1
|
Nakanishi N, Kaikita K, Oimatsu Y, Ishii M, Kuyama N, Arima Y, Araki S, Nakamura T, Yamamoto E, Tsujita K. 5-Aminolevulinic acid combined with ferrous iron ameliorates myocardial ischemia/reperfusion injury by increasing heme oxygenase-1. Heart Vessels 2025; 40:545-556. [PMID: 39499265 DOI: 10.1007/s00380-024-02480-9] [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: 04/09/2024] [Accepted: 10/23/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND 5-Aminolevulinic acid (5-ALA) is a naturally occurring metabolic precursor of heme, and 5-ALA combined with ferrous iron can induce heme oxygenase-1 (HO-1) in various cells. In this study, we investigated the cardioprotective effect of 5-ALA after myocardial ischemia/reperfusion (I/R) injury using a murine model. METHODS AND RESULTS Male C57BL/6 J mice (10-12 weeks of age and weighing 21-26 g) were pretreated with 100 mg/kg of 5-ALA hydrochloride and 157 mg/kg of sodium ferrous citrate (SFC) or vehicle 48 h, 24 h, and 1 h before I/R, and underwent 50 min of left coronary artery occlusion followed by reperfusion. Infarct area (IA) and area at risk (AAR) were determined by Evans blue and triphenyltetrazolium chloride double staining after reocclusion. Pre-administration with 5-ALA/SFC significantly reduced IA/AAR compared with placebo (34.0% vs. 51.7%, respectively; p = 0.001). Real-time PCR assay after reperfusion showed that mRNA expressions of TNF-α, IL-1β, and BNP were significantly lower, and that of HO-1 was significantly higher in the 5-ALA/SFC group than in the vehicle group in ischemic sites. An inhibition experiment revealed that zinc protoporphyrin IX, an inhibitor of HO-1, inhibited the cardioprotective effects of 5-ALA/SFC. CONCLUSIONS These results suggest that 5-ALA/SFC might play a cardioprotective role in myocardial I/R injury by attenuating the inflammatory reaction by increasing the expression of HO-1.
Collapse
Affiliation(s)
- Nobuhiro Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Division of Cardiovascular Medicine and Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
| | - Yu Oimatsu
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoto Kuyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Nakamura
- Department of Medical Information Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
2
|
Chen J, Wang B, Meng T, Li C, Liu C, Liu Q, Wang J, Liu Z, Zhou Y. Oxidative Stress and Inflammation in Myocardial Ischemia-Reperfusion Injury: Protective Effects of Plant-Derived Natural Active Compounds. J Appl Toxicol 2024. [PMID: 39482870 DOI: 10.1002/jat.4719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of death among patients with cardiovascular diseases. Percutaneous coronary intervention (PCI) has been the preferred clinical treatment for AMI due to its safety and efficiency. However, research indicates that the rapid restoration of myocardial oxygen supply following PCI can lead to secondary myocardial injury, termed myocardial ischemia-reperfusion injury (MIRI), posing a grave threat to patient survival. Despite ongoing efforts, the mechanisms underlying MIRI are not yet fully elucidated. Among them, oxidative stress and inflammation stand out as critical pathophysiological mechanisms, playing significant roles in MIRI. Natural compounds have shown strong clinical therapeutic potential due to their high efficacy, availability, and low side effects. Many current studies indicate that natural compounds can mitigate MIRI by reducing oxidative stress and inflammatory responses. Therefore, this paper reviews the mechanisms of oxidative stress and inflammation during MIRI and the role of natural compounds in intervening in these processes, aiming to provide a basis and reference for future research and development of drugs for treating MIRI.
Collapse
Affiliation(s)
- Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Wang Y, Han J, Zhan S, Guo C, Yin S, Zhan L, Zhou Q, Liu R, Yan H, Wang X, Yan D. Fucoidan alleviates doxorubicin-induced cardiotoxicity by inhibiting ferroptosis via Nrf2/GPX4 pathway. Int J Biol Macromol 2024; 276:133792. [PMID: 38992539 DOI: 10.1016/j.ijbiomac.2024.133792] [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: 02/09/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Doxorubicin (Dox), a chemotherapeutic agent frequently used to treat cancer, elicits cardiotoxicity, a condition referred to as Dox-induced cardiotoxicity (DIC), and ferroptosis plays a contributory role in its pathophysiology. Fucoidan, a polysaccharide with various biological activities and safety profile, has potential therapeutic and pharmaceutical applications. This study aimed to investigate the protective effects and underlying mechanisms of fucoidan in DIC. Echocardiography, biomarkers of cardiomyocyte injury, serum creatine kinase, creatine kinase isoenzyme and lactate dehydrogenase, as well as histological staining results, revealed that fucoidan significantly reduced myocardial damage and improved cardiac function in DIC mice. Transmission electron microscopy; levels of lipid reactive oxygen species, glutathione, and malondialdehyde; ferroptosis-related markers; and regulatory factors such as glutathione peroxidase 4 (GPX4), transferrin receptor protein-1, ferritin heavy chain-1, heme oxygenase-1 in the heart tissue were measured to explore the effect of fucoidan on Dox-induced ferroptosis. These results suggested that fucoidan could inhibit cardiomyocyte ferroptosis caused by Dox. In vitro experiments revealed that silencing nuclear factor-erythroid 2-related factor 2 (Nrf2) in cardiomyocytes reduced the inhibitory effect of fucoidan on ferroptosis. Hence, fucoidan has the potential to ameliorate DIC by inhibiting ferroptosis via the Nrf2/GPX4 pathway.
Collapse
Affiliation(s)
- Yizhi Wang
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Jiawen Han
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Shifang Zhan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Chenyu Guo
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Shuangneng Yin
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Lin Zhan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Qianyi Zhou
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Ruiying Liu
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Hua Yan
- Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan 430022, Hubei, China
| | - Xiaoyan Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, Hainan, China.
| | - Dan Yan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China; Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan 430022, Hubei, China; Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China.
| |
Collapse
|
4
|
Payne FM, Dabb AR, Harrison JC, Sammut IA. Inhibitors of NLRP3 Inflammasome Formation: A Cardioprotective Role for the Gasotransmitters Carbon Monoxide, Nitric Oxide, and Hydrogen Sulphide in Acute Myocardial Infarction. Int J Mol Sci 2024; 25:9247. [PMID: 39273196 PMCID: PMC11395567 DOI: 10.3390/ijms25179247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Myocardial ischaemia reperfusion injury (IRI) occurring from acute coronary artery disease or cardiac surgical interventions such as bypass surgery can result in myocardial dysfunction, presenting as, myocardial "stunning", arrhythmias, infarction, and adverse cardiac remodelling, and may lead to both a systemic and a localised inflammatory response. This localised cardiac inflammatory response is regulated through the nucleotide-binding oligomerisation domain (NACHT), leucine-rich repeat (LRR)-containing protein family pyrin domain (PYD)-3 (NLRP3) inflammasome, a multimeric structure whose components are present within both cardiomyocytes and in cardiac fibroblasts. The NLRP3 inflammasome is activated via numerous danger signals produced by IRI and is central to the resultant innate immune response. Inhibition of this inherent inflammatory response has been shown to protect the myocardium and stop the occurrence of the systemic inflammatory response syndrome following the re-establishment of cardiac circulation. Therapies to prevent NLRP3 inflammasome formation in the clinic are currently lacking, and therefore, new pharmacotherapies are required. This review will highlight the role of the NLRP3 inflammasome within the myocardium during IRI and will examine the therapeutic value of inflammasome inhibition with particular attention to carbon monoxide, nitric oxide, and hydrogen sulphide as potential pharmacological inhibitors of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Fergus M Payne
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Alisha R Dabb
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Joanne C Harrison
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ivan A Sammut
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
5
|
Padda I, Sethi Y, Das M, Fabian D, Ralhan T, Aziz D, Sexton J, Johal G. Heme Oxygenase-1, Cardiac Senescence, and Myocardial Infarction: A Critical Review of the Triptych. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07590-0. [PMID: 38940935 DOI: 10.1007/s10557-024-07590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE Heme oxygenase-1 (HO-1) is a crucial enzyme in heme metabolism, facilitating the breakdown of heme into biliverdin, carbon monoxide, and free iron. Renowned for its potent cytoprotective properties, HO-1 showcases notable antioxidant, anti-inflammatory, and anti-apoptotic effects. In this review, the authors aim to explore the profound impact of HO-1 on cardiac senescence and its potential implications in myocardial infarction (MI). RESULTS Recent research has unveiled the intricate role of HO-1 in cellular senescence, characterized by irreversible growth arrest and functional decline. Notably, cardiac senescence has emerged as a pivotal factor in the development of various cardiovascular conditions, including MI. Notably, cardiac senescence has emerged as an important factor in the development of various cardiovascular conditions, including myocardial infarction (MI). The accumulation of senescent cells, spanning vascular endothelial cells, vascular smooth muscle cells, cardiomyocytes, and progenitor cells, poses a significant risk for cardiovascular diseases such as vascular aging, atherosclerosis, myocardial infarction, and ventricular remodeling. Inhibition of cardiomyocyte senescence not only reduces senescence-associated inflammation but also impacts other myocardial lineages, hinting at a broader mechanism of propagation in pathological remodeling. HO-1 has been shown to improve heart function and mitigate cardiomyocyte senescence induced by ischemic injury and aging. Furthermore, HO-1 induction has been found to alleviate H2O2-induced cardiomyocyte senescence. As we grow in our understanding of antiproliferative, antiangiogenic, anti-aging, and vascular effects of HO-1, we see the potential to exploit potential links between individual susceptibility to cardiac senescence and myocardial infarction. CONCLUSIONS This review investigates strategies for upregulating HO-1, including gene targeting and pharmacological agents, as potential therapeutic approaches. By synthesizing compelling evidence from diverse experimental models and clinical investigations, this study elucidates the therapeutic potential of targeting HO-1 as an innovative strategy to mitigate cardiac senescence and improve outcomes in myocardial infarction, emphasizing the need for further research in this field.
Collapse
Affiliation(s)
- Inderbir Padda
- Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA
- PearResearch, Dehradun, India
| | - Yashendra Sethi
- PearResearch, Dehradun, India.
- Government Doon Medical College, Dehradun, Uttarakhand, India.
| | - Maumita Das
- School of Medicine, St. George's University, True Blue, Grenada
| | - Daniel Fabian
- Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA
| | - Tushar Ralhan
- Department of Internal Medicine, Robert Wood Johnson Medical School, RutgersNew Brunswick, NJ, USA
| | - Daniel Aziz
- Department of Internal Medicine, Robert Wood Johnson Medical School, RutgersNew Brunswick, NJ, USA
| | - Jaime Sexton
- Department of Internal Medicine, Robert Wood Johnson Medical School, RutgersNew Brunswick, NJ, USA
| | - Gurpreet Johal
- Valley Medical Center, University of Washington, Seattle, USA
| |
Collapse
|
6
|
Wu A, Zhong C, Song X, Yuan W, Tang M, Shu T, Huang H, Yang P, Liu Q. The activation of LBH-CRYAB signaling promotes cardiac protection against I/R injury by inhibiting apoptosis and ferroptosis. iScience 2024; 27:109510. [PMID: 38660406 PMCID: PMC11039335 DOI: 10.1016/j.isci.2024.109510] [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: 09/10/2023] [Revised: 12/29/2023] [Accepted: 03/13/2024] [Indexed: 04/26/2024] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury stands out among cardiovascular diseases, and current treatments are considered unsatisfactory. For cardiomyocytes (CMs) in ischemic tissues, the upregulation of Limb-bud and Heart (LBH) and αB-crystallin (CRYAB) and their subsequent downregulation in the context of cardiac fibrosis have been verified in our previous research. Here, we focused on the effects and mechanisms of activated LBH-CRYAB signaling on damaged CMs during I/R injury, and confirmed the occurrence of mitochondrial apoptosis and ferroptosis during I/R injury. The application of inhibitors, ectopic expression vectors, and knockout mouse models uniformly verified the role of LBH in alleviating both apoptosis and ferroptosis of CMs. p53 was identified as a mutual downstream effector for both LBH-CRYAB-modulated apoptosis and ferroptosis inhibition. In mouse models, LBH overexpression was confirmed to exert enhanced cardiac protection against I/R-induced apoptosis and ferroptosis, suggesting that LBH could serve as a promising target for the development of I/R therapy.
