1
|
Raj Murthi S, Petry A, Shashikadze B, Stöckl JB, Schmid M, Santamaria G, Klingel K, Kračun D, Chen X, Bauer S, Schmitt JP, Flenkenthaler F, Gorham J, Toepfer CN, Potěšil D, Hruška P, Zdráhal Z, Mayer Z, Klop M, Lehmann L, Qin Y, Papanakli L, Spielmann N, Moretti A, Fröhlich T, Ewert P, Holdenrieder S, Seidman JG, Seidman CE, Görlach A, Wolf CM. Contribution of hypoxia-inducible factor 1alpha to pathogenesis of sarcomeric hypertrophic cardiomyopathy. Sci Rep 2025; 15:2132. [PMID: 39820339 PMCID: PMC11739497 DOI: 10.1038/s41598-025-85187-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) caused by autosomal-dominant mutations in genes coding for structural sarcomeric proteins, is the most common inherited heart disease. HCM is associated with myocardial hypertrophy, fibrosis and ventricular dysfunction. Hypoxia-inducible transcription factor-1α (Hif-1α) is the central master regulators of cellular hypoxia response and associated with HCM. Yet its exact role remains to be elucidated. Therefore, the effect of a cardiomyocyte-specific Hif-1a knockout (cHif1aKO) was studied in an established α-MHC719/+ HCM mouse model that exhibits the classical features of human HCM. The results show that Hif-1α protein and HIF targets were upregulated in left ventricular tissue of α-MHC719/+ mice. Cardiomyocyte-specific abolishment of Hif-1a blunted the disease phenotype, as evidenced by decreased left ventricular wall thickness, reduced myocardial fibrosis, disordered SRX/DRX state and ROS production. cHif1aKO induced normalization of pro-hypertrophic and pro-fibrotic left ventricular remodeling signaling evidenced on whole transcriptome and proteomics analysis in α-MHC719/+ mice. Proteomics of serum samples from patients with early onset HCM revealed significant modulation of HIF. These results demonstrate that HIF signaling is involved in mouse and human HCM pathogenesis. Cardiomyocyte-specific knockout of Hif-1a attenuates disease phenotype in the mouse model. Targeting Hif-1α might serve as a therapeutic option to mitigate HCM disease progression.
Collapse
Affiliation(s)
- Sarala Raj Murthi
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis LAFUGA Gene Center, LMU Munich, Munich, Germany
| | - Jan B Stöckl
- Laboratory for Functional Genome Analysis LAFUGA Gene Center, LMU Munich, Munich, Germany
| | - Manuel Schmid
- Department of Genetics, Harvard Medical School, Boston, USA
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gianluca Santamaria
- First Department of Medicine and Regenerative Medicine in Cardiovascular Diseases, Klinikum rechts der Isar, School of Medicine & Health, Technical University of Munich, Munich, Germany
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| | - Damir Kračun
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
- University Hospital Balgrist, University of Zurich and Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Xinpei Chen
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Sabine Bauer
- Experimental Cardiology, Department of Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Joachim P Schmitt
- Institute of Pharmacology, University Hospital Düsseldorf and Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine-University, Düsseldorf, Germany
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis LAFUGA Gene Center, LMU Munich, Munich, Germany
| | - Josh Gorham
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Christopher N Toepfer
- Department of Genetics, Harvard Medical School, Boston, USA
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - David Potěšil
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Pavel Hruška
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zsuzsanna Mayer
- Institute for Laboratory Medicine, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Mathieu Klop
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Luisa Lehmann
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Yishi Qin
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Laura Papanakli
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alessandra Moretti
- First Department of Medicine and Regenerative Medicine in Cardiovascular Diseases, Klinikum rechts der Isar, School of Medicine & Health, Technical University of Munich, Munich, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA Gene Center, LMU Munich, Munich, Germany
| | - Peter Ewert
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Stefan Holdenrieder
- Institute for Laboratory Medicine, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
| | | | | | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Cordula M Wolf
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, TUM University Hospital, School of Medicine & Health, Technical University of Munich, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
2
|
Zhu L, Liu Y, Wang K, Wang N. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications. Ageing Res Rev 2024; 104:102629. [PMID: 39644925 DOI: 10.1016/j.arr.2024.102629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute myocardial infarction (AMI), primarily caused by coronary atherosclerosis, initiates a series of events that culminate in the obstruction of coronary arteries, resulting in severe myocardial ischemia and hypoxia. The subsequent myocardial ischemia/reperfusion (I/R) injury further aggravates cardiac damage, leading to a decline in heart function and the risk of life-threatening complications. The complex interplay of multiple regulated cell death (RCD) pathways plays a pivotal role in the pathogenesis of AMI. Each RCD pathway is orchestrated by a symphony of molecular regulatory mechanisms, highlighting the dynamic changes and critical roles of key effector molecules. Strategic disruption or inhibition of these molecular targets offers a tantalizing prospect for mitigating or even averting the onset of RCD, thereby limiting the extensive loss of cardiomyocytes and the progression of detrimental myocardial fibrosis. This review systematically summarizes the mechanisms underlying various forms of RCD, provides an in-depth exploration of the pathogenesis of AMI through the lens of RCD, and highlights a range of promising therapeutic targets that hold the potential to revolutionize the management of AMI.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiyang Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Wang J, Li Y, Deng L, Zha Y, Zhang S. FTO suppresses cardiac fibrosis after myocardial infarction via m 6A-mediated epigenetic modification of EPRS. Mol Med 2024; 30:213. [PMID: 39538146 PMCID: PMC11562098 DOI: 10.1186/s10020-024-00985-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Cardiac fibrosis is common in myocardial infarction (MI), leading to progressive cardiac dysfunction. Studies suggested that the abnormal N6-methyladenosine (m6A) modification induced by fat mass and obesity protein (FTO) is vital in MI. However, the effects of FTO on post-infarction cardiac fibrosis have not been detected. METHODS Western blot and quantitative real-time PCR were performed to detect the expression of FTO in the fibrotic tissue of rats. The functions of FTO on collagen biosynthesis were analyzed in vitro and in vivo. The underlying targets of FTO were selected through RNA-seq with m6A-seq. The following dual luciferase reporter assay and RNA stability assay were conducted to investigate the mechanisms of FTO-mediated m6A regulation. RESULTS The expression of FTO was decreased in the fibrotic tissue of post-infarction rats. The HIF-1 signal pathway was enriched after MI. HIF-1α could bind to the promoter of FTO and inhibit its expression. Functionally, FTO inhibited collagen synthesis after MI in vitro and in vivo. Mechanistically, EPRS was selected as the underlying target of FTO-induced m6A regulation. IGF2BP3 recognized and bound to the m6A sites of EPRS mRNA, which improved its stability. EPRS was required for cardiac fibrosis induced by FTO silencing. CONCLUSIONS FTO, identified as a cardioprotective factor, suppressed collagen synthesis in post-infarction cardiac fibrosis via m6A modification, which provided a new therapeutic strategy for cardiac fibrosis.
