1
|
Yang X, Wu H, Liu D, Zhou G, Zhang D, Yang Q, Liu Y, Li Y. The link between ferroptosis and autophagy in myocardial ischemia/reperfusion injury: new directions for therapy. J Cardiovasc Transl Res 2025; 18:408-423. [PMID: 39885084 DOI: 10.1007/s12265-025-10590-6] [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: 07/04/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Myocardial ischemia/reperfusion (I/R)-induced cell death, such as autophagy and ferroptosis, is a major contributor to cardiac injury. Regulating cell death may be key to mitigating myocardial ischemia/reperfusion injury (MI/RI). Autophagy is a crucial physiological process involving cellular self-digestion and compensation, responsible for degrading excess or malfunctioning long-lived proteins and organelles. During MI/RI, autophagy plays both "survival" and "death" roles. A growing body of research indicates that ferroptosis is a type of autophagy-dependent cell death. This article provides a comprehensive review of the functions of autophagy and ferroptosis in MI/RI, as well as the molecules mediating their interaction. Understanding the link between autophagy and ferroptosis may offer new therapeutic directions for MI/RI, bearing significant clinical implications.
Collapse
Affiliation(s)
- Xiaoting Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China.
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China.
| | - Di Liu
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huang Shi, HuBei Province, China
| | - Gang Zhou
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Dong Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Qingzhuo Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yanfang Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Yi Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| |
Collapse
|
2
|
Johnson E, Albakri JS, Allemailem KS, Sultan A, Alwanian WM, Alrumaihi F, Almansour NM, Aldakheel FM, Khalil FMA, Abduallah AM, Smith O. Mitochondrial dysfunction and calcium homeostasis in heart failure: Exploring the interplay between oxidative stress and cardiac remodeling for future therapeutic innovations. Curr Probl Cardiol 2025; 50:102968. [PMID: 39653095 DOI: 10.1016/j.cpcardiol.2024.102968] [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: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Heart failure (HF) is a multifaceted clinical syndrome characterized by the heart's inability to pump sufficient blood to meet the body's metabolic demands. It arises from various etiologies, including myocardial injury, hypertension, and valvular heart disease. A critical aspect of HF pathophysiology involves mitochondrial dysfunction, particularly concerning calcium (Ca2+) homeostasis and oxidative stress. This review highlights the pivotal role of excess mitochondrial Ca2+ in exacerbating oxidative stress, contributing significantly to HF progression. Novel insights are provided regarding the mechanisms by which mitochondrial Ca2+ overload leads to increased production of reactive oxygen species (ROS) and impaired cellular function. Despite this understanding, key gaps in research remain, particularly in elucidating the complex interplay between mitochondrial dynamics and oxidative stress across different HF phenotypes. Furthermore, therapeutic strategies targeting mitochondrial dysfunction are still in their infancy, with limited applications in clinical practice. By summarizing recent findings and identifying these critical research gaps, this review aims to pave the way for innovative therapeutic approaches that improve the management of heart failure, ultimately enhancing patient outcomes through targeted interventions.
Collapse
Affiliation(s)
- Emily Johnson
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | | | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Abdulaziz Sultan
- Family Medicine Senior Registrar, Ministry of Health, Saudi Arabia
| | - Wanian M Alwanian
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Fahad M Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Fatma Mohamed Ameen Khalil
- King Khalid University, Applied College, Unit of health specialties, basic sciences and their applications, Mohayil Asir Abha, 61421, Saudi Arabia
| | - Alduwish Manal Abduallah
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Alkarj 11942, Saudi Arabia
| | - Oliver Smith
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
3
|
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] [Grants] [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
|
4
|
Gladka MM, Kohela A, de Leeuw AE, Molenaar B, Versteeg D, Kooijman L, van Geldorp M, van Ham WB, Caliandro R, Haigh JJ, van Veen TAB, van Rooij E. Hypoxia-responsive zinc finger E-box-binding homeobox 2 (ZEB2) regulates a network of calcium-handling genes in the injured heart. Cardiovasc Res 2024; 120:1869-1883. [PMID: 39308239 PMCID: PMC11630050 DOI: 10.1093/cvr/cvae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/16/2024] [Accepted: 05/11/2024] [Indexed: 12/11/2024] Open
Abstract
AIMS Intracellular calcium (Ca2+) overload is known to play a critical role in the development of cardiac dysfunction. Despite the remarkable improvement in managing the progression of heart disease, developing effective therapies for heart failure (HF) remains a challenge. A better understanding of molecular mechanisms that maintain proper Ca2+ levels and contractility in the injured heart could be of therapeutic value. METHODS AND RESULTS Here, we report that transcription factor zinc finger E-box-binding homeobox 2 (ZEB2) is induced by hypoxia-inducible factor 1-alpha (HIF1α) in hypoxic cardiomyocytes and regulates a network of genes involved in Ca2+ handling and contractility during ischaemic heart disease. Gain- and loss-of-function studies in genetic mouse models revealed that ZEB2 expression in cardiomyocytes is necessary and sufficient to protect the heart against ischaemia-induced diastolic dysfunction and structural remodelling. Moreover, RNA sequencing of ZEB2-overexpressing (Zeb2 cTg) hearts post-injury implicated ZEB2 in regulating numerous Ca2+-handling and contractility-related genes. Mechanistically, ZEB2 overexpression increased the phosphorylation of phospholamban at both serine-16 and threonine-17, implying enhanced activity of sarcoplasmic reticulum Ca2+-ATPase (SERCA2a), thereby augmenting SR Ca2+ uptake and contractility. Furthermore, we observed a decrease in the activity of Ca2+-dependent calcineurin/NFAT signalling in Zeb2 cTg hearts, which is the main driver of pathological cardiac remodelling. On a post-transcriptional level, we showed that ZEB2 expression can be regulated by the cardiomyocyte-specific microRNA-208a (miR-208a). Blocking the function of miR-208a with anti-miR-208a increased ZEB2 expression in the heart and effectively protected from the development of pathological cardiac hypertrophy. CONCLUSION Together, we present ZEB2 as a central regulator of contractility and Ca2+-handling components in the mammalian heart. Further mechanistic understanding of the role of ZEB2 in regulating Ca2+ homeostasis in cardiomyocytes is an essential step towards the development of improved therapies for HF.
Collapse
Affiliation(s)
- Monika M Gladka
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Arwa Kohela
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- School of Biotechnology, Nile University, Giza, Egypt
| | - Anne E de Leeuw
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Bas Molenaar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Lieneke Kooijman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Mariska van Geldorp
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Willem B van Ham
- Department of Medical Physiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Rocco Caliandro
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jody J Haigh
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht (UMCU), Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Centre Utrecht (UMCU), Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
5
|
Zhang X, Gao YP, Dong WS, Li K, Hu YX, Ye YJ, Hu C. FNDC4 alleviates cardiac ischemia/reperfusion injury through facilitating HIF1α-dependent cardiomyocyte survival and angiogenesis in male mice. Nat Commun 2024; 15:9667. [PMID: 39516487 PMCID: PMC11549404 DOI: 10.1038/s41467-024-53564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Fibronectin type III domain-containing (FNDC) proteins play critical roles in cellular homeostasis and cardiac injury, and our recent findings define FNDC5 as a promising cardioprotectant against doxorubicin- and aging-related cardiac injury. FNDC4 displays a high homology with FNDC5; however, its role and mechanism in cardiac ischemia/reperfusion (I/R) injury remain elusive. Here, we show that cardiac and plasma FNDC4 levels are elevated during I/R injury in a hypoxia-inducible factor 1α (HIF1α)-dependent manner. Cardiac-specific FNDC4 overexpression facilitates, while cardiac-specific FNDC4 knockdown inhibits cardiomyocyte survival and angiogenesis in I/R-stressed hearts of male mice through regulating the proteasomal degradation of HIF1α. Interestingly, FNDC4 does not directly stimulate angiogenesis of endothelial cells, but increases the expression and secretion of fibroblast growth factor 1 from cardiomyocytes to enhance angiogenesis in a paracrine manner. Moreover, therapeutic administration of recombinant FNDC4 protein is sufficient to alleviate cardiac I/R injury in male mice, without resulting in significant side effects. In this work, we reveal that FNDC4 alleviates cardiac I/R injury through facilitating HIF1α-dependent cardiomyocyte survival and angiogenesis, and define FNDC4 as a promising predictive and therapeutic target of cardiac I/R injury.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| | - Yi-Peng Gao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Wen-Sheng Dong
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Kang Li
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Yu-Xin Hu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Can Hu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
6
|
Jiao J, Zhao Y, Li Q, Jin S, Liu Z. LncRNAs in tumor metabolic reprogramming and tumor microenvironment remodeling. Front Immunol 2024; 15:1467151. [PMID: 39539540 PMCID: PMC11557318 DOI: 10.3389/fimmu.2024.1467151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem composed of tumor cells, immune cells, supporting cells, and the extracellular matrix. Typically, the TME is characterized by an immunosuppressive state. To meet the demands of rapid proliferation, cancer cells undergo metabolic reprogramming, which enhances their biosynthesis and bioenergy supply. Immune cells require similar nutrients for activation and proliferation, leading to competition and immunosuppression within the TME. Additionally, tumor metabolites inhibit immune cell activation and function. Consequently, an immunosuppressed and immune-tolerant TME promotes cancer cell proliferation and metastasis. Long non-coding RNAs (lncRNAs), a category of non-coding RNA longer than 200 nucleotides, regulate tumor metabolic reprogramming by interacting with key enzymes, transporters, and related signaling pathways involved in tumor metabolism. Furthermore, lncRNAs can interact with both cellular and non-cellular components in the TME, thereby facilitating tumor growth, metastasis, drug resistance, and inducing immunosuppression. Recent studies have demonstrated that lncRNAs play a crucial role in reshaping the TME by regulating tumor metabolic reprogramming. In this discussion, we explore the potential mechanisms through which lncRNAs regulate tumor metabolic reprogramming to remodel the TME. Additionally, we examine the prospects of lncRNAs as targets for anti-tumor therapy and as biomarkers for tumor prognosis.
Collapse
Affiliation(s)
- Jianhang Jiao
- Department of Orthopedics, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Yangzhi Zhao
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Qimei Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, Jilin University, Changchun, China
| | - Zhongshan Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Mialet-Perez J, Belaidi E. Interplay between hypoxia inducible Factor-1 and mitochondria in cardiac diseases. Free Radic Biol Med 2024; 221:13-22. [PMID: 38697490 DOI: 10.1016/j.freeradbiomed.2024.04.239] [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: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Ischemic heart diseases and cardiomyopathies are characterized by hypoxia, energy starvation and mitochondrial dysfunction. HIF-1 acts as a cellular oxygen sensor, tuning the balance of metabolic and oxidative stress pathways to provide ATP and sustain cell survival. Acting on mitochondria, HIF-1 regulates different processes such as energy substrate utilization, oxidative phosphorylation and mitochondrial dynamics. In turn, mitochondrial homeostasis modifications impact HIF-1 activity. This underlies that HIF-1 and mitochondria are tightly interconnected to maintain cell homeostasis. Despite many evidences linking HIF-1 and mitochondria, the mechanistic insights are far from being understood, particularly in the context of cardiac diseases. Here, we explore the current understanding of how HIF-1, reactive oxygen species and cell metabolism are interconnected, with a specific focus on mitochondrial function and dynamics. We also discuss the divergent roles of HIF in acute and chronic cardiac diseases in order to highlight that HIF-1, mitochondria and oxidative stress interaction deserves to be deeply investigated. While the strategies aiming at stabilizing HIF-1 have provided beneficial effects in acute ischemic injury, some deleterious effects were observed during prolonged HIF-1 activation. Thus, deciphering the link between HIF-1 and mitochondria will help to optimize HIF-1 modulation and provide new therapeutic perspectives for the treatment of cardiovascular pathologies.