Collapse
Affiliation(s)
- Anbiao Wu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
- Beijing Institute of Basic Medical Sciences, Beijing 100850, People’s Republic of China
| | - Chongbin Zhong
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Xudong Song
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Wen Yuan
- Experimental Animal Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Mintian Tang
- Experimental Animal Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Tao Shu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Houda Huang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Pingzhen Yang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| | - Qicai Liu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Laboratory of Heart Center, Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, People’s Republic of China
| |
Collapse
|
7
|
Kiliç Y, Guzel Erdogan D, Baykul M, Nas K. Examining the functions of the vascular endothelial growth factor/hypoxia-inducible factor signaling pathway in psoriatic arthritis. Arch Rheumatol 2023; 38:579-589. [PMID: 38125055 PMCID: PMC10728743 DOI: 10.46497/archrheumatol.2023.9898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/18/2023] [Indexed: 12/23/2023] Open
Abstract
Objectives The present study aimed to examine the roles of the vascular endothelial growth factor (VEGF), hypoxia-inducible factor (HIF), and heme oxygenase-1 (HO-1) in psoriatic arthritis (PsA). Patients and methods In this cross-sectional study conducted between November 2020 and May 2021, 64 patients (43 female, 21 male; mean age: 43.2±10.4 years; range, 22 to 60 years) with active PsA were included in the patient group, and 64 healthy volunteers (43 female, 21 male; mean age: 42.8±10.5 years; range, 23 to 61 years) were included in the control group. The demographic features of all cases were recorded. The following indices were used to assess the activity of PsA: Bath Ankylosing Spondylitis Disease Activity Index, Disease Activity Score in 28 joints (DAS28), and Visual Analog Scale. Additionally, Disease Activity in Psoriatic Arthritis (DAPSA) and Psoriasis Area and Severity Index (PASI) were used to evaluate the patients. The biochemical parameters of the patients were calculated. The serum levels of VEGF, HIF, and HO-1 were determined using an enzyme-linked immunosorbent assay. Results When the molecule levels and clinical features of the groups were evaluated, it was found that the VEGF and HIF-1 levels were higher in the patient group compared to the control group (p<0.05). No difference was observed in the comparison of the HO-1 levels of the patient group and the control group (p<0.05). A positive correlation was found between VEGF, HIF-1, and HO-1 (p<0.05). A positive relationship was found between VEGF and HIF-1 and erythrocyte sedimentation rate, C-reactive protein, DAPSA score, and PASI score (p<0.05). It was also determined that there was a positive relationship between the HIF molecule and DAS28 (p<0.05). Conclusion According to the results obtained in the present study, VEGF and HIF play a role in the etiology of PsA, and the observation of intermolecular correlation suggests that these molecules move together in pathogenesis.
Collapse
Affiliation(s)
- Yavuz Kiliç
- Department of Physiotherapy and Rehabilitation, Sakarya University of Applied Sciences, Vocational School of Health Services, Sakarya, Türkiye
| | - Derya Guzel Erdogan
- Department of Physiology, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| | - Merve Baykul
- Department of Physical Medicine and Rehabilitation, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| | - Kemal Nas
- Department of Physical Medicine and Rehabilitation, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| |
Collapse
|
8
|
Wei X, Li Y, Luo P, Dai Y, Jiang T, Xu M, Hao Y, Zhang C, Liu Y. Development and Validation of Robust Ferroptosis-Related Genes in Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Dev Dis 2023; 10:344. [PMID: 37623357 PMCID: PMC10455596 DOI: 10.3390/jcdd10080344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
(1) Background: Despite the evidence that ferroptosis is involved in myocardial ischemia-reperfusion (MIR), the critical regulator of ferroptosis in MIR remains unclear. (2) Methods: We included three GEO datasets and a set of ferroptosis-related genes with 259 genes. Following the identification of the differentially expressed ferroptosis-related genes (DEFRGs) and hub genes, we performed the functional annotation, protein-protein interaction network, and immune infiltration analysis. The GSE168610 dataset, a cell model, and an animal model were then used to verify key genes. (3) Results: We identified 17 DEFRGs and 9 hub genes in the MIR samples compared to the control. Heme oxygenase 1 (Hmox1), activating transcription factor 3 (Atf3), epidermal growth factor receptor (Egfr), and X-box binding protein 1 (Xbp1) were significantly upregulated in response to ischemic and hypoxic stimuli. In contrast, glutathione peroxidase 4 (Gpx4) and vascular endothelial growth factor A (Vegfa) were consistently decreased in either the oxygen and glucose deprivation/reoxygenation cell or the MIR mouse model. (4) Conclusions: This study emphasized the relevance of ferroptosis in MIR. It has been successfully demonstrated that nine ferroptosis-related genes (Hmox1, Atf3, Egfr, Gpx4, Cd44, Vegfa, asparagine synthetase (Asns), Xbp1, and bromodomain containing 4 (Brd4)) are involved in the process. Additional studies are needed to explore potential therapeutic targets for MIR.
Collapse
Affiliation(s)
- Xiuxian Wei
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Li
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengcheng Luo
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Dai
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mulin Xu
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of General Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Hao
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Liu
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Y.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
9
|
Li X, Ou W, Xie M, Yang J, Li Q, Li T. Nanomedicine-Based Therapeutics for Myocardial Ischemic/Reperfusion Injury. Adv Healthc Mater 2023; 12:e2300161. [PMID: 36971662 PMCID: PMC11468948 DOI: 10.1002/adhm.202300161] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/05/2023] [Indexed: 03/29/2023]
Abstract
Myocardial ischemic/reperfusion (IR) injury is a global cardiovascular disease with high mortality and morbidity. Therapeutic interventions for myocardial ischemia involve restoring the occluded coronary artery. However, reactive oxygen species (ROS) inevitably impair the cardiomyocytes during the ischemic and reperfusion phases. Antioxidant therapy holds great promise against myocardial IR injury. The current therapeutic methodologies for ROS scavenging depend predominantly on administering antioxidants. Nevertheless, the intrinsic drawbacks of antioxidants limit their further clinical transformation. The use of nanoplatforms with versatile characteristics greatly benefits drug delivery in myocardial ischemic therapy. Nanoplatform-mediated drug delivery significantly improves drug bioavailability, increases therapeutic index, and reduces systemic toxicity. Nanoplatforms can be specifically and reasonably designed to enhance molecule accumulation at the myocardial site. The present review initially summarizes the mechanism of ROS generation during the process of myocardial ischemia. The understanding of this phenomenon will facilitate the advancement of innovative therapeutic strategies against myocardial IR injury. The latest developments in nanomedicine for treating myocardial ischemic injury are then discussed. Finally, the current challenges and perspectives in antioxidant therapy for myocardial IR injury are addressed.
Collapse
Affiliation(s)
- Xi Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Wei Ou
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
- Department of AnesthesiologyNanchong Central HospitalNanchong637000P. R. China
| | - Maodi Xie
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Jing Yang
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Qian Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Tao Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| |
Collapse
|
10
|
Xu Y, Bu G. Identification of two novel ferroptosis-associated targets in sepsis-induced cardiac injury: Hmox1 and Slc7a11. Front Cardiovasc Med 2023; 10:1185924. [PMID: 37424906 PMCID: PMC10326630 DOI: 10.3389/fcvm.2023.1185924] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose Sepsis-induced cardiac injury is a severe complication of sepsis and has a high mortality. Recent research has implicated ferroptosis as a contributing factor to myocardial cell death. This study is aimed at finding novel ferroptosis-associated targets in sepsis-induced cardiac injury. Methods and results In our study, a total of two Gene expression omnibus datasets (GSE185754 and GSE171546) were obtained for bioinformatics analysis. GSEA enrichment analysis demonstrated that ferroptosis pathway Z-score rapidly increased in the first 24 h and decreased gradually in the following 24-72 h. Fuzzy analysis was then used to obtain distinct clusters of temporal patterns and find genes in cluster 4 that exhibited the same trend with ferroptosis progression during the time points. After intersecting the differentially expressed genes, genes in cluster 4, and ferroptosis-related genes, three ferroptosis-associated targets were finally selected: Ptgs2, Hmox1, and Slc7a11. While Ptgs2 has been previously reported to be involved in the regulation of septic cardiomyopathy, this study is the first to demonstrate that downregulation of Hmox1 and Slc7a11 can alleviate ferroptosis in sepsis-induced cardiac injury. Conclusion This study reports Hmox1 and Slc7a11 as ferroptosis-associated targets in sepsis-induced cardiac injury, and both of them may become key therapeutic and diagnostic targets for this complication in the future.
Collapse
Affiliation(s)
- Yushun Xu
- Department of Cardiology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Gang Bu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Liu Y, Ji X, Zhou Z, Zhang J, Zhang J. Myocardial ischemia-reperfusion injury; Molecular mechanisms and prevention. Microvasc Res 2023:104565. [PMID: 37307911 DOI: 10.1016/j.mvr.2023.104565] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Cardiovascular diseases are one of the leading causes of mortality in developed countries. Among cardiovascular disorders, myocardial infarction remains a life-threatening problem predisposing to the development and progression of ischemic heart failure. Ischemia/reperfusion (I/R) injury is a critical cause of myocardial injury. In recent decades, many efforts have been made to find the molecular and cellular mechanisms underlying the development of myocardial I/R injury and post-ischemic remodeling. Some of these mechanisms are mitochondrial dysfunction, metabolic alterations, inflammation, high production of ROS, and autophagy deregulation. Despite continuous efforts, myocardial I/R injury remains a major challenge in medical treatments of thrombolytic therapy, heart disease, primary percutaneous coronary intervention, and coronary arterial bypass grafting. The development of effective therapeutic strategies to reduce or prevent myocardial I/R injury is of great clinical significance.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Xiang Ji
- Department of Integrative, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Zhou Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Jingwen Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Juan Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China; First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250011, China.
| |
Collapse
|
12
|
Zheng T, Huang Z, Ling H, Li J, Cheng H, Chen D, Lu Q, Zhao J, Su W. The mechanism of the Nfe2l2/Hmox1 signaling pathway in ferroptosis regulation in acute compartment syndrome. J Biochem Mol Toxicol 2023; 37:e23228. [PMID: 36193742 PMCID: PMC10078270 DOI: 10.1002/jbt.23228] [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: 10/30/2021] [Revised: 07/30/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
Acute compartment syndrome (ACS) is a life-threatening orthopedic emergency, which can even result in amputation. Ferroptosis is an iron-dependent form of nonapoptotic cell death. This study investigated the mechanism of ferroptosis in ACS, explored candidate markers, and determined effective treatments. This study identified pathways involved in the development of ACS through gene set enrichment analysis (GSEA), Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG), and GSEA of heme oxygenase 1 (Hmox1). Bioinformatics methods, combined with real-time quantitative polymerase chain reaction, western blot analysis, and iron staining, were applied to determine whether ferroptosis was involved in the progression of ACS and to explore the mechanism of nuclear factor erythroid-2-related factor 2 (Nfe2l2)/Hmox1 in ferroptosis regulation. Optimal drugs for the treatment of ACS were also investigated using Connectivity Map. The ferroptosis pathway was enriched in GSEA, KEGG of DEGs, and GSEA of Hmox1. After ACS, the reactive oxygen species content, tissue iron content, and oxidative stress level increased, whereas glutathione peroxidase 4 protein expression decreased. The skeletal muscle was swollen and necrotized; the number of mitochondrial cristae became fewer or even disappeared, and Nfe2l2/Hmox1 expression increased at the transcriptional and protein levels. Hmox1 was highly expressed in ACS, indicating that Hmox1 is a possible marker for ACS. we could predict 12 potential target drugs for the treatment of ACS. In conclusion, Hmox1 was a potential candidate marker for ACS diagnosis. Ferroptosis was involved in the progression of ACS. It was speculated that ferroptosis is inhibited by the Nfe2l2/Hmox1 signaling pathway.