Collapse
Affiliation(s)
- Jian Wang
- Department of Emergency, Renji Hospital, Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Yanyan Li
- Department of Cardiology, Xinhua Hospital, Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Lijie Deng
- Department of Emergency, Renji Hospital, Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Yafang Zha
- Department of Emergency, Renji Hospital, Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Song Zhang
- Department of Emergency, Renji Hospital, Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
4
|
Zhang F, Zhang L, Hu G, Chen X, Liu H, Li C, Guo X, Huang C, Sun F, Li T, Cui Z, Guo Y, Yan W, Xia Y, Liu Z, Lin Z, Duan W, Lu L, Wang X, Wang Z, Wang S, Tao L. Rectifying METTL4-Mediated N 6-Methyladenine Excess in Mitochondrial DNA Alleviates Heart Failure. Circulation 2024; 150:1441-1458. [PMID: 38686562 DOI: 10.1161/circulationaha.123.068358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/08/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Myocardial mitochondrial dysfunction underpins the pathogenesis of heart failure (HF), yet therapeutic options to restore myocardial mitochondrial function are scarce. Epigenetic modifications of mitochondrial DNA (mtDNA), such as methylation, play a pivotal role in modulating mitochondrial homeostasis. However, their involvement in HF remains unclear. METHODS Experimental HF models were established through continuous angiotensin II and phenylephrine (AngII/PE) infusion or prolonged myocardial ischemia/reperfusion injury. The landscape of N6-methyladenine (6mA) methylation within failing cardiomyocyte mtDNA was characterized using high-resolution mass spectrometry and methylated DNA immunoprecipitation sequencing. A tamoxifen-inducible cardiomyocyte-specific Mettl4 knockout mouse model and adeno-associated virus vectors designed for cardiomyocyte-targeted manipulation of METTL4 (methyltransferase-like protein 4) expression were used to ascertain the role of mtDNA 6mA and its methyltransferase METTL4 in HF. RESULTS METTL4 was predominantly localized within adult cardiomyocyte mitochondria. 6mA modifications were significantly more abundant in mtDNA than in nuclear DNA. Postnatal cardiomyocyte maturation presented with a reduction in 6mA levels within mtDNA, coinciding with a decrease in METTL4 expression. However, an increase in both mtDNA 6mA level and METTL4 expression was observed in failing adult cardiomyocytes, suggesting a shift toward a neonatal-like state. METTL4 preferentially targeted mtDNA promoter regions, which resulted in interference with transcription initiation complex assembly, mtDNA transcriptional stalling, and ultimately mitochondrial dysfunction. Amplifying cardiomyocyte mtDNA 6mA through METTL4 overexpression led to spontaneous mitochondrial dysfunction and HF phenotypes. The transcription factor p53 was identified as a direct regulator of METTL4 transcription in response to HF-provoking stress, thereby revealing a stress-responsive mechanism that controls METTL4 expression and mtDNA 6mA. Cardiomyocyte-specific deletion of the Mettl4 gene eliminated mtDNA 6mA excess, preserved mitochondrial function, and mitigated the development of HF upon continuous infusion of AngII/PE. In addition, specific silencing of METTL4 in cardiomyocytes restored mitochondrial function and offered therapeutic relief in mice with preexisting HF, irrespective of whether the condition was induced by AngII/PE infusion or myocardial ischemia/reperfusion injury. CONCLUSIONS Our findings identify a pivotal role of cardiomyocyte mtDNA 6mA and the corresponding methyltransferase, METTL4, in the pathogenesis of mitochondrial dysfunction and HF. Targeted suppression of METTL4 to rectify mtDNA 6mA excess emerges as a promising strategy for developing mitochondria-focused HF interventions.
Collapse
Affiliation(s)
- Fuyang Zhang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Zhang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guangyu Hu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiyao Chen
- Geriatrics (X.C.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Liu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Congye Li
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiong Guo
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chong Huang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fangfang Sun
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tongzheng Li
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhe Cui
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongzhen Guo
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjun Yan
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunlong Xia
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiyuan Liu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen Lin
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weixun Duan
- Cardiovascular Surgery (W.D., L.L.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Linhe Lu
- Cardiovascular Surgery (W.D., L.L.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyi Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhengyang Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shan Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Tao
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
5
|
Shen L, Huang S, Fan H, Zhai C. Colchicine Prevents Cardiac Rupture in Mice with Myocardial Infarction by Inhibiting P53-Dependent Apoptosis. Int Heart J 2024; 65:905-912. [PMID: 39261032 DOI: 10.1536/ihj.23-448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Cardiac rupture is a fatal complication following myocardial infarction (MI) and there are currently no effective pharmacological strategies for preventing this condition. In this study, we investigated the effect of colchicine on post-infarct cardiac rupture in mice and its underlying mechanisms.We induced MI in mice by permanently ligating the left anterior descending artery. Oral colchicine or vehicle was administered at a dose of 0.1 mg/kg/day from day 1 to day 7 after MI. Cultured neonatal cardiomyocytes and fibroblasts were exposed to normoxia or anoxia and treated with colchicine.Colchicine significantly improved the survival rate (colchicine, n = 46: 82.6% versus vehicle, n = 42: 61.9%, P < 0.05) at 1 week after MI. Histological analysis revealed colchicine significantly reduced the infarct size and the number of macrophages around the infarct area. Colchicine decreased apoptosis in the myocardium of the border zone and cultured cardiomyocytes and fibroblasts as assessed by TUNEL assay. Colchicine also attenuated the activation of p53 and decreased the expression of cleaved-caspase 3 and bax, as assessed by Western blotting.Colchicine prevents cardiac rupture via inhibition of apoptosis, which is attributable to the downregulation of p53 activity. Our findings suggest that colchicine may be a prospective preventive medicine for cardiac rupture, however, large clinical trials are required.
Collapse
Affiliation(s)
- Liang Shen
- Department of Cardiology, Affiliated Hospital of Jiaxing University
| | - Shaodai Huang
- Department of Cardiology, Affiliated Hospital of Jiaxing University
| | - Hongyan Fan
- Department of Cardiology, Affiliated Hospital of Jiaxing University
| | - Changlin Zhai
- Department of Cardiology, Affiliated Hospital of Jiaxing University
| |
Collapse
|
6
|
Zaruba MM, Staggl S, Ghadge SK, Maurer T, Gavranovic-Novakovic J, Jeyakumar V, Schönherr P, Wimmer A, Pölzl G, Bauer A, Messner M. Roxadustat Attenuates Adverse Remodeling Following Myocardial Infarction in Mice. Cells 2024; 13:1074. [PMID: 38994928 PMCID: PMC11240812 DOI: 10.3390/cells13131074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Activation of the CXCL12/CXCR4/ACKR3 axis is known to aid myocardial repair through ischemia-triggered hypoxia-inducible factor-1α (HIF-1α). To enhance the upregulation of HIF-1α, we administered roxadustat, a novel prolyl hydroxylase inhibitor (PHI) clinically approved by the European Medicines Agency 2021 for the treatment of renal anemia, with the purpose of improving LV function and attenuating ischemic cardiomyopathy. METHODS We evaluated roxadustat's impact on HIF-1 stimulation, cardiac remodeling, and function after MI. Therefore, we analyzed nuclear HIF-1 expression, the mRNA and protein expression of key HIF-1 target genes (RT-PCR, Western blot), inflammatory cell infiltration (immunohistochemistry), and apoptosis (TUNEL staining) 7 days after MI. Additionally, we performed echocardiography in male and female C57BL/6 mice 28 days post-MI. RESULTS We found a substantial increase in nuclear HIF-1, associated with an upregulation of HIF-1α target genes like CXCL12/CXCR4/ACKR3 at the mRNA and protein levels. Roxadustat increased the proportion of myocardial reparative M2 CD206+ cells, suggesting beneficial alterations in immune cell migration and a trend towards reduced apoptosis. Echocardiography showed that roxadustat treatment significantly preserved ejection fraction and attenuated subsequent ventricular dilatation, thereby reducing adverse remodeling. CONCLUSIONS Our findings suggest that roxadustat is a promising clinically approved treatment option to preserve myocardial function by attenuating adverse remodeling.