Collapse
Affiliation(s)
- Jeanne Mialet-Perez
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, France
| | - Elise Belaidi
- Univ. Lyon 1, Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, LBTI UMR 5305, 69367, Lyon, France.
| |
Collapse
|
8
|
Troise D, Infante B, Mercuri S, Piccoli C, Lindholm B, Stallone G. Hypoxic Inducible Factor Stabilization in Pericytes beyond Erythropoietin Production: The Good and the Bad. Antioxidants (Basel) 2024; 13:537. [PMID: 38790642 PMCID: PMC11118908 DOI: 10.3390/antiox13050537] [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: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The paracrine signaling pathways for the crosstalk between pericytes and endothelial cells are essential for the coordination of cell responses to challenges such as hypoxia in both healthy individuals and pathological conditions. Ischemia-reperfusion injury (IRI), one of the causes of cellular dysfunction and death, is associated with increased expression of genes involved in cellular adaptation to a hypoxic environment. Hypoxic inducible factors (HIFs) have a central role in the response to processes initiated by IRI not only linked to erythropoietin production but also because of their participation in inflammation, angiogenesis, metabolic adaptation, and fibrosis. While pericytes have an essential physiological function in erythropoietin production, a lesser-known role of HIF stabilization during IRI is that pericytes' HIF expression could influence vascular remodeling, cell loss and organ fibrosis. Better knowledge of mechanisms that control functions and consequences of HIF stabilization in pericytes beyond erythropoietin production is advisable for the development of therapeutic strategies to influence disease progression and improve treatments. Thus, in this review, we discuss the dual roles-for good or bad-of HIF stabilization during IRI, focusing on pericytes, and consequences in particular for the kidneys.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Bengt Lindholm
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
9
|
Cheng Z, Xue K, Xiong C, Zheng Z, Li J, Qiao X. MRPS16 promotes lung adenocarcinoma growth via the PI3K/AKT/Frataxin signalling axis. J Cell Mol Med 2024; 28:e18166. [PMID: 38506080 PMCID: PMC10951875 DOI: 10.1111/jcmm.18166] [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: 06/05/2023] [Revised: 12/26/2023] [Accepted: 01/24/2024] [Indexed: 03/21/2024] Open
Abstract
Although MRPS16 is involved in cancer development, its mechanisms in developing LAUD remain unclear. Herein, qRT-PCR, WB and IHC were utilized for evaluating MRPS16 expression levels, while functional assays besides animal experiments were performed to measure MRPS16 effect on LAUD progression. Using WB, the MRPS16 effect on PI3K/AKT/Frataxin signalling pathway was tested. According to our study, MRPS16 was upregulated in LAUD and was correlated to the advanced TNM stage as well as poor clinical outcomes, which represent an independent prognostic factor. Based on functional assays, MRPS16 is involved in promoting LAUD growth, migration and invasion, which was validated further in subsequent analyses through PI3K/AKT/Frataxin pathway activation. Moreover, MRPS16-knockdown-mediated Frataxin overexpression was shown to restore the reduction in tumour cells proliferation, migration and invasion. Our results revealed that MRPS16 caused an aggressive phenotype to LAUD and was a poor prognosticator; thus, targeting MRPS16 may be effectual in LAUD treatment.
Collapse
Affiliation(s)
- Zaixing Cheng
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Kaming Xue
- Department of Traditional Chinese MedicineUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Cui Xiong
- Department of EndocrinologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Zhikun Zheng
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Jinsong Li
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xinwei Qiao
- Department of Thoracic SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
10
|
Zhang Q, Luo Y, Peng L, Rong X, Liu Y, Li J, Luo J. Ferroptosis in cardiovascular diseases: role and mechanism. Cell Biosci 2023; 13:226. [PMID: 38102663 PMCID: PMC10724928 DOI: 10.1186/s13578-023-01169-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023] Open
Abstract
In multicellular organisms, regulatory cell death is a crucial aspect of growth and development. Ferroptosis, which was postulated roughly ten years ago, is a mode of cell death that differs from apoptosis, autophagy, and pyrodeath. This distinct pattern of cell death is triggered by an imbalance between oxidants and antioxidants and strongly associated with the metabolism of iron, lipids, amino acids, and glutathione. A growing body of research has implicated ferroptosis in the incidence and progression of many organ traumas and degenerative diseases. Recently, ferroptosis has gained attention as a crucial regulatory mechanism underlying the initiation and development of a variety of cardiovascular diseases, including myocardial ischemia/reperfusion injury, cardiomyopathy, arrhythmia, chemotherapy, and Corona Virus-2-induced cardiac injury. Pharmacological therapies that inhibit ferroptosis have great potential for the management of cardiovascular disorders. This review discusses the prevalence and regulatory mechanisms of ferroptosis, effect of ferroptosis on the immune system, significance of ferroptosis in cardiovascular diseases, and potential therapeutic value of regulating ferroptosis in a variety of heart diseases.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Peng
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xi Rong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingxue Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Collaborative Innovation Centre for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.
| | - Jing Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Collaborative Innovation Centre for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.
| |
Collapse
|
11
|
Jiang M, Fan X, Wang Y, Sun X. Effects of hypoxia in cardiac metabolic remodeling and heart failure. Exp Cell Res 2023; 432:113763. [PMID: 37726046 DOI: 10.1016/j.yexcr.2023.113763] [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: 06/11/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
Aerobic cellular respiration requires oxygen, which is an essential part of cardiomyocyte metabolism. Thus, oxygen is required for the physiologic metabolic activities and development of adult hearts. However, the activities of metabolic pathways associated with hypoxia in cardiomyocytes (CMs) have not been conclusively described. In this review, we discuss the role of hypoxia in the development of the hearts metabolic system, and the metabolic remodeling associated with the hypoxic adult heart. Hypoxia-inducible factors (HIFs), the signature transcription factors in hypoxic environments, is also investigated for their potential to modulate hypoxia-induced metabolic changes. Metabolic remodeling existing in hypoxic hearts have also been shown to occur in chronic failing hearts, implying that novel therapeutic options for heart failure (HF) may exist from the hypoxic perspective. The pressure overload-induced HF and diabetes-induced HF are also discussed to demonstrate the effects of HIF factor-related pathways to control the metabolic remodeling of failing hearts.
Collapse
Affiliation(s)
- Mingzhou Jiang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Xi Fan
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Yiqing Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China.
| | - Xiaotian Sun
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Vagnozzi RJ, Robinson EL. Frataxin Deacetylation in Macrophages: Avoiding SIRTain Myocyte Death. Circ Res 2023; 133:648-650. [PMID: 37708247 PMCID: PMC10506396 DOI: 10.1161/circresaha.123.323503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Affiliation(s)
- Ronald J. Vagnozzi
- Department of Medicine, Division of Cardiology, University
of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Consortium for Fibrosis Research & Translation,
University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Institute, University of Colorado Anschutz Medical
Campus, Aurora, CO, USA
| | - Emma L. Robinson
- Department of Medicine, Division of Cardiology, University
of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Consortium for Fibrosis Research & Translation,
University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
13
|
Qi Y, Hu M, Wang Z, Shang W. Mitochondrial iron regulation as an emerging target in ischemia/reperfusion injury during kidney transplantation. Biochem Pharmacol 2023; 215:115725. [PMID: 37524207 DOI: 10.1016/j.bcp.2023.115725] [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: 06/05/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
The injury caused by ischemia and subsequent reperfusion (I/R) is inevitable during kidney transplantation and its current management remains unsatisfactory. Iron is considered to play a remarkable pathologic role in the initiation or progression of tissue damage induced by I/R, whereas the effects of iron-related therapy remain controversial owing to the complicated nature of iron's involvement in multiple biological processes. A significant portion of the cellular iron is located in the mitochondria, which exerts a central role in the development and progression of I/R injury. Recent studies of iron regulation associated with mitochondrial function represents a unique opportunity to improve our knowledge on the pathophysiology of I/R injury. However, the molecular mechanisms linking mitochondria to the iron homeostasis remain unclear. In this review, we provide a comprehensive analysis of the alterations to iron metabolism in I/R injury during kidney transplantation, analyze the current understanding of mitochondrial regulation of iron homeostasis and discussed its potential application in I/R injury. The elucidation of regulatory mechanisms regulating mitochondrial iron homeostasis will offer valuable insights into potential therapeutic targets for alleviating I/R injury with the ultimate aim of improving kidney graft outcomes, with potential implications that could also extend to acute kidney injury or other I/R injuries.
Collapse
Affiliation(s)
- Yuanbo Qi
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Mingyao Hu
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Zhigang Wang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Wenjun Shang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
14
|
Li N, Gu X, Liu F, Zhang Y, Sun Y, Gao S, Wang B, Zhang C. Network pharmacology-based analysis of potential mechanisms of myocardial ischemia-reperfusion injury by total salvianolic acid injection. Front Pharmacol 2023; 14:1202718. [PMID: 37680709 PMCID: PMC10482107 DOI: 10.3389/fphar.2023.1202718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023] Open
Abstract
In this review, we investigated the potential mechanism of Total Salvianolic Acid Injection (TSI) in protecting against myocardial ischemia reperfusion injury (MI/RI). To achieve this, we predicted the component targets of TSI using Pharmmapper and identified the disease targets of MI/RI through GeneCards, DisGenNET, and OMIM databases. We constructed protein-protein interaction networks by analyzing the overlapping targets and performed functional enrichment analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. Our analysis yielded 90 targets, which were implicated in the potential therapeutic effects of TSI on MI/RI. Seven critical signaling pathways significantly contributed to TSI's protective effects, namely, PI3K signaling, JAK-STAT signaling, Calcium signaling, HIF-1 signaling, Nuclear receptor signaling, Cell Cycle, and Apoptosis. Subsequently, we conducted a comprehensive literature review of these seven key signaling pathways to gain further insights into their role in the TSI-mediated treatment of MI/RI. By establishing these connections, our study lays a solid foundation for future research endeavours to elucidate the molecular mechanisms through which TSI exerts its beneficial effects on MI/RI.