Collapse
Affiliation(s)
- Tiejun Zheng
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Zhao Huang
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - He Ling
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Junfeng Li
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Cheng
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Dingquan Chen
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Qinzhen Lu
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Wei Su
- Department of Orthopaedic Traumatology and Hand Surgery, The First Affiliated Hospital to Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
13
|
Tarasov KV, Chakir K, Riordon DR, Lyashkov AE, Ahmet I, Perino MG, Silvester AJ, Zhang J, Wang M, Lukyanenko YO, Qu JH, Barrera MCR, Juhaszova M, Tarasova YS, Ziman B, Telljohann R, Kumar V, Ranek M, Lammons J, Bychkov R, de Cabo R, Jun S, Keceli G, Gupta A, Yang D, Aon MA, Adamo L, Morrell CH, Otu W, Carroll C, Chambers S, Paolocci N, Huynh T, Pacak K, Weiss R, Field L, Sollott SJ, Lakatta EG. A remarkable adaptive paradigm of heart performance and protection emerges in response to marked cardiac-specific overexpression of ADCY8. eLife 2022; 11:e80949. [PMID: 36515265 PMCID: PMC9822292 DOI: 10.7554/elife.80949] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Adult (3 month) mice with cardiac-specific overexpression of adenylyl cyclase (AC) type VIII (TGAC8) adapt to an increased cAMP-induced cardiac workload (~30% increases in heart rate, ejection fraction and cardiac output) for up to a year without signs of heart failure or excessive mortality. Here, we show classical cardiac hypertrophy markers were absent in TGAC8, and that total left ventricular (LV) mass was not increased: a reduced LV cavity volume in TGAC8 was encased by thicker LV walls harboring an increased number of small cardiac myocytes, and a network of small interstitial proliferative non-cardiac myocytes compared to wild type (WT) littermates; Protein synthesis, proteosome activity, and autophagy were enhanced in TGAC8 vs WT, and Nrf-2, Hsp90α, and ACC2 protein levels were increased. Despite increased energy demands in vivo LV ATP and phosphocreatine levels in TGAC8 did not differ from WT. Unbiased omics analyses identified more than 2,000 transcripts and proteins, comprising a broad array of biological processes across multiple cellular compartments, which differed by genotype; compared to WT, in TGAC8 there was a shift from fatty acid oxidation to aerobic glycolysis in the context of increased utilization of the pentose phosphate shunt and nucleotide synthesis. Thus, marked overexpression of AC8 engages complex, coordinate adaptation "circuity" that has evolved in mammalian cells to defend against stress that threatens health or life (elements of which have already been shown to be central to cardiac ischemic pre-conditioning and exercise endurance cardiac conditioning) that may be of biological significance to allow for proper healing in disease states such as infarction or failure of the heart.
Collapse
Affiliation(s)
- Kirill V Tarasov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Khalid Chakir
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Daniel R Riordon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Alexey E Lyashkov
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Ismayil Ahmet
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Maria Grazia Perino
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Allwin Jennifa Silvester
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Jing Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Yevgeniya O Lukyanenko
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Jia-Hua Qu
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Miguel Calvo-Rubio Barrera
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Magdalena Juhaszova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Yelena S Tarasova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Bruce Ziman
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Richard Telljohann
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Vikas Kumar
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Mark Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - John Lammons
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Rostislav Bychkov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Rafael de Cabo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Seungho Jun
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ashish Gupta
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Dongmei Yang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Christopher H Morrell
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Walter Otu
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Cameron Carroll
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Shane Chambers
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Thanh Huynh
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Robert Weiss
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Loren Field
- Kraennert Institute of Cardiology, Indiana University School of MedicineIdianapolisUnited States
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| |
Collapse
|
14
|
Hindmarch CCT, Tian L, Xiong PY, Potus F, Bentley RET, Al-Qazazi R, Prins KW, Archer SL. An integrated proteomic and transcriptomic signature of the failing right ventricle in monocrotaline induced pulmonary arterial hypertension in male rats. Front Physiol 2022; 13:966454. [PMID: 36388115 PMCID: PMC9664166 DOI: 10.3389/fphys.2022.966454] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/19/2022] [Indexed: 01/25/2023] Open
Abstract
Aim: Pulmonary arterial hypertension (PAH) is an obstructive pulmonary vasculopathy that results in death from right ventricular failure (RVF). There is limited understanding of the molecular mechanisms of RVF in PAH. Methods: In a PAH-RVF model induced by injection of adult male rats with monocrotaline (MCT; 60 mg/kg), we performed mass spectrometry to identify proteins that change in the RV as a consequence of PAH induced RVF. Bioinformatic analysis was used to integrate our previously published RNA sequencing data from an independent cohort of PAH rats. Results: We identified 1,277 differentially regulated proteins in the RV of MCT rats compared to controls. Integration of MCT RV transcriptome and proteome data sets identified 410 targets that are concordantly regulated at the mRNA and protein levels. Functional analysis of these data revealed enriched functions, including mitochondrial metabolism, cellular respiration, and purine metabolism. We also prioritized 15 highly enriched protein:transcript pairs and confirmed their biological plausibility as contributors to RVF. We demonstrated an overlap of these differentially expressed pairs with data published by independent investigators using multiple PAH models, including the male SU5416-hypoxia model and several male rat strains. Conclusion: Multiomic integration provides a novel view of the molecular phenotype of RVF in PAH which includes dysregulation of pathways involving purine metabolism, mitochondrial function, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Charles Colin Thomas Hindmarch
- QCPU, Queen’s Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Lian Tian
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Ping Yu Xiong
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Francois Potus
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et Pneumologie de Quebec, Quebec City, QC, Canada
| | | | - Ruaa Al-Qazazi
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Kurt W. Prins
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Stephen L. Archer
- QCPU, Queen’s Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada,Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Stephen L. Archer,
| |
Collapse
|
15
|
Grün B, Tirre M, Pyschny S, Singh V, Kehl HG, Jux C, Drenckhahn JD. Inhibition of mitochondrial respiration has fundamentally different effects on proliferation, cell survival and stress response in immature versus differentiated cardiomyocyte cell lines. Front Cell Dev Biol 2022; 10:1011639. [PMID: 36211452 PMCID: PMC9538794 DOI: 10.3389/fcell.2022.1011639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Myocardial tissue homeostasis is critically important for heart development, growth and function throughout the life course. The loss of cardiomyocytes under pathological conditions ultimately leads to cardiovascular disease due to the limited regenerative capacity of the postnatal mammalian heart. Inhibition of electron transport along the mitochondrial respiratory chain causes cellular stress characterized by ATP depletion as well as excessive generation of reactive oxygen species. Adult cardiomyocytes are highly susceptible to mitochondrial dysfunction whereas embryonic cardiomyocytes in the mouse heart have been shown to be resistant towards mitochondrial complex III inhibition. To functionally characterize the molecular mechanisms mediating this stress tolerance, we used H9c2 cells as an in vitro model for immature cardiomyoblasts and treated them with various inhibitors of mitochondrial respiration. The complex I inhibitor rotenone rapidly induced cell cycle arrest and apoptosis whereas the complex III inhibitor antimycin A (AMA) had no effect on proliferation and only mildly increased cell death. HL-1 cells, a differentiated and contractile cardiomyocyte cell line from mouse atrium, were highly susceptible to AMA treatment evident by cell cycle arrest and death. AMA induced various stress response mechanisms in H9c2 cells, such as the mitochondrial unfolded protein response (UPRmt), integrated stress response (ISR), heat shock response (HSR) and antioxidative defense. Inhibition of the UPR, ISR and HSR by siRNA mediated knock down of key components does not impair growth of H9c2 cells upon AMA treatment. In contrast, knock down of NRF2, an important transcriptional regulator of genes involved in detoxification of reactive oxygen species, reduces growth of H9c2 cells upon AMA treatment. Various approaches to activate cell protective mechanisms and alleviate oxidative stress in HL-1 cells failed to rescue them from AMA induced growth arrest and death. In summary, these data show that the site of electron transport interruption along the mitochondrial respiratory chain determines cell fate in immature cardiomyoblasts. The study furthermore points to fundamental differences in stress tolerance and cell survival between immature and differentiated cardiomyocytes which may underlie the growth plasticity of embryonic cardiomyocytes during heart development but also highlight the obstacles of cardioprotective therapies in the adult heart.
Collapse
Affiliation(s)
- Bent Grün
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Michaela Tirre
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Simon Pyschny
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Vijay Singh
- Department of Pediatric Hematology and Oncology, Justus Liebig University, Gießen, Germany
| | - Hans-Gerd Kehl
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Christian Jux
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
- Department of Pediatric Cardiology, Justus Liebig University, Gießen, Germany
| | - Jörg-Detlef Drenckhahn
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
- Department of Pediatric Cardiology, Justus Liebig University, Gießen, Germany
- *Correspondence: Jörg-Detlef Drenckhahn,
| |
Collapse
|
16
|
Wang Z, He Z, Xuan Q, Zhang Y, Xu J, Lin J, Li H, Chen W, Jiang T. Analysis of the potential ferroptosis mechanism and multitemporal expression change of central ferroptosis-related genes in cardiac ischemia–reperfusion injury. Front Physiol 2022; 13:934901. [PMID: 36091399 PMCID: PMC9461145 DOI: 10.3389/fphys.2022.934901] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/29/2022] [Indexed: 12/15/2022] Open
Abstract
Acute myocardial infraction is the most severe type of coronary artery disease and remains a substantial burden to the health care system globally. Although myocardial reperfusion is critical for ischemic cardiac tissue survival, the reperfusion itself could cause paradoxical injury. This paradoxical phenomenon is known as ischemia–reperfusion injury (IRI), and the exact molecular mechanism of IRI is still far from being elucidated and is a topic of controversy. Meanwhile, ferroptosis is a nonapoptotic form of cell death that has been reported to be associated with various cardiovascular diseases. Thus, we explored the potential ferroptosis mechanism and target in cardiac IRI via bioinformatics analysis and experiment. GSE4105 data were obtained from the GEO database and consist of a rat IRI model and control. After identifying differentially expressed ferroptosis-related genes (DEFRGs) and hub genes of cardiac IRI, we performed enrichment analysis, coexpression analysis, drug–gene interaction prediction, and mRNA–miRNA regulatory network construction. Moreover, we validated and explored the multitemporal expression of hub genes in a hypoxia/reoxygenation (H/R)-induced H9C2 cell injury model under different conditions via RT-qPCR. A total of 43 DEFRGs and 7 hub genes (tumor protein p53 [Tp53], tumor necrosis factor [Tnf], hypoxia-inducible factor 1 subunit alpha [Hif1a], interleukin 6 [Il6], heme oxygenase 1 [Hmox1], X-box binding protein 1 [Xbp1], and caspase 8 [Casp8]) were screened based on bioinformatics analysis. The functional annotation of these genes revealed apoptosis, and the related signaling pathways could have association with the pathogenesis of ferroptosis in cardiac IRI. In addition, the expression of the seven hub genes in IRI models were found higher than that of control under different H/R conditions and time points. In conclusion, the analysis of 43 DEFRGs and 7 hub genes could reveal the potential biological pathway and mechanism of ferroptosis in cardiac IRI. In addition, the multitemporal expression change of hub genes in H9C2 cells under different H/R conditions could provide clues for further ferroptosis mechanism exploring, and the seven hub genes could be potential biomarkers or therapeutic targets in cardiac IRI.
Collapse
Affiliation(s)
- Zuoxiang Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Medicine, Soochow University, Suzhou, Jiangsu, China
| | - Zhisong He
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qinkao Xuan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yue Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jialiang Xu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jia Lin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- *Correspondence: Weixiang Chen, ; Tingbo Jiang,
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- *Correspondence: Weixiang Chen, ; Tingbo Jiang,
| |
Collapse
|
17
|
Miyamoto HD, Ikeda M, Ide T, Tadokoro T, Furusawa S, Abe K, Ishimaru K, Enzan N, Sada M, Yamamoto T, Matsushima S, Koumura T, Yamada KI, Imai H, Tsutsui H. Iron Overload via Heme Degradation in the Endoplasmic Reticulum Triggers Ferroptosis in Myocardial Ischemia-Reperfusion Injury. JACC Basic Transl Sci 2022; 7:800-819. [PMID: 36061338 PMCID: PMC9436815 DOI: 10.1016/j.jacbts.2022.03.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/16/2022] [Accepted: 03/16/2022] [Indexed: 10/25/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a promising therapeutic target to improve clinical outcomes after acute myocardial infarction. Ferroptosis, triggered by iron overload and excessive lipid peroxides, is reportedly involved in I/R injury. However, its significance and mechanistic basis remain unclear. Here, we show that glutathione peroxidase 4 (GPx4), a key endogenous suppressor of ferroptosis, determines the susceptibility to myocardial I/R injury. Importantly, ferroptosis is a major mode of cell death in I/R injury, distinct from mitochondrial permeability transition (MPT)-driven necrosis. This suggests that the use of therapeutics targeting both modes is an effective strategy to further reduce the infarct size and thereby ameliorate cardiac remodeling after I/R injury. Furthermore, we demonstrate that heme oxygenase 1 up-regulation in response to hypoxia and hypoxia/reoxygenation degrades heme and thereby induces iron overload and ferroptosis in the endoplasmic reticulum (ER) of cardiomyocytes. Collectively, ferroptosis triggered by GPx4 reduction and iron overload in the ER is distinct from MPT-driven necrosis in both in vivo phenotype and in vitro mechanism for I/R injury. The use of therapeutics targeting ferroptosis in conjunction with cyclosporine A can be a promising strategy for I/R injury.
