Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Roddy Hiram
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montreal, QC, Canada
| | - Feng Xiong
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montreal, QC, Canada
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Xiaoling Su
- Department of Cardiology, Qinghai Provincial People's Hospital, No.2 Gong He Road, Xining, 810007, People's Republic of China
| | - Rong Chang
- Department of Cardiology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, No. 187 Guanlan Road, Longhua District, Shenzhen, 518109, China
| | - Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Min Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
61
|
Li W, Li Y, Jiang F, Liu H. Correlation between serum levels of microRNA-21 and inflammatory factors in patients with chronic heart failure. Medicine (Baltimore) 2022; 101:e30596. [PMID: 36197244 PMCID: PMC9509079 DOI: 10.1097/md.0000000000030596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
As the leading cause of hospitalization and mortality worldwide, heart failure (HF) has caused significant burden on both individuals and the whole society. Thus, increasing knowledge about the phytopathology of HF is in demand for both diagnosis and treatment. Previous studies have shown that both microRNA 21 (miRNA-21) and inflammatory factors are closely related to the development of cardiac fibrosis, hypertrophy, and HF. However, whether there is any crosstalk between the 2 has not been examined. The current study evaluated the correlation between serum levels of miRNA-21 and critical inflammatory factors during the progress of chronic heart failure (CHF), providing new insights in understanding the physiopathology of CHF and identifying CHF biomarkers. In the presented study, serum level of miR-21, cardiac neurohormone, and critical inflammatory factors were measured and compared on 120 (67 male/53 female) CHF patients and 100 (58 male/42 female) health people with non-failing hearts. Echocardiography was also conducted to assess the severity of CHF. Correlations between different factors were calculated and tested for statistical significance. From our results, CHF patients showed significantly decreased serum levels of miR-21 while increased levels of inflammatory factors and cardiac neurohormone (P < .05). Levels of miR-21 negatively correlate with cardiac function while positively correlates with myocardial remodeling (P < .05). Levels of miR-21 negatively correlate with inflammation in CHF (P < .05). These findings indicate the potential crosstalk between serum miR-21 and inflammation during CHF progression, suggesting the potential of miR-21 in CHF diagnosis, severity indication, and treatment.
Collapse
Affiliation(s)
- Weiwei Li
- Emergency Department, Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Weiwei Li, Emergency Department, Second Hospital of Lanzhou University, No. 82 Cuiyingmen, Lanzhou, Gansu 730000, China (e-mail: )
| | - Yanan Li
- Emergency Department, Second Hospital of Lanzhou University, Lanzhou, China
| | - Feng Jiang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
62
|
Novel Therapies for the Treatment of Cardiac Fibrosis Following Myocardial Infarction. Biomedicines 2022; 10:biomedicines10092178. [PMID: 36140279 PMCID: PMC9496565 DOI: 10.3390/biomedicines10092178] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
Cardiac fibrosis is a common pathological consequence of most myocardial diseases. It is associated with the excessive accumulation of extracellular matrix proteins as well as fibroblast differentiation into myofibroblasts in the cardiac interstitium. This structural remodeling often results in myocardial dysfunctions such as arrhythmias and impaired systolic function in patients with heart conditions, ultimately leading to heart failure and death. An understanding of the precise mechanisms of cardiac fibrosis is still limited due to the numerous signaling pathways, cells, and mediators involved in the process. This review article will focus on the pathophysiological processes associated with the development of cardiac fibrosis. In addition, it will summarize the novel strategies for anti-fibrotic therapies such as epigenetic modifications, miRNAs, and CRISPR technologies as well as various medications in cellular and animal models.
Collapse
|
63
|
Shao J, Liu J, Zuo S. Roles of Epigenetics in Cardiac Fibroblast Activation and Fibrosis. Cells 2022; 11:cells11152347. [PMID: 35954191 PMCID: PMC9367448 DOI: 10.3390/cells11152347] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiac fibrosis is a common pathophysiologic process associated with numerous cardiovascular diseases, resulting in cardiac dysfunction. Cardiac fibroblasts (CFs) play an important role in the production of the extracellular matrix and are the essential cell type in a quiescent state in a healthy heart. In response to diverse pathologic stress and environmental stress, resident CFs convert to activated fibroblasts, referred to as myofibroblasts, which produce more extracellular matrix, contributing to cardiac fibrosis. Although multiple molecular mechanisms are implicated in CFs activation and cardiac fibrosis, there is increasing evidence that epigenetic regulation plays a key role in this process. Epigenetics is a rapidly growing field in biology, and provides a modulated link between pathological stimuli and gene expression profiles, ultimately leading to corresponding pathological changes. Epigenetic modifications are mainly composed of three main categories: DNA methylation, histone modifications, and non-coding RNAs. This review focuses on recent advances regarding epigenetic regulation in cardiac fibrosis and highlights the effects of epigenetic modifications on CFs activation. Finally, we provide some perspectives and prospects for the study of epigenetic modifications and cardiac fibrosis.
Collapse
Affiliation(s)
- Jingrong Shao
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China;
| | - Jiao Liu
- Tianjin Key Laboratory of Inflammatory Biology, Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China;
| | - Shengkai Zuo
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China;
- Correspondence:
| |
Collapse
|
64
|
Targeting Myocardial Fibrosis—A Magic Pill in Cardiovascular Medicine? Pharmaceutics 2022; 14:pharmaceutics14081599. [PMID: 36015225 PMCID: PMC9414721 DOI: 10.3390/pharmaceutics14081599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Fibrosis, characterized by an excessive accumulation of extracellular matrix, has long been seen as an adaptive process that contributes to tissue healing and regeneration. More recently, however, cardiac fibrosis has been shown to be a central element in many cardiovascular diseases (CVDs), contributing to the alteration of cardiac electrical and mechanical functions in a wide range of clinical settings. This paper aims to provide a comprehensive review of cardiac fibrosis, with a focus on the main pathophysiological pathways involved in its onset and progression, its role in various cardiovascular conditions, and on the potential of currently available and emerging therapeutic strategies to counteract the development and/or progression of fibrosis in CVDs. We also emphasize a number of questions that remain to be answered, and we identify hotspots for future research.
Collapse
|
65
|
Braidotti N, Chen SN, Long CS, Cojoc D, Sbaizero O. Piezo1 Channel as a Potential Target for Hindering Cardiac Fibrotic Remodeling. Int J Mol Sci 2022; 23:8065. [PMID: 35897650 PMCID: PMC9330509 DOI: 10.3390/ijms23158065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibrotic tissues share many common features with neoplasms where there is an increased stiffness of the extracellular matrix (ECM). In this review, we present recent discoveries related to the role of the mechanosensitive ion channel Piezo1 in several diseases, especially in regulating tumor progression, and how this can be compared with cardiac mechanobiology. Based on recent findings, Piezo1 could be upregulated in cardiac fibroblasts as a consequence of the mechanical stress and pro-inflammatory stimuli that occurs after myocardial injury, and its increased activity could be responsible for a positive feedback loop that leads to fibrosis progression. The increased Piezo1-mediated calcium flow may play an important role in cytoskeleton reorganization since it induces actin stress fibers formation, a well-known characteristic of fibroblast transdifferentiation into the activated myofibroblast. Moreover, Piezo1 activity stimulates ECM and cytokines production, which in turn promotes the phenoconversion of adjacent fibroblasts into new myofibroblasts, enhancing the invasive character. Thus, by assuming the Piezo1 involvement in the activation of intrinsic fibroblasts, recruitment of new myofibroblasts, and uncontrolled excessive ECM production, a new approach to blocking the fibrotic progression can be predicted. Therefore, targeted therapies against Piezo1 could also be beneficial for cardiac fibrosis.
Collapse
Affiliation(s)
- Nicoletta Braidotti
- Department of Physics, University of Trieste, Via A. Valerio 2, 34127 Trieste, Italy;
- Institute of Materials, National Research Council of Italy (CNR-IOM), Area Science Park Basovizza, Strada Statale 14, Km 163,5, 34149 Trieste, Italy;
| | - Suet Nee Chen
- CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, 12700 East 19th Ave., Aurora, CO 80045, USA;
| | - Carlin S. Long
- Center for the Prevention of Heart and Vascular Disease, University of California, 555 Mission Bay Blvd South, Rm 352K, San Francisco, CA 94143, USA;
| | - Dan Cojoc
- Institute of Materials, National Research Council of Italy (CNR-IOM), Area Science Park Basovizza, Strada Statale 14, Km 163,5, 34149 Trieste, Italy;
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6/A, 34127 Trieste, Italy
| |
Collapse
|
66
|
Fan J, Ren M, Adhikari BK, Wang H, He Y. The NLRP3 Inflammasome as a Novel Therapeutic Target for Cardiac Fibrosis. J Inflamm Res 2022; 15:3847-3858. [PMID: 35836721 PMCID: PMC9273832 DOI: 10.2147/jir.s370483] [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: 04/12/2022] [Accepted: 07/01/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiac fibrosis often has adverse cardiovascular effects, including heart failure, sudden death, and malignant arrhythmias. However, there is no targeted therapy for cardiac fibrosis. Inflammation is known to play a crucial role in the disorder, and the NLR pyrin domain-containing-3 (NLRP3) inflammasome is closely associated with innate immunity. Therefore, further understanding the pathophysiological role of the inflammasome in cardiac fibrosis may provide novel strategies for the prevention and treatment of the disorder. The aim of this review was to summarize the present knowledge of NLRP3 inflammasome-related mechanisms underlying cardiac fibrosis and to suggest potential targeted therapy that could be used to treat the condition.