Collapse
Affiliation(s)
- Nan Li
- Tianjin University of Chinese Medicine, Tianjin, China
| | - Xufang Gu
- The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fanqi Liu
- The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yao Zhang
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanjun Sun
- Tianjin University of Chinese Medicine, Tianjin, China
| | - Shengwei Gao
- Tianjin University of Chinese Medicine, Tianjin, China
| | - Baohe Wang
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chen Zhang
- The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
15
|
Zhang Z, Jiang W, Zhang C, Yin Y, Mu N, Wang Y, Yu L, Ma H. Frataxin inhibits the sensitivity of the myocardium to ferroptosis by regulating iron homeostasis. Free Radic Biol Med 2023; 205:305-317. [PMID: 37343689 DOI: 10.1016/j.freeradbiomed.2023.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 06/23/2023]
Abstract
RATIONALE Myocardial ischemia/reperfusion (I/R) injury is characterized by cell death via various cellular mechanisms upon reperfusion. As a new type of cell death, ferroptosis provides new opportunities to reduce myocardial cell death. Ferroptosis is known to be more active during reperfusion than ischemia. However, the mechanisms regulating ferroptosis during ischemia and reperfusion remain largely unknown. METHODS The contribution of ferroptosis in ischemic and reperfused myocardium were detected by administered of Fer-1, a ferroptosis inhibitor to C57BL/6 mice, followed by left anterior descending (LAD) ligation surgery. Ferroptosis was evaluated by measurement of cell viability, ptgs2 mRNA level, iron production, malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) levels. H9C2 cells were exposed to hypoxia/reoxygenation to mimic in vivo I/R. We used LC-MS/MS to identify potential E3 ligases that interacted with frataxin in heart tissue. Cardiac-specific overexpression of frataxin in whole heart was achieved by intracardiac injection of frataxin, carried by adeno-associated virus serotype 9 (AAV9) containing cardiac troponin T (cTnT) promoter. RESULTS We showed that regulators of iron metabolism, especially iron regulatory protein activity, were increased in the ischemic myocardium or hypoxia cardiomyocytes. In addition, we found that frataxin, which is involved in iron metabolism, is differentially expressed in the ischemic and reperfused myocardium and involved in the regulation of cardiomyocytes ferroptosis. Furthermore, we identified an E3 ligase, NHL repeat-containing 1 (NHLRC1), that mediates frataxin ubiquitination degradation. Cardiac-specific overexpression of frataxin ameliorated myocardial I/R injury through ferroptosis inhibition. CONCLUSIONS Through a multi-level study from molecule to animal model, these findings uncover the key role of frataxin in inhibiting cardiomyocyte ferroptosis and provide new strategies and perspectives for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Zihui Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Wenhua Jiang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Chan Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Heng Ma
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China; Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China.
| |
Collapse
|
16
|
Culley MK, Rao RJ, Mehta M, Zhao J, El Khoury W, Harvey LD, Perk D, Tai YY, Tang Y, Shiva S, Rabinovitch M, Gu M, Bertero T, Chan SY. Frataxin deficiency disrupts mitochondrial respiration and pulmonary endothelial cell function. Vascul Pharmacol 2023; 151:107181. [PMID: 37164245 PMCID: PMC10524929 DOI: 10.1016/j.vph.2023.107181] [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: 08/22/2022] [Revised: 04/19/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Deficiency of iron‑sulfur (FeS) clusters promotes metabolic rewiring of the endothelium and the development of pulmonary hypertension (PH) in vivo. Joining a growing number of FeS biogenesis proteins critical to pulmonary endothelial function, recent data highlighted that frataxin (FXN) reduction drives Fe-S-dependent genotoxic stress and senescence across multiple types of pulmonary vascular disease. Trinucleotide repeat mutations in the FXN gene cause Friedreich's ataxia, a disease characterized by cardiomyopathy and neurodegeneration. These tissue-specific phenotypes have historically been attributed to mitochondrial reprogramming and oxidative stress. Whether FXN coordinates both nuclear and mitochondrial processes in the endothelium is unknown. Here, we aim to identify the mitochondria-specific effects of FXN deficiency in the endothelium that predispose to pulmonary hypertension. Our data highlight an Fe-S-driven metabolic shift separate from previously described replication stress whereby FXN knockdown diminished mitochondrial respiration and increased glycolysis and oxidative species production. In turn, FXN-deficient endothelial cells had increased vasoconstrictor production (EDN1) and decreased nitric oxide synthase expression (NOS3). These data were observed in primary pulmonary endothelial cells after pharmacologic inhibition of FXN, mice carrying a genetic endothelial deletion of FXN, and inducible pluripotent stem cell-derived endothelial cells from patients with FXN mutations. Altogether, this study indicates FXN is an upstream driver of pathologic aberrations in metabolism and genomic stability. Moreover, our study highlights FXN-specific vasoconstriction in vivo, prompting future studies to investigate available and novel PH therapies in contexts of FXN deficiency.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rashmi J Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Monica Mehta
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lloyd D Harvey
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Dror Perk
- Medical Scientist Training Program, Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yi Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, USA
| | - Marlene Rabinovitch
- Stanford Children's Health Betty Irene Moore Children's Heart Center, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, UMR7275, IPMC, Valbonne, France
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
Iske J, Cao Y, Roesel MJ, Shen Z, Nian Y. Metabolic reprogramming of myeloid-derived suppressor cells in the context of organ transplantation. Cytotherapy 2023; 25:789-797. [PMID: 37204374 DOI: 10.1016/j.jcyt.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 05/20/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are naturally occurring leukocytes that develop from immature myeloid cells under inflammatory conditions that were discovered initially in the context of tumor immunity. Because of their robust immune inhibitory activities, there has been growing interest in MDSC-based cellular therapies for transplant tolerance induction. Indeed, various pre-clinical studies have introduced in vivo expansion or adoptive transfer of MDSC as a promising therapeutic strategy leading to a profound extension of allograft survival due to suppression of alloreactive T cells. However, several limitations of cellular therapies using MDSCs remain to be addressed, including their heterogeneous nature and limited expansion capacity. Metabolic reprogramming plays a crucial role for differentiation, proliferation and effector function of immune cells. Notably, recent reports have focused on a distinct metabolic phenotype underlying the differentiation of MDSCs in an inflammatory microenvironment representing a regulatory target. A better understanding of the metabolic reprogramming of MDSCs may thus provide novel insights for MDSC-based treatment approaches in transplantation. In this review, we will summarize recent, interdisciplinary findings on MDSCs metabolic reprogramming, dissect the underlying molecular mechanisms and discuss the relevance for potential treatment approaches in solid-organ transplantation.
Collapse
Affiliation(s)
- Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yu Cao
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Maximilian J Roesel
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Zhongyang Shen
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yeqi Nian
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China.
| |
Collapse
|
18
|
Yan X, Xie Y, Liu H, Huang M, Yang Z, An D, Jiang G. Iron accumulation and lipid peroxidation: implication of ferroptosis in diabetic cardiomyopathy. Diabetol Metab Syndr 2023; 15:161. [PMID: 37468902 DOI: 10.1186/s13098-023-01135-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023] Open
Abstract
Diabetic cardiomyopathy (DC) is a serious heart disease caused by diabetes. It is unrelated to hypertension and coronary artery disease and can lead to heart insufficiency, heart failure and even death. Currently, the pathogenesis of DC is unclear, and clinical intervention is mainly symptomatic therapy and lacks effective intervention objectives. Iron overdose mediated cell death, also known as ferroptosis, is widely present in the physiological and pathological processes of diabetes and DC. Iron is a key trace element in the human body, regulating the metabolism of glucose and lipids, oxidative stress and inflammation, and other biological processes. Excessive iron accumulation can lead to the imbalance of the antioxidant system in DC and activate and aggravate pathological processes such as excessive autophagy and mitochondrial dysfunction, resulting in a chain reaction and accelerating myocardial and microvascular damage. In-depth understanding of the regulating mechanisms of iron metabolism and ferroptosis in cardiovascular vessels can help improve DC management. Therefore, in this review, we summarize the relationship between ferroptosis and the pathogenesis of DC, as well as potential intervention targets, and discuss and analyze the limitations and future development prospects of these targets.
Collapse
Affiliation(s)
- Xuehua Yan
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Xinjiang, China
| | - Yang Xie
- Affiliated Hospital of Traditional Chinese Medicine of Xinjiang Medical University, Xinjiang, China
| | - Hongbing Liu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Meng Huang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Zhen Yang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China
| | - Dongqing An
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China.
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Xinjiang, China.
- Affiliated Hospital of Traditional Chinese Medicine of Xinjiang Medical University, Xinjiang, China.
| | - Guangjian Jiang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang, China.
| |
Collapse
|
19
|
Liu J, Chen H, Lin H, Peng S, Chen L, Cheng X, Yao P, Tang Y. Iron-frataxin involved in the protective effect of quercetin against alcohol-induced liver mitochondrial dysfunction. J Nutr Biochem 2023; 114:109258. [PMID: 36587874 DOI: 10.1016/j.jnutbio.2022.109258] [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: 01/04/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Emerging evidence supports the beneficial effect of quercetin on liver mitochondrial disorders. However, the molecular mechanism by which quercetin protects mitochondria is limited, especially in alcoholic liver disease. In this study, C57BL/6N mice were fed with Lieber De Carli liquid diet (28% ethanol-derived calories) for 12 weeks plus a single binge ethanol and intervened with quercetin (100 mg/kg.bw). Moreover, HepG2CYP2E1+/+ were stimulated with ethanol (100 mM) and quercetin (50 µM) to investigate the effects of mitochondrial protein frataxin. The results indicated that quercetin alleviated alcohol-induced histopathological changes and mitochondrial functional disorders in mice livers. Consistent with increased PINK1, Parkin, Bnip3 and LC3II as well as decreased p62, TOM20 and VDAC1 expression, the inhibition of mitophagy by ethanol was blocked by quercetin. Additionally, quercetin improved the imbalance of iron metabolism-related proteins expression in alcohol-fed mice livers. Compared with ethanol-treated Lv-empty HepG2CYP2E1+/+ cells, frataxin deficiency further exacerbated the inhibition of mitochondrial function. Conversely, restoration of frataxin expression ameliorated the effect of ethanol. Furthermore, frataxin deficiency reduced the protective effects of quercetin on mitochondria disordered by ethanol. Attentively, ferric ammonium citrate (FAC) and deferiprone decreased or increased frataxin expression in HepG2CYP2E1+/+, respectively. Notably, we further found FAC reversed the increasing effect of quercetin on frataxin expression. Ultimately, silencing NCOA4 attenuated the inhibition of quercetin on LDH release and mitochondrial membrane potential increase, and similar results were observed by adding FAC. Collectively, these findings demonstrated quercetin increased frataxin expression through regulating iron level, thereby mitigating ethanol-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Huimin Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongkun Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shufen Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xueer Cheng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
20
|
Hazra R, Novelli EM, Hu X. Astrocytic mitochondrial frataxin-A promising target for ischemic brain injury. CNS Neurosci Ther 2023; 29:783-788. [PMID: 36550598 PMCID: PMC9928550 DOI: 10.1111/cns.14068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
In the ischemic brain, hypoxia leads to mitochondrial dysfunction, insufficient energy production, and astrocyte activation. Yet, most studies investigating mitochondrial dysfunction in cerebral ischemia have focused exclusively on neurons. This review will highlight the importance of the morphological, molecular, and functional heterogeneity of astrocytes in their role in brain injuries and explore how activated astrocytes exhibit calcium imbalance, reactive oxygen species overproduction, and apoptosis. In addition, special focus will be given to the role of the mitochondrial protein frataxin in activated astrocytes during ischemia and its putative role in the pharmacological management of cerebral ischemia.