Collapse
Key Words
- AMI, acute myocardial infarction
- CsA, cyclosporine A
- CypD, cyclophilin D
- DXZ, dexrazoxane
- ER, endoplasmic reticulum
- Fer-1, ferrostatin-1
- GPx4, glutathione peroxidase 4
- H/R, hypoxia-reoxygenation
- HF, heart failure
- HO-1, heme oxygenase 1
- I/R, ischemia-reperfusion
- LP, lipid peroxide
- MPT, mitochondrial permeability transition
- MPT-driven necrosis
- RCD, regulated cell death
- STEMI, ST-segment elevation myocardial infarction
- cyclosporine A
- ferroptosis
- glutathione peroxidase 4
- ischemia-reperfusion injury
Collapse
Affiliation(s)
- Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shun Furusawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ko Abe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosei Ishimaru
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuyuki Enzan
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Sada
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taishi Yamamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoko Koumura
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Imai
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
18
|
Machado SE, Spangler D, Stacks DA, Darley-Usmar V, Benavides GA, Xie M, Balla J, Zarjou A. Counteraction of Myocardial Ferritin Heavy Chain Deficiency by Heme Oxygenase-1. Int J Mol Sci 2022; 23:8300. [PMID: 35955444 PMCID: PMC9368247 DOI: 10.3390/ijms23158300] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
Given the abundance of heme proteins (cytochromes) in the mitochondrion, it is evident that a meticulously orchestrated iron metabolism is essential for cardiac health. Here, we examined the functional significance of myocardial ferritin heavy chain (FtH) in a model of acute myocardial infarction. We report that FtH deletion did not alter either the mitochondrial regulatory and surveillance pathways (fission and fusion) or mitochondrial bioenergetics in response to injury. Furthermore, deletion of myocardial FtH did not affect cardiac function, assessed by measurement of left ventricular ejection fraction, on days 1, 7, and 21 post injury. To identify the modulated pathways providing cardiomyocyte protection coincident with FtH deletion, we performed unbiased transcriptomic analysis. We found that following injury, FtH deletion was associated with upregulation of several genes with anti-ferroptotic properties, including heme oxygenase-1 (HO-1) and the cystine/glutamate anti-porter (Slc7a11). These results suggested that HO-1 overexpression mitigates ferroptosis via upregulation of Slc7a11. Indeed, using transgenic mice with HO-1 overexpression, we demonstrate that overexpressed HO-1 is coupled with increased Slc7a11 expression. In conclusion, we demonstrate that following injury, myocardial FtH deletion leads to a compensatory upregulation in a number of anti-ferroptotic genes, including HO-1. Such HO-1 induction leads to overexpression of Slc7a11 and protects the heart against ischemia-reperfusion-mediated ferroptosis, preserves mitochondrial function, and overall function of the myocardium.
Collapse
Affiliation(s)
- Sarah E. Machado
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Daryll Spangler
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Delores A. Stacks
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Victor Darley-Usmar
- Mitochondrial Medicine Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.D.-U.); (G.A.B.)
| | - Gloria A. Benavides
- Mitochondrial Medicine Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.D.-U.); (G.A.B.)
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - József Balla
- ELKH-UD Vascular Pathophysiology Research Group 11003, Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Abolfazl Zarjou
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| |
Collapse
|
19
|
Yang CY, Tinhofer IE, Cheng MH. Long-term outcomes of arterial ischemia or venous occlusion on vascularized groin lymph nodes in a rat model. J Surg Oncol 2022; 126:633-639. [PMID: 35678754 DOI: 10.1002/jso.26980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 05/28/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND This study investigated the long-term effects of arterial ischemia and venous occlusion on lymph node drainage function in a rat model. METHODS Bilateral groin lymph node flaps of 18 Lewis rats were dissected. The pedicle artery was clamped for 4, 5, and 6 h (A4, A5, and A6 groups), and the vein for 3, 4, and 5 h (V3, V4, and V5 groups) in six flaps. At 4 weeks, the evaluations included gross morphomics, indocyanine green (ICG) lymphography, histological section, immunofluorescence of terminal deoxynucleotidyl transferase assay, and heme oxygenase-1 (HO-1) stain. RESULTS The lymph node flaps developed shrinkage and partial necrosis in A5, A6, V4, and V5 groups. Hemorrhage in the lymph node cortex and medulla was observed histologically in A5, A6, and V5 groups. ICG lymphography showed loss of lymphatic drainage function in 2 of 6 flaps in A6 and V5 groups. Cell death was shown partly in cortical follicles in A5 and V4 groups and completely in A6 and V5 groups. The HO-1 expression was statistically increased in A5 and V5 groups, respectively (p < 0.05). CONCLUSIONS The critical arterial ischemia and venous occlusion time were 4 h at 4 weeks of follow-up.
Collapse
Affiliation(s)
- Chin-Yu Yang
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ines E Tinhofer
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Hospital of Divine Savior, Vienna, Austria
| | - Ming-Huei Cheng
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung University and Medical College, Taoyuan, Taiwan
- Center for Lymphedema Microsurgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Division of Plastic Surgery, Ann The University of Michigan, Arbor, Michigan, USA
| |
Collapse
|
20
|
Heme Oxygenase-1: An Anti-Inflammatory Effector in Cardiovascular, Lung, and Related Metabolic Disorders. Antioxidants (Basel) 2022; 11:antiox11030555. [PMID: 35326205 PMCID: PMC8944973 DOI: 10.3390/antiox11030555] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/24/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
The heme oxygenase (HO) enzyme system catabolizes heme to carbon monoxide (CO), ferrous iron, and biliverdin-IXα (BV), which is reduced to bilirubin-IXα (BR) by biliverdin reductase (BVR). HO activity is represented by two distinct isozymes, the inducible form, HO-1, and a constitutive form, HO-2, encoded by distinct genes (HMOX1, HMOX2, respectively). HO-1 responds to transcriptional activation in response to a wide variety of chemical and physical stimuli, including its natural substrate heme, oxidants, and phytochemical antioxidants. The expression of HO-1 is regulated by NF-E2-related factor-2 and counter-regulated by Bach-1, in a heme-sensitive manner. Additionally, HMOX1 promoter polymorphisms have been associated with human disease. The induction of HO-1 can confer protection in inflammatory conditions through removal of heme, a pro-oxidant and potential catalyst of lipid peroxidation, whereas iron released from HO activity may trigger ferritin synthesis or ferroptosis. The production of heme-derived reaction products (i.e., BV, BR) may contribute to HO-dependent cytoprotection via antioxidant and immunomodulatory effects. Additionally, BVR and BR have newly recognized roles in lipid regulation. CO may alter mitochondrial function leading to modulation of downstream signaling pathways that culminate in anti-apoptotic, anti-inflammatory, anti-proliferative and immunomodulatory effects. This review will present evidence for beneficial effects of HO-1 and its reaction products in human diseases, including cardiovascular disease (CVD), metabolic conditions, including diabetes and obesity, as well as acute and chronic diseases of the liver, kidney, or lung. Strategies targeting the HO-1 pathway, including genetic or chemical modulation of HO-1 expression, or application of BR, CO gas, or CO donor compounds show therapeutic potential in inflammatory conditions, including organ ischemia/reperfusion injury. Evidence from human studies indicate that HO-1 expression may represent a biomarker of oxidative stress in various clinical conditions, while increases in serum BR levels have been correlated inversely to risk of CVD and metabolic disease. Ongoing human clinical trials investigate the potential of CO as a therapeutic in human disease.
Collapse
|
21
|
Lu Y, An L, Taylor MRG, Chen QM. Nrf2 signaling in heart failure: expression of Nrf2, Keap1, antioxidant, and detoxification genes in dilated or ischemic cardiomyopathy. Physiol Genomics 2022; 54:115-127. [PMID: 35073209 PMCID: PMC8897001 DOI: 10.1152/physiolgenomics.00079.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased levels of oxidative stress have been found with heart failure. Whether failing hearts express antioxidant and detoxification enzymes have not been addressed systematically. Nrf2 gene encodes a transcription factor that regulates the expression of antioxidant and detoxification genes. Using RNA-Seq data set from explanted hearts of 37 patients with dilated cardiomyopathy (DCM), 13 patients with ischemic cardiomyopathy (ICM), and 14 nonfailure (NF) donors as a control, we addressed whether failing hearts change the expression of Nrf2, its negative regulator Keap1, and antioxidant or detoxification genes. Significant increases in the ratio of Nrf2 to Keap1 were found to associate with DCM or ICM. Antioxidant genes showed decreased expression in both types of heart failure, including NQO1, SOD1, GPX3, GPX4, GSR, PRDX1, and TXNRD1. Detoxification enzymes, GCLM and EPHX1, also showed decreased expression, whereas the CYP1B1 transcript was elevated in both DCM and ICM. The genes encoding metal-binding protein ferritin were decreased, whereas five out of 12 metallothionein genes showed elevated expression. Our finding on Nrf2 gene expression has been validated by meta-analysis of seven independent data sets of microarray or RNA-Seq for differential gene expression in DCM and ICM from NF controls. In conclusion, minor elevation of Nrf2 gene expression is not coupled to increases in antioxidant and detoxification genes, supporting an impairment of Nrf2 signaling in patients with heart failure. Decreases in multiple antioxidant and detoxification genes are consistent with the observed increases of oxidative stress in failing hearts.
Collapse
Affiliation(s)
- Yingying Lu
- 1Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, Arizona,2Interdisciplanary Program in Statistics and Data Science, University of Arizona, Tucson, Arizona
| | - Lingling An
- 3Department of Biosystems Engineering, University of Arizona, Tucson, Arizona
| | - Matthew R. G. Taylor
- 4Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Qin M. Chen
- 1Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, Arizona
| |
Collapse
|
22
|
Effects of Heme Oxygenase-1 on c-Kit-Positive Cardiac Cells. Int J Mol Sci 2021; 22:ijms222413448. [PMID: 34948245 PMCID: PMC8704354 DOI: 10.3390/ijms222413448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 01/02/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is one of the most powerful cytoprotective proteins known. The goal of this study was to explore the effects of HO-1 in c-kit-positive cardiac cells (CPCs). LinNEG/c-kitPOS CPCs were isolated and expanded from wild-type (WT), HO-1 transgenic (TG), or HO-1 knockout (KO) mouse hearts. Compared with WT CPCs, cell proliferation was significantly increased in HO-1TG CPCs and decreased in HO-1KO CPCs. HO-1TG CPCs also exhibited a marked increase in new DNA synthesis during the S-phase of cell division, not only under normoxia (21% O2) but after severe hypoxia (1% O2 for 16 h). These properties of HO-1TG CPCs were associated with nuclear translocation (and thus activation) of Nrf2, a key transcription factor that regulates antioxidant genes, and increased protein expression of Ec-SOD, the only extracellular antioxidant enzyme. These data demonstrate that HO-1 upregulates Ec-SOD in CPCs and suggest that this occurs via activation of Nrf2, which thus is potentially involved in the crosstalk between two antioxidants, HO-1 in cytoplasm and Ec-SOD in extracellular matrix. Overexpression of HO-1 in CPCs may improve the survival and reparative ability of CPCs after transplantation and thus may have potential clinical application to increase efficacy of cell therapy.