Collapse
Affiliation(s)
- Jiwen Fan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Meng Ren
- Department of Medical Oncology, Jilin Provincial Cancer Hospital, Changchun, People's Republic of China
| | - Binay Kumar Adhikari
- Department of Cardiology, Nepal Armed Police Force (APF) Hospital, Kathmandu, Nepal
| | - Haodong Wang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
67
|
Cunha PS, Laranjo S, Heijman J, Oliveira MM. The Atrium in Atrial Fibrillation - A Clinical Review on How to Manage Atrial Fibrotic Substrates. Front Cardiovasc Med 2022; 9:879984. [PMID: 35859594 PMCID: PMC9289204 DOI: 10.3389/fcvm.2022.879984] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/03/2022] [Indexed: 12/27/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in the population and is associated with a significant clinical and economic burden. Rigorous assessment of the presence and degree of an atrial arrhythmic substrate is essential for determining treatment options, predicting long-term success after catheter ablation, and as a substrate critical in the pathophysiology of atrial thrombogenesis. Catheter ablation of AF has developed into an essential rhythm-control strategy. Nowadays is one of the most common cardiac ablation procedures performed worldwide, with its success inversely related to the extent of atrial structural disease. Although atrial substrate evaluation remains complex, several diagnostic resources allow for a more comprehensive assessment and quantification of the extent of left atrial structural remodeling and the presence of atrial fibrosis. In this review, we summarize the current knowledge on the pathophysiology, etiology, and electrophysiological aspects of atrial substrates promoting the development of AF. We also describe the risk factors for its development and how to diagnose its presence using imaging, electrocardiograms, and electroanatomic voltage mapping. Finally, we discuss recent data regarding fibrosis biomarkers that could help diagnose atrial fibrotic substrates.
Collapse
Affiliation(s)
- Pedro Silva Cunha
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Sérgio Laranjo
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Mário Martins Oliveira
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
68
|
Fan J, Ren M, He Y. Diagnostic and Therapeutic Properties of Exosomes in Cardiac Fibrosis. Front Cell Dev Biol 2022; 10:931082. [PMID: 35859903 PMCID: PMC9289295 DOI: 10.3389/fcell.2022.931082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiac fibrosis results from both the differentiation of cardiac fibroblasts and excessive accumulation of extracellular matrix (ECM), leading to myocardial stiffness and reduced compliance of the ventricular wall. The conversion of cardiac fibroblasts to myofibroblasts is the most important initiating step in the process of this pathological cardiac remodeling. It occurs during the progression of many cardiovascular diseases, adversely influencing both the clinical course and outcome of the disease. The pathogenesis is complex and there is no effective treatment. Exosomes are extracellular vesicles that mediate intercellular communication through delivering specific cargoes of functional nucleic acids and proteins derived from particular cell types. Recent studies have found that exosomes play an important role in the diagnosis and treatment of cardiac fibrosis, and is a potential biotherapeutics and drug delivery vectors for the treatment of cardiac fibrosis. The present review aimed to summarize the current knowledge of exosome-related mechanisms underlying cardiac fibrosis and to suggest potential therapy that could be used to treat the condition.
Collapse
Affiliation(s)
- Jiwen Fan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Ren
- Department of Medical Oncology, Jilin Provincial Cancer Hospital, Changchun, China
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Yuquan He,
| |
Collapse
|
69
|
Saleh S, George J, Kott KA, Meikle PJ, Figtree GA. The Translation and Commercialisation of Biomarkers for Cardiovascular Disease—A Review. Front Cardiovasc Med 2022; 9:897106. [PMID: 35722087 PMCID: PMC9201254 DOI: 10.3389/fcvm.2022.897106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
As a leading cause of mortality and morbidity worldwide, cardiovascular disease and its diagnosis, quantification, and stratification remain significant health issues. Increasingly, patients present with cardiovascular disease in the absence of known risk factors, suggesting the presence of yet unrecognized pathological processes and disease predispositions. Fortunately, a host of emerging cardiovascular biomarkers characterizing and quantifying ischaemic heart disease have shown great promise in both laboratory settings and clinical trials. These have demonstrated improved predictive value additional to widely accepted biomarkers as well as providing insight into molecular phenotypes beneath the broad umbrella of cardiovascular disease that may allow for further personalized treatment regimens. However, the process of translation into clinical practice – particularly navigating the legal and commercial landscape – poses a number of challenges. Practical and legal barriers to the biomarker translational pipeline must be further considered to develop strategies to bring novel biomarkers into the clinical sphere and apply these advances at the patient bedside. Here we review the progress of emerging biomarkers in the cardiovascular space, with particular focus on those relevant to the unmet needs in ischaemic heart disease.
Collapse
Affiliation(s)
- Soloman Saleh
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Katharine A. Kott
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Gemma A. Figtree
| |
Collapse
|
70
|
Yan Z, Cheng X, Wang T, Hong X, Shao G, Fu C. Therapeutic potential for targeting Annexin A1 in fibrotic diseases. Genes Dis 2022; 9:1493-1505. [PMID: 36157506 PMCID: PMC9485289 DOI: 10.1016/j.gendis.2022.05.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/30/2022] [Indexed: 11/23/2022] Open
Abstract
Annexin A1, a well-known endogenous anti-inflammatory mediator, plays a critical role in a variety of pathological processes. Fibrosis is described by a failure of tissue regeneration and contributes to the development of many diseases. Accumulating evidence supports that Annexin A1 participates in the progression of tissue fibrosis. However, the fundamental mechanisms by which Annexin A1 regulates fibrosis remain elusive, and even the functions of Annexin A1 in fibrotic diseases are still paradoxical. This review focuses on the roles of Annexin A1 in the development of fibrosis of lung, liver, heart, and other tissues, with emphasis on the therapy potential of Annexin A1 in fibrosis, and presents future research interests and directions in fibrotic diseases.
Collapse
|
71
|
Bachmann JC, Baumgart SJ, Uryga AK, Bosteen MH, Borghetti G, Nyberg M, Herum KM. Fibrotic Signaling in Cardiac Fibroblasts and Vascular Smooth Muscle Cells: The Dual Roles of Fibrosis in HFpEF and CAD. Cells 2022; 11:1657. [PMID: 35626694 PMCID: PMC9139546 DOI: 10.3390/cells11101657] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) and atherosclerosis-driven coronary artery disease (CAD) will have ongoing fibrotic remodeling both in the myocardium and in atherosclerotic plaques. However, the functional consequences of fibrosis differ for each location. Thus, cardiac fibrosis leads to myocardial stiffening, thereby compromising cardiac function, while fibrotic remodeling stabilizes the atherosclerotic plaque, thereby reducing the risk of plaque rupture. Although there are currently no drugs targeting cardiac fibrosis, it is a field under intense investigation, and future drugs must take these considerations into account. To explore similarities and differences of fibrotic remodeling at these two locations of the heart, we review the signaling pathways that are activated in the main extracellular matrix (ECM)-producing cells, namely human cardiac fibroblasts (CFs) and vascular smooth muscle cells (VSMCs). Although these signaling pathways are highly overlapping and context-dependent, effects on ECM remodeling mainly act through two core signaling cascades: TGF-β and Angiotensin II. We complete this by summarizing the knowledge gained from clinical trials targeting these two central fibrotic pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kate M. Herum
- Research and Early Development, Novo Nordisk A/S, Novo Nordisk Park, 2760 Maaloev, Denmark; (J.C.B.); (S.J.B.); (A.K.U.); (M.H.B.); (G.B.); (M.N.)