Collapse
Affiliation(s)
- Rimi Hazra
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Enrico M Novelli
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaoming Hu
- Center of Cerebrovascular Disease Research, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
21
|
Gao LT, Yuan JQ, Zhang ZY, Zhao HM, Gao L. Hypermethylation of the Bmp4 promoter dampens binding of HIF-1α and impairs its cardiac protective effects from oxidative stress in prenatally GC-exposed offspring. Cell Mol Life Sci 2023; 80:58. [PMID: 36746787 PMCID: PMC9902418 DOI: 10.1007/s00018-023-04703-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/30/2022] [Accepted: 01/22/2023] [Indexed: 02/08/2023]
Abstract
The exposure to an unhealthy environment in utero can lead to the occurrence of cardiovascular diseases in the offspring. Glucocorticoids (GC) are essential for normal development and maturation of fetal organs and is a first-line treatment for pregnant women affected by autoimmune diseases. However, excess prenatal GC exposure might program the development of fetal organs and cause a number of chronic diseases in later life. Our previous studies indicated that cardiac functions were significantly compromised in rat offspring prenatally exposed to the synthetic glucocorticoid dexamethasone (DEX), only after ischemia-reperfusion. In the present study, we further observed that DNA hypermethylation of bone morphogenetic protein 4 (Bmp4) promoter in cardiomyocytes caused by prenatal DEX exposure substantially dampened the binding activity of transcription factor HIF-1α induced by cardiac ischemia. Therefore, prenatal DEX exposure inhibits the induction of BMP4 upon I/R and attenuates the protective effects of BMP4 in cardiomyocytes, which eventually manifests as malfunction of the adult heart. Moreover, we employed two cardiac-specific Bmp4 knock-in mouse models and found that in vivo BMP4 overexpression could rescue the cardiac dysfunction caused by prenatal GC exposure. In depth mechanistic research revealed that BMP4 protects the cardiomyocytes from mitophagy and apoptosis by attenuating mitochondrial PGC-1α expression in a p-Smad and Parkin-dependent manner. These findings suggest that prenatal GC exposure increases the susceptibility of the offspring's heart to a "second strike" after birth, due to the failure of hypoxia-induced HIF-1α transactivation of the hypermethylated Bmp4 promoter in cardiomyocytes. Pretreatment with the DNA methylation inhibitor, 5-Aza-2'-deoxycytidine, could be a potential therapeutic method for this programming effect of GC exposure during pregnancy on neonatal cardiac dysfunction.
Collapse
Affiliation(s)
- Ling-Tong Gao
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China
| | - Jian-Qiang Yuan
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China
| | - Zhi-Yu Zhang
- Department of Health Management, Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Hou-Ming Zhao
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China
| | - Lu Gao
- Department of Physiology, Naval Medical University, 800 Xiangyin Rd., Shanghai, 200433, People's Republic of China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China. .,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, People's Republic of China.
| |
Collapse
|
22
|
Zhou Y, Zhang H, Huang Y, Wu S, Liu Z. Tanshinone IIA regulates expression of glucose transporter 1 via activation of the HIF‑1α signaling pathway. Mol Med Rep 2022; 26:328. [PMID: 36069225 PMCID: PMC9727584 DOI: 10.3892/mmr.2022.12844] [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: 05/05/2022] [Accepted: 07/29/2022] [Indexed: 12/30/2022] Open
Abstract
Tanshinone IIA (Tan 2A) is a lipid‑soluble compound extracted from the Chinese herb Danshen (Salvia miltiorrhiza Bunge). It protects neuron and microvascular endothelial cells against hypoxia/ischemia both in vitro and in vivo however the mechanism is not fully known. Glucose transporter 1 (GLUT‑1) is ubiquitously expressed in all types of tissue in the human body and serves important physiological functions due to its glucose uptake ability. The present study evaluated the role of Tan 2A in regulating GLUT‑1 expression and its potential mechanism. RT‑PCR and western Blot were used to detect the expression of GLUT‑1. Si RNA mediated knockdown and CHIP assay were used to explore the mechanism of Tan 2A on GLUT‑1expression. Tan 2A treatment induced expression of GLUT‑1 and subsequently increased glucose uptake in endothelial cells (ECs). Furthermore, mRNA expression levels of vascular endothelial cell growth factor, BCL2 interacting protein 3 and enolase 2, which are target genes for hypoxia‑inducible factor‑1α (HIF‑1α), were significantly upregulated by Tan 2A. Co‑immunoprecipitation demonstrated that Tan 2A markedly increased the association of HIF‑1α with recombination signal‑binding protein for immunoglobulin κJ region (RBPJκ). Moreover, knockdown of HIF‑1α and RBPJκ significantly reversed the regulatory effect of Tan 2A on mRNA expression levels of these genes in ECs. The results of the present study suggested that HIF‑1α partially mediated the regulatory effect of Tan 2A on GLUT‑1 expression in ECs. Therefore, GLUT‑1 may be a potential therapeutic target for Tan 2A.
Collapse
Affiliation(s)
- Yanyun Zhou
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China,Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Hong Zhang
- Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Yitong Huang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Shengyun Wu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zongjun Liu
- Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China,Correspondence to: Dr Zongjun Liu, Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Putuo, Shanghai 200062, P.R. China, E-mail:
| |
Collapse
|
23
|
Niu N, Li H, Du X, Wang C, Li J, Yang J, Liu C, Yang S, Zhu Y, Zhao W. Effects of NRF-1 and PGC-1α cooperation on HIF-1α and rat cardiomyocyte apoptosis under hypoxia. Gene 2022; 834:146565. [PMID: 35569770 DOI: 10.1016/j.gene.2022.146565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hypoxia is a primary inducer of cardiomyocyte injury, its significant marker being hypoxia-induced cardiomyocyte apoptosis. Nuclear respiratory factor-1 (NRF-1) and hypoxia-inducible factor-1α (HIF-1α) are transcriptional regulatory elements implicated in multiple biological functions, including oxidative stress response. However, their roles in hypoxia-induced cardiomyocyte apoptosis remain unknown. The effect HIF-1α, together with NRF-1, exerts on cardiomyocyte apoptosis also remains unclear. METHODS We established a myocardial hypoxia model and investigated the effects of these proteins on the proliferation and apoptosis of rat cardiomyocytes (H9C2) under hypoxia. Further, we examined the association between NRF-1 and HIF-1α to improve the current understanding of NRF-1 anti-apoptotic mechanisms. RESULTS The results show that NRF-1 and HIF-1α are important anti-apoptotic molecules in H9C2 cells under hypoxia, although their regulatory mechanisms differ. NRF-1 could bind to the promoter region of Hif1a and negatively regulate its expression. Additionally, HIF-1β exhibited competitive binding with NRF-1 and HIF-1α, demonstrating a synergism between NRF-1 and the peroxisome proliferator-activated receptor-gamma coactivator-1α. CONCLUSION These results indicate that cardiomyocytes can regulate different molecular patterns to tolerate hypoxia, providing a novel methodological framework for studying cardiomyocyte apoptosis under hypoxia.
Collapse
Affiliation(s)
- Nan Niu
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Hui Li
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Xiancai Du
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Chan Wang
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Junliang Li
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Jihui Yang
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Cheng Liu
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Songhao Yang
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Yazhou Zhu
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China
| | - Wei Zhao
- School of Basic Medicine, Ningxia Medical University, 1160 Shengli South Street, Xingqing District, Yinchuan City, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
24
|
Wang R, Liu F, Huang P, Zhang Y, He J, Pang X, Zhang D, Guan Y. Ozone preconditioning protects rabbit heart against global ischemia-reperfusion injury in vitro by up-regulating HIF-1α. Biomed Pharmacother 2022; 150:113033. [PMID: 35658224 DOI: 10.1016/j.biopha.2022.113033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 12/07/2022] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a major factor that leads to cardiac dysfunction in cardiovascular surgery during extracorporeal circulation. Recent studies have found that ozone (O3) has protective effect on MIRI caused by the anterior descending branch of the ligated left coronary artery. However, whether O3 preconditioning has the same protective effect on global MIRI and the mechanism underlying this clinical treatment remains elusive. Here, we hypothesized that O3 preconditioning (O3P) could protect rabbit heart against global MIRI in vitro by up-regulating HIF-1α. Rabbits were treated intraperitoneally with O2/O3 mixture with different concentrations and then injected with YC-1 (inhibitor of HIF-1α) before the establishment of the global MIRI model using the Langendorff isolated heart perfusion apparatus. We investigated the effects of O3 preconditioning on cardiac systolic function, myocardial infarction, inflammatory response, mitochondrial function, myocardial pathological changes and arrhythmias. We found that the heart with O3 preconditioning significantly increased HR, LVDP and IL-10 expression, and decreased IL-6 expression, CK-MB, cTnT and cTnI concentration, myocardial infarction area, myocardial pathological injury and the occurrence of ventricular tachycardia and ventricular fibrillation. Meanwhile, the level of HIF-1α was significantly increased. However, after treatment of specific inhibitor of HIF-1α, the protective effect of O3 preconditioning was reversed completely. Our data indicates that O3 preconditioning has protective effect on MIRI and this protective effect is positively associated with dosage of O3. Energy metabolism disorder is the initial stage of MIRI and up-regulation of HIF-1α plays an important role in reducing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Rui Wang
- Anesthesia and Operation Center, The First Medical Center, Chinese PLA General Hospital, Beijing, China; School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Fengjin Liu
- Department of Emergency, Yantai Yuhuangding Hospital, Shandong, China
| | - Puxidan Huang
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yu Zhang
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jun He
- Department of Anesthesiology, Beijing Huaxin Hospital, First Hospital of Tsinghua University, Beijing, China
| | - Xiaolin Pang
- Department of Anesthesiology, Beijing Huaxin Hospital, First Hospital of Tsinghua University, Beijing, China
| | - Dongya Zhang
- Department of Anesthesiology, Beijing Huaxin Hospital, First Hospital of Tsinghua University, Beijing, China.
| | - Yuan Guan
- Department of Anesthesiology, Beijing Huaxin Hospital, First Hospital of Tsinghua University, Beijing, China.
| |
Collapse
|
25
|
Monfort B, Want K, Gervason S, D’Autréaux B. Recent Advances in the Elucidation of Frataxin Biochemical Function Open Novel Perspectives for the Treatment of Friedreich’s Ataxia. Front Neurosci 2022; 16:838335. [PMID: 35310092 PMCID: PMC8924461 DOI: 10.3389/fnins.2022.838335] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is the most prevalent autosomic recessive ataxia and is associated with a severe cardiac hypertrophy and less frequently diabetes. It is caused by mutations in the gene encoding frataxin (FXN), a small mitochondrial protein. The primary consequence is a defective expression of FXN, with basal protein levels decreased by 70–98%, which foremost affects the cerebellum, dorsal root ganglia, heart and liver. FXN is a mitochondrial protein involved in iron metabolism but its exact function has remained elusive and highly debated since its discovery. At the cellular level, FRDA is characterized by a general deficit in the biosynthesis of iron-sulfur (Fe-S) clusters and heme, iron accumulation and deposition in mitochondria, and sensitivity to oxidative stress. Based on these phenotypes and the proposed ability of FXN to bind iron, a role as an iron storage protein providing iron for Fe-S cluster and heme biosynthesis was initially proposed. However, this model was challenged by several other studies and it is now widely accepted that FXN functions primarily in Fe-S cluster biosynthesis, with iron accumulation, heme deficiency and oxidative stress sensitivity appearing later on as secondary defects. Nonetheless, the biochemical function of FXN in Fe-S cluster biosynthesis is still debated. Several roles have been proposed for FXN: iron chaperone, gate-keeper of detrimental Fe-S cluster biosynthesis, sulfide production stimulator and sulfur transfer accelerator. A picture is now emerging which points toward a unique function of FXN as an accelerator of a key step of sulfur transfer between two components of the Fe-S cluster biosynthetic complex. These findings should foster the development of new strategies for the treatment of FRDA. We will review here the latest discoveries on the biochemical function of frataxin and the implication for a potential therapeutic treatment of FRDA.