Collapse
|
23
|
Costa Silva RCM, Correa LHT. Heme Oxygenase 1 in Vertebrates: Friend and Foe. Cell Biochem Biophys 2021; 80:97-113. [PMID: 34800278 DOI: 10.1007/s12013-021-01047-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/07/2021] [Indexed: 10/19/2022]
Abstract
HO-1 is the inducible form of the enzyme heme-oxygenase. HO-1 catalyzes heme breakdown, reducing the levels of this important oxidant molecule and generating antioxidant, anti-inflammatory, and anti-apoptotic byproducts. Thus, HO-1 has been described as an important stress response mechanism during both physiologic and pathological processes. Interestingly, some findings are demonstrating that uncontrolled levels of HO-1 byproducts can be associated with cell death and tissue destruction as well. Furthermore, HO-1 can be located in the nucleus, influencing gene transcription, cellular proliferation, and DNA repair. Here, we will discuss several studies that approach HO-1 effects as a protective or detrimental mechanism in different pathological conditions. In this sense, as the major organs of vertebrates will deal specifically with distinct types of stresses, we discuss the HO-1 role in each of them, exposing the contradictions associated with HO-1 expression after different insults and circumstances.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Leonardo Holanda Travassos Correa
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Kuninaka Y, Ishida Y, Nosaka M, Ishigami A, Taruya A, Shimada E, Kimura A, Yamamoto H, Ozaki M, Furukawa F, Kondo T. Forensic significance of intracardiac heme oxygenase-1 expression in acute myocardial ischemia. Sci Rep 2021; 11:21828. [PMID: 34750390 PMCID: PMC8575909 DOI: 10.1038/s41598-021-01102-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022] Open
Abstract
Heme oxygenase-1 (HO-1), an inducible stress-response protein, exerts anti-oxidant and anti-apoptotic effects. However, its significance in forensic diagnosis of acute ischemic heart diseases (AIHD) such as myocardial infarction (MI) is still unknown. We examined the immunohistochemical expression of HO-1 in the heart samples to discuss their forensic significance to determine acute cardiac ischemia. The heart samples were obtained from 23 AIHD cases and 33 non-AIHD cases as controls. HO-1 positive signals in cardiomyocyte nuclear were detected in 78.2% of AIHD cases, however, that were detected in only 24.2% control cases with statistical difference between AIHD and non-AIHD groups. In contrast to HO-1 protein expression, there was no significant difference in the appearance of myoglobin pallor regions and leukocyte infiltration in the hearts between AIHD and non-AIHD groups. From the viewpoints of forensic pathology, intracardiac HO-1 expression would be considered a valuable marker to diagnose AIHD as the cause of death.
Collapse
Affiliation(s)
- Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Akiko Ishigami
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Akira Taruya
- Department of Cardiovascular Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Emi Shimada
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Hiroki Yamamoto
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Mitsunori Ozaki
- Department of Neurological Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Fukumi Furukawa
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
- Takatsuki Red Cross Hospital, 1-1-1 Abuno, Takatsuki-shi, Osaka, 569-1096, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
25
|
Wächter K, Navarrete Santos A, Großkopf A, Baldensperger T, Glomb MA, Szabó G, Simm A. AGE-Rich Bread Crust Extract Boosts Oxidative Stress Interception via Stimulation of the NRF2 Pathway. Nutrients 2021; 13:nu13113874. [PMID: 34836129 PMCID: PMC8622267 DOI: 10.3390/nu13113874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/30/2022] Open
Abstract
Advanced glycation end products (AGEs) result from a non-enzymatic reaction of proteins with reactive carbohydrates. Heat-processed food, such as bread, contains high amounts of AGEs. The activation of the NF-κB signaling pathway by bread crust extract (BCE) is well understood. However, it is largely unknown whether NRF2, the master regulator of oxidative stress resistance in mammalian cells, is affected by BCE. We have investigated the molecular mechanisms by which BCE induces antioxidant gene expression in cellular models. Our data showed that soluble extracts from bread crust are capable of stimulating the NRF2 signaling pathway. Furthermore, NRF2 pathway activation was confirmed by microarray and reporter-cell analyses. QRT-PCR measurements and Western blot analyses indicated an induction of antioxidative genes such as HMOX1, GCLM and NQO1 upon BCE treatment. Moreover, BCE pretreated cells had a survival advantage compared to control cells when exposed to oxidative stress. BCE induces phosphorylation of AKT and ERK kinase in EA.hy926 cells. By mass spectrometry, several new, potentially active modifications in BCE were identified. Our findings indicate that BCE activates NRF2-dependent antioxidant gene expression, thus provoking a protection mechanism against oxidative stress-mediated tissue injury. Hence, BCE can be considered as functional food with antioxidative and cardioprotective potential.
Collapse
Affiliation(s)
- Kristin Wächter
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.G.); (G.S.); (A.S.)
- Correspondence: ; Tel.: +49-345-557-7068
| | - Alexander Navarrete Santos
- Center for Medical Basic Research, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Anne Großkopf
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.G.); (G.S.); (A.S.)
| | - Tim Baldensperger
- German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany;
- Institute of Chemistry, Food Chemistry, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Marcus A. Glomb
- Institute of Chemistry, Food Chemistry, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Gábor Szabó
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.G.); (G.S.); (A.S.)
| | - Andreas Simm
- Department for Cardiac Surgery, University Hospital Halle (Saale), Martin-Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.G.); (G.S.); (A.S.)
- Center for Medical Basic Research, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| |
Collapse
|
26
|
Hofmann A, Müglich M, Wolk S, Khorzom Y, Sabarstinski P, Kopaliani I, Egorov D, Horn F, Brunssen C, Giebe S, Hamann B, Deussen A, Morawietz H, Poitz DM, Reeps C. Induction of Heme Oxygenase-1 Is Linked to the Severity of Disease in Human Abdominal Aortic Aneurysm. J Am Heart Assoc 2021; 10:e022747. [PMID: 34622673 PMCID: PMC8751892 DOI: 10.1161/jaha.121.022747] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Rupture of abdominal aortic aneurysm (rAAA) is associated with high case fatality rates, and risk of rupture increases with the AAA diameter. Heme oxygenase‐1 (gene HMOX1, protein HO‐1) is a stress‐induced protein and induction has protective effects in the vessel wall. HMOX1−/− mice are more susceptible to angiotensin II‐induced AAA formation, but the regulation in human nonruptured and ruptured AAA is only poorly understood. Our hypothesis proposed that HO‐1 is reduced in AAA and lowering is inversely associated with the AAA diameter. Methods and Results AAA walls from patients undergoing elective open repair (eAAA) or surgery because of rupture (rAAA) were analyzed for aortic HMOX1/HO‐1 expression by quantitative real‐time polymerase chain reaction and Western blot. Aortas from patients with aortic occlusive disease served as controls. HMOX1/HO‐1 expression was 1.1‐ to 7.6‐fold upregulated in eAAA and rAAA. HO‐1 expression was 3‐fold higher in eAAA specimen with a diameter >84.4 mm, whereas HO‐1 was not different in rAAA. Other variables that are known for associations with AAA and HO‐1 induction were tested. In eAAA, HO‐1 expression was negatively correlated with aortic collagen content and oxidative stress parameters H2O2 release, oxidized proteins, and thiobarbituric acid reactive substances. Serum HO‐1 concentrations were analyzed in patients with eAAA, and maximum values were found in an aortic diameter of 55 to 70 mm with no further increase >70 mm, compared with <55 mm. Conclusions Aortic HO‐1 expression was increased in eAAA and rAAA. HO‐1 increased with the severity of disease but was additionally connected to less oxidative stress and vasoprotective mechanisms.
Collapse
Affiliation(s)
- Anja Hofmann
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Margarete Müglich
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Steffen Wolk
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Yazan Khorzom
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Pamela Sabarstinski
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Irakli Kopaliani
- Department of Physiology Medical Faculty Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Dmitry Egorov
- Department of Physiology Medical Faculty Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Franziska Horn
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation Department of Medicine III University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Sindy Giebe
- Division of Vascular Endothelium and Microcirculation Department of Medicine III University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Bianca Hamann
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Andreas Deussen
- Department of Physiology Medical Faculty Carl Gustav Carus Dresden Technische Universität Dresden Dresden Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation Department of Medicine III University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery Department of Visceral, Thoracic and Vascular Surgery University Hospital and Medical Faculty Carl Gustav Carus Technische Universität Dresden Dresden Germany
| |
Collapse
|
27
|
Liang KW, Lee WJ, Lee WL, Wu JP, Sheu WHH. Shorter (GT) n repeats in the haem-oxygenase 1 gene promoter are associated with better mid-term survival in subjects with coronary artery disease and abnormal ejection fraction. Biomarkers 2021; 26:732-736. [PMID: 34634993 DOI: 10.1080/1354750x.2021.1992650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Haem oxygenase (HO)-1 is a rate-limiting enzyme for degrading haem into carbon monoxide. Subjects with longer GT repeats in the HO-1 gene (HMOX1) promoter are more likely to have coronary artery disease (CAD) and cardiovascular events. METHODS We retrospectively enrolled CAD subjects with an abnormal ejection fraction (EF) <50% from our catheterisation data (N = 670). Polymerase chain reactions were performed for amplifying the HMOX1 promoter GT repeating segment to determine the number of repeats. RESULTS In a median follow-up period of 40 months, 213 patients died. The distribution of genotype for HMOX1 promoter GT repeating segments SS, SL, and LL were significantly different (p < 0.001) between the dead (44.6%, 36.2%, 19.2%, respectively) and the survived (53.8%, 37.4%, 8.8%, respectively) (S allele: ≤30 repeats, L allele: >30 repeats). In Cox regression analysis, carrier of S allele (hazard ratio 0.665, p = 0.027), a higher EF (hazard ratio 0.037, p = 0.001), and revascularization with PCI were all negatively associated with all-cause death in subjects with CAD and abnormal EF. CONCLUSIONS Carrier of shorter (GT)n repeats of HMOX1 gene promoter was negatively correlated with death events in CAD patients with abnormal EF.
Collapse
Affiliation(s)
- Kae-Woei Liang
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,College of Life Science, National Chung Hsing University, Taichung, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Social Work, Tunghai University, Taichung, Taiwan
| | - Wen-Lieng Lee
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jen-Pey Wu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wayne H-H Sheu
- Department of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,College of Life Science, National Chung Hsing University, Taichung, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
28
|
McClung JA, Levy L, Garcia V, Stec DE, Peterson SJ, Abraham NG. Heme-oxygenase and lipid mediators in obesity and associated cardiometabolic diseases: Therapeutic implications. Pharmacol Ther 2021; 231:107975. [PMID: 34499923 DOI: 10.1016/j.pharmthera.2021.107975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity-mediated metabolic syndrome remains the leading cause of death worldwide. Among many potential targets for pharmacological intervention, a promising strategy involves the heme oxygenase (HO) system, specifically its inducible form, HO-1. This review collects and updates much of the current knowledge relevant to pharmacology and clinical medicine concerning HO-1 in metabolic diseases and its effect on lipid metabolism. HO-1 has pleotropic effects that collectively reduce inflammation, while increasing vasodilation and insulin and leptin sensitivity. Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Similarly, HO-1and bilirubin are potential therapeutic targets in the treatment of fat-induced liver diseases. HO-1-mediated upregulation of EET is capable not only of reversing endothelial dysfunction and hypertension, but also of reversing cardiac remodeling, a hallmark of the metabolic syndrome. This process involves browning of white fat tissue (i.e. formation of healthy adipocytes) and reduced lipotoxicity, which otherwise will be toxic to the heart. More importantly, this review examines the activity of EET in biological systems and a series of pathways that explain its mechanism of action and discusses how these might be exploited for potential therapeutic use. We also discuss the link between cardiac ectopic fat deposition and cardiac function in humans, which is similar to that described in obese mice and is regulated by HO-1-EET-PGC1α signaling, a potent negative regulator of the inflammatory adipokine NOV.