| |
Collapse
|
72
|
Li X, Shi S, Yang Y, Lou Y, Chen S, Guo Y, Qi X, Wang Y, Liu Z, Cheng Y. The anticardiac fibrosis of total alkaloids of Plumula nelumbinis by regulating circulating lipidomic profile: In vivo study. J Food Biochem 2022; 46:e14194. [PMID: 35502470 DOI: 10.1111/jfbc.14194] [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: 12/05/2021] [Revised: 03/08/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
Abstract
Plumula nelumbinis has great medicinal potential as a herbal tea and traditional drug in China. This study was aimed to evaluate the anticardiac fibrosis of the total alkaloids of P. nelumbinis (TAP). TAP at 50 mg/kg/day significantly ameliorated isoproterenol-induced cardiac fibrosis in mice (p < .05). The circulating lipidomics study revealed that TAP improved the lipid metabolism dysfunction in cardiac fibrosis. Meanwhile, TAP suppressed the lipid accumulation, decreased MDA level (p < .01) in heart, and increased FFA level (p < .01). Furthermore, integrating lipidomics, chemical profiles and pharmacology network analysis found that AMPK and PI3K/Akt signaling pathways were the potential targeted pathway by TAP to regulate lipid metabolism dysfunction including glycerophospholipid metabolism. Above all, TAP provided a potential anticardiac fibrosis effect partly through regulation of lipid profiles. PRACTICAL APPLICATIONS: The total alkaloids of Plumula nelumbinis (TAP) suppressed ISO-induced cardiac fibrosis in mice. Network pharmacology analysis and experiments revealed that TAP-regulated AMPK and PI3K/Akt signaling pathway to improve lipid metabolism disorder in cardiac fibrosis. This study provides evidence to the therapeutic potential of TAP in the treatment of ISO-induced cardiac fibrosis and could be a drug candidate for prevention and treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Xuping Li
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuotao Shi
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Yang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanmei Lou
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sixuan Chen
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yixin Guo
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoxiao Qi
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanyuan Cheng
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
73
|
Han X, Zhang Y, Zhang X, Ji H, Wang W, Qiao O, Li X, Wang J, Liu C, Huang L, Gao W. Targeting adipokines: A new strategy for the treatment of myocardial fibrosis. Pharmacol Res 2022; 181:106257. [DOI: 10.1016/j.phrs.2022.106257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/26/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
|
74
|
Nemir M, Kay M, Maison D, Berthonneche C, Sarre A, Plaisance I, Pedrazzini T. Inhibition of the NOTCH1 Pathway in the Stressed Heart Limits Fibrosis and Promotes Recruitment of Non-Myocyte Cells into the Cardiomyocyte Fate. J Cardiovasc Dev Dis 2022; 9:jcdd9040111. [PMID: 35448087 PMCID: PMC9024539 DOI: 10.3390/jcdd9040111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiac pathologies lead to an acute or gradual loss of cardiomyocytes. Because of the limited regenerative capacity of the mammalian heart, cardiomyocytes are only replaced by fibrotic tissue. Excessive fibrosis contributes to the deterioration of cardiac function and the transition to heart failure, which is the leading cause of morbidity and mortality worldwide. Currently, no treatments can promote replenishment of the injured heart with newly formed cardiomyocytes. In this context, regenerative strategies explore the possibility to promote recovery through induction of cardiomyocyte production from pre-existing cardiomyocytes. On the other hand, cardiac non-myocyte cells can be directly reprogrammed into induced cardiac precursor cells and cardiomyocytes, suggesting that these cells could be exploited to produce cardiomyocytes in vivo. Here, we provide evidence that the sequential activation and inhibition of the NOTCH1 signaling pathway in the stressed heart decreases fibrosis and improves cardiac function in the stressed heart. This is accompanied by the emergence of new cardiomyocytes from non-myocyte origin. Overall, our data show how a developmental pathway such as the NOTCH pathway can be manipulated to provide therapeutic benefit in the damaged heart.
Collapse
Affiliation(s)
- Mohamed Nemir
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, 1011 Lausanne, Switzerland; (M.N.); (M.K.); (D.M.); (I.P.)
| | - Maryam Kay
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, 1011 Lausanne, Switzerland; (M.N.); (M.K.); (D.M.); (I.P.)
| | - Damien Maison
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, 1011 Lausanne, Switzerland; (M.N.); (M.K.); (D.M.); (I.P.)
| | - Corinne Berthonneche
- Cardiovascular Assessment Facility, University of Lausanne, 1011 Lausanne, Switzerland; (C.B.); (A.S.)
| | - Alexandre Sarre
- Cardiovascular Assessment Facility, University of Lausanne, 1011 Lausanne, Switzerland; (C.B.); (A.S.)
| | - Isabelle Plaisance
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, 1011 Lausanne, Switzerland; (M.N.); (M.K.); (D.M.); (I.P.)
| | - Thierry Pedrazzini
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, 1011 Lausanne, Switzerland; (M.N.); (M.K.); (D.M.); (I.P.)
- Correspondence: ; Tel.: +41-21-314-0765
| |
Collapse
|
75
|
MiRNA-122-5p inhibitor abolishes angiotensin II-mediated loss of autophagy and promotion of apoptosis in rat cardiofibroblasts by modulation of the apelin-AMPK-mTOR signaling. In Vitro Cell Dev Biol Anim 2022; 58:136-148. [PMID: 35133561 DOI: 10.1007/s11626-022-00651-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 11/05/2022]
Abstract
MicroRNAs (miRNAs) have emerged as essential regulators that could have pivotal roles in cardiac homeostasis and pathological remodeling of various cardiovascular diseases. We previously demonstrated that miRNA-122-5p overexpression exacerbated the process of vascular hypertrophy, fibrosis, and dysfunction in hypertensive rats and rat aortic adventitial fibroblasts. However, the exact roles and underlying mechanisms of miRNA-122-5p in myocardial fibroblasts remain largely unknown. In this work, neonatal rat cardiofibroblasts (CFs) were isolated and primarily cultured from the hearts of 2- to 3-d-old Sprague-Dawley rats. Stimulation of angiotensin II (Ang II) resulted in marked increases in cellular proliferation and migration and levels of collagen I, collagen III, CTGF, and TGF-β1 in cultured CFs. Furthermore, Ang II led to promoted expression of P62, Bax, and phosphorylated mTOR as well as downregulation of LC3II, beclin-1, and AMPK-phosphorylated levels, thereby contributing to imbalance of autophagy and apoptosis, and cellular injury in CFs, which were significantly ameliorated by treatment with miRNA-122-5p inhibitor. These changes were associated with decreased levels of collagen I, collagen III, CTGF, and TGF-β1. Furthermore, Ang II-induced loss of autophagy and promotion of apoptosis in CFs were prevented by the treatment with Pyr1-apelin-13 or AMPK agonist AICAR or mTOR inhibitor rapamycin, respectively. In contrast, administration of miRNA-122-5p mimics and autophagy inhibitor 3-methylademine reversed beneficial roles of Pyr1-apelin-13. Collectively, these data indicated that miRNA-122-5p is an essential regulator of autophagy and apoptosis in rat CFs via the apelin/AMPK/mTOR signaling pathway, which may be potentially used as a therapeutic target in myocardial fibrosis and related diseases.
Collapse
|
76
|
Wang J, Li H, Lv Z, Luo X, Deng W, Zou T, Zhang Y, Sang W, Wang X. The miR-214-3p/c-Ski axis modulates endothelial-mesenchymal transition in human coronary artery endothelial cells in vitro and in mice model in vivo. Hum Cell 2022; 35:486-497. [PMID: 34978047 DOI: 10.1007/s13577-021-00653-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/20/2021] [Indexed: 01/08/2023]
Abstract
Cardiovascular disease (CVD) is a leading non-communicable disease with a high fatality rate worldwide. Hypertension, a common cardiovascular condition, is a significant risk factor for the development of heart failure because the activation of the renin-angiotensin system (RAS) is considered to be the major promoting reason behind myocardial fibrosis (MF). In this study, Angiotensin II (Ang II) stimulation-induced endothelial to mesenchymal transition (End-MT) in HCAECs, including the decrease of CD31 level, the increase of α-SMA, collagen I, slug, snail, and TGF-β1 levels, and the promotion of Smad2/3 phosphorylation. Meanwhile, the c-Ski level was reduced in Ang II-stimulated HCAECs. In HCAECs, Ang II-induced changes could be partially attenuated by c-Ski overexpression. miR-214-3p directly targeted c-Ski and inhibited c-Ski expression. Moreover, miR-214-3p inhibition reduced Ang II-caused End-MT in HCAECs. miR-214-3p overexpression further enhanced Ang II-induced End-MT, while c-Ski overexpression could markedly reverse the effects of miR-214-3p overexpression. In the Ang II-induced mouse cardiac hypertrophic model, Ang II-caused increase of cellular cross-sectional area and cardiac fibrosis were partially ameliorated by LV-c-Ski; when mice were co-treated with LV-c-Ski and agomir-214-3p, the beneficial effects of LV-c-Ski were reversed. In conclusion, the miR-214-3p/c-Ski axis modulated Ang II-induced End-MT in HCAECs and cardiac hypertrophy and fibrosis in the mice model.
Collapse
Affiliation(s)
- Juan Wang
- Department of Cardiology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Hongjian Li
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China.
| | - Zhongying Lv
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Xiaomei Luo
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting Zou
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Yue Zhang
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Wanyue Sang
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Xuehua Wang
- Department of Hypertension, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| |
Collapse
|
77
|
Chignon A, Argaud D, Boulanger MC, Mkannez G, Bon-Baret V, Li Z, Thériault S, Bossé Y, Mathieu P. Genome-wide chromatin contacts of super-enhancer-associated lncRNA identify LINC01013 as a regulator of fibrosis in the aortic valve. PLoS Genet 2022; 18:e1010010. [PMID: 35041643 PMCID: PMC8797204 DOI: 10.1371/journal.pgen.1010010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 01/28/2022] [Accepted: 12/22/2021] [Indexed: 12/22/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is characterized by a fibrocalcific process. The regulatory mechanisms that drive the fibrotic response in the aortic valve (AV) are poorly understood. Long noncoding RNAs derived from super-enhancers (lncRNA-SE) control gene expression and cell fate. Herein, multidimensional profiling including chromatin immunoprecipitation and sequencing, transposase-accessible chromatin sequencing, genome-wide 3D chromatin contacts of enhancer-promoter identified LINC01013 as an overexpressed lncRNA-SE during CAVD. LINC01013 is within a loop anchor, which has contact with the promoter of CCN2 (CTGF) located at ~180 kb upstream. Investigation showed that LINC01013 acts as a decoy factor for the negative transcription elongation factor E (NELF-E), whereby it controls the expression of CCN2. LINC01013-CCN2 is part of a transforming growth factor beta 1 (TGFB1) network and exerts a control over fibrogenesis. These findings illustrate a novel mechanism whereby a dysregulated lncRNA-SE controls, through a looping process, the expression of CCN2 and fibrogenesis of the AV. Calcific aortic valve disease is the most common heart valve disorder characterized by a thickening of the aortic valve resulting from fibrotic and calcific processes. Because the aortic valve replacement is currently the only therapeutic option, the identification of key molecular processes that control the progression of the disease could lead to the development of novel noninvasive therapies. Growing evidence suggests that long noncoding RNAs (lncRNAs) fine tune gene expression in health and disease states. By using a multidimensional profiling including genome-wide 3D enhancer-promoter looping data, we identified LINC01013, a lncRNA, as a regulator of fibrogenesis. Specifically, we found that LINC01013 is located in a cluster of distant enhancers (super-enhancer) in aortic valve interstitial cells and has significant long-range looping with the promoter of CCN2, a gene that orchestrates fibrogenesis. We discovered that LINC01013 is acting as a decoy factor for a negative transcription elongation factor, whereby it controls the transcription of CCN2. In turn, higher expression of LINC01013 during calcific aortic valve disease promoted the expression of CCN2 and a fibrogenic program. These findings provide evidence that LINC01013 is a key regulator of fibrogenesis in CAVD.