Collapse
Affiliation(s)
- Marc-Michael Zaruba
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Simon Staggl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Santhosh Kumar Ghadge
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030 Vienna, Austria
| | - Thomas Maurer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Jasmina Gavranovic-Novakovic
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Vivek Jeyakumar
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Patric Schönherr
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Andreas Wimmer
- Department of Surgery, Kardinal Schwarzenberg Klinikum GmbH, 5620 Salzburg, Austria;
| | - Gerhard Pölzl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Axel Bauer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Moritz Messner
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| |
Collapse
|
7
|
Shen C, Chen X, Lin Y, Yang Y. Hypoxia triggers cardiomyocyte apoptosis via regulating the m 6A methylation-mediated LncMIAT/miR-708-5p/p53 axis. Heliyon 2024; 10:e32455. [PMID: 38961902 PMCID: PMC11219354 DOI: 10.1016/j.heliyon.2024.e32455] [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: 12/02/2023] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Long-time hypoxia induced cardiomyocyte apoptosis is an important mechanism of myocardial ischemia (MI) injury. Interestingly, long noncoding RNA myocardial infarction-associated transcript (LncMIAT) has been involved in the regulation of MI injury; however, the underlying mechanism by which LncMIAT affects the progression of hypoxia-induced cardiomyocyte apoptosis remains unclear. In the present study, hypoxia was found to promote cardiomyocyte apoptosis through an increased expression of LncMIAT in vitro. Biological investigations and dual-luciferase gene reporter assay further revealed that LncMIAT was able to bind with miR-708-5p to upregulate the p53-mediated cell death of the cardiomyocytes. Silencing of LncMIAT or overexpression of miR-708-5p led to a significant reduction in p53-mediated cardiomyocyte apoptosis. The methylated RNA immunoprecipitation (MeRIP)-qPCR results showed that hypoxia exerted its effects on LncMIAT through AKLBH5-N6-methyladenosine (m6A) methylation and therefore hypoxia was shown to trigger HL-1 cardiomyocyte apoptosis via the m6A methylation-mediated LncMIAT/miR-708-5p/p53 axis. Silencing of AKLBH5 significantly alleviated the m6A methylation-mediated LncMIAT upregulation and p53-mediated cardiomyocyte apoptosis, while promoted miR-708-5p expression. Taken together, the present study highlighted that LncMIAT could act as a key biological target during hypoxia-induced cardiomyocyte apoptosis. In addition, it was shown that hypoxia could promote cardiomyocyte apoptosis through regulation of the m6A methylation-mediated LncMIAT/miR-708-5p/p53 signaling axis.
Collapse
Affiliation(s)
- Chuqiao Shen
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, 230012, PR China
| | - Xiaoqi Chen
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| | - Yixuan Lin
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, 230012, PR China
| |
Collapse
|
8
|
Najjar RS, Roy RK, Stern JE, Feresin RG. Raspberry polyphenols target molecular pathways of heart failure. J Nutr Biochem 2024; 124:109535. [PMID: 37984734 DOI: 10.1016/j.jnutbio.2023.109535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Approximately 650,000 new cases of heart failure (HF) are diagnosed annually with a 50% five-year mortality rate. HF is characterized by reduced left ventricular (LV) ejection fraction and hypertrophy of the LV wall. The pathophysiological remodeling of the heart is mediated by increased oxidative stress and inflammation. Raspberries are rich in polyphenols which may favorably impact enzymes involved in redox homeostasis while also targeting inflammatory signaling. Thus, the objective of this study was to investigate whether raspberry polyphenols could attenuate HF. Sprague Dawley rats consumed a 10% (w/w) raspberry diet for 7 weeks. At week 3, HF was surgically induced via coronary artery ligation. Hemodynamics and morphology of the heart were assessed. Expression of cardiac proteins involved in oxidative stress, inflammation, apoptosis, and remodeling were examined, and histological analysis was conducted. Additionally, human cardiomyocytes were treated with raspberry polyphenol extract (RBPE) followed by CoCl2 to chemically induce hypoxia. Redox status, apoptosis, and mitochondrial dysfunction were measured. Raspberries attenuated reductions in cardiac function and reduced morphological changes which coincided with reduced toll-like receptor (TLR)4 signaling. Reductions in oxidative stress, apoptosis, and remodeling occurred in vivo. Incubation of cardiomyocytes with RBPE attenuated CoCl2-induced oxidative stress and apoptosis despite pronounced hypoxia-inducible factor (HIF)-1α expression. These data indicate that consumption of raspberries can reduce the underlying molecular drivers of HF; thus, leading to the observed improvements in cardiac functional capacity and morphology. This dietary strategy may be an effective alternative strategy for treating HF. However, further investigation into alternative models of HF is warranted.
Collapse
Affiliation(s)
- Rami S Najjar
- Department of Nutrition, Georgia State University, Atlanta, Georgia, USA; Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA
| | - Rafaela G Feresin
- Department of Nutrition, Georgia State University, Atlanta, Georgia, USA; Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
9
|
Pan J, Zhang L, Li D, Li Y, Lu M, Hu Y, Sun B, Zhang Z, Li C. Hypoxia-inducible factor-1: Regulatory mechanisms and drug therapy in myocardial infarction. Eur J Pharmacol 2024; 963:176277. [PMID: 38123007 DOI: 10.1016/j.ejphar.2023.176277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Myocardial infarction (MI), an acute cardiovascular disease characterized by coronary artery blockage, inadequate blood supply, and subsequent ischemic necrosis of the myocardium, is one of the leading causes of death. The cellular, physiological, and pathological responses following MI are complex, involving multiple intertwined pathological mechanisms. Hypoxia-inducible factor-1 (HIF-1), a crucial regulator of hypoxia, plays a significant role in of the development of MI by modulating the behavior of various cells such as cardiomyocytes, endothelial cells, macrophages, and fibroblasts under hypoxic conditions. HIF-1 regulates various post-MI adaptive reactions to acute ischemia and hypoxia through various mechanisms. These mechanisms include angiogenesis, energy metabolism, oxidative stress, inflammatory response, and ventricular remodeling. With its crucial role in MI, HIF-1 is expected to significantly influence the treatment of MI. However, the drugs available for the treatment of MI targeting HIF-1 are currently limited, and most contain natural compounds. The development of precision-targeted drugs modulating HIF-1 has therapeutic potential for advancing MI treatment research and development. This study aimed to summarize the regulatory role of HIF-1 in the pathological responses of various cells following MI, the diverse mechanisms of action of HIF-1 in MI, and the potential drugs targeting HIF-1 for treating MI, thus providing the theoretical foundations for potential clinical therapeutic targets.