Collapse
Affiliation(s)
- John A McClung
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Lior Levy
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, United States of America.
| | - Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, United States of America; New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, United States of America
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America; Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America.
| |
Collapse
|
29
|
Oliveira BC, Santos PP, Figueiredo AM, Rafacho BPM, Ishikawa L, Zanati SG, Fernandes AAH, Azevedo PS, Polegato BF, Zornoff LAM, Minicucci MF, Paiva SAR. Influence of Consumption of Orange Juice (Citrus Sinensis) on Cardiac Remodeling of Rats Submitted to Myocardial Infarction. Arq Bras Cardiol 2021; 116:1127-1136. [PMID: 34133599 PMCID: PMC8288524 DOI: 10.36660/abc.20190397] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Fundamento O suco de laranja (SL) é rico em polifenóis com propriedades anti-inflamatórias e antioxidantes. Após o infarto do miocárdio (IM), mudanças complexas ocorrem na estrutura e na função cardíacas, processo conhecido como remodelação cardíaca (RC). O estresse oxidativo e a inflamação podem modular esse processo. Nossa hipótese foi a de que o consumo de SL atenua a RC após o IM. Objetivos Avaliar a influência do SL sobre a RC após IM pela análise de variáveis funcionais, morfológicas, de estresse oxidativo, de inflação, e de metabolismo energético. Métodos Um total de 242 ratos machos pesando entre 200 e 250g foram submetidos a um procedimento cirúrgico (ligação da artéria coronária ou cirurgia simulada). Sete dia após a cirurgia, os animais sobreviventes foram divididos para um dos quatro grupos: 1) SM, animais sham que receberam água e maltodextrina (n= 20); 2) SSL, animais sham que receberam SL (n= 20); 3) IM, animais infartados que receberam água e maltodextrina (n= 40); e 4) ISL, animais infartados que receberam SL (n = 40). A análise estatística foi realizada pelo teste de ANOVA com dois fatores com o teste de Holm-Sidak. Os resultados foram apresentados em média ± desvio padrão, e o nível de significância adotado foi de 5%. Resultados Três meses depois, o IM levou à hipertrofia do ventrículo esquerdo (VE), com disfunção sistólica e diastólica, e aumento nos mediadores inflamatórios e de estresse oxidativo. Os animais que consumiram SL apresentaram menor atividade da glutationa peroxidase e maior expressão da heme-oxigenase-1 (HO-1). Conclusão O SL atenuou a RC, e a HO-1 pode exercer um importante papel nesse processo.
Collapse
Affiliation(s)
- Bruna C Oliveira
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Priscila P Santos
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Amanda M Figueiredo
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Bruna P M Rafacho
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Larissa Ishikawa
- Instituto de Biociências Campus de Botucatu (UNESP), Botucatu , SP - Brasil
| | - Silméia G Zanati
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Ana A H Fernandes
- Instituto de Biociências Campus de Botucatu (UNESP), Botucatu , SP - Brasil
| | - Paula S Azevedo
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Bertha F Polegato
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Leonardo A M Zornoff
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Marcos F Minicucci
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil
| | - Sergio A R Paiva
- Universidade Estadual Paulista Júlio de Mesquita Filho Campus de Botucatu - Faculdade de Medicina de Botucatu , Botucatu , SP - Brasil.,Food Research Center FoRC , São Paulo , SP - Brasil
| |
Collapse
|
30
|
Li S, Lei Z, Zhao M, Hou Y, Wang D, Xu X, Lin X, Li J, Tang S, Yu J, Meng T. Propofol Inhibits Ischemia/Reperfusion-Induced Cardiotoxicity Through the Protein Kinase C/Nuclear Factor Erythroid 2-Related Factor Pathway. Front Pharmacol 2021; 12:655726. [PMID: 34054535 PMCID: PMC8155638 DOI: 10.3389/fphar.2021.655726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022] Open
Abstract
Both hydrogen peroxide (H2O2, H) and ischemia/reperfusion (I/R) can damage cardiomyocytes, which was inhibited by propofol (P). The present research was designed to examine whether propofol can reduce myocardial I/R injury by activating protein kinase C (PKC)/nuclear factor erythroid-2-related factor 2 (NRF2) pathway in H9C2 cells and rat Langendorff models. H9C2 cells were disposed of no reagents (C), H2O2 for 24 h (H), propofol for 1 h before H2O2 (H+P), and chelerythrine (CHE, PKC inhibitor) for 1 h before propofol and H2O2 (H+P+CHE). N = 3. The PKC gene of H9C2 was knocked down by siRNA and overexpressed by phorbol 12-myristate 13-acetate (PMA, PKC agonist). The cell viability and the expressions of PKC, NRF2, or heme oxygenase-1(HO-1) were evaluated. Propofol significantly reduced H9C2 cell mortality induced by H2O2, and significantly increased NRF2 nuclear location and HO-1 expression, which were restrained by siRNA knockout of PKC and promoted by PMA. Rat hearts were treated with KrebsHenseleit solution for 120 min (C), with (I/R+P) or without (I/R) propofol for 20 min before stopping perfusion for 30 min and reperfusion for 60 min, and CHE for 10 min before treated with propofol. N = 6. The levels of lactate dehydrogenase (LDH), superoxide dismutase (SOD), and creatine kinase-MB (CK-MB) in perfusion fluid and antioxidant enzymes in the myocardium were assessed. I/R, which increased LDH and CK-MB expression and reduced SOD expression, boosted the pathological damage and infarcts of the myocardium after reperfusion. However, propofol restrained all these effects, an activity that was antagonized by CHE. The results suggest that propofol pretreatment protects against I/R injury by activating of PKC/NRF2 pathway.
Collapse
Affiliation(s)
- Shengqiang Li
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhen Lei
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Zhao
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yonghao Hou
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Di Wang
- Department of Anesthesiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xingli Xu
- Department of Cardiovascular Ultrasound and Non-invasive Cardiology, Sichuan People's Hospital, Chengdu, China
| | - Xiaowen Lin
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingxin Li
- Department of Physiology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuhai Tang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingui Yu
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Meng
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
31
|
Zhai Z, Zou P, Liu F, Xia Z, Li J. Ferroptosis Is a Potential Novel Diagnostic and Therapeutic Target for Patients With Cardiomyopathy. Front Cell Dev Biol 2021; 9:649045. [PMID: 33869204 PMCID: PMC8047193 DOI: 10.3389/fcell.2021.649045] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiomyocyte death is a fundamental progress in cardiomyopathy. However, the mechanism of triggering the death of myocardial cells remains unclear. Ferroptosis, which is the nonapoptotic, iron-dependent, and peroxidation-driven programmed cell death pathway, that is abundant and readily accessible, was not discovered until recently with a pharmacological approach. New researches have demonstrated the close relationship between ferroptosis and the development of many cardiovascular diseases, and several ferroptosis inhibitors, iron chelators, and small antioxidant molecules can relieve myocardial injury by blocking the ferroptosis pathways. Notably, ferroptosis is gradually being considered as an important cell death mechanism in the animal models with multiple cardiomyopathies. In this review, we will discuss the mechanism of ferroptosis and the important role of ferroptosis in cardiomyopathy with a special emphasis on the value of ferroptosis as a potential novel diagnostic and therapeutic target for patients suffering from cardiomyopathy in the future.
Collapse
Affiliation(s)
- Zhenyu Zhai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Pengtao Zou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fuxiang Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zirong Xia
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
32
|
Khalfin B, Lichtenstein A, Albeck A, Nathan I. Targeting Necrosis: Elastase-like Protease Inhibitors Curtail Necrotic Cell Death Both In Vitro and in Three In Vivo Disease Models. J Med Chem 2021; 64:1510-1523. [PMID: 33522230 DOI: 10.1021/acs.jmedchem.0c01683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Necrosis is the main mode of cell death, which leads to multiple clinical conditions affecting hundreds of millions of people worldwide. Its molecular mechanisms are poorly understood, hampering therapeutics development. Here, we identify key proteolytic activities essential for necrosis using various biochemical approaches, enzymatic assays, medicinal chemistry, and siRNA library screening. These findings provide strategies to treat and prevent necrosis, including known medicines used for other indications, siRNAs, and establish a platform for the design of new inhibitory molecules. Indeed, inhibitors of these pathways demonstrated protective activity in vitro and in vivo in animal models of traumatic brain injury, acute myocardial infarction, and drug-induced liver toxicity. Consequently, this study may pave the way for the development of novel therapies for the treatment, inhibition, or prevention of a large number of hitherto untreatable diseases.
Collapse
Affiliation(s)
- Boris Khalfin
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Alexandra Lichtenstein
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Amnon Albeck
- The Julius Spokojny Bioorganic Chemistry Laboratory, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Ilana Nathan
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
- Soroka University Medical Center, Beer Sheva 8457108, Israel
| |
Collapse
|
33
|
Zhao X, Wang Y, Liu C, Zhou P, Sheng Z, Li J, Zhou J, Chen R, Chen Y, Zhao H, Yan H. Prognostic Value of Total Bilirubin in Patients With ST-Segment Elevation Acute Myocardial Infarction Undergoing Primary Coronary Intervention. Front Cardiovasc Med 2021; 7:615254. [PMID: 33392275 PMCID: PMC7773653 DOI: 10.3389/fcvm.2020.615254] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Bilirubin, a natural product of heme catabolism, has antioxidant and anti-inflammatory activities and is inversely associated with stable coronary artery disease. However, the relationship between the bilirubin levels and long-term outcomes in patients with ST-segment elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PPCI) remains unknown. This study aimed to establish a score model based on bilirubin for predicting major adverse cardiovascular events (MACEs) and stratify patients to the level of care. Methods and Results: Data of 4,151 consecutive patients with STEMI who underwent PPCI were evaluated, and 3,708 cases were analyzed. The total bilirubin (TBil) levels were measured during admission, and the study population was divided into two groups. The high TBil group (n = 143) comprised patients who had a TBil level of ≥22 μmmol/L, and the low TBil group (n = 3,565) comprised patients who had a TBil level of <22 μmmol/L. The median follow-up period was 754 days (2.066 years). The MACE was significantly lower in the high TBil group than in the low TBil group (3.5% vs. 11.0%, p = 0.001). In the multivariate Cox regression analysis, a significant association was noted between the TBil levels and adjusted risk of MACE (hazard ratio, 0.279; 95% confidence interval, 0.088-0.877; p = 0.029). A prediction score model composed of TBil, age, hypertension history, and other eight variables was developed, with scores ranging from 0 to 500. The scores categorized patients into low-, medium-, and high-risk categories. The cumulative survival rate was significantly higher in the low-risk group than in the medium- and high-risk groups for MACE, all-cause death, cardiac death, recurrent myocardial infarction, and ischemic stroke (p < 0.001, p < 0.001, p < 0.001, p = 0.030, and p = 0.001, respectively). The area under the curve of the TBil score was 0.768; this was significantly greater in the pairwise comparison with the Global Registry of Acute Coronary Events score (p = 0.0012). Conclusion: The new prediction score model based on TBil could be used in clinical practice to support risk stratification as recommended in the clinical guidelines.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Ying Wang
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Chen Liu
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Peng Zhou
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Zhaoxue Sheng
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Jiannan Li
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Jinying Zhou
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Runzhen Chen
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Yi Chen
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Hanjun Zhao
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Hongbing Yan
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China.,Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
| |
Collapse
|
34
|
Liang KW, Lee WJ, Lee WL, Wu JP, Lee IT, Wang JS, Sheu WHH. Subjects with coronary artery disease and reduced ejection fraction have longer (GT) n repeats in the heme-oxygenase 1 gene promoter. Heart Vessels 2021; 36:615-620. [PMID: 33388910 DOI: 10.1007/s00380-020-01733-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 11/13/2020] [Indexed: 11/29/2022]
Abstract
Heme oxygenase (HO)-1 is a rate-limiting enzyme for degrading heme into carbon monoxide. Longer (GT)n repeat of the HO-1 gene (HMOX1) promoter has a lower transcription rate. Subjects with longer GT repeats in the HMOX1 promoter are more likely to have coronary artery disease (CAD) and cardiovascular events. We retrospectively enrolled CAD subjects with an abnormal ejection fraction (EF) < 50% from our catheterization data (N = 670). Polymerase chain reactions were performed for amplifying the HMOX1 promoter GT repeating segment to determine the number of repeats. Two subgroups, reduced EF < 40% (N = 256), and mid-range EF 40-49% (N = 414), were compared. The distribution of genotypes of SS, SL and LL were significantly different in reduced EF (29%, 48%, 23%) vs. mid-range EF CAD (64%, 30%, 5%) (S allele: ≤ 30 repeats, L allele: > 30 repeats) (p < 0.001). The patients with reduced EF had a significantly longer average (GT)n (median 27.5 vs. 26.5, p = 0.004) than those with the mid-range EF. In multivariate analysis, the carrier of L allele (odds ratio 4.437, p < 0.001) was a significant predictor for the diagnosis of reduced vs. mid-range EF CAD. In conclusion, CAD patients with reduced EF had longer HMOX1 promoter (GT)n repeats than those with mid-range EF.