Collapse
Affiliation(s)
- Arnaud Chignon
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
| | - Déborah Argaud
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
| | - Marie-Chloé Boulanger
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
| | - Ghada Mkannez
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
| | - Valentin Bon-Baret
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
| | - Zhonglin Li
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Quebec, Canada
| | - Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Quebec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Quebec, Canada
- * E-mail:
| |
Collapse
|
78
|
Graham C, Sethu P. Myocardial Fibrosis: Cell Signaling and In Vitro Modeling. CARDIOVASCULAR SIGNALING IN HEALTH AND DISEASE 2022:287-321. [DOI: 10.1007/978-3-031-08309-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
79
|
Rui H, Zhao F, Yuhua L, Hong J. Suppression of SMOC2 alleviates myocardial fibrosis via the ILK/p38 pathway. Front Cardiovasc Med 2022; 9:951704. [PMID: 36935650 PMCID: PMC10017443 DOI: 10.3389/fcvm.2022.951704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/13/2022] [Indexed: 03/06/2023] Open
Abstract
Background Fibrosis of the myocardium is one of the main pathological changes of adverse cardiac remodeling, which is associated with unsatisfactory outcomes in patients with heart disease. Further investigations into the precise molecular mechanisms of cardiac fibrosis are urgently required to seek alternative therapeutic strategies for individuals suffering from heart failure. SMOC2 has been shown to be essential to exert key pathophysiological roles in various physiological processes in vivo, possibly contributing to the pathogenesis of fibrosis. A study investigating the relationship between SMOC2 and myocardial fibrosis has yet to be conducted. Methods Mice received a continuous ISO injection subcutaneously to induce cardiac fibrosis, and down-regulation of SMOC2 was achieved by adeno-associated virus-9 (AAV9)-mediated shRNA knockdown. Neonatal fibroblasts were separated and cultured in vitro with TGFβ to trigger fibrosis and infected with either sh-SMOC2 or sh-RNA as a control. The role and mechanisms of SMOC2 in myocardial fibrosis were further examined and analyzed. Results SMOC2 knockdown partially reversed cardiac functional impairment and cardiac fibrosis in vivo after 21 consecutive days of ISO injection. We further demonstrated that targeting SMOC2 expression effectively slowed down the trans-differentiation and collagen deposition of cardiac fibroblasts stimulated by TGFβ. Mechanistically, targeting SMOC2 expression inhibited the induction of ILK and p38 in vivo and in vitro, and ILK overexpression increased p38 phosphorylation activity and compromised the protective effects of sh-SMOC2-mediated cardiac fibrosis. Conclusion Therapeutic SMOC2 silencing alleviated cardiac fibrosis through inhibition of the ILK/p38 signaling, providing a preventative and control strategy for cardiac remodeling management in clinical practice.
Collapse
Affiliation(s)
- Huang Rui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Fang Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lei Yuhua
- Department of Cardiology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi City, China
| | - Jiang Hong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Jiang Hong,
| |
Collapse
|
80
|
Lv SL, Zeng ZF, Gan WQ, Wang WQ, Li TG, Hou YF, Yan Z, Zhang RX, Yang M. Lp-PLA2 inhibition prevents Ang II-induced cardiac inflammation and fibrosis by blocking macrophage NLRP3 inflammasome activation. Acta Pharmacol Sin 2021; 42:2016-2032. [PMID: 34226664 PMCID: PMC8632984 DOI: 10.1038/s41401-021-00703-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022]
Abstract
Macrophage-mediated inflammation plays an important role in hypertensive cardiac remodeling, whereas effective pharmacological treatments targeting cardiac inflammation remain unclear. Lipoprotein-associated phospholipase A2 (Lp-PLA2) contributes to vascular inflammation-related diseases by mediating macrophage migration and activation. Darapladib, the most advanced Lp-PLA2 inhibitor, has been evaluated in phase III trials in atherosclerosis patients. However, the role of darapladib in inhibiting hypertensive cardiac fibrosis remains unknown. Using a murine angiotensin II (Ang II) infusion-induced hypertension model, we found that Pla2g7 (the gene of Lp-PLA2) was the only upregulated PLA2 gene detected in hypertensive cardiac tissue, and it was primarily localized in heart-infiltrating macrophages. As expected, darapladib significantly prevented Ang II-induced cardiac fibrosis, ventricular hypertrophy, and cardiac dysfunction, with potent abatement of macrophage infiltration and inflammatory response. RNA sequencing revealed that darapladib strongly downregulated the expression of genes and signaling pathways related to inflammation, extracellular matrix, and proliferation. Moreover, darapladib substantially reduced the Ang II infusion-induced expression of nucleotide-binding oligomerization domain-like receptor with pyrin domain 3 (NLRP3) and interleukin (IL)-1β and markedly attenuated caspase-1 activation in cardiac tissues. Furthermore, darapladib ameliorated Ang II-stimulated macrophage migration and IL-1β secretion in macrophages by blocking NLRP3 inflammasome activation. Darapladib also effectively blocked macrophage-mediated transformation of fibroblasts into myofibroblasts by inhibiting the activation of the NLRP3 inflammasome in macrophages. Overall, our study identifies a novel anti-inflammatory and anti-cardiac fibrosis role of darapladib in Lp-PLA2 inhibition, elucidating the protective effects of suppressing NLRP3 inflammasome activation. Lp-PLA2 inhibition by darapladib represents a novel therapeutic strategy for hypertensive cardiac damage treatment.
Collapse
Affiliation(s)
- Si-Lin Lv
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zi-Fan Zeng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Qiang Gan
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wei-Qi Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tie-Gang Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yu-Fang Hou
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zheng Yan
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ri-Xin Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Min Yang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
81
|
Is the Macrophage Phenotype Determinant for Fibrosis Development? Biomedicines 2021; 9:biomedicines9121747. [PMID: 34944564 PMCID: PMC8698841 DOI: 10.3390/biomedicines9121747] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/16/2022] Open
Abstract
Fibrosis is a pathophysiological process of wound repair that leads to the deposit of connective tissue in the extracellular matrix. This complication is mainly associated with different pathologies affecting several organs such as lung, liver, heart, kidney, and intestine. In this fibrotic process, macrophages play an important role since they can modulate fibrosis due to their high plasticity, being able to adopt different phenotypes depending on the microenvironment in which they are found. In this review, we will try to discuss whether the macrophage phenotype exerts a pivotal role in the fibrosis development in the most important fibrotic scenarios.
Collapse
|
82
|
Zhang QJ, He Y, Li Y, Shen H, Lin L, Zhu M, Wang Z, Luo X, Hill JA, Cao D, Luo RL, Zou R, McAnally J, Liao J, Bajona P, Zang QS, Yu Y, Liu ZP. Matricellular Protein Cilp1 Promotes Myocardial Fibrosis in Response to Myocardial Infarction. Circ Res 2021; 129:1021-1035. [PMID: 34610755 DOI: 10.1161/circresaha.121.319482] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Qing-Jun Zhang
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Yu He
- Department of Clinical Laboratory, First Affiliated hospital of Guangxi Medical University, China (Y.H.)
| | - Yongnan Li
- The Sixth General Surgery, Biliary & Vascular surgery, Shengjing Hospital of China Medical University, China (Y.L.)
| | - Huali Shen
- Institutes of Biochemical Science, Fudan University, China (H.S., L.L.)
| | - Ling Lin
- Institutes of Biochemical Science, Fudan University, China (H.S., L.L.)
| | - Min Zhu
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Zhaoning Wang
- Department of Molecular Biology (Z.W., J.A.H., J.M., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Xiang Luo
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Joseph A Hill
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology (Z.W., J.A.H., J.M., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Dian Cao
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Richard L Luo
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Raymond Zou
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - John McAnally
- Department of Molecular Biology (Z.W., J.A.H., J.M., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (J.L.)
| | - Pietro Bajona
- Department of Cardiovascular and Thoracic Surgery (P.B.), UT Southwestern Medical Center, Dallas, TX.,Allegheny Health Network-Drexel University College of Medicine, Pittsburgh, PA (P.B.)
| | - Qun S Zang
- Department of Surgery, Stritch School of Medicine, Burn & Shock Trauma Research Institute, Loyola University, Maywood, IL (Q.S.Z.)