Collapse
Affiliation(s)
- Jinyuan Pan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Dongxiao Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanlong Hu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Bowen Sun
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chao Li
- Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, 266000, China.
| |
Collapse
|
10
|
Ishimaru K, Ikeda M, Miyamoto HD, Furusawa S, Abe K, Watanabe M, Kanamura T, Fujita S, Nishimura R, Toyohara T, Matsushima S, Koumura T, Yamada K, Imai H, Tsutsui H, Ide T. Deferasirox Targeting Ferroptosis Synergistically Ameliorates Myocardial Ischemia Reperfusion Injury in Conjunction With Cyclosporine A. J Am Heart Assoc 2024; 13:e031219. [PMID: 38158218 PMCID: PMC10863836 DOI: 10.1161/jaha.123.031219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Ferroptosis, an iron-dependent form of regulated cell death, is a major cell death mode in myocardial ischemia reperfusion (I/R) injury, along with mitochondrial permeability transition-driven necrosis, which is inhibited by cyclosporine A (CsA). However, therapeutics targeting ferroptosis during myocardial I/R injury have not yet been developed. Hence, we aimed to investigate the therapeutic efficacy of deferasirox, an iron chelator, against hypoxia/reoxygenation-induced ferroptosis in cultured cardiomyocytes and myocardial I/R injury. METHODS AND RESULTS The effects of deferasirox on hypoxia/reoxygenation-induced iron overload in the endoplasmic reticulum, lipid peroxidation, and ferroptosis were examined in cultured cardiomyocytes. In a mouse model of I/R injury, the infarct size and adverse cardiac remodeling were examined after treatment with deferasirox, CsA, or both in combination. Deferasirox suppressed hypoxia- or hypoxia/reoxygenation-induced iron overload in the endoplasmic reticulum, lipid peroxidation, and ferroptosis in cultured cardiomyocytes. Deferasirox treatment reduced iron levels in the endoplasmic reticulum and prevented increases in lipid peroxidation and ferroptosis in the I/R-injured myocardium 24 hours after I/R. Deferasirox and CsA independently reduced the infarct size after I/R injury to a similar degree, and combination therapy with deferasirox and CsA synergistically reduced the infarct size (infarct area/area at risk; control treatment: 64±2%; deferasirox treatment: 48±3%; CsA treatment: 48±4%; deferasirox+CsA treatment: 37±3%), thereby ameliorating adverse cardiac remodeling on day 14 after I/R. CONCLUSIONS Combination therapy with deferasirox and CsA may be a clinically feasible and effective therapeutic approach for limiting I/R injury and ameliorating adverse cardiac remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Kosei Ishimaru
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shun Furusawa
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Ko Abe
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masatsugu Watanabe
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takuya Kanamura
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Satoshi Fujita
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Ryohei Nishimura
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takayuki Toyohara
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - Tomoko Koumura
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical SciencesKitasato UniversityTokyoJapan
| | - Ken‐ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical SciencesKyushu UniversityFukuokaJapan
| | - Hirotaka Imai
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical SciencesKitasato UniversityTokyoJapan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- School of Medicine and Graduate SchoolInternational University of Health and WelfareFukuokaJapan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
11
|
Rihan M, Sharma SS. Inhibition of Pyruvate kinase M2 (PKM2) by shikonin attenuates isoproterenol-induced acute myocardial infarction via reduction in inflammation, hypoxia, apoptosis, and fibrosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:145-159. [PMID: 37382601 DOI: 10.1007/s00210-023-02593-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Myocardial infarction (MI) is a major cause of mortality and disability globally. MI results from acute or chronic myocardial ischemia characterized by an imbalance of oxygen demand and supply, leading to irreversible myocardial injury. Despite several significant efforts in the understanding of MI, the therapy of MI is not satisfactory due to its complicated pathophysiology. Recently, therapeutic potential of targeting pyruvate kinase M2 (PKM2) has been postulated in several cardiovascular diseases. PKM2 gene knockout and expression studies implicated the role of PKM2 in MI. However, the effects of pharmacological interventions targeting PKM2 have not been investigated in MI. Therefore, in the present study, effect of PKM2 inhibitor has been investigated in the MI along with elucidation of possible mechanism(s). MI in rats was induced by administrations of isoproterenol (ISO) at a dose of 100 mg/kg s.c. for two consecutives days at 24-h interval. At the same time, shikonin (PKM2 inhibitor) was administered at 2 and 4 mg/kg in ISO-induced MI rats. After the shikonin treatment, the ventricular functions were measured using a PV-loop system. Plasma MI injury markers, cardiac histology, and immunoblotting were performed to elucidate the molecular mechanism. Treatment of shikonin 2 and 4 mg/kg ameliorated cardiac injury, reduced infarct size, biochemical alterations, ventricular dysfunction, and cardiac fibrosis in ISO-induced MI. Expression of PKM2 in the ventricle was reduced while PKM1 expression increased in the shikonin treated group, indicating PKM2 inhibition restores PKM1 expression. In addition, PKM splicing protein (hnRNPA2B1 & PTBP1), HIF-1α, and caspase-3 expression were reduced after shikonin treatment. Our findings suggest that pharmacological inhibition of PKM2 with shikonin could be a potential therapeutic strategy to treat MI.
Collapse
Affiliation(s)
- Mohd Rihan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar (Mohali), 160062, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S Nagar (Mohali), 160062, Punjab, India.
| |
Collapse
|
12
|
Basuthakur P, Roy A, Patra CR, Chakravarty S. Therapeutic potentials of terbium hydroxide nanorods for amelioration of hypoxia-reperfusion injury in cardiomyocytes. BIOMATERIALS ADVANCES 2023; 153:213531. [PMID: 37429046 DOI: 10.1016/j.bioadv.2023.213531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/06/2023] [Accepted: 06/17/2023] [Indexed: 07/12/2023]
Abstract
Myocardial hypoxia reperfusion (H/R) injury is the paradoxical exacerbation of myocardial damage, caused by the sudden restoration of blood flow to hypoxia affected myocardium. It is a critical contributor of acute myocardial infarction, which can lead to cardiac failure. Despite the current pharmacological advancements, clinical translation of cardioprotective therapies have proven challenging. As a result, researchers are looking for alternative approaches to counter the disease. In this regard, nanotechnology, with its versatile applications in biology and medicine, can confer broad prospects for treatment of myocardial H/R injury. Herein, we attempted to explore whether a well-established pro-angiogenic nanoparticle, terbium hydroxide nanorods (THNR) can ameliorate myocardial H/R injury. For this study, in vitro H/R-injury model was established in rat cardiomyocytes (H9c2 cells). Our investigations demonstrated that THNR enhance cardiomyocyte survival against H/R-induced cell death. This pro-survival effect of THNR is associated with reduction of oxidative stress, lipid peroxidation, calcium overload, restoration of cytoskeletal integrity and mitochondrial membrane potential as well as augmentation of cellular anti-oxidant enzymes such as glutathione-s-transferase (GST) and superoxide dismutase (SOD) to counter H/R injury. Molecular analysis revealed that the above observations are traceable to the predominant activation of PI3K-AKT-mTOR and ERK-MEK signalling pathways by THNR. Concurrently, THNR also exhibit apoptosis inhibitory effects mainly by suppression of pro-apoptotic proteins like Cytochrome C, Caspase 3, Bax and p53 with simultaneous restoration of anti-apoptotic protein, Bcl-2 and Survivin. Thus, considering the above attributes, we firmly believe that THNR have the potential to be developed as an alternative approach for amelioration of H/R injury in cardiomyocytes.