Collapse
Affiliation(s)
- Kae-Woei Liang
- Cardiovascular Center, Taichung Veterans General Hospital, 1650 Taiwan Boulevard, Sec. 4, Taichung, 40705, Taiwan. .,Department of Medicine, School of Medicine, National Yang Ming University, Taipei, Taiwan.
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Social Work, Tunghai University, Taichung, Taiwan
| | - Wen-Lieng Lee
- Cardiovascular Center, Taichung Veterans General Hospital, 1650 Taiwan Boulevard, Sec. 4, Taichung, 40705, Taiwan.,Department of Medicine, School of Medicine, National Yang Ming University, Taipei, Taiwan
| | - Jen-Pey Wu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Te Lee
- Department of Medicine, School of Medicine, National Yang Ming University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jun-Sing Wang
- Department of Medicine, School of Medicine, National Yang Ming University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wayne H-H Sheu
- Department of Medicine, School of Medicine, National Yang Ming University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,School of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
35
|
Fernández-Fierro A, Funes SC, Rios M, Covián C, González J, Kalergis AM. Immune Modulation by Inhibitors of the HO System. Int J Mol Sci 2020; 22:ijms22010294. [PMID: 33396647 PMCID: PMC7794909 DOI: 10.3390/ijms22010294] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
The heme oxygenase (HO) system involves three isoforms of this enzyme, HO-1, HO-2, and HO-3. The three of them display the same catalytic activity, oxidating the heme group to produce biliverdin, ferrous iron, and carbon monoxide (CO). HO-1 is the isoform most widely studied in proinflammatory diseases because treatments that overexpress this enzyme promote the generation of anti-inflammatory products. However, neonatal jaundice (hyperbilirubinemia) derived from HO overexpression led to the development of inhibitors, such as those based on metaloproto- and meso-porphyrins inhibitors with competitive activity. Further, non-competitive inhibitors have also been identified, such as synthetic and natural imidazole-dioxolane-based, small synthetic molecules, inhibitors of the enzyme regulation pathway, and genetic engineering using iRNA or CRISPR cas9. Despite most of the applications of the HO inhibitors being related to metabolic diseases, the beneficial effects of these molecules in immune-mediated diseases have also emerged. Different medical implications, including cancer, Alzheimer´s disease, and infections, are discussed in this article and as to how the selective inhibition of HO isoforms may contribute to the treatment of these ailments.
Collapse
Affiliation(s)
- Ayleen Fernández-Fierro
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; (A.F.-F.); (M.R.); (C.C.); (J.G.)
| | - Samanta C. Funes
- Instituto Multidisciplinario de Investigaciones Biológicas-San Luis, Consejo Nacional de Investigaciones Científicas y Técnicas—Universidad Nacional de San Luis, 5700 San Luis, Argentina;
| | - Mariana Rios
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; (A.F.-F.); (M.R.); (C.C.); (J.G.)
| | - Camila Covián
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; (A.F.-F.); (M.R.); (C.C.); (J.G.)
| | - Jorge González
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; (A.F.-F.); (M.R.); (C.C.); (J.G.)
| | - Alexis M. Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; (A.F.-F.); (M.R.); (C.C.); (J.G.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- Correspondence: ; Tel.: +56-22-686-2842
| |
Collapse
|
36
|
Oshima Y, Otsuki A, Endo R, Nakasone M, Harada T, Takahashi S, Inagaki Y. The Effects of Volatile Anesthetics on Lung Ischemia-Reperfusion Injury: Basic to Clinical Studies. J Surg Res 2020; 260:325-344. [PMID: 33373852 DOI: 10.1016/j.jss.2020.11.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Case reports from as early as the 1970s have shown that intravenous injection of even a small dose of volatile anesthetics result in fatal lung injury. Direct contact between volatile anesthetics and pulmonary vasculature triggers chemical damage in the vessel walls. A wide variety of factors are involved in lung ischemia-reperfusion injury (LIRI), such as pulmonary endothelial cells, alveolar epithelial cells, alveolar macrophages, neutrophils, mast cells, platelets, proinflammatory cytokines, and surfactant. With a constellation of factors involved, the assessment of the protective effect of volatile anesthetics in LIRI is difficult. Multiple animal studies have reported that with regards to LIRI, sevoflurane demonstrates an anti-inflammatory effect in immunocompetent cells and an anti-apoptotic effect on lung tissue. Scattered studies have dismissed a protective effect of desflurane against LIRI. While a single-center randomized controlled trial (RCT) found that volatile anesthetics including desflurane demonstrated a lung-protective effect in thoracic surgery, a multicenter RCT did not demonstrate a lung-protective effect of desflurane. LIRI is common in lung transplantation. One study, although limited due to its small sample size, found that the use of volatile anesthetics in organ procurement surgery involving "death by neurologic criteria" donors did not improve lung graft survival. Future studies on the protective effect of volatile anesthetics against LIRI must examine not only the mechanism of the protective effect but also differences in the effects of different types of volatile anesthetics, their optimal dosage, and the appropriateness of their use in the event of marked alveolar capillary barrier damage.
Collapse
Affiliation(s)
- Yoshiaki Oshima
- Department of Anesthesiology, Yonago Medical Center, Yonago, Tottori, Japan.
| | - Akihiro Otsuki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Ryo Endo
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Masato Nakasone
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomomi Harada
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Shunsaku Takahashi
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yoshimi Inagaki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
37
|
Stępniewski J, Tomczyk M, Andrysiak K, Kraszewska I, Martyniak A, Langrzyk A, Kulik K, Wiśniewska E, Jeż M, Florczyk-Soluch U, Polak K, Podkalicka P, Kachamakova-Trojanowska N, Józkowicz A, Jaźwa-Kusior A, Dulak J. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes, in Contrast to Adipose Tissue-Derived Stromal Cells, Efficiently Improve Heart Function in Murine Model of Myocardial Infarction. Biomedicines 2020; 8:biomedicines8120578. [PMID: 33297443 PMCID: PMC7762393 DOI: 10.3390/biomedicines8120578] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/23/2022] Open
Abstract
Cell therapies are extensively tested to restore heart function after myocardial infarction (MI). Survival of any cell type after intracardiac administration, however, may be limited due to unfavorable conditions of damaged tissue. Therefore, the aim of this study was to evaluate the therapeutic effect of adipose-derived stromal cells (ADSCs) and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) overexpressing either the proangiogenic SDF-1α or anti-inflammatory heme oxygenase-1 (HO-1) in a murine model of MI. ADSCs and hiPSCs were transduced with lentiviral vectors encoding luciferase (Luc), GFP and either HO-1 or SDF-1α. hiPSCs were then differentiated to hiPSC-CMs using small molecules modulating the WNT pathway. Genetically modified ADSCs were firstly administered via intracardiac injection after MI induction in Nude mice. Next, ADSCs-Luc-GFP and genetically modified hiPSC-CMs were injected into the hearts of the more receptive NOD/SCID strain to compare the therapeutic effect of both cell types. Ultrasonography, performed on days 7, 14, 28 and 42, revealed a significant decrease of left ventricular ejection fraction (LVEF) in all MI-induced groups. No improvement of LVEF was observed in ADSC-treated Nude and NOD/SCID mice. In contrast, administration of hiPSC-CMs resulted in a substantial increase of LVEF, occurring between 28 and 42 days after MI, and decreased fibrosis, regardless of genetic modification. Importantly, bioluminescence analysis, as well as immunofluorescent staining, confirmed the presence of hiPSC-CMs in murine tissue. Interestingly, the luminescence signal was strongest in hearts treated with hiPSC-CMs overexpressing HO-1. Performed experiments demonstrate that hiPSC-CMs, unlike ADSCs, are effective in improving heart function after MI. Additionally, long-term evaluation of heart function seems to be crucial for proper assessment of the effect of cell administration.
Collapse
Affiliation(s)
- Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Correspondence: (J.S.); (J.D.); Tel.: +48-12-664-6375 (J.D.)
| | - Mateusz Tomczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Izabela Kraszewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Alicja Martyniak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Agnieszka Langrzyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Kardio-Med Silesia, Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland
| | - Klaudia Kulik
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Kardio-Med Silesia, Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland
| | - Ewa Wiśniewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Kardio-Med Silesia, Curie-Skłodowskiej 10C, 41-800 Zabrze, Poland
| | - Mateusz Jeż
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | | | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Agnieszka Jaźwa-Kusior
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (K.A.); (I.K.); (A.M.); (A.L.); (K.K.); (E.W.); (M.J.); (U.F.-S.); (K.P.); (P.P.); (A.J.); (A.J.-K.)
- Correspondence: (J.S.); (J.D.); Tel.: +48-12-664-6375 (J.D.)
| |
Collapse
|
38
|
Teoh JP, Li X, Simoncini T, Zhu D, Fu X. Estrogen-Mediated Gaseous Signaling Molecules in Cardiovascular Disease. Trends Endocrinol Metab 2020; 31:773-784. [PMID: 32682630 DOI: 10.1016/j.tem.2020.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/07/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Gender difference is well recognized as a key risk factor for cardiovascular disease (CVD). Estrogen, the primary female sex hormone, improves cardiovascular functions through receptor (ERα, ERβ, or G protein-coupled estrogen receptor)-initiated genomic or non-genomic mechanisms. Gaseous signaling molecules, including nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO), are important regulators of cardiovascular function. Recent studies have demonstrated that estrogen regulates the production of these signaling molecules in cardiovascular cells to exert its cardiovascular protective effects. We discuss current understanding of gaseous signaling molecules in cardiovascular disease (CVD), the underlying mechanisms through which estrogen exerts cardiovascular protective effects by regulating these molecules, and how these findings can be translated to improve the health of postmenopausal women.
Collapse
Affiliation(s)
- Jian-Peng Teoh
- Department of Gynecology and Obstetrics, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511518, P.R. China; Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, P.R. China
| | - Xiaosa Li
- Department of Gynecology and Obstetrics, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511518, P.R. China; Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, P.R. China
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa 56100, Italy
| | - Dongxing Zhu
- Department of Gynecology and Obstetrics, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511518, P.R. China; Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, P.R. China.
| | - Xiaodong Fu
- Department of Gynecology and Obstetrics, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511518, P.R. China; Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, P.R. China.
| |
Collapse
|
39
|
MZe786 Rescues Cardiac Mitochondrial Activity in High sFlt-1 and Low HO-1 Environment. Antioxidants (Basel) 2020; 9:antiox9070598. [PMID: 32660064 PMCID: PMC7402164 DOI: 10.3390/antiox9070598] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Hypertensive disorder in pregnancy is a major cause of maternal and perinatal mortality worldwide. Women who have had preeclampsia are at three to four times higher risk in later life of developing high blood pressure and heart disease. Soluble Flt-1 (sFlt-1) is elevated in preeclampsia and may remain high postpartum in women with a history of preeclampsia. Heme oxygenase-1 (Hmox1/HO-1) exerts protective effects against oxidative stimuli and is compromised in the placenta of pregnant women with preeclampsia. We hypothesized that sFlt-1 inhibits cardiac mitochondrial activity in HO-1 deficient mice. HO-1 haplo-insufficient mice (Hmox1+/−) were injected with adenovirus encoding sFlt-1 (Ad-sFlt-1) or control virus (Ad-CMV). Subsequently, they were treated daily with either placebo or MZe786 for six days, when the heart tissue was harvested to assess cardiac mitochondrial activity. Here, we show that the loss of HO-1 disturbed cardiac mitochondrial respiration and reduced mitochondrial biogenesis. The overexpression of sFlt-1 resulted in the inhibition of the cardiac mitochondrial activity in Hmox1+/− mice. The present study demonstrates that the hydrogen sulfide (H2S) releasing molecule, MZe786, rescues mitochondrial activity by stimulating cardiac mitochondrial biogenesis and antioxidant defense in Hmox1−/− mice and in Hmox1+/− mice exposed to a high sFlt-1 environment.