| | - Yonghao Yu
- Department of Biochemistry (Y.Y.), UT Southwestern Medical Center, Dallas, TX
| | - Zhi-Ping Liu
- Internal Medicine-Cardiology Division (Q.-J.Z., M.Z., X.L., J.A.H., D.C., R.L.L., R.Z., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology (Z.W., J.A.H., J.M., Z.-P.L.), UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
83
|
Mainardi A, Carminati F, Ugolini GS, Occhetta P, Isu G, Robles Diaz D, Reid G, Visone R, Rasponi M, Marsano A. A dynamic microscale mid-throughput fibrosis model to investigate the effects of different ratios of cardiomyocytes and fibroblasts. LAB ON A CHIP 2021; 21:4177-4195. [PMID: 34545378 PMCID: PMC8547330 DOI: 10.1039/d1lc00092f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/03/2021] [Indexed: 05/26/2023]
Abstract
Cardiac fibrosis is a maladaptive remodeling of the myocardium hallmarked by contraction impairment and excessive extracellular matrix deposition (ECM). The disease progression, nevertheless, remains poorly understood and present treatments are not capable of controlling the scarring process. This is partly due to the absence of physiologically relevant, easily operable, and low-cost in vitro models, which are of the utmost importance to uncover pathological mechanisms and highlight possible targets for anti-fibrotic therapies. In classic models, fibrotic features are usually obtained using substrates with scar mimicking stiffness and/or supplementation of morphogens such as transforming growth factor β1 (TGF-β1). Qualities such as the interplay between activated fibroblasts (FBs) and cardiomyocytes (CMs), or the mechanically active, three-dimensional (3D) environment, are, however, neglected or obtained at the expense of the number of experimental replicates achievable. To overcome these shortcomings, we engineered a micro-physiological system (MPS) where multiple 3D cardiac micro-tissues can be subjected to cyclical stretching simultaneously. Up to six different biologically independent samples are incorporated in a single device, increasing the experimental throughput and paving the way for higher yielding drug screening campaigns. The newly developed MPS was used to co-culture different ratios of neonatal rat CMs and FBs, investigating the role of CMs in the modulation of fibrosis traits, without the addition of morphogens, and in soft substrates. The expression of contractile stress fibers and of degradative enzymes, as well as the deposition of fibronectin and type I collagen were superior in microtissues with a low amount of CMs. Moreover, high CM-based microconstructs simulating a ratio similar to that of healthy tissues, even if subjected to both cyclic stretch and TGF-β1, did not show any of the investigated fibrotic signs, indicating a CM fibrosis modulating effect. Overall, this in vitro fibrosis model could help to uncover new pathological aspects studying, with mid-throughput and in a mechanically active, physiologically relevant environment, the crosstalk between the most abundant cell types involved in fibrosis.
Collapse
Affiliation(s)
- Andrea Mainardi
- Departments of Biomedicine and Surgery, University Basel and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Francesca Carminati
- Departments of Biomedicine and Surgery, University Basel and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Giovanni Stefano Ugolini
- Departments of Biomedicine and Surgery, University Basel and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
- BiomimX S.r.l., Via Giovanni Durando 38/A, 20158 Milano, Italy
| | - Giuseppe Isu
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Diana Robles Diaz
- Departments of Biomedicine and Surgery, University Basel and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Gregory Reid
- Departments of Biomedicine and Surgery, University Basel and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Roberta Visone
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Anna Marsano
- Departments of Biomedicine and Surgery, University Basel and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| |
Collapse
|
84
|
Sadoughi F, Hallajzadeh J, Mirsafaei L, Asemi Z, Zahedi M, Mansournia MA, Yousefi B. Cardiac fibrosis and curcumin: a novel perspective on this natural medicine. Mol Biol Rep 2021; 48:7597-7608. [PMID: 34648140 DOI: 10.1007/s11033-021-06768-1] [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: 05/22/2021] [Accepted: 09/10/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND According to WHO statistics, cardiovascular disease are the leading causes of death in the world. One of the main factors which is causing heart failure, systolic and diastolic dysfunction, and arrythmias is a condition named cardiac fibrosis. This condition is defined by the accumulation of fibroblast-produced ECM in myocardium layer of the heart. OBJECTIVE Accordingly, the current review aims to depict the role of curcumin in the regulation of different signaling pathways that are involved in cardiac fibrosis. RESULTS A great number of cellular and molecular mechanisms such as oxidative stress, inflammation, and mechanical stress are acknowledged to be involved in cardiac fibrosis. Despite the available therapeutic procedures which are designed to target these mechanisms in order to prevent cardiac fibrosis, still, effective therapeutic methods are needed. Curcumin is a natural Chinese medicine which currently has been declared to have therapeutic properties such as anti-oxidant and immunomodulatory activities. In this review, we have gathered several experimental studies in order to represent diverse impacts of this turmeric derivative on pathogenic factors of cardiac fibrosis. CONCLUSION Curcumin might open new avenues in the field of cardiovascular treatment.
Collapse
Affiliation(s)
- Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Liaosadat Mirsafaei
- Department of Cardiology, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mahdi Zahedi
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgān, Iran.
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
85
|
Sun B, Zhao C, Mao Y. MiR-218-5p Mediates Myocardial Fibrosis after Myocardial Infarction by Targeting CX43. Curr Pharm Des 2021; 27:4504-4512. [PMID: 34587879 DOI: 10.2174/1381612827666210929111622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Myocardial fibrosis after myocardial infarction (MI) has been considered a core factor in the deterioration of cardiac function. Previous studies have shown that miRNA plays an important role in various pathophysiological processes of the heart. However, the role of miRNA in myocardial fibrosis regulation after MI remains unclear. In the present study, we documented that miR-218-5p was significantly decreased in myocardial fibroblasts after MI. METHODS The miRNA expression profiles of MI were downloaded from GEO Datasets. The expression of a fibrosis-related gene in vivo and in vitro was analyzed by RT-PCR, western blotting, and immunohistochemical staining. RESULTS Total 7 up- and 9 downregulated common miRNAs were found in the two profiles. Among these common genes, miR-218-5p was downregulated in the MI mice. MiR-218-5p mediated the myocardial fibrosis in vivo and in vitro. Mechanistically, we found that GJA1 (CX43) may be the target of miR218-5p, and overexpressed CX43 can partly block the function of miR-218-5p in fibrosis inhibition. CONCLUSION Our results suggested that miR-218-5p plays an important role in myocardial fibrosis after MI by targeting CX43. Thus, miR-218-5p promises to be a potential diagnosis and treatment of myocardial fibrosis after MI.
Collapse
Affiliation(s)
- Bing Sun
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Cuimei Zhao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Yu Mao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
86
|
MSCs-Derived Extracellular Vesicles Carrying miR-212-5p Alleviate Myocardial Infarction-Induced Cardiac Fibrosis via NLRC5/VEGF/TGF-β1/SMAD Axis. J Cardiovasc Transl Res 2021; 15:302-316. [PMID: 34508321 DOI: 10.1007/s12265-021-10156-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
The purpose of the present study was to define the role of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) in the progression of myocardial infarction (MI)-induced cardiac fibrosis. An in vitro cell model of hypoxia-induced cardiac fibrosis was constructed in cardiac fibroblasts (CFs). miR-212-5p was poorly expressed in clinical pathological samples and animal models of cardiac fibrosis caused by MI, while miR-212-5p expression was enriched in EVs released from MSCs. EVs from MSCs were isolated, evaluated, and co-cultured with CFs. Dual-luciferase reporter gene assay revealed that miR-212-5p negatively targeted NLRC5 progression of cardiac fibrosis. Following loss- and gain-function assay, EVs expressing miR-212-5p protected against cardiac fibrosis evidenced by reduced levels of α-SMA, Collagen I, TGF-β1, and IL-1β. In vivo experiments further confirmed the above research results. Collectively, EVs from MSCs expressing miR-212-5p may attenuate MI by suppressing the NLRC5/VEGF/TGF-β1/SMAD axis.