Collapse
Affiliation(s)
- Papia Basuthakur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arpita Roy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
13
|
Furusawa S, Ikeda M, Ide T, Kanamura T, Miyamoto HD, Abe K, Ishimaru K, Watanabe M, Tsutsui Y, Miyake R, Fujita S, Tohyama T, Matsushima S, Baba Y, Tsutsui H. Cardiac Autoantibodies Against Cardiac Troponin I in Post-Myocardial Infarction Heart Failure: Evaluation in a Novel Murine Model and Applications in Therapeutics. Circ Heart Fail 2023; 16:e010347. [PMID: 37522180 DOI: 10.1161/circheartfailure.122.010347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/05/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Cardiac autoantibodies (cAAbs) are involved in the progression of adverse cardiac remodeling in heart failure (HF). However, our understanding of cAAbs in HF is limited owing to the absence of relevant animal models. Herein, we aimed to establish and characterize a murine model of cAAb-positive HF after myocardial infarction (MI), thereby facilitating the development of therapeutics targeting cAAbs in post-MI HF. METHODS MI was induced in BALB/c mice. Plasma cAAbs were evaluated using modified Western blot-based methods. Prognosis, cardiac function, inflammation, and fibrosis were compared between cAAb-positive and cAAb-negative MI mice. Rapamycin was used to inhibit cAAb production. RESULTS Common cAAbs in BALB/c MI mice targeted cTnI (cardiac troponin I). Herein, 71% (24/34) and 44% (12/27) of the male and female MI mice, respectively, were positive for cAAbs against cTnI (cTnIAAb). Germinal centers were formed in the spleens and mediastinal lymph nodes of cTnIAAb-positive MI mice. cTnIAAb-positive MI mice showed progressive cardiac remodeling with a worse prognosis (P=0.014, by log-rank test), which was accompanied by cardiac inflammation, compared with that in cTnIAAb-negative MI mice. Rapamycin treatment during the first 7 days after MI suppressed cTnIAAb production (cTnIAAb positivity, 59% [29/49] and 7% [2/28] in MI mice treated with vehicle and rapamycin, respectively; P<0.001, by Pearson χ2 test), consequently improving the survival and ameliorating cardiac inflammation, cardiac remodeling, and HF in MI mice. CONCLUSIONS The present post-MI HF model may accelerate our understanding of cTnIAAb and support the development of therapeutics against cTnIAAbs in post-MI HF.
Collapse
Affiliation(s)
- Shun Furusawa
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Kanamura
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ko Abe
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosei Ishimaru
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatsugu Watanabe
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (M.W.)
| | - Yoshitomo Tsutsui
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Miyake
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Fujita
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Tohyama
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Clinical and Translational Research of Kyushu University Hospital, Fukuoka, Japan (T.T.)
| | - Shouji Matsushima
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Baba
- Department of Molecular Genetics, Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan (Y.B.)
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, T.T., S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (S. Furusawa, M.I., T.I., T.K., H.D.M., K.A., K.I., M.W., Y.T., R.M., S. Fujita, S.M., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- School of Medicine and Graduate School, International University of Health and Welfare, Fukuoka, Japan (H.T.)
| |
Collapse
|
14
|
Zhang Y, Hong L, Li X, Li Y, Zhang X, Jiang J, Shi F, Diao H. M1 macrophage-derived exosomes promote autoimmune liver injury by transferring long noncoding RNA H19 to hepatocytes. MedComm (Beijing) 2023; 4:e303. [PMID: 37398637 PMCID: PMC10310975 DOI: 10.1002/mco2.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 07/04/2023] Open
Abstract
Exosomes mediate intercellular communication by transmitting active molecules. The function of long noncoding RNA (lncRNA) H19 in autoimmune liver injury is unclear. Concanavalin A (ConA)-induced liver injury is well-characterized immune-mediated hepatitis. Here, we showed that lncRNA H19 expression was increased in the liver after ConA treatment, accompanied by increased exosome secretion. Moreover, injection of AAV-H19 aggravated ConA-induced hepatitis, with an increase in hepatocyte apoptosis. However, GW4869, an exosome inhibitor, alleviated ConA-induced liver injury and inhibited the upregulation of lncRNA H19. Intriguingly, lncRNA H19 expression in the liver was significantly downregulated, after macrophage depletion. Importantly, the lncRNA H19 was primarily expressed in type I macrophage (M1) and encapsulated in M1-derived exosomes. Furthermore, H19 was transported from M1 to hepatocytes via exosomes, and exosomal H19 dramatically induced hepatocytes apoptosis both in vitro and vivo. Mechanistically, H19 upregulated the transcription of hypoxia-inducible factor-1 alpha (HIF-1α), which accumulated in the cytoplasm and mediated hepatocyte apoptosis by upregulating p53. M1-derived exosomal lncRNA H19 plays a pivotal role in ConA-induced hepatitis through the HIF-1α-p53 signaling pathway. These findings identify M1 macrophage-derived exosomal H19 as a novel target for the treatment of autoimmune liver diseases.
Collapse
Affiliation(s)
- Yongting Zhang
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Liang Hong
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Xuehui Li
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Yuyu Li
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Xujun Zhang
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Jingjing Jiang
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Fan Shi
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Hongyan Diao
- State Key Laboratory for Diagnosis & Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseaseCollaborative Innovation Center for Diagnosis & Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
15
|
Sharma S, Rana AK, Rahmatkar SN, Patial V, Singh D. Protective effect of Nardostachys jatamansi extract against lithium-pilocarpine-induced spontaneous recurrent seizures and associated cardiac irregularities in a rat model. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116280. [PMID: 36813245 DOI: 10.1016/j.jep.2023.116280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nardostachys jatamansi (D.Don) DC. is a perennial herbaceous medicinal plant widely used for the ethnomedical treatment of various ailments. The underground parts of the plants are used in traditional medicine to manage epilepsy and other cardiovascular conditions. AIM OF THE STUDY The present study was undertaken to investigate the efficacy of a characterized hydroalcoholic extract (NJET) of Nardostachys jatamansi in the lithium-pilocarpine rat model of spontaneous recurrent seizures (SRS) and associated cardiac irregularities. MATERIALS AND METHODS NJET was prepared by percolation using 80% ethanol. The dried NEJT was subjected to UHPLC-qTOF-MS/MS for chemical characterization. Molecular docking studies were performed using the characterized compounds to understand mTOR interactions. The animals showing SRS following lithium-pilocarpine administration were treated with NJET for 6 weeks. Afterward, seizure severity, cardiac parameters, serum biochemistry, and histopathological parameters were studied. The cardiac tissue was processed for specific protein and gene expression studies. RESULTS The UHPLC-qTOF-MS/MS characterized 13 compounds in NJET. The identified compounds subjected to molecular docking showed promising binding affinities toward mTOR. There was a dose-dependent decrease in the severity of SRS following the extract administration. A reduction in mean arterial pressure and serum biochemical markers (lactate dehydrogenase and creatine kinase) was also observed following NJET treatment in epileptic animals. Histopathological investigations revealed reduced degenerative changes and decreased fibrosis following the extract treatment. The cardiac mRNA level of Mtor, Rps6, Hif1a, and Tgfb3 was reduced in the extract-treated groups. Further, a similar reduction in the protein expression of p-mTOR and HIF-1α was also observed following NJET treatment in the cardiac tissue. CONCLUSIONS The results concluded that NJET treatment reduces lithium-pilocarpine-induced recurrent seizures and associated cardiac irregularities via downregulation of the mTOR signalling pathway.