Collapse
|
26
|
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: 25] [Impact Index Per Article: 8.3] [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
|
27
|
Calpain-Mediated Mitochondrial Damage: An Emerging Mechanism Contributing to Cardiac Disease. Cells 2021; 10:cells10082024. [PMID: 34440793 PMCID: PMC8392834 DOI: 10.3390/cells10082024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Calpains belong to the family of calcium-dependent cysteine proteases expressed ubiquitously in mammals and many other organisms. Activation of calpain is observed in diseased hearts and is implicated in cardiac cell death, hypertrophy, fibrosis, and inflammation. However, the underlying mechanisms remain incompletely understood. Recent studies have revealed that calpains target and impair mitochondria in cardiac disease. The objective of this review is to discuss the role of calpains in mediating mitochondrial damage and the underlying mechanisms, and to evaluate whether targeted inhibition of mitochondrial calpain is a potential strategy in treating cardiac disease. We expect to describe the wealth of new evidence surrounding calpain-mediated mitochondrial damage to facilitate future mechanistic studies and therapy development for cardiac disease.
Collapse
|
28
|
Fu Y, Zhao C, Saxu R, Yao C, Zhao L, Zheng W, Yu P, Teng Y. Anastatin Derivatives Alleviate Myocardial Ischemia-Reperfusion Injury via Antioxidative Properties. Molecules 2021; 26:4779. [PMID: 34443365 PMCID: PMC8399290 DOI: 10.3390/molecules26164779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/22/2021] [Accepted: 08/02/2021] [Indexed: 11/23/2022] Open
Abstract
(±)-Anastatins A and B are flavonoids isolated from Anastatica hierochuntica. In a previous study, twenty-four di- and tri-substituted novel derivatives of anastatins were designed and their preliminary antioxidant activities were evaluated. In the present study, the protective effect of myocardial ischemia-reperfusion (I/R) and the systematic antioxidant capacity of 24 derivatives were further studied. Compound 13 was the most potent among all the compounds studied, which increased the survival of H9c2 cells to 80.82%. The antioxidant capability of compound 13 was evaluated in ferric reducing antioxidant power, 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) radical scavenging, and 2,2-diphenyl-1-picrylhydrazyl assays. It was observed that compound 13 significantly reduced infarcted areas and improved histopathological and electrocardiogram changes in rats with myocardial I/R injury. Moreover, compound 13 decreased the leakage rates of serum lactate dehydrogenase, creatine kinase, and malonyldialdehyde from rat myocardial tissues and increased the level of glutathione and superoxide dismutase activities following myocardial I/R injury in rats. Taken together, we concluded that compound 13 had potent cardioprotective effects against myocardial I/R injury both in vitro and in vivo owing to its extensive antioxidant activities.
Collapse
Affiliation(s)
- Ying Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| | - Cai Zhao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| | - Rengui Saxu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| | - Chaoran Yao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| | - Lianbo Zhao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| | - Weida Zheng
- Medical College, Yanbian University, No.977 Gongyuan Road, Yanji 133002, China;
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (C.Z.); (R.S.); (C.Y.); (L.Z.)
| |
Collapse
|
29
|
Liu XW, Lu MK, Zhong HT, Liu JJ, Fu YP. Panax Notoginseng Saponins Protect H9c2 Cells From Hypoxia-reoxygenation Injury Through the Forkhead Box O3a Hypoxia-inducible Factor-1 Alpha Cell Signaling Pathway. J Cardiovasc Pharmacol 2021; 78:e681-e689. [PMID: 34354001 PMCID: PMC8584197 DOI: 10.1097/fjc.0000000000001120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/14/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Panax notoginseng saponins (PNS) are commonly used in the treatment of cardiovascular diseases. Whether PNS can protect myocardial ischemia-reperfusion injury by regulating the forkhead box O3a hypoxia-inducible factor-1 alpha (FOXO3a/HIF-1α) cell signaling pathway remains unclear. The purpose of this study was to investigate the protective effect of PNS on H9c2 cardiomyocytes through the FOXO3a/HIF-1α cell signaling pathway. Hypoxia and reoxygenation of H9C2 cells were used to mimic MIRI in vitro, and the cells were treated with PNS, 2-methoxyestradiol (2ME2), and LY294002." Cell proliferation, lactate dehydrogenase, and malonaldehyde were used to evaluate the degree of cell injury. The level of reactive oxygen species was detected with a fluorescence microscope. The apoptosis rate was detected by flow cytometry. The expression of autophagy-related proteins and apoptosis-related proteins was detected by western blot assay. PNS could reduce H9c2 hypoxia-reoxygenation injury by promoting autophagy and inhibiting apoptosis through the HIF-1α/FOXO3a cell signaling pathway. Furthermore, the protective effects of PNS were abolished by HIF-1α inhibitor 2ME2 and PI3K/Akt inhibitor LY294002. PNS could reduce H9c2 hypoxia-reoxygenation injury by promoting autophagy and inhibiting apoptosis through the HIF-1α/FOXO3a cell signaling pathway.
Collapse
Affiliation(s)
- Xin-Wen Liu
- Department of Pharmacy, Affiliated Hospital of Shaoxing University, Shaoxing, PR China;
| | - Meng-Kai Lu
- Department of Pharmacy, Affiliated Hospital of Shaoxing University, Shaoxing, PR China;
| | - Hui-Ting Zhong
- Department of Research, Affiliated Hospital of Shaoxing University, Shaoxing, PR China; and
| | - Jing-Jing Liu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, PR China.
| | - Yong-Ping Fu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, PR China.
| |
Collapse
|
30
|
Culley MK, Zhao J, Tai YY, Tang Y, Perk D, Negi V, Yu Q, Woodcock CSC, Handen A, Speyer G, Kim S, Lai YC, Satoh T, Watson AM, Aaraj YA, Sembrat J, Rojas M, Goncharov D, Goncharova EA, Khan OF, Anderson DG, Dahlman JE, Gurkar AU, Lafyatis R, Fayyaz AU, Redfield MM, Gladwin MT, Rabinovitch M, Gu M, Bertero T, Chan SY. Frataxin deficiency promotes endothelial senescence in pulmonary hypertension. J Clin Invest 2021; 131:136459. [PMID: 33905372 PMCID: PMC8159699 DOI: 10.1172/jci136459] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
The dynamic regulation of endothelial pathophenotypes in pulmonary hypertension (PH) remains undefined. Cellular senescence is linked to PH with intracardiac shunts; however, its regulation across PH subtypes is unknown. Since endothelial deficiency of iron-sulfur (Fe-S) clusters is pathogenic in PH, we hypothesized that a Fe-S biogenesis protein, frataxin (FXN), controls endothelial senescence. An endothelial subpopulation in rodent and patient lungs across PH subtypes exhibited reduced FXN and elevated senescence. In vitro, hypoxic and inflammatory FXN deficiency abrogated activity of endothelial Fe-S-containing polymerases, promoting replication stress, DNA damage response, and senescence. This was also observed in stem cell-derived endothelial cells from Friedreich's ataxia (FRDA), a genetic disease of FXN deficiency, ataxia, and cardiomyopathy, often with PH. In vivo, FXN deficiency-dependent senescence drove vessel inflammation, remodeling, and PH, whereas pharmacologic removal of senescent cells in Fxn-deficient rodents ameliorated PH. These data offer a model of endothelial biology in PH, where FXN deficiency generates a senescent endothelial subpopulation, promoting vascular inflammatory and proliferative signals in other cells to drive disease. These findings also establish an endothelial etiology for PH in FRDA and left heart disease and support therapeutic development of senolytic drugs, reversing effects of Fe-S deficiency across PH subtypes.