Collapse
|
87
|
Li M, Qi C, Song R, Xiong C, Zhong X, Song Z, Ning Z, Song X. Inhibition of Long Noncoding RNA SNHG20 Improves Angiotensin II-Induced Cardiac Fibrosis and Hypertrophy by Regulating the MicroRNA 335/ Galectin-3 Axis. Mol Cell Biol 2021; 41:e0058020. [PMID: 34228494 PMCID: PMC8384070 DOI: 10.1128/mcb.00580-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 11/29/2020] [Accepted: 06/12/2021] [Indexed: 11/20/2022] Open
Abstract
Cardiac fibrosis is a hallmark of various heart diseases and ultimately leads to heart failure. Although long noncoding RNA (lncRNA) SNHG20 has been reported to play important roles in various cancers, its function in cardiac fibrosis remains unclear. The expression of SNHG20 and microRNA 335 (miR-335) in heart tissues of angiotensin II-induced mice and angiotensin II-stimulated mouse cardiomyocyte cell line HL-1 were detected by quantitative real-time PCR (qRT-PCR). Cell viability was evaluated by cell counting kit-8 assay. The expression of galectin-3, fibrosis-related proteins (fibronectin, collagen IaI, and α-SMA), and apoptosis-related proteins [cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase (PARP)] was detected by Western blotting. Bioinformatics prediction, luciferase reporter assay, and RNA pulldown assay were performed to determine the relationship between SNHG20 and miR-335 as well as miR-335 and Galectin-3. Gain- and loss-function assays were performed to determine the role of SNHG20/miR-335/Galectin-3 in cardiac fibrosis. SNHG20 was significantly upregulated and miR-335 was downregulated in heart tissues of angiotensin II-treated mice and angiotensin II-stimulated HL-1 cells. Downregulation of SNHG20 effectively enhanced cell viability and decreased cell size of HL-1 cells and the expression levels of fibrosis-related proteins (fibronectin, collagen IaI, and α-SMA) and apoptosis-related proteins (cleaved caspase-3 and cleaved PARP), which were induced by angiotensin II treatment. Furthermore, SNHG20 elevated the expression levels of Galectin-3 by directly regulating miR-335. Our study revealed that downregulation of SNHG20 improved angiotensin II-induced cardiac fibrosis by targeting the miR-335/Galectin-3 axis, suggesting that SNHG20 is a therapeutic target for cardiac fibrosis and hypertrophy.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Chunli Qi
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Renxing Song
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Chunming Xiong
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Xiao Zhong
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Ziguang Song
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Zhongping Ning
- Shanghai University of Medicine & Health Sciences, Zhoupu Hospital, Shanghai, China
| | - Xiang Song
- Shanghai University of Medicine & Health Sciences, Zhoupu Hospital, Shanghai, China
| |
Collapse
|
88
|
Li X, Li L, Lei W, Chua HZ, Li Z, Huang X, Wang Q, Li N, Zhang H. Traditional Chinese medicine as a therapeutic option for cardiac fibrosis: Pharmacology and mechanisms. Biomed Pharmacother 2021; 142:111979. [PMID: 34358754 DOI: 10.1016/j.biopha.2021.111979] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death worldwide and cardiac fibrosis is a common pathological process for cardiac remodeling in cardiovascular diseases. Cardiac fibrosis not only accelerates the deterioration progress of diseases but also becomes a pivotal contributor for futile treatment in clinical cardiovascular trials. Although cardiac fibrosis is common and prevalent, effective medicines to provide sufficient clinical intervention for cardiac fibrosis are still unavailable. Traditional Chinese medicine (TCM) is the natural essence experienced boiling, fry, and other processing methods, including active ingredients, extracts, and herbal formulas, which have been applied to treat human diseases for a long history. Recently, research has increasingly focused on the great potential of TCM for the prevention and treatment of cardiac fibrosis. Here, we aim to clarify the identified pro-fibrotic mechanisms and intensively summarize the application of TCM in improving cardiac fibrosis by working on these mechanisms. Through comprehensively analyzing, TCM mainly regulates the following pathways during ameliorating cardiac fibrosis: attenuation of inflammation and oxidative stress, inhibition of cardiac fibroblasts activation, reduction of extracellular matrix accumulation, modulation of the renin-angiotensin-aldosterone system, modulation of autophagy, regulation of metabolic-dependent mechanisms, and targeting microRNAs. We also discussed the deficiencies and the development direction of anti-fibrotic therapies on cardiac fibrosis. The data reviewed here demonstrates that TCM shows a robust effect on alleviating cardiac fibrosis, which provides us a rich source of new drugs or drug candidates. Besides, we also hope this review may give some enlightenment for treating cardiac fibrosis in clinical practice.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Wei Lei
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hui Zi Chua
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zining Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xianglong Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China.
| | - Qilong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Nan Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
89
|
Moukette B, Barupala NP, Aonuma T, Sepulveda M, Kawaguchi S, Kim IM. Interactions between noncoding RNAs as epigenetic regulatory mechanisms in cardiovascular diseases. Methods Cell Biol 2021; 166:309-348. [PMID: 34752338 DOI: 10.1016/bs.mcb.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cardiovascular diseases (CVDs) represent the foremost cause of mortality in the United States and worldwide. It is estimated that CVDs account for approximately 17.8 million deaths each year. Despite the advances made in understanding cellular mechanisms and gene mutations governing the pathophysiology of CVDs, they remain a significant cause of mortality and morbidity. A major segment of mammalian genomes encodes for genes that are not further translated into proteins. The roles of the majority of such noncoding ribonucleic acids (RNAs) have been puzzling for a long time. However, it is becoming increasingly clear that noncoding RNAs (ncRNAs) are dynamically expressed in different cell types and have a comprehensive selection of regulatory roles at almost every step involved in DNAs, RNAs and proteins. Indeed, ncRNAs regulate gene expression through epigenetic interactions, through direct binding to target sequences, or by acting as competing endogenous RNAs. The profusion of ncRNAs in the cardiovascular system suggests that they may modulate complex regulatory networks that govern cardiac physiology and pathology. In this review, we summarize various functions of ncRNAs and highlight the recent literature on interactions between ncRNAs with an emphasis on cardiovascular disease regulation. Furthermore, as the broad-spectrum of ncRNAs potentially establishes new avenues for therapeutic development targeting CVDs, we discuss the innovative prospects of ncRNAs as therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bruno Moukette
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nipuni P Barupala
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tatsuya Aonuma
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Marisa Sepulveda
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Satoshi Kawaguchi
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Il-Man Kim
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States; Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
90
|
Yanai K, Ishibashi K, Morishita Y. Systematic Review and Meta-Analysis of Renin-Angiotensin-Aldosterone System Blocker Effects on the Development of Cardiovascular Disease in Patients With Chronic Kidney Disease. Front Pharmacol 2021; 12:662544. [PMID: 34276363 PMCID: PMC8283791 DOI: 10.3389/fphar.2021.662544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Cardiovascular events are one of the most serious complications that increase the risk of mortality and morbidity in pre-dialysis and on-dialysis chronic kidney disease (CKD) patients. Activation of the renin-angiotensin-aldosterone system (RAAS) is considered to contribute to the development of cardiovascular events in these populations. Therefore, several kinds of RAAS blockers have been frequently prescribed to prevent cardiovascular events in patients with CKD; however, their effectiveness remains controversial. This systematic review focuses on whether RAAS blockers prevent cardiovascular events in patients with CKD. Method: PubMed were searched to retrieve reference lists of eligible trials and related reviews. Randomized prospective controlled trials that investigated the effects on cardiovascular events in CKD patients that were published in English from 2010 to 2020 were included. Results: Among 167 identified studies, 11 eligible studies (n = 8,322 subjects) were included in the meta-analysis. The meta-analysis showed that RAAS blockers significantly reduced cardiovascular events in on-dialysis patients with CKD [three studies; odds ratio (OR), 0.52; 95% confidence interval (CI), 0.36 to 0.74; p = 0.0003], but there was no significant difference in pre-dialysis patients with CKD because of the heterogeneity in each study (eight studies). We also investigated the effects of each kind of RAAS blocker on cardiovascular events in CKD patients. Among the RAAS blockers, mineralocorticoid receptor antagonists significantly decreased cardiovascular events in pre-dialysis or on-dialysis patients with CKD (four studies; OR, 0.60; 95%CI, 0.50 to 0.73, p < 0.0001). However, angiotensin receptor blockers did not show significant effects (four studies; OR, 0.65; 95%CI, 0.42 to 1.01; p = 0.0529). The effects of angiotensin converting enzyme inhibitors and direct renin inhibitors on cardiovascular events in patients with CKD could not be analyzed because there were too few studies. Conclusion: Mineralocorticoid receptor antagonists may decrease cardiovascular events in pre-dialysis or on-dialysis patients with CKD.
Collapse
Affiliation(s)
- Katsunori Yanai
- First Department of Integrated Medicine, Division of Nephrology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Kenichi Ishibashi
- Department of Medical Physiology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yoshiyuki Morishita
- First Department of Integrated Medicine, Division of Nephrology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| |
Collapse
|
91
|
Passaro F, Tocchetti CG, Spinetti G, Paudice F, Ambrosone L, Costagliola C, Cacciatore F, Abete P, Testa G. Targeting fibrosis in the failing heart with nanoparticles. Adv Drug Deliv Rev 2021; 174:461-481. [PMID: 33984409 DOI: 10.1016/j.addr.2021.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/15/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) is a clinical syndrome characterized by typical symptoms and signs caused by a structural and/or functional cardiac abnormality, resulting in a reduced cardiac output and/or elevated intracardiac pressures at rest or during stress. Due to increasing incidence, prevalence and, most importantly mortality, HF is a healthcare burden worldwide, despite the improvement of treatment options and effectiveness. Acute and chronic cardiac injuries trigger the activation of neurohormonal, inflammatory, and mechanical pathways ultimately leading to fibrosis, which plays a key role in the development of cardiac dysfunction and HF. The use of nanoparticles for targeted drug delivery would greatly improve therapeutic options to identify, prevent and treat cardiac fibrosis. In this review we will highlight the mechanisms of cardiac fibrosis development to depict the pathophysiological features for passive and active targeting of acute and chronic cardiac fibrosis with nanoparticles. Then we will discuss how cardiomyocytes, immune and inflammatory cells, fibroblasts and extracellular matrix can be targeted with nanoparticles to prevent or restore cardiac dysfunction and to improve the molecular imaging of cardiac fibrosis.