Collapse
Affiliation(s)
- Supriya Sharma
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anil Kumar Rana
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shubham Nilkanth Rahmatkar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
16
|
Sato T, Takeda N. The roles of HIF-1α signaling in cardiovascular diseases. J Cardiol 2023; 81:202-208. [PMID: 36127212 DOI: 10.1016/j.jjcc.2022.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 12/29/2022]
Abstract
Oxygen is essential for living organisms. Molecular oxygen binds to hemoglobin and is delivered to every organ in the body. In several cardiovascular diseases or anemia, local oxygen tension drops below its physiological level and tissue hypoxia develops. In such conditions, the expression of hypoxia-responsive genes increases to alleviate the respective condition. The hypoxia-responsive genes include the genes coding erythropoietin (EPO), vascular endothelial growth factor-A, and glycolytic enzymes. Hypoxia-inducible factor (HIF)-1α, HIF-2α, and HIF-3α are transcription factors that regulate the hypoxia-responsive genes. The HIF-α proteins are continuously degraded by an oxygen-dependent degrading pathway involving HIF-prolyl hydroxylases (HIF-PHs) and von Hippel-Lindau tumor suppressor protein. However, upon hypoxia, this degradation ceases and the HIF-α proteins form heterodimers with HIF-1β (a constitutive subunit of HIF), which results in the induction of hypoxia responsive genes. HIF-1α and HIF-2α are potential therapeutic targets for renal anemia, where EPO production is impaired due to chronic kidney diseases. Small molecule HIF-PH inhibitors are currently used to activate HIF-α signaling and to increase plasma hemoglobin levels by restoring EPO production. In this review, we will discuss the current understanding of the roles of the HIF-α signaling pathway in cardiovascular diseases. This will include the roles of HIF-1α in cardiomyocytes as well as in stromal cells including macrophages.
Collapse
Affiliation(s)
- Tatsuyuki Sato
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
17
|
Zhou F, Zhang Z, Wang M, Zhu W, Ruan J, Long H, Zhang Y, Gu N. Guanxin V attenuates myocardial ischaemia reperfusion injury through regulating iron homeostasis. PHARMACEUTICAL BIOLOGY 2022; 60:1884-1898. [PMID: 36215067 PMCID: PMC9553176 DOI: 10.1080/13880209.2022.2123934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 06/16/2023]
Abstract
CONTEXT Guanxin V (GX), a traditional Chinese medicine formula, is safe and effective in the treatment of coronary artery disease. However, its protective effect on myocardial ischaemia reperfusion injury (MIRI) is unclear. OBJECTIVE To investigate the cardioprotective effect of GX on MIRI and explore the potential mechanism. MATERIALS AND METHODS Sprague-Dawley male rats were divided into Sham, MIRI and MIRI + GX groups. GX (6 g/kg) was administered to rats via intragastric administration for seven days before ischaemia reperfusion (IR) surgery. The infarct size, histopathology, serum enzyme activities, ultrastructure of the cardiac mitochondria were assessed. H9c2 cells were pre-treated with GX (0.5 mg/mL), and then exposed to hypoxia/reoxygenation (HR). The cell viability and LDH levels were measured. Network pharmacology was conducted to predict the potential mechanism. The related targets of GX were predicted using the TCMSP database, DrugBank database, etc. Finally, pharmacological experiments were used to validate the predicted results. RESULTS In vivo, GX significantly reduced the myocardial infarct size from 56.33% to 17.18%, decreased the levels of AST (239.32 vs. 369.18 U/L), CK-MB (1324.61 vs. 2066.47 U/L) and LDH (1245.26 vs. 1969.62 U/L), and reduced mitochondrial damage. In vitro, GX significantly increased H9c2 cell viability (IC50 = 3.913 mg/mL) and inhibited the release of LDH (207.35 vs. 314.33). In addition, GX could maintain iron homeostasis and reduce oxidative stress level by regulating iron metabolism-associated proteins. CONCLUSIONS GX can attenuate MIRI via regulating iron homeostasis, indicating that GX may act as a potential candidate for the treatment of MIRI.
Collapse
Affiliation(s)
- Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengguang Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meiyuan Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weina Zhu
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Ruan
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongyan Long
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Gu
- Department of Cardiovascular Disease, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
18
|
Abe K, Ikeda M, Ide T, Tadokoro T, Miyamoto HD, Furusawa S, Tsutsui Y, Miyake R, Ishimaru K, Watanabe M, Matsushima S, Koumura T, Yamada KI, Imai H, Tsutsui H. Doxorubicin causes ferroptosis and cardiotoxicity by intercalating into mitochondrial DNA and disrupting Alas1-dependent heme synthesis. Sci Signal 2022; 15:eabn8017. [PMID: 36318618 DOI: 10.1126/scisignal.abn8017] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Clinical use of doxorubicin (DOX) is limited because of its cardiotoxicity, referred to as DOX-induced cardiomyopathy (DIC). Mitochondria-dependent ferroptosis, which is triggered by iron overload and excessive lipid peroxidation, plays a pivotal role in the progression of DIC. Here, we showed that DOX accumulated in mitochondria by intercalating into mitochondrial DNA (mtDNA), inducing ferroptosis in an mtDNA content-dependent manner. In addition, DOX disrupted heme synthesis by decreasing the abundance of 5'-aminolevulinate synthase 1 (Alas1), the rate-limiting enzyme in this process, thereby impairing iron utilization, resulting in iron overload and ferroptosis in mitochondria in cultured cardiomyocytes. Alas1 overexpression prevented this outcome. Administration of 5-aminolevulinic acid (5-ALA), the product of Alas1, to cultured cardiomyocytes and mice suppressed iron overload and lipid peroxidation, thereby preventing DOX-induced ferroptosis and DIC. Our findings reveal that the accumulation of DOX and iron in mitochondria cooperatively induces ferroptosis in cardiomyocytes and suggest that 5-ALA can be used as a potential therapeutic agent for DIC.
Collapse
Affiliation(s)
- Ko Abe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Immunoregulatory Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shun Furusawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshitomo Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryo Miyake
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kosei Ishimaru
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Masatsugu Watanabe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomoko Koumura
- Departments of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hirotaka Imai
- Departments of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
19
|
Ikeda M, Ide T, Matsushima S, Ikeda S, Okabe K, Ishikita A, Tadokoro T, Sada M, Abe K, Sato M, Hanada A, Arai S, Ohtani K, Nonami A, Mizuno S, Morimoto S, Motohashi S, Akashi K, Taniguchi M, Tsutsui H. Immunomodulatory Cell Therapy Using αGalCer-Pulsed Dendritic Cells Ameliorates Heart Failure in a Murine Dilated Cardiomyopathy Model. Circ Heart Fail 2022; 15:e009366. [PMID: 36268712 PMCID: PMC9760469 DOI: 10.1161/circheartfailure.122.009366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a life-threatening disease, resulting in refractory heart failure. An immune disorder underlies the pathophysiology associated with heart failure progression. Invariant natural killer T (iNKT) cell activation is a prospective therapeutic strategy for ischemic heart disease. However, its efficacy in nonischemic cardiomyopathy, such as DCM, remains to be elucidated, and the feasible modality for iNKT cell activation in humans is yet to be validated. METHODS Dendritic cells isolated from human volunteers were pulsed with α-galactosylceramide ex vivo, which were used as α-galactosylceramide-pulsed dendritic cells (αGCDCs). We treated DCM mice harboring mutated troponin TΔK210/ΔK210 with αGCDCs and evaluated the efficacy of iNKT cell activation on heart failure in DCM mice. Furthermore, we investigated the molecular basis underlying its therapeutic effects in these mice and analyzed primary cardiac cells under iNKT cell-secreted cytokines. RESULTS The number of iNKT cells in the spleens of DCM mice was reduced compared with that in wild-type mice, whereas αGCDC treatment activated iNKT cells, prolonged survival of DCM mice, and prevented decline in the left ventricular ejection fraction for 4 weeks, accompanied by suppressed interstitial fibrosis. Mechanistically, αGCDC treatment suppressed TGF (transforming growth factor)-β signaling and expression of fibrotic genes and restored vasculature that was impaired in DCM hearts by upregulating angiopoietin 1 (Angpt1) expression. Consistently, IFNγ (interferon gamma) suppressed TGF-β-induced Smad2/3 signaling and the expression of fibrotic genes in cardiac fibroblasts and upregulated Angpt1 expression in cardiomyocytes via Stat1. CONCLUSIONS Immunomodulatory cell therapy with αGCDCs is a novel therapeutic strategy for heart failure in DCM.
Collapse
Affiliation(s)
- Masataka Ikeda
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Immunoregulatory Cardiovascular Medicine (M.I., T.I.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Immunoregulatory Cardiovascular Medicine (M.I., T.I.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Ikeda
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Okabe
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihito Ishikita
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Sada
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ko Abe
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Midori Sato
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiko Hanada
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinobu Arai
- Department of Early Childhood and Elementary Education, Faculty of Education, Nakamura Gakuen University, Fukuoka, Japan (S.A.)
| | - Kisho Ohtani
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Nonami
- Center for Advanced Medical Innovation, Kyushu University Hospital, Fukuoka, Japan (A.N.)
| | - Shinichi Mizuno
- Department of Health Sciences (S. Mizuno), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sachio Morimoto
- Department of Health Sciences at Fukuoka, International University of Health and Welfare, Japan (S. Morimoto)
| | - Shinichiro Motohashi
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medical Immunology, Graduate School of Medicine, Chiba University, Japan (S. Motohashi)
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science (K. Akashi), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Taniguchi
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan (M.T.)
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology (M.I., T.I., S.M., S.I., K.O., A.I., T.T., M.S., K. Abe, M.S., A.H., K.O., H.T.), Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
20
|
Miyamoto HD, Ikeda M, Ide T, Tadokoro T, Furusawa S, Abe K, Ishimaru K, Enzan N, Sada M, Yamamoto T, Matsushima S, Koumura T, Yamada KI, Imai H, Tsutsui H. Iron Overload via Heme Degradation in the Endoplasmic Reticulum Triggers Ferroptosis in Myocardial Ischemia-Reperfusion Injury. JACC Basic Transl Sci 2022; 7:800-819. [PMID: 36061338 PMCID: PMC9436815 DOI: 10.1016/j.jacbts.2022.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/16/2022] [Accepted: 03/16/2022] [Indexed: 10/25/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a promising therapeutic target to improve clinical outcomes after acute myocardial infarction. Ferroptosis, triggered by iron overload and excessive lipid peroxides, is reportedly involved in I/R injury. However, its significance and mechanistic basis remain unclear. Here, we show that glutathione peroxidase 4 (GPx4), a key endogenous suppressor of ferroptosis, determines the susceptibility to myocardial I/R injury. Importantly, ferroptosis is a major mode of cell death in I/R injury, distinct from mitochondrial permeability transition (MPT)-driven necrosis. This suggests that the use of therapeutics targeting both modes is an effective strategy to further reduce the infarct size and thereby ameliorate cardiac remodeling after I/R injury. Furthermore, we demonstrate that heme oxygenase 1 up-regulation in response to hypoxia and hypoxia/reoxygenation degrades heme and thereby induces iron overload and ferroptosis in the endoplasmic reticulum (ER) of cardiomyocytes. Collectively, ferroptosis triggered by GPx4 reduction and iron overload in the ER is distinct from MPT-driven necrosis in both in vivo phenotype and in vitro mechanism for I/R injury. The use of therapeutics targeting ferroptosis in conjunction with cyclosporine A can be a promising strategy for I/R injury.
Collapse
Key Words
- AMI, acute myocardial infarction
- CsA, cyclosporine A
- CypD, cyclophilin D
- DXZ, dexrazoxane
- ER, endoplasmic reticulum
- Fer-1, ferrostatin-1
- GPx4, glutathione peroxidase 4
- H/R, hypoxia-reoxygenation
- HF, heart failure
- HO-1, heme oxygenase 1
- I/R, ischemia-reperfusion
- LP, lipid peroxide
- MPT, mitochondrial permeability transition
- MPT-driven necrosis
- RCD, regulated cell death
- STEMI, ST-segment elevation myocardial infarction
- cyclosporine A
- ferroptosis
- glutathione peroxidase 4
- ischemia-reperfusion injury
Collapse
Affiliation(s)
- Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shun Furusawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ko Abe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosei Ishimaru
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuyuki Enzan
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Sada
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taishi Yamamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoko Koumura
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Imai
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
21
|
Zeng X, Lin C, Sun Y, Zhang J. Serum TP53 Protein Level as a Sensitive Biomarker for the Diagnosis of Myocardial Damage in Children. MEDICAL SCIENCE MONITOR : INTERNATIONAL MEDICAL JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022; 28:e936248. [PMID: 35751366 PMCID: PMC9241449 DOI: 10.12659/msm.936248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND High levels of TP53 protein can lead to apoptosis of myocardial cells. However, TP53 protein influence of myocardial damage remains unclear. This prospective study investigated the involvement of TP53 protein in secondary myocardial damage in children up to 18 years of age. MATERIAL AND METHODS Serum TP53 protein, N-terminal prohormone B-type natriuretic peptide (NT-ProBNP), cardiac troponin-I (cTnI), and creatine kinase isoenzyme MB (CK-MB) concentrations were measured in 50 hospitalized patients with secondary myocardial damage, 50 hospitalized patients without myocardial damage, and 50 healthy individuals (control). Cardiac damage was diagnosed based on cTnI, NT-ProBNP, and CK-MB levels, with electrocardiographic evidence as the reference. The appropriate cut-off value of TP53 protein for secondary myocardial damage was analyzed by receiver operating characteristic (ROC) curves. RESULTS The serum TP53 protein, NT-ProBNP, cTnI, and CK-MB concentrations of the patients with and without myocardial damage were 10.20±1.20 and 0.30±0.10 ng/L, 505.30 and 107.8 ng/L, 0.23±0.13 and 0.02±0.01 μg/L, and 28.30±5.13 and 12.24±4.29 IU/L, respectively. For the 50 patients with myocardial damage, the area under the ROC curve for serum TP53 protein, NT-ProBNP, cTnI, and CK-MB concentrations were 0.89 (95% CI: 0.81-0.95), 0.83 (95% CI: 0.77-0.91), 0.92 (95% CI: 0.84-0.97), and 0.85 (95% CI: 0.78-0.93), respectively, and the diagnostic cut-off values were 12.00 ng/L, 500.00 ng/L, 0.16 μg/L, and 27.00 IU/L, respectively, with positive likelihood ratios of 20.8, 13.2, 24.6, and 15.6. CONCLUSIONS TP53 protein is a valid biomarker of secondary myocardial damage in pediatric patients and can be diagnostic.