Collapse
Affiliation(s)
- Miranda K. Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yi Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Dror Perk
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Qiujun Yu
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Chen-Shan C. Woodcock
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Adam Handen
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Gil Speyer
- Research Computing, Arizona State University, Tempe, Arizona, USA
| | - Seungchan Kim
- Center for Computational Systems Biology, Department of Electrical and Computer Engineering, College of Engineering, Prairie View A&M University, Prairie View, Texas, USA
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Taijyu Satoh
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Annie M.M. Watson
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - John Sembrat
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Dmitry Goncharov
- Lung Center, Pulmonary Vascular Disease Program, Division of Pulmonary, Critical Care and Sleep Medicine, University of California Davis School of Medicine, Davis, California, USA
| | - Elena A. Goncharova
- Lung Center, Pulmonary Vascular Disease Program, Division of Pulmonary, Critical Care and Sleep Medicine, University of California Davis School of Medicine, Davis, California, USA
| | - Omar F. Khan
- Institute of Biomedical Engineering, Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Daniel G. Anderson
- Department of Chemical Engineering, Institute of Medical Engineering and Science, Harvard-MIT Division of Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - James E. Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Aditi U. Gurkar
- Aging Institute, Division of Geriatric Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, GRECC VA, Pittsburgh, Pennsylvania, USA
| | - Robert Lafyatis
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ahmed U. Fayyaz
- Department of Cardiovascular Medicine and
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesotta, USA
| | | | - Mark T. Gladwin
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Marlene Rabinovitch
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Thomas Bertero
- Université Côte d’Azur, CNRS, UMR7275, IPMC, Valbonne, France
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Fu X, Eggert M, Yoo S, Patel N, Zhong J, Steinke I, Govindarajulu M, Turumtay EA, Mouli S, Panizzi P, Beyers R, Denney T, Arnold R, Amin RH. The Cardioprotective Mechanism of Phenylaminoethyl Selenides (PAESe) Against Doxorubicin-Induced Cardiotoxicity Involves Frataxin. Front Pharmacol 2021; 11:574656. [PMID: 33912028 PMCID: PMC8072348 DOI: 10.3389/fphar.2020.574656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/09/2020] [Indexed: 11/28/2022] Open
Abstract
Doxorubicin (DOX) is an anthracycline cancer chemotherapeutic that exhibits cumulative dose-limiting cardiotoxicity and limits its clinical utility. DOX treatment results in the development of morbid cardiac hypertrophy that progresses to congestive heart failure and death. Recent evidence suggests that during the development of DOX mediated cardiac hypertrophy, mitochondrial energetics are severely compromised, thus priming the cardiomyocyte for failure. To mitigate cumulative dose (5 mg/kg, QIW x 4 weeks with 2 weeks recovery) dependent DOX, mediated cardiac hypertrophy, we applied an orally active selenium based compound termed phenylaminoethyl selenides (PAESe) (QIW 10 mg/kg x 5) to our animal model and observed that PAESe attenuates DOX-mediated cardiac hypertrophy in athymic mice, as observed by MRI analysis. Mechanistically, we demonstrated that DOX impedes the stability of the iron-sulfur cluster biogenesis protein Frataxin (FXN) (0.5 fold), resulting in enhanced mitochondrial free iron accumulation (2.5 fold) and reduced aconitase activity (0.4 fold). Our findings further indicate that PAESe prevented the reduction of FXN levels and the ensuing elevation of mitochondrial free iron levels. PAESe has been shown to have anti-oxidative properties in part, by regeneration of glutathione levels. Therefore, we observed that PAESe can mitigate DOX mediated cardiac hypertrophy by enhancing glutathione activity (0.4 fold) and inhibiting ROS formation (1.8 fold). Lastly, we observed that DOX significantly reduced cellular respiration (basal (5%) and uncoupled (10%)) in H9C2 cardiomyoblasts and that PAESe protects against the DOX-mediated attenuation of cellular respiration. In conclusion, the current study determined the protective mechanism of PAESe against DOX mediated myocardial damage and that FXN is implicitly involved in DOX-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Xiaoyu Fu
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Mathew Eggert
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Sieun Yoo
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Nikhil Patel
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Juming Zhong
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Ian Steinke
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Manoj Govindarajulu
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | | | - Shravanthi Mouli
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Peter Panizzi
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Ronald Beyers
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, United States.,Auburn University M.R.I. Research Center, Auburn, AL, United States
| | - Thomas Denney
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, United States.,Auburn University M.R.I. Research Center, Auburn, AL, United States
| | - Robert Arnold
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| | - Rajesh H Amin
- Department of Drug, Discovery and Development, Harrison School of Pharmacy, Auburn University, Alabama, AL, United States
| |
Collapse
|
32
|
Zheng J, Chen P, Zhong J, Cheng Y, Chen H, He Y, Chen C. HIF‑1α in myocardial ischemia‑reperfusion injury (Review). Mol Med Rep 2021; 23:352. [PMID: 33760122 PMCID: PMC7974458 DOI: 10.3892/mmr.2021.11991] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a severe injury to the ischemic myocardium following the recovery of blood flow. Currently, there is no effective treatment for MIRI in clinical practice. Over the past two decades, biological studies of hypoxia and hypoxia-inducible factor-1α (HIF-1α) have notably improved understanding of oxygen homeostasis. HIF-1α is an oxygen-sensitive transcription factor that mediates adaptive metabolic responses to hypoxia and serves a pivotal role in MIRI. In particular, previous studies have demonstrated that HIF-1α improves mitochondrial function, decreases cellular oxidative stress, activates cardioprotective signaling pathways and downstream protective genes and interacts with non-coding RNAs. The present review summarizes the roles and associated mechanisms of action of HIF-1α in MIRI. In addition, HIF-1α-associated MIRI intervention, including natural compounds, exosomes, ischemic preconditioning and ischemic post-processing are presented. The present review provides evidence for the roles of HIF-1α activation in MIRI and supports its use as a therapeutic target.
Collapse
Affiliation(s)
- Jie Zheng
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Peier Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Jianfeng Zhong
- Guangdong Key Laboratory of Age‑related Cardiac and Cerebral Diseases, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yu Cheng
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Hao Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524000, P.R. China
| | - Can Chen
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524003, P.R. China
| |
Collapse
|
33
|
Liu J, He H, Wang J, Guo X, Lin H, Chen H, Jiang C, Chen L, Yao P, Tang Y. Oxidative stress-dependent frataxin inhibition mediated alcoholic hepatocytotoxicity through ferroptosis. Toxicology 2020; 445:152584. [PMID: 33017621 DOI: 10.1016/j.tox.2020.152584] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022]
Abstract
Alcoholic liver disease (ALD) is one of the severe liver diseases, resulting in high morbidity and mortality. However, frataxin, a mitochondrial protein mainly participating in iron homeostasis and oxidative stress, remains uncertain in the pathogenesis of ALD. In the present study, the role of frataxin in ALD was investigated. Ethanol (100 mM) decreased frataxin expression at 48 and 72 h in HepG2. Dramatically, in HepG2 overexpressing cytochrome P450 2E1 (HepG2CYP2E1+/+), frataxin level was down-regulated with ethanol stimulation at 12 h. Moreover, chronically feeding ethanol to mice via Lieber-DeCarli liquid diet (30 % of total calories) for 15 weeks significantly inhibited frataxin expression. Ferroptosis signature proteins were dysregulated, accompanied by mitochondrial damage of morphology, enhanced malondialdehyde and decreased glutathione in the liver, as well as accumulation of reactive oxygen species and mitochondrial labile iron pool in primary hepatocytes. Notably, proteomics screening of frataxin deficient-HepG2 further suggested frataxin was associated with ferroptosis. Furthermore, the ferroptosis inhibitor ferrostatin-1 blocked the increase of lactate dehydrogenase release by ethanol in HepG2CYP2E1+/+. Most importantly, frataxin deficiency enhanced ferroptosis driven by ethanol via evaluating the levels of lactate dehydrogenase, cell morphological changes, mitochondrial labile iron pool, and lipid peroxidation. Conversely, restoring frataxin alleviated the sensitivity to ferroptosis. In addition, frataxin overexpression mitigated the sensitivity of ethanol-induced ferroptosis in HepG2CYP2E1+/+. Collectively, our study revealed that frataxin-mediated ferroptosis contributed to ALD, highlighting a potential therapeutic strategy for ALD.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hui He
- Department of Preventive Medicine, Changzhi Medical College, Changzhi 046000, China
| | - Jing Wang
- Preventive Medicine Experimental Teaching Center, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongkun Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huimin Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunjie Jiang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
34
|
Chen X, Wang C, Yang P, Shi L, Wang H. Ube2s-stabilized β-catenin protects against myocardial ischemia/reperfusion injury by activating HIF-1α signaling. Aging (Albany NY) 2020; 12:5716-5732. [PMID: 32250966 PMCID: PMC7185123 DOI: 10.18632/aging.102960] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 02/22/2020] [Indexed: 04/12/2023]
Abstract
The activation of hypoxia-inducible factor (HIF) is an important event for mediating the adaptive response to myocardial ischemia/reperfusion (MI/R) injury. The ubiquitin-conjugating enzyme E2S (Ube2s) catalyzes ubiquitin conjugation to target proteins. Here, we report the positive regulation of HIF-1α signaling by Ube2s via stabilizing β-catenin, by which Ube2s acts to protect against MI/R injury. We show that Ube2s expression is upregulated in the hearts of mice subjected to MI/R injury. Functionally, Ube2s depletion exacerbates and its overexpression ameliorates MI/R injury. In addition, Ube2s augments the activation of HIF-1α and reduces myocardial apoptosis. Moreover, Ube2s induces the accumulation of β-Catenin through increasing its stabilization. Importantly, β-Catenin knockdown abrogates Ube2s-augmented HIF-1α activation, and meanwhile, diminishes the protective effect of Ube2s on MI/R injury, thus establishing a causal link between Ube2s-stabilized β-catenin and HIF-1α-mediated myocardial protection. Altogether, this study identifies the Ube2s/β-catenin/HIF-1α axis as a novel protective regulator involved in MI/R injury, and also implies that it might represent a potential therapeutic target for ameliorating MI/R injury.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Chiyao Wang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Pei Yang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Lei Shi
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Haiyan Wang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| |
Collapse
|
35
|
Packer M. Critical examination of mechanisms underlying the reduction in heart failure events with SGLT2 inhibitors: identification of a molecular link between their actions to stimulate erythrocytosis and to alleviate cellular stress. Cardiovasc Res 2020; 117:74-84. [PMID: 32243505 DOI: 10.1093/cvr/cvaa064] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/10/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Sodium-glucose co-transporter 2 (SGLT2) inhibitors reduce the risk of serious heart failure events, even though SGLT2 is not expressed in the myocardium. This cardioprotective benefit is not related to an effect of these drugs to lower blood glucose, promote ketone body utilization or enhance natriuresis, but it is linked statistically with their action to increase haematocrit. SGLT2 inhibitors increase both erythropoietin and erythropoiesis, but the increase in red blood cell mass does not directly prevent heart failure events. Instead, erythrocytosis is a biomarker of a state of hypoxia mimicry, which is induced by SGLT2 inhibitors in manner akin to cobalt chloride. The primary mediators of the cellular response to states of energy depletion are sirtuin-1 and hypoxia-inducible factors (HIF-1α/HIF-2α). These master regulators promote the cellular adaptation to states of nutrient and oxygen deprivation, promoting mitochondrial capacity and minimizing the generation of oxidative stress. Activation of sirtuin-1 and HIF-1α/HIF-2α also stimulates autophagy, a lysosome-mediated degradative pathway that maintains cellular homoeostasis by removing dangerous constituents (particularly unhealthy mitochondria and peroxisomes), which are a major source of oxidative stress and cardiomyocyte dysfunction and demise. SGLT2 inhibitors can activate SIRT-1 and stimulate autophagy in the heart, and thereby, favourably influence the course of cardiomyopathy. Therefore, the linkage between erythrocytosis and the reduction in heart failure events with SGLT2 inhibitors may be related to a shared underlying molecular mechanism that is triggered by the action of these drugs to induce a perceived state of oxygen and nutrient deprivation.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, 621 N. Hall Street, Dallas, TX 75226, USA.,Imperial College, London, UK
| |
Collapse
|
36
|
Investigating the potential effects of selective histone deacetylase 6 inhibitor ACY1215 on infarct size in rats with cardiac ischemia-reperfusion injury. BMC Pharmacol Toxicol 2020; 21:21. [PMID: 32178737 PMCID: PMC7077123 DOI: 10.1186/s40360-020-0400-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 02/28/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Despite the fact that histone deacetylase (HDAC) inhibitors have been tested to treat various cardiovascular diseases, the effects of selective HDAC6 inhibitor ACY1215 on infarct size during cardiac ischemia-reperfusion (IR) injury still remain unknown. In the present study we aimed to investigate the effects of ACY1215 on infarct size in rats with cardiac IR injury, as well as to examine the association between HDAC6 inhibitors and the gene expression of hypoxia inducible factor-1α (HIF-1α), a key regulator of cellular responses to hypoxia. METHODS By using computational analysis of high-throughput expression profiling dataset, the association between HDAC inhibitors (pan-HDAC inhibitors panobinostat and vorinostat, and HDAC6 inhibitor ISOX) and their effects on HIF-1α gene-expression were evaluated. The male Wistar rats treated with ligation of left coronary artery followed by reperfusion were used as a cardiac IR model. ACY1215 (50 mg/kg), pan-HDAC inhibitor MPT0E028 (25 mg/kg), and vehicle were intraperitoneally injected within 5 min before reperfusion. The infarct size in rat myocardium was determined by 2,3,5-triphenyltetrazolium chloride staining. The serum levels of transforming growth factor-β (TGF-β) and C-reactive protein (CRP) were also determined. RESULTS The high-throughput gene expression assay showed that treatment of ISOX was associated with a more decreased gene expression of HIF-1α than that of panobinostat and vorinostat. Compared to control rats, ACY1215-treated rats had a smaller infarct size (49.75 ± 9.36% vs. 19.22 ± 1.70%, p < 0.05), while MPT0E028-treated rats had a similar infarct size to control rats. ACY-1215- and MPT0E028-treated rats had a trend in decreased serum TGF-β levels, but not statistically significant. ACY1215-treated rats also had higher serum CRP levels compared to control rats (641.6 μg/mL vs. 961.37 ± 64.94 μg/mL, p < 0.05). CONCLUSIONS Our research indicated that HDAC6 inhibition by ACY1215 might reduce infarct size in rats with cardiac IR injury possibly through modulating HIF-1α expression. TGF-β and CRP should be useful biomarkers to monitor the use of ACY1215 in cardiac IR injury.