Collapse
|
40
|
Li X, Zhang Q, Nasser MI, Xu L, Zhang X, Zhu P, He Q, Zhao M. Oxygen homeostasis and cardiovascular disease: A role for HIF? Biomed Pharmacother 2020; 128:110338. [PMID: 32526454 DOI: 10.1016/j.biopha.2020.110338] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 12/17/2022] Open
Abstract
Hypoxia, the decline of tissue oxygen stress, plays a role in mediating cellular processes. Cardiovascular disease, relatively widespread with increased mortality, is closely correlated with oxygen homeostasis regulation. Besides, hypoxia-inducible factor-1(HIF-1) is reported to be a crucial component in regulating systemic hypoxia-induced physiological and pathological modifications like oxidative stress, damage, angiogenesis, vascular remodeling, inflammatory reaction, and metabolic remodeling. In addition, HIF1 controls the movement, proliferation, apoptosis, differentiation and activity of numerous core cells, such as cardiomyocytes, endothelial cells (ECs), smooth muscle cells (SMCs), and macrophages. Here we review the molecular regulation of HIF-1 in cardiovascular diseases, intended to improve therapeutic approaches for clinical diagnoses. Better knowledge of the oxygen balance control and the signal mechanisms involved is important to advance the development of hypoxia-related diseases.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, China
| | - Quyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, China
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Linyong Xu
- Xiangya School of Life Science, Central South University, Changsha, Hunan Province 410013, China
| | - Xueyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China.
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China.
| |
Collapse
|
41
|
Vitverova B, Najmanova I, Vicen M, Tripska K, Sa ICI, Hyspler R, Pericacho M, Nachtigal P. Long term effects of soluble endoglin and mild hypercholesterolemia in mice hearts. PLoS One 2020; 15:e0233725. [PMID: 32470058 PMCID: PMC7259503 DOI: 10.1371/journal.pone.0233725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/11/2020] [Indexed: 01/15/2023] Open
Abstract
Soluble endoglin (sEng) released into the circulation was suggested to be related to cardiovascular based pathologies. It was demonstrated that a combination of high sEng levels and long-term exposure (six months) to high fat diet (HFD) resulted in aggravation of endothelial dysfunction in the aorta. Thus, in this study, we hypothesized that a similar experimental design would affect the heart morphology, TGFβ signaling, inflammation, fibrosis, oxidative stress and eNOS signaling in myocardium in transgenic mice overexpressing human sEng. Three-month-old female transgenic mice overexpressing human sEng in plasma (Sol-Eng+ high) and their age-matched littermates with low levels of human sEng (Sol-Eng+ low) were fed a high-fat diet containing 1.25% of cholesterol and 40% of fat for six months. A blood analysis was performed, and the heart samples were analyzed by qRT-PCR and Western blot. The results of this study showed no effects of sEng and HFD on myocardial morphology/hypertrophy/fibrosis. However, the expression of pSmad2/3 and p-eNOS was reduced in Sol-Eng+ high mice. On the other hand, sEng and HFD did not significantly affect the expression of selected members of TGFβ signaling (membrane endoglin, TGFβRII, ALK-5, ALK-1, Id-1, PAI-1), inflammation (VCAM-1, ICAM-1), oxidative stress (NQO1, HO-1) and heart remodeling (PDGFβ, COL1A1, β-MHC). In conclusion, the results of this study confirmed that sEng, even combined with a high-fat diet inducing hypercholesterolemia administered for six months, does not affect the structure of the heart with respect to hypertrophy, fibrosis, inflammation and oxidative stress. Interestingly, pSmad2/3/p-eNOS signaling was reduced in both the heart in this study and the aorta in the previous study, suggesting a possible alteration of NO metabolism caused by six months exposure to high sEng levels and HFD. Thus, we might conclude that sEng combined with a high-fat diet might be related to the alteration of NO production due to altered pSmad2/3/p-eNOS signaling in the heart and aorta.
Collapse
Affiliation(s)
- Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Iveta Najmanova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radek Hyspler
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, and the Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
- * E-mail:
| |
Collapse
|
42
|
de Azevedo JTC, Malmegrim KCR. Immune mechanisms involved in sickle cell disease pathogenesis: current knowledge and perspectives. Immunol Lett 2020; 224:1-11. [PMID: 32437728 DOI: 10.1016/j.imlet.2020.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/02/2020] [Accepted: 04/18/2020] [Indexed: 12/18/2022]
Abstract
Sickle cell disease (SCD) is caused by a single point mutation in the β-chain of the hemoglobin gene that results in the replacement of glutamic acid with valine in the hemoglobin protein. However, recent studies have demonstrated that alterations in several other genes, especially immune related genes, may be associated with complications of SCD. In fact, higher chronic inflammatory status is related to more severe clinical symptoms in SCD patients, suggesting crucial roles of the immune system in SCD physiopathology. Nevertheless, although participation of innate immune cells in SCD pathogenesis has been broadly and extensively described, little is known about the roles of the adaptive immune system in this disease. In addition, the influence of treatments on the immune system of SCD patients and their complications (such as alloimmunization) are not yet completely understood. Thus, we reviewed the current knowledge about the immune mechanisms involved in SCD pathogenesis. We suggest recommendations for future studies to allow for a broader understanding of SCD pathogenesis, helping in the development of new therapies and improvement in the life quality and expectancy of patients.
Collapse
Affiliation(s)
- Júlia Teixeira Cottas de Azevedo
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
43
|
Zhong B, Ma S, Wang DH. Protease-activated receptor 2 protects against myocardial ischemia-reperfusion injury through the lipoxygenase pathway and TRPV1 channels. Exp Ther Med 2019; 18:3636-3642. [PMID: 31602241 DOI: 10.3892/etm.2019.7987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 07/22/2019] [Indexed: 12/13/2022] Open
Abstract
This study tests the hypothesis that the lipoxygenase (LOX) pathway mediates protease-activated receptor (PAR) 2-induced activation of the transient receptor potential vanilloid receptor 1 (TRPV1) to protect the heart from ischemia/reperfusion (I/R) injury. SLIGRL, a PAR2 activating peptide, was administered prior to reperfusion following left anterior descending coronary artery ligation in wild type (WT) and TRPV1 knockout (TRPV1-/-) mice. In a Langendorffly perfused heart I/R model, hemodynamic parameters, including left ventricular end-diastolic pressure, left ventricular developed pressure, coronary blood flow and left ventricular peak +dP/dt were evaluated after I/R. SLIGRL reduced the cardiac infarct size in WT and TRPV1-/- mice with a greater effect in the former strain (P<0.05). SLIGRL increased plasma levels of calcitonin gene-related peptide (CGRP) and substance P in WT (both P<0.05) but not in TRPV1-/- mice. Pretreatment with CGRP8-37 (a CGRP receptor antagonist) or RP67580 (a neurokinin-1 receptor antagonist) alone had no effect on SLIGRL-induced cardiac protection in either strain. However, combined administration of CGRP8-37 and RP67580 abolished SLIGRL-induced cardiac protection in WT but not in TRPV1-/- mice. Nordihydroguaiaretic acid (a general LOX inhibitor) and baicalein (a 12-LOX inhibitor), but not indomethacin (a cyclooxygenase inhibitor) and hexanamide (a selective cytochrome P450 epoxygenase inhibitor), abolished the protective effects of SLIGRL in WT (all P<0.05) but not in TRPV1-/- hearts. These data suggested that PAR2, possibly via 12-LOX, activates TRPV1 and leads to CGRP and substance P release to prevent I/R injury in the heart, indicating that the 12-LOX-TRPV1 pathway conveys cardiac protection to alleviate myocardial infarction.
Collapse
Affiliation(s)
- Beihua Zhong
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Shuangtao Ma
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Donna H Wang
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI 48824, USA.,Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
44
|
Andreas M, Oeser C, Kainz FM, Shabanian S, Aref T, Bilban M, Messner B, Heidtmann J, Laufer G, Kocher A, Wolzt M. Intravenous Heme Arginate Induces HO-1 (Heme Oxygenase-1) in the Human Heart. Arterioscler Thromb Vasc Biol 2019; 38:2755-2762. [PMID: 30354231 DOI: 10.1161/atvbaha.118.311832] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- HO-1 (heme oxygenase-1) induction may prevent or reduce ischemia-reperfusion injury. We previously evaluated its in vivo induction after a single systemic administration of heme arginate in peripheral blood mononuclear cells. The current trial was designed to assess the pharmacological tissue induction of HO-1 in the human heart with heme arginate in vivo. Approach and Results- Patients planned for conventional aortic valve replacement received placebo (n=8), 1 mg/kg (n=7) or 3 mg/kg (n=9) heme arginate infused intravenously 24 hours before surgery. A biopsy of the right ventricle was performed directly before aortic cross-clamping and after cross-clamp release. In addition, the right atrial appendage was partially removed for analysis. HO-1 protein and mRNA concentrations were measured in tissue samples and in peripheral blood mononuclear cells before to and up to 72 hours after surgery. No study medication-related adverse events occurred. A strong, dose-dependent effect on myocardial HO-1 mRNA levels was observed (right ventricle: 7.9±5.0 versus 88.6±49.1 versus 203.6±148.7; P=0.002 and right atrium: 10.8±8.8 versus 229.8±173.1 versus 392.7±195.7; P=0.001). This was paralleled by a profound increase of HO-1 protein concentration in atrial tissue (8401±3889 versus 28 585±10 692 versus 29 022±8583; P<0.001). Surgery and heme arginate infusion significantly increased HO-1 mRNA concentration in peripheral blood mononuclear cells ( P<0.001). HO-1 induction led to a significant increase of postoperative carboxyhemoglobin (1.7% versus 1.4%; P=0.041). No effect on plasma HO-1 protein levels could be detected. Conclusions- Myocardial HO-1 mRNA and protein can be dose-dependently induced by heme arginate. Protective effects of this therapeutic strategy should be evaluated in upcoming clinical trials. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT02314780.
Collapse
Affiliation(s)
- Martin Andreas
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Claudia Oeser
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Frieda-Maria Kainz
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Shiva Shabanian
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Tandis Aref
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine (M.B.), Medical University of Vienna, Austria
- Department of Clinical Pharmacology (M.B., M.W.), Medical University of Vienna, Austria
| | - Barbara Messner
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Julian Heidtmann
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Guenther Laufer
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Alfred Kocher
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Michael Wolzt
- Department of Clinical Pharmacology (M.B., M.W.), Medical University of Vienna, Austria
| |
Collapse
|
45
|
The Protective Role of Heme Oxygenase-1 in Atherosclerotic Diseases. Int J Mol Sci 2019; 20:ijms20153628. [PMID: 31344980 PMCID: PMC6695885 DOI: 10.3390/ijms20153628] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. These products have anti-inflammatory, anti-oxidant, anti-apoptotic, and anti-thrombotic properties. Although HO-1 is expressed at low levels in most tissues under basal conditions, it is highly inducible in response to various pathophysiological stresses/stimuli. HO-1 induction is thus thought to be an adaptive defense system that functions to protect cells and tissues against injury in many disease settings. In atherosclerosis, HO-1 may play a protective role against the progression of atherosclerosis, mainly due to the degradation of pro-oxidant heme, the generation of anti-oxidants biliverdin and bilirubin and the production of vasodilator CO. In animal models, a lack of HO-1 was shown to accelerate atherosclerosis, whereas HO-1 induction reduced atherosclerosis. It was also reported that HO-1 induction improved the cardiac function and postinfarction survival in animal models of heart failure or myocardial infarction. Recently, we and others examined blood HO-1 levels in patients with atherosclerotic diseases, e.g., coronary artery disease (CAD) and peripheral artery disease (PAD). Taken together, these findings to date support the notion that HO-1 plays a protective role against the progression of atherosclerotic diseases. This review summarizes the roles of HO-1 in atherosclerosis and focuses on the clinical studies that examined the relationships between HO-1 levels and atherosclerotic diseases.
Collapse
|
46
|
Drummond HA, Mitchell ZL, Abraham NG, Stec DE. Targeting Heme Oxygenase-1 in Cardiovascular and Kidney Disease. Antioxidants (Basel) 2019; 8:antiox8060181. [PMID: 31216709 PMCID: PMC6617021 DOI: 10.3390/antiox8060181] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/13/2019] [Accepted: 06/15/2019] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase (HO) plays an important role in the cardiovascular system. It is involved in many physiological and pathophysiological processes in all organs of the cardiovascular system. From the regulation of blood pressure and blood flow to the adaptive response to end-organ injury, HO plays a critical role in the ability of the cardiovascular system to respond and adapt to changes in homeostasis. There have been great advances in our understanding of the role of HO in the regulation of blood pressure and target organ injury in the last decade. Results from these studies demonstrate that