Collapse
Affiliation(s)
- Xianglin Zeng
- Department of Pediatrics, Shunde Women's and Children's Hospital of Guangdong Medical University, Foshan, Guangdong, China (mainland)
| | - Chunwang Lin
- Department of Pediatrics, Shunde Women's and Children's Hospital of Guangdong Medical University, Foshan, Guangdong, China (mainland)
| | - Yanna Sun
- Department of Pediatrics, Shunde Women's and Children's Hospital of Guangdong Medical University, Foshan, Guangdong, China (mainland)
| | - Jianping Zhang
- Department of Pediatrics, Shunde Women's and Children's Hospital of Guangdong Medical University, Foshan, Guangdong, China (mainland)
| |
Collapse
|
22
|
Deng R, Wang Y, Bu Y, Wu H. BNIP3 mediates the different adaptive responses of fibroblast-like synovial cells to hypoxia in patients with osteoarthritis and rheumatoid arthritis. Mol Med 2022; 28:64. [PMID: 35690741 PMCID: PMC9188199 DOI: 10.1186/s10020-022-00490-9] [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: 12/01/2021] [Accepted: 05/25/2022] [Indexed: 01/01/2023] Open
Abstract
Background Hypoxia is one of the important characteristics of synovial microenvironment in rheumatoid arthritis (RA), and plays an important role in synovial hyperplasia. In terms of cell survival, fibroblast-like synovial cells (FLSs) are relatively affected by hypoxia. In contrast, fibroblast-like synovial cells from patients with RA (RA-FLSs) are particularly resistant to hypoxia-induced cell death. The purpose of this study was to evaluate whether fibroblast-like synovial cells in patients with osteoarthritis (OA-FLSs) and RA-FLSs have the same adaptation to hypoxia. Methods CCK-8, flow cytometry and BrdU were used to detect the proliferation of OA-FLSs and RA-FLSs under different oxygen concentrations. Apoptosis was detected by AV/PI, TUNEL and Western blot, mitophagy was observed by electron microscope, laser confocal microscope and Western blot, the state of mitochondria was detected by ROS and mitochondrial membrane potential by flow cytometry, BNIP3 and HIF-1α were detected by Western blot and RT-qPCR. The silencing of BNIP3 was achieved by stealth RNA system technology. Results After hypoxia, the survival rate of OA-FLSs decreased, while the proliferation activity of RA-FLSs further increased. Hypoxia induced an increase in apoptosis and inhibition of mitophagy in OA-FLSs, but not in RA-FLSs. Hypoxia led to a more lasting adaptive response. RA-FLSs displayed a more significant increase in the expression of genes transcriptionally regulated by HIF-1α. Interestingly, they showed higher BNIP3 expression than OA-FLSs, and showed stronger mitophagy and proliferation activities. BNIP3 siRNA experiment confirmed the potential role of BNIP3 in the survival of RA-FLSs. Inhibition of BNIP3 resulted in the decrease of cell proliferation, mitophagy and the increase of apoptosis. Conclusion In summary, RA-FLSs maintained intracellular redox balance through mitophagy to promote cell survival under hypoxia. The mitophagy of OA-FLSs was too little to maintain the redox balance of mitochondria, resulting in apoptosis. The difference of mitophagy between OA-FLSs and RA-FLSs under hypoxia is mediated by the level of BNIP3 expression.
Collapse
Affiliation(s)
- Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China. .,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China. .,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China. .,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
23
|
Sazonova EN, Gusev IA, Malofey YB, Lanshakova AV, Vdovenko SV. Effects of Neonatal Administration of Non-Opiate Analogues of Leu-Enkephalin to Heart Tissue Homeostasis of Prepubertal Albino Rats Exposed to Hypoxia. Bull Exp Biol Med 2022; 173:188-192. [PMID: 35737163 DOI: 10.1007/s10517-022-05516-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Indexed: 10/17/2022]
Abstract
Hypobaric hypoxia (pO2 65 mm Hg, duration 4 h) induced a significant increase in the number of cardiomyocytes expressing р53, beclin-1, endothelial NO synthase and accumulation and degranulation of mast cells in the epicardium in hearts of prepubertal female rats (age 45-47 days); the number of cardiomyocytes with nucleoli decreased, while the number of single-nucleolar cardiomyocytes increased after this exposure. Five-fold administration of non-opiate analogue of leu-enkephalin (NALE peptide: Phe-D-Ala-Gly-Phe-Leu-Arg; 100 μg/kg) during the neonatal period reduced the severity of the post-hypoxic changes in the heart. Neonatal administration of NALE (100 μg/kg) against the background of NO synthase blockade with L-NAME (50 mg/kg) did not abolish the cardioprotective effects of the peptide. A similar correction of posthypoxic changes in the heart was observed after neonatal administration of original peptide G (Phe-D-Ala-Gly-Phe-Leu-Gly; 100 μg/kg). Thus, NO synthase-NO system and C-terminal amino acid Arg in the molecule of non-opiate analogue of leu-enkephalin are not required for the cardioprotective effects of peptides. Non-opiate leu-enkephalin analogs, peptides NALE and G, can be considered as promising substances for increasing heart resistance to hypoxia during later age periods.
Collapse
Affiliation(s)
- E N Sazonova
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia.
- Khabarovsk Branch of the Far Eastern Research Center for Physiology and Pathology of Respiration - Research Institute for the Protection of Motherhood and Childhood, Khabarovsk, Russia.
| | - I A Gusev
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| | - Yu B Malofey
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| | - A V Lanshakova
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| | - S V Vdovenko
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
| |
Collapse
|
24
|
Bouhamida E, Morciano G, Perrone M, Kahsay AE, Della Sala M, Wieckowski MR, Fiorica F, Pinton P, Giorgi C, Patergnani S. The Interplay of Hypoxia Signaling on Mitochondrial Dysfunction and Inflammation in Cardiovascular Diseases and Cancer: From Molecular Mechanisms to Therapeutic Approaches. BIOLOGY 2022; 11:biology11020300. [PMID: 35205167 PMCID: PMC8869508 DOI: 10.3390/biology11020300] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary The regulation of hypoxia has recently emerged as having a central impact in mitochondrial function and dysfunction in various diseases, including the major disorders threatening worldwide: cardiovascular diseases and cancer. Despite the studies in this matter, its effective role in protection and disease progression even though its direct molecular mechanism in both disorders is still to be elucidated. This review aims to cover the current knowledge about the effect of hypoxia on mitochondrial function and dysfunction, and inflammation, in cardiovascular diseases and cancer, and reports further therapeutic strategies based on the modulation of hypoxic pathways. Abstract Cardiovascular diseases (CVDs) and cancer continue to be the primary cause of mortality worldwide and their pathomechanisms are a complex and multifactorial process. Insufficient oxygen availability (hypoxia) plays critical roles in the pathogenesis of both CVDs and cancer diseases, and hypoxia-inducible factor 1 (HIF-1), the main sensor of hypoxia, acts as a central regulator of multiple target genes in the human body. Accumulating evidence demonstrates that mitochondria are the major target of hypoxic injury, the most common source of reactive oxygen species during hypoxia and key elements for inflammation regulation during the development of both CVDs and cancer. Taken together, observations propose that hypoxia, mitochondrial abnormality, oxidative stress, inflammation in CVDs, and cancer are closely linked. Based upon these facts, this review aims to deeply discuss these intimate relationships and to summarize current significant findings corroborating the molecular mechanisms and potential therapies involved in hypoxia and mitochondrial dysfunction in CVDs and cancer.
Collapse
Affiliation(s)
- Esmaa Bouhamida
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
| | - Mariasole Perrone
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
| | - Asrat E. Kahsay
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
| | - Mario Della Sala
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland;
| | - Francesco Fiorica
- Department of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, Ospedale Mater Salutis di Legnago, 37045 Verona, Italy;
| | - Paolo Pinton
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Correspondence: (C.G.); (S.P.)
| | - Simone Patergnani
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy; (E.B.); (G.M.); (M.P.); (A.E.K.); (M.D.S.); (P.P.)
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48022 Cotignola, Italy
- Correspondence: (C.G.); (S.P.)
| |
Collapse
|