Collapse
|
37
|
Panax Notoginseng Saponins Attenuate Myocardial Ischemia-Reperfusion Injury Through the HIF-1α/BNIP3 Pathway of Autophagy. J Cardiovasc Pharmacol 2020; 73:92-99. [PMID: 30531436 DOI: 10.1097/fjc.0000000000000640] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Panax Notoginseng Saponins (PNS) is a formula of Chinese medicine commonly used for treating ischemia myocardial in China. However, its mechanism of action is yet unclear. This study investigated the effect and the mechanism of PNS on myocardial ischemia-reperfusion injury (MIRI) through the hypoxia-inducible factor 1α (HIF-1α)/bcl-2/adenovirus E1B19kDa-interacting protein3 (BNIP3) pathway of autophagy. METHODS We constructed a rat model of myocardial injury and compared among 4 groups (n = 10, each): the sham-operated group (Sham), the ischemia-reperfusion group (IR), the PNS low-dose group, and the PNS high-dose group were pretreated with PNS (30 and 60 mg/kg, respectively). Serum creatine kinase, malonaldehyde (MDA), lactate dehydrogenase, myocardial tissue superoxide dismutase, and reactive oxygen species were detected in rats with myocardial ischemia-reperfusion after the intervention of PNS. The rat myocardial tissue was examined using hematoxylin and eosin (H&E) staining, and the mitochondria of myocardial cells were observed using transmission electron microscopy. The expressions of microtubule-associated protein light chain 3 (LC3), HIF-1α, BNIP3, Beclin-1, and autophagy-related gene-5 (Atg5) in rat myocardial tissue were detected using Western blotting. RESULTS The results showed that PNS was significantly protected against MIRI, as evidenced by the decreasing in the concentration of serum CK, MDA, lactate dehydrogenase, and myocardial tissue superoxide dismutase, reactive oxygen species, the attenuation of myocardial tissue histopathological changes and the mitochondrial damages of myocardial cells, and the increase of mitochondria autophagosome in myocardial cells. In addition, PNS significantly increased the expression of LC3 and the ratio of LC3II/LC3I in rat myocardial tissue. Moreover, PNS significantly increased the expression of HIF-1α, BNIP3, Atg5, and Beclin-1 in rat myocardial tissue. CONCLUSIONS The protective effect of PNS on MIRI was mainly due to its ability to enhance the mitochondrial autophagy of myocardial tissue through the HIF-1α/BNIP3 pathway.
Collapse
|
38
|
Liu ZG, Li Y, Jiao JH, Long H, Xin ZY, Yang XY. MicroRNA regulatory pattern in spinal cord ischemia-reperfusion injury. Neural Regen Res 2020; 15:2123-2130. [PMID: 32394971 PMCID: PMC7716024 DOI: 10.4103/1673-5374.280323] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
After spinal cord injury, dysregulated miRNAs appear and can participate in inflammatory responses, as well as the inhibition of apoptosis and axon regeneration through multiple pathways. However, the functions of miRNAs in spinal cord ischemia-reperfusion injury progression remain unclear. miRCURY LNATM Arrays were used to analyze miRNA expression profiles of rats after 90 minutes of ischemia followed by reperfusion for 24 and 48 hours. Furthermore, subsequent construction of aberrantly expressed miRNA regulatory patterns involved cell survival, proliferation, and apoptosis. Remarkably, the mitogen-activated protein kinase (MAPK) signaling pathway was the most significantly enriched pathway among 24- and 48-hour groups. Bioinformatics analysis and quantitative reverse transcription polymerase chain reaction confirmed the persistent overexpression of miR-22-3p in both groups. These results suggest that the aberrant miRNA regulatory network is possibly regulated MAPK signaling and continuously affects the physiological and biochemical status of cells, thus participating in the regulation of spinal cord ischemia-reperfusion injury. As such, miR-22-3p may play sustained regulatory roles in spinal cord ischemia-reperfusion injury. All experimental procedures were approved by the Animal Ethics Committee of Jilin University, China [approval No. 2020 (Research) 01].
Collapse
Affiliation(s)
- Zhi-Gang Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yin Li
- School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Jian-Hang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Hao Long
- Pain Clinic, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zhuo-Yuan Xin
- The Key Laboratory of Zoonosis Search, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xiao-Yu Yang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
39
|
Monnier V, Llorens JV, Navarro JA. Impact of Drosophila Models in the Study and Treatment of Friedreich's Ataxia. Int J Mol Sci 2018; 19:E1989. [PMID: 29986523 PMCID: PMC6073496 DOI: 10.3390/ijms19071989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Drosophila melanogaster has been for over a century the model of choice of several neurobiologists to decipher the formation and development of the nervous system as well as to mirror the pathophysiological conditions of many human neurodegenerative diseases. The rare disease Friedreich’s ataxia (FRDA) is not an exception. Since the isolation of the responsible gene more than two decades ago, the analysis of the fly orthologue has proven to be an excellent avenue to understand the development and progression of the disease, to unravel pivotal mechanisms underpinning the pathology and to identify genes and molecules that might well be either disease biomarkers or promising targets for therapeutic interventions. In this review, we aim to summarize the collection of findings provided by the Drosophila models but also to go one step beyond and propose the implications of these discoveries for the study and cure of this disorder. We will present the physiological, cellular and molecular phenotypes described in the fly, highlighting those that have given insight into the pathology and we will show how the ability of Drosophila to perform genetic and pharmacological screens has provided valuable information that is not easily within reach of other cellular or mammalian models.
Collapse
Affiliation(s)
- Véronique Monnier
- Unité de Biologie Fonctionnelle et Adaptative (BFA), Sorbonne Paris Cité, Université Paris Diderot, UMR8251 CNRS, 75013 Paris, France.
| | - Jose Vicente Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, 96100 Valencia, Spain.
| | - Juan Antonio Navarro
- Lehrstuhl für Entwicklungsbiologie, Universität Regensburg, 93040 Regensburg, Germany.
| |
Collapse
|
40
|
Niu G, Zhu D, Zhang X, Wang J, Zhao Y, Wang X. Role of Hypoxia-Inducible Factors 1α (HIF1α) in SH-SY5Y Cell Autophagy Induced by Oxygen-Glucose Deprivation. Med Sci Monit 2018; 24:2758-2766. [PMID: 29724989 PMCID: PMC5954843 DOI: 10.12659/msm.905140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background HIF-1α plays an important role in hypoxia-ischemia brain damage. Accumulating evidences demonstrates that HIF-1α can contribute to cell autophagy. Oxygen-glucose deprivation (OGD) is a commonly used ischemic model in vitro. Our study was performed to investigate the influences of HIF-1α on autophagy in SH-SY5Y cells under OGD treatment. Material/Methods An OGD model was constructed in SH-SY5Y cells. PI method and MTT assay were used to test cell death and viability, respectively. Western blot assay was used to estimate the protein levels of HIF-1α and LC3. Quantitative GFP-LC3 light microscopy autophagy assay was performed for SH-SY5Y cells. 2ME2 and siRNA-HIF-1α were applied to investigate the effects of HIF-1α-knockdown on LC3 expression. Additionally, 3-MA (autophagy inhibitor) and autophagy inducer rapamycin (Rapa) were used to investigate the effects of autophagy on cell survival under OGD condition. Results Under OGD, the apoptosis of SH-SY5Ycells was increased while cell viability rate was decreased. The expression of HIF-1α was increased along with the advancement of OGD treatment and achieved the highest level at 24 h. However, inhibiting HIF-1α expression decreased the cell apoptosis and increased cell viability. LC3-II expression was gradually increased with the duration of OGD condition and knockdown of HIF-1α resulted in decreased expression of LC3. Inhibiting autophagy significantly enhanced the viability and reduced the apoptosis of SH-SY5Y cells, while enhancing autophagy showed the opposite effects. Conclusions Enhanced expression of HIF-1α may be related to autophagy activation in SH-SY5Y cells, thus contributing to ischemic/hypoxic brain damage.
Collapse
Affiliation(s)
- Guohui Niu
- Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Dengna Zhu
- Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Xiaoli Zhang
- Department of Pediatric Internal Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Jun Wang
- Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Yunxia Zhao
- Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Xin Wang
- Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
41
|
Wang S, Lv Y, Wang Y, Du P, Tan W, Lammi MJ, Guo X. Network Analysis of Se-and Zn-related Proteins in the Serum Proteomics Expression Profile of the Endemic Dilated Cardiomyopathy Keshan Disease. Biol Trace Elem Res 2018; 183:40-48. [PMID: 28819918 DOI: 10.1007/s12011-017-1063-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/23/2017] [Indexed: 11/26/2022]
Abstract
Keshan disease (KD) is an endemic cardiomyopathy with high mortality. Selenium (Se) and zinc (Zn) deficiencies are closely related to KD. The molecular mechanism of KD pathogenesis is still unclear. There are only few studies on the interaction of trace elements and proteins associated with the pathogenesis of KD. In this study, isobaric tags for relative and absolute quantitation (iTRAQ)-coupled two-dimensional liquid chromatography tandem mass spectrometry (2DLC-MS/MS) technique analysis was used to analyze the differential expression of proteins from serum samples. Comparative Toxicogenomics Database (CTD) was used to screen Se- and Zn-associated proteins. Then, pathway and network analyses of Se- and Zn-associated proteins were constituted by Cytoscape ClueGO and GeneMANIA plugins. One hundred and five differentially expressed proteins were obtained by 2DLC-MS/MS, among them 19 Se- and 3 Zn-associated proteins. Fifty-two pathways were identified from ClueGO and 1 network from GeneMANIA analyses. The results showed that Se-associated proteins STAT3 and MAPK1 and Zn-associated proteins HIF1A and PARP1, the proteins involved in HIF-1 signaling pathway and apoptosis pathway, may play significant roles in the pathogenesis of KD. The approach of this study would be also beneficial for further dissecting molecular mechanism of other trace element-associated disease.