Collapse
|
92
|
Hernández-Aguilar AI, Luciano-Villa CA, Tello-Flores VA, Beltrán-Anaya FO, Zubillaga-Guerrero MI, Flores-Alfaro E. Dysregulation of lncRNA-H19 in cardiometabolic diseases and the molecular mechanism involved : a systematic review. Expert Rev Mol Diagn 2021; 21:809-821. [PMID: 34133256 DOI: 10.1080/14737159.2021.1944808] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Cardiometabolic diseases are a global public health problem, with significant increases in their prevalence. Different epigenetic factors involved in the progression of metabolic alterations have been described, such as long non-coding RNAs (lncRNAs). H19 is a multifunctional lncRNA expressed from the maternal allele, with low expression after birth, except in the skeletal muscle and heart. Recent studies have linked its dysregulation to alterations in cell metabolism.Areas covered: H19 plays a role in the pathogenesis of coronary artery disease, nonalcoholic fatty liver disease, hepatic and renal fibrosis, insulin resistance, type 2 diabetes, and inflammation. H19 acts mainly as a competitive endogenous RNA of molecules involved in pathways that regulate cell metabolism. In this review, we analyzed the dysregulation of H19 in cardiometabolic diseases and its relationship with molecular alterations in different signaling pathways.Expert opinion: The association of H19 with the development of cardiometabolic diseases, indicates that H19 could be a therapeutic target and prognostic biomarker for these diseases. Controversies have been reported regarding the expression of H19 in some metabolic diseases, therefore, it is necessary to continue research to clarify its pathogenic effect in different organs.
Collapse
Affiliation(s)
- Ana Iris Hernández-Aguilar
- Faculty of Chemical‑Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| | | | | | - Fredy Omar Beltrán-Anaya
- Faculty of Chemical‑Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| | | | - Eugenia Flores-Alfaro
- Faculty of Chemical‑Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero, Mexico
| |
Collapse
|
93
|
Wang X, Huang T, Xie H. CTRP12 Alleviates Isoproterenol Induced Cardiac Fibrosis via Inhibiting the Activation of P38 Pathway. Chem Pharm Bull (Tokyo) 2021; 69:178-184. [PMID: 33518600 DOI: 10.1248/cpb.c19-01109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
C1q/tumor necrosis factor (TNF)-related protein 12 (CTRP12) plays a crucial part in cardiovascular diseases especially the coronary artery disease. Nonetheless, it is unrevealed that whether the CTRP12 participates in the progress of cardiac fibrosis. In this study, we investigated whether CTRP12 regulates pathological myocardial fibrosis. We isolated neonatal rat cardiac fibroblasts were cultured with recombination CTRP12 followed by stimulating with Isoproterenol (ISO, 100 µM) for 24 h. Then the adenovirus were used to achieve the CTRP12-overexpressed fibroblasts. In vivo, the C57/B6 mice were subjected to recombinant human CTRP12 (0.2 µg/g/d) for 2 weeks after injected with Isoproterenol (ISO, 10 mg/kg/d for 3 d then 5 mg/kg/d for 11 d, subcutaneously (s.c.), 2 weeks) and mice were also subjected to adenovirus with P38 overexpressing system to explore the mechanism. As a result, CTRP12 significantly inhibit the transformation of cardiac fibroblasts to myofibroblasts and the transcription of cardiac fibrosis-related proteins induced by ISO in vitro. The administration of CTRP12 can effectively reduce the cardiac fibrosis and enhance the cardiac function in mice hearts. The treatment with CTRP12 did not change the expression level of phosphorylated (p)-smad2, smad4, p-extracellular regulated protein kinases 1/2 and c-Jun N-terminal kinase 1/2, but it suppressed the activation of p38. Cardiac overexpression of p38 could abolish this kind of cardioprotective effects by CTRP12. In summary, the CTRP12 protect against the ISO induced cardiac fibrosis via suppressing the p38 signal pathway.
Collapse
Affiliation(s)
- Xiaoling Wang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical Collage, Huazhong University of Science and Technology
| | - Ting Huang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical Collage, Huazhong University of Science and Technology
| | - Han Xie
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical Collage, Huazhong University of Science and Technology
| |
Collapse
|
94
|
Zhao H, Yang H, Geng C, Chen Y, Tang Y, Li Z, Pang J, Shu T, Nie Y, Liu Y, Jia K, Wang J. Elevated IgE promotes cardiac fibrosis by suppressing miR-486a-5p. Theranostics 2021; 11:7600-7615. [PMID: 34158869 PMCID: PMC8210611 DOI: 10.7150/thno.47845] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Cardiac fibrosis is an important feature of cardiac remodeling and is a hallmark of heart failure. Recent studies indicate that elevated IgE plays a causal role in pathological cardiac remodeling. However, the underlying mechanism of how IgE promotes cardiac fibrosis has not been fully elucidated. Methods and Results: To explore the function of IgE in cardiac fibrosis, we stimulated mouse primary cardiac fibroblasts (CFs) with IgE and found that both IgE receptor (FcεR1) and fibrosis related proteins were increased after IgE stimulation. Specific deletion of FcεR1 in CFs alleviated angiotensin II (Ang II)-induced cardiac fibrosis in mice. To investigate the mechanisms underlying the IgE-mediated cardiac fibrosis, deep miRNA-seq was performed. Bioinformatics and signaling pathway analysis revealed that IgE upregulated Col1a1 and Col3a1 expression in CFs by repressing miR-486a-5p, with Smad1 participating downstream of miR-486a-5p in this process. Lentivirus-mediated overexpression of miR-486a-5p was found to alleviate Ang II-induced myocardial interstitial fibrosis in mice. Moreover, miR-486-5p serum levels were lower in patients with heart failure than in healthy controls, and were negatively correlated with NT-proBNP levels. Conclusions: Our study demonstrates that elevated IgE promotes pathological cardiac fibrosis by modulating miR-486a-5p and downstream factors, such as Smad1. These findings suggest new targets for pathological cardiac fibrosis intervention.
Collapse
Affiliation(s)
- Hongmei Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Hongqin Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Chi Geng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Yang Chen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Yaqin Tang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Zhiwei Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Junling Pang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Ting Shu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102308, China
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Kegang Jia
- Department of Clinical Laboratory, TEDA International Cardiovascular Hospital, Tianjin 300457, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
95
|
Huo JL, Jiao L, An Q, Chen X, Qi Y, Wei B, Zheng Y, Shi X, Gao E, Liu HM, Chen D, Wang C, Zhao W. Myofibroblast Deficiency of LSD1 Alleviates TAC-Induced Heart Failure. Circ Res 2021; 129:400-413. [PMID: 34078090 DOI: 10.1161/circresaha.120.318149] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jin-Ling Huo
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Lemin Jiao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Qi An
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Xiuying Chen
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Yuruo Qi
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Bingfei Wei
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Yichao Zheng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Xiaojing Shi
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (E.G.)
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Dong Chen
- Department of Pathology, Beijing Anzhen Hospital, Capital Medical University, China (D.C.)
| | - Cong Wang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University (J.-L.H., L.J., Q.A., X.C., Y.Q., B.W., Y.Z., X.S., H.-M.L., C.W., W.Z.)
| |
Collapse
|
96
|
Miwa S, Yokota M, Ueyama Y, Maeda K, Ogoshi Y, Seki N, Ogawa N, Nishihata J, Nomura A, Adachi T, Kitao Y, Nozawa K, Ishikawa T, Ukaji Y, Shiozaki M. Discovery of Selective Transforming Growth Factor β Type II Receptor Inhibitors as Antifibrosis Agents. ACS Med Chem Lett 2021; 12:745-751. [PMID: 34055221 DOI: 10.1021/acsmedchemlett.0c00679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/19/2021] [Indexed: 12/20/2022] Open
Abstract
Historically, modulation of transforming growth factor β (TGF-β) signaling has been deemed a rational strategy to treat many disorders, though few successful examples have been reported to date. This difficulty could be partially attributed to the challenges of achieving good specificity over many closely related enzymes that are implicated in distinct phenotypes in organ development and in tissue homeostasis. Recently, fresolimumab and disitertide, two peptidic TGF-β blockers, demonstrated significant therapeutic effects toward human skin fibrosis. Therefore, the selective blockage of TGF-β signaling assures a viable treatment option for fibrotic skin disorders such as systemic sclerosis (SSc). In this report, we disclose selective TGF-β type II receptor (TGF-βRII) inhibitors that exhibited high functional selectivity in cell-based assays. The representative compound 29 attenuated collagen type I alpha 1 chain (COL1A1) expression in a mouse fibrosis model, which suggests that selective inhibition of TGF-βRII-dependent signaling could be a new treatment for fibrotic disorders.
Collapse
Affiliation(s)
- Shohei Miwa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
- Division of Material Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Masahiro Yokota
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yoshifumi Ueyama
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Katsuya Maeda
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yosuke Ogoshi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Noriyoshi Seki
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Naoki Ogawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Jun Nishihata
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Akihiro Nomura
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Tsuyoshi Adachi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yuki Kitao
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Keisuke Nozawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Tomohiro Ishikawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Yutaka Ukaji
- Division of Material Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Makoto Shiozaki
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| |
Collapse
|
97
|
Ghosh MC, Zhang DL, Ollivierre WH, Noguchi A, Springer DA, Linehan WM, Rouault TA. Therapeutic inhibition of HIF-2α reverses polycythemia and pulmonary hypertension in murine models of human diseases. Blood 2021; 137:2509-2519. [PMID: 33512384 PMCID: PMC8109019 DOI: 10.1182/blood.2020009138] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
Polycythemia and pulmonary hypertension are 2 human diseases for which better therapies are needed. Upregulation of hypoxia-inducible factor-2α (HIF-2α) and its target genes, erythropoietin (EPO) and endothelin-1, causes polycythemia and pulmonary hypertension in patients with Chuvash polycythemia who are homozygous for the R200W mutation in the von Hippel Lindau (VHL) gene and in a murine mouse model of Chuvash polycythemia that bears the same homozygous VhlR200W mutation. Moreover, the aged VhlR200W mice developed pulmonary fibrosis, most likely due to the increased expression of Cxcl-12, another Hif-2α target. Patients with mutations in iron regulatory protein 1 (IRP1) also develop polycythemia, and Irp1-knockout (Irp1-KO) mice exhibit polycythemia, pulmonary hypertension, and cardiac fibrosis attributable to translational derepression of Hif-2α, and the resultant high expression of the Hif-2α targets EPO, endothelin-1, and Cxcl-12. In this study, we inactivated Hif-2α with the second-generation allosteric HIF-2α inhibitor MK-6482 in VhlR200W, Irp1-KO, and double-mutant VhlR200W;Irp1-KO mice. MK-6482 treatment decreased EPO production and reversed polycythemia in all 3 mouse models. Drug treatment also decreased right ventricular pressure and mitigated pulmonary hypertension in VhlR200W, Irp1-KO, and VhlR200W;Irp1-KO mice to near normal wild-type levels and normalized the movement of the cardiac interventricular septum in VhlR200Wmice. MK-6482 treatment reduced the increased expression of Cxcl-12, which, in association with CXCR4, mediates fibrocyte influx into the lungs, potentially causing pulmonary fibrosis. Our results suggest that oral intake of MK-6482 could represent a new approach to treatment of patients with polycythemia, pulmonary hypertension, pulmonary fibrosis, and complications caused by elevated expression of HIF-2α.