Collapse
Affiliation(s)
- Sen Wang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yanyan Lv
- Department of Rheumatology, Xi'an No.5 Hospital, Xi'an, Shaanxi, China
| | - Yingting Wang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Peiru Du
- School of Public Health, Health Science Center of Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Wuhong Tan
- School of Public Health, Health Science Center of Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| | - Mikko J Lammi
- School of Public Health, Health Science Center of Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Department of Integrative Medical Biology, University of Umeå, Umeå, Sweden.
| | - Xiong Guo
- School of Public Health, Health Science Center of Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
42
|
Yu Y, Yu Y, Zhang Y, Zhang Z, An W, Zhao X. Treatment with D-β-hydroxybutyrate protects heart from ischemia/reperfusion injury in mice. Eur J Pharmacol 2018; 829:121-128. [PMID: 29679541 DOI: 10.1016/j.ejphar.2018.04.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/20/2022]
Abstract
The present study was designed to examine the protection of D-β-hydroxybutyrate (BHB) against ischemia/reperfusion (I/R) injury in heart and investigate its underlying mechanism. Male adult mice were exposed to 30 min of ischemia and 24 h of reperfusion. Osmotic pumps were implanted subcutaneously 5 min before reperfusion for continuous delivery of the exogenous BHB (1.6 mmol/kg/24 h). Treatment with BHB reduced infarct size and levels of cardiac troponin I, creatine kinase and lactate dehydrogenase in serum, attenuated apoptosis in myocardium, and preserved cardiac function of I/R mice. Importantly, treatment of I/R mice with BHB promoted autophagic flux, evidenced by reduced the ratio of LC3-II/LC3-I and protein expression of p62 and enhanced protein expression of lysosome associated membrane protein-2 (Lamp2) in myocardium. Treatment of I/R mice with BHB reduced mitochondrial formation of reactive oxygen species, enhanced adenosine triphosphate production, attenuated mitochondrial swelling, and partly restored mitochondrial membrane potential in myocardium. Furthermore, treatment of I/R mice with BHB abated oxidative stress and attenuated endoplasmic reticulum stress in myocardium. Our results indicated that treatment with exogenous BHB protected heart from I/R injury in mice.
Collapse
Affiliation(s)
- Yongsheng Yu
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Changhai Road 168, Shanghai 200433, China
| | - Yunhua Yu
- Department of Geriatrics, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Yuefan Zhang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Zhigang Zhang
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Weishuai An
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Changhai Road 168, Shanghai 200433, China
| | - Xianxian Zhao
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Changhai Road 168, Shanghai 200433, China.
| |
Collapse
|
43
|
Nazari-Shafti TZ, Xu Z, Bader AM, Henke G, Klose K, Falk V, Stamm C. Mesenchymal Stromal Cells Cultured in Serum from Heart Failure Patients Are More Resistant to Simulated Chronic and Acute Stress. Stem Cells Int 2018; 2018:5832460. [PMID: 29760728 PMCID: PMC5901835 DOI: 10.1155/2018/5832460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/14/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022] Open
Abstract
Despite regulatory issues surrounding the use of animal-derived cell culture supplements, most clinical cardiac cell therapy trials using mesenchymal stromal cells (MSCs) still rely on fetal bovine serum (FBS) for cell expansion before transplantation. We sought to investigate the effect of human serum from heart failure patients (HFS) on cord blood MSCs (CB-MSCs) during short-term culture under regular conditions and during simulated acute and chronic stress. Cell survival, proliferation, metabolic activity, and apoptosis were quantified, and gene expression profiles of selected apoptosis and cell cycle regulators were determined. Compared to FBS, HFS and serum from healthy donors (CS) showed similar effects by substantially increasing cell survival during chronic and acute stress and by increasing cell yields 5 days after acute stress. Shortly after the termination of acute stress, both HFS and CS resulted in a marked decrease in apoptotic cells. Transcriptome analysis suggested a decrease in TNF-mediated induction of caspases and decreased activation of mitochondrial apoptosis. Our data confirm that human serum from both healthy donors and heart failure patients results in increased cell yields and increased resistance to cellular stress signals. Therefore, we consider autologous serum a valid alternative to FBS in cell-based therapies addressing severe heart disease.
Collapse
Affiliation(s)
- Timo Z. Nazari-Shafti
- Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
- Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Zhiyi Xu
- Berlin Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | - Georg Henke
- Berlin Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Kristin Klose
- Berlin Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Volkmar Falk
- Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
- Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Christof Stamm
- Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
- Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Center for Regenerative Therapies (BCRT), Berlin, Germany
| |
Collapse
|
44
|
Ghanei N, Amin RH. Commentary: Neutral Commentary on Frontiers Article "Cobalt Chloride Upregulates Impaired HIF-1α Expression to Restore Sevoflurane Post-conditioning-Dependent Myocardial Protection in Diabetic Rats". Front Physiol 2017; 8:926. [PMID: 29209228 PMCID: PMC5702356 DOI: 10.3389/fphys.2017.00926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/31/2017] [Indexed: 11/15/2022] Open
Affiliation(s)
- Nila Ghanei
- Cardio-Metabolic Research Lab, Department of Drug Discovery and Development, The Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Rajesh H Amin
- Cardio-Metabolic Research Lab, Department of Drug Discovery and Development, The Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| |
Collapse
|
45
|
Zhang W, Ren H, Xu C, Zhu C, Wu H, Liu D, Wang J, Liu L, Li W, Ma Q, Du L, Zheng M, Zhang C, Liu J, Chen Q. Hypoxic mitophagy regulates mitochondrial quality and platelet activation and determines severity of I/R heart injury. eLife 2016; 5. [PMID: 27995894 PMCID: PMC5214169 DOI: 10.7554/elife.21407] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/18/2016] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial dysfunction underlies many prevalent diseases including heart disease arising from acute ischemia/reperfusion (I/R) injury. Here, we demonstrate that mitophagy, which selectively removes damaged or unwanted mitochondria, regulated mitochondrial quality and quantity in vivo. Hypoxia induced extensive mitochondrial degradation in a FUNDC1-dependent manner in platelets, and this was blocked by in vivo administration of a cell-penetrating peptide encompassing the LIR motif of FUNDC1 only in wild-type mice. Genetic ablation of Fundc1 impaired mitochondrial quality and increased mitochondrial mass in platelets and rendered the platelets insensitive to hypoxia and the peptide. Moreover, hypoxic mitophagy in platelets protected the heart from worsening of I/R injury. This represents a new mechanism of the hypoxic preconditioning effect which reduces I/R injury. Our results demonstrate a critical role of mitophagy in mitochondrial quality control and platelet activation, and suggest that manipulation of mitophagy by hypoxia or pharmacological approaches may be a novel strategy for cardioprotection.
Collapse
Affiliation(s)
- Weilin Zhang
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - He Ren
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Chunling Xu
- Department of Physiology, Peking University School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chongzhuo Zhu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hao Wu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Dong Liu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jun Wang
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lei Liu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- Center for Medical Genetics, Beijing Children's Hospital, Capital Medical University, Beijing, China.,Beijing Pediatric Research Institute, Beijing, China.,MOE Key Laboratory of Major Diseases in Children, Beijing, China
| | - Qi Ma
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Lei Du
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ming Zheng
- Department of Physiology, Peking University School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiaotong University, Shanghai, China
| | - Quan Chen
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
46
|
Huang X, Zuo L, Lv Y, Chen C, Yang Y, Xin H, Li Y, Qian Y. Asiatic Acid Attenuates Myocardial Ischemia/Reperfusion Injury via Akt/GSK-3β/HIF-1α Signaling in Rat H9c2 Cardiomyocytes. Molecules 2016; 21:molecules21091248. [PMID: 27657024 PMCID: PMC6273770 DOI: 10.3390/molecules21091248] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 12/11/2022] Open
Abstract
Myocardial ischemic/reperfusion injury results from severe impairment of coronary blood supply and leads to irreversible cell death, with limited therapeutic possibilities. Asiatic acid is a pentacyclic triterpenoid derived from the tropical medicinal plant Centella asiatica and serves a variety of bioactivities. In this study, we determined the effect of asiatic acid on myocardial ischemia/reperfusion injury and investigated the underlying mechanisms, using an in vitro rat H9c2 cardiomyocytes model of oxygen-glucose deprivation/reoxygenation (OGD/R) injury. Results showed that pre-treatment with asiatic acid significantly augmented cell viability and prevented lactate dehydrogenase (LDH) release in a concentration-dependent manner after OGD/R exposure. Asiatic acid at 10 μM effectively inhibited apoptotic cell death, suppressed the activities of caspase-3 and caspase-9, and reversed Bax/Bcl-2 ratio in hypoxic H9c2 cells. In addition, asiatic acid improved mitochondrial function, as evidenced by reduced reactive oxygen species (ROS) accumulation, enhanced mitochondrial membrane potential and decreased intracellular calcium concentration. Using Western blot assay, we found that asiatic acid promoted the phosphorylation of Akt and subsequent inactivation of glycogen synthase kinase-3β (GSK-3β), and induced the expression of hypoxia-inducible factor 1α (HIF-1α) after OGD/R. The cardioprotective effects of asiatic acid were attenuated by the Akt or HIF-1α inhibitor. Taken together, these data suggested that asiatic acid exerted protective effects against OGD/R-induced apoptosis in cardiomyocytes, at least partly via the Akt/GSK-3β/HIF-1α pathway.
Collapse
Affiliation(s)
- Xiang Huang
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Nanchang 330031, China.
| | - Li Zuo
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Nanchang 330031, China.
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang 330006, China.
| | - Chuqiao Chen
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Nanchang 330031, China.
| | - Yaqin Yang
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Nanchang 330031, China.
| | - Hongbo Xin
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Nanchang 330031, China.
| | - Yunman Li
- Department of Physiology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | - Yisong Qian
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Nanchang 330031, China.
| |
Collapse
|
47
|
Pearson JT. Cardiac responses to hypoxia and reoxygenation in Drosophila. New insights into evolutionarily conserved gene responses. Focus on "Cardiac responses to hypoxia and reoxygenation in Drosophila". Am J Physiol Regul Integr Comp Physiol 2015; 309:R1344-6. [PMID: 26447213 DOI: 10.1152/ajpregu.00419.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 11/22/2022]
Affiliation(s)
- James T Pearson
- Monash Biomedical Imaging Facility, Melbourne, Australia, Department of Physiology, Monash University, Melbourne, Australia; and Australian Synchrotron, Melbourne, Australia
| |
Collapse
|
48
|
Amaral N, Okonko DO. Mitigation of myocardial ischemia-reperfusion injury via HIF-1α-frataxin signaling. Am J Physiol Heart Circ Physiol 2015; 309:H728-30. [PMID: 26209059 PMCID: PMC4591396 DOI: 10.1152/ajpheart.00553.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Nelson Amaral
- Cardiovascular Division, British Heart Foundation Centre of Excellence, James Black Centre, King's College London, London, United Kingdom
| | - Darlington O Okonko
- Cardiovascular Division, British Heart Foundation Centre of Excellence, James Black Centre, King's College London, London, United Kingdom
| |
Collapse
|