Collapse
Affiliation(s)
- Manik C Ghosh
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development
| | - De-Liang Zhang
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development
| | - Wade H Ollivierre
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development
| | - Audrey Noguchi
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, and
| | | | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tracey A Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development
| |
Collapse
|
98
|
Wang G, Wu H, Liang P, He X, Liu D. Fus knockdown inhibits the profibrogenic effect of cardiac fibroblasts induced by angiotensin II through targeting Pax3 thereby regulating TGF-β1/Smad pathway. Bioengineered 2021; 12:1415-1425. [PMID: 33896391 PMCID: PMC8806211 DOI: 10.1080/21655979.2021.1918522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Angiotensin II/transforming growth factor-β1 (AngII/TGF-β1) signal axis is an important regulatory pathway for atrial fibrosis, which can contribute to atrial fibrillation (AF). Fused in sarcoma (FUS) was recently found to regulate cardiac diseases. This study aimed to investigate whether FUS could regulate AngII induced fibrosis and uncover the possible mechanisms. The expression of FUS in AF patients and AngII-induced cardiac fibroblasts was measured by RT-qPCR and western blot assays. Fus was silenced in cells using short hairpin RNA (shRNA), then cell proliferation, migration, collagen synthesis and TGF-β1/Smad signaling were detected by CCK-8, wound healing and western blot assays, respectively. The possible target for Fus was predicted by searching Starbase database and verified by RNA-binding protein immunoprecipitation (RIP) and RNA pull down. Cells were overexpressed with Pax3 in the presence of Fus silence and AngII stimulation, then the above cellular processes were further evaluated. Results showed that FUS was upregulated in AF patients and AngII-induced cardiac fibroblasts. Fus knockdown inhibited AngII-enhanced cell proliferation, migration, collagen synthesis and TGF-β1/Smad signaling activation. Furthermore, Fus functions as an RNA-binding protein to bind to Pax3 mRNA and positively regulate its expression. Further studies demonstrated that Pax3 overexpression canceled the above effects of Fus knockdown on cell proliferation, migration, collagen synthesis, and TGF-β1/Smad signaling activation in AngII-induced cells. In conclusion, Fus could target Pax3 to increase the pro-fibrotic effect of AngII in cardiac fibroblasts via activating TGF-β1/Smad signaling. Knockdown of Fus/Pax3 axis may provide a potential therapy for relieving AF.
Collapse
Affiliation(s)
- Guoqiang Wang
- Department of Cardiology, Chongqing Kanghua Zhonglian Cardiovascular Hospital, Chongqing, China
| | - Hong Wu
- Department of Obstetrics and Gynecology, The People's Hospital, Chongqing, China
| | - Peng Liang
- Department of Cardiology, The People's Hospital, Chongqing, China
| | - Xiaojiao He
- Department of Cardiology, The People's Hospital, Chongqing, China
| | - Dong Liu
- Department of Cardiology, The People's Hospital, Chongqing, China
| |
Collapse
|
99
|
Tan Z, Jiang X, Zhou W, Deng B, Cai M, Deng S, Xu Y, Ding W, Chen G, Chen R, Zhang S, Zhou Y, Liu B, Zhang J. Taohong siwu decoction attenuates myocardial fibrosis by inhibiting fibrosis proliferation and collagen deposition via TGFBR1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113838. [PMID: 33460756 DOI: 10.1016/j.jep.2021.113838] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 05/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myocardial fibrosis after myocardial infarction (MI) leads to cardiac remodeling and loss of function. Taohong siwu decoction (THSWD), a well-known traditional Chinese medicinal prescription, has been clinically used to treat various cardiovascular and cerebrovascular diseases, but its potential functions in myocardial fibrosis after MI remain uncharacterized. AIM OF THE STUDY The purpose of current study was to explore the potential mechanism action and anti-myocardial fibrosis effects of treatment with THSWD in vivo and in vitro. MATERIALS AND METHODS Mouse underwent ligation of coronary artery to induce MI and divided equally into the sham group, model group and THSWD treatment groups. After 4 weeks, the effects of THSWD treatment on cardiac function were estimated by echocardiography. HE staining was used to detect the pathologic changes and Masson trichrome staining was used to estimate tissue fibrosis. To further explore the regulatory molecular mechanisms of THSWD, transcriptome analysis was performed. Furthermore, in vitro, we investigated the effect of THSWD on cell proliferation and collagen deposition in primary cardiac fibrosis cells and its possible mechanism of action. Overexpression of TGFBR1 was achieved by infection with an adenovirus vector encoding TGFBR1. RESULTS Treatment with THSWD significantly decreased myocardial fibrosis and recovered cardiac function in the post-MI mouse. The transcriptomics data imply that the TGF-β pathway might be a target in the anti-fibrosis effect of THSWD. THSWD inhibits TGF-β1-induced proliferation of primary cardiac fibroblasts. THSWD decreased collagen expression and TGFBR1 and Smad2/3 phosphorylation. Moreover, the inhibitory effect of THSWD on CFs proliferation and collagen deposition, as well as TGFBR1 signaling pathway-associated proteins expression was partially abrogated by overexpression of TGFBR1. CONCLUSION Collectively, the results implicate that THSWD attenuates myocardial fibrosis by inhibiting fibrosis proliferation and collagen deposition via inhibiting TGFBR1, and might be a potential therapeutic agent for treatment of myocardial fibrosis post-MI.
Collapse
Affiliation(s)
- Zhangbin Tan
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Xiaoli Jiang
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenyi Zhou
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Bo Deng
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Min Cai
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Suihui Deng
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Youcai Xu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenjun Ding
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Guanghong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Ruixue Chen
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Shuangwei Zhang
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Yingchun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Bin Liu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China.
| | - Jingzhi Zhang
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
100
|
Su C, Xue J, Ye C, Chen A. Role of the central renin‑angiotensin system in hypertension (Review). Int J Mol Med 2021; 47:95. [PMID: 33846799 PMCID: PMC8041481 DOI: 10.3892/ijmm.2021.4928] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/25/2021] [Indexed: 12/16/2022] Open
Abstract
Present in more than one billion adults, hypertension is the most significant modifiable risk factor for mortality resulting from cardiovascular disease. Although its pathogenesis is not yet fully understood, the disruption of the renin-angiotensin system (RAS), consisting of the systemic and brain RAS, has been recognized as one of the primary reasons for several types of hypertension. Therefore, acquiring sound knowledge of the basic science of RAS and the under- lying mechanisms of the signaling pathways associated with RAS may facilitate the discovery of novel therapeutic targets with which to promote the management of patients with cardiovascular and kidney disease. In total, 4 types of angiotensin II receptors have been identified (AT1R-AT4R), of which AT1R plays the most important role in vasoconstriction and has been most extensively studied. It has been found in several regions of the brain, and its distribution is highly associated with that of angiotensin-like immunoreactivity in nerve terminals. The effect of AT1R involves the activation of multiple media and signaling pathways, among which the most important signaling pathways are considered to be AT1R/JAK/STAT and Ras/Raf/MAPK pathways. In addition, the regulation of the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and cyclic AMP response element-binding (CREB) pathways is also closely related to the effect of ATR1. Their mechanisms of action are related to pro-inflammatory and sympathetic excitatory effects. Central AT1R is involved in almost all types of hypertension, including spontaneous hypertension, salt-sensitive hypertension, obesity-induced hypertension, renovascular hypertension, diabetic hypertension, L-NAME-induced hypertension, stress-induced hypertension, angiotensin II-induced hyper- tension and aldosterone-induced hypertension. There are 2 types of central AT1R blockade, acute blockade and chronic blockade. The latter can be achieved by chemical blockade or genetic engineering. The present review article aimed to high- light the prevalence, functions, interactions and modulation means of central AT-1R in an effort to assist in the treatment of several pathological conditions. The identification of angiotensin-derived peptides and the development of AT-2R agonists may provide a wider perspective on RAS, as well as novel therapeutic strategies.
Collapse
Affiliation(s)
- Chuanxin Su
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Jinhua Xue
- Research Center for Cardiovascular and Cerebrovascular Diseases, The University of Duisburg‑Essen, Duisburg‑Essen University, D-45122 Essen, Germany
| | - Chao Ye
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Aidong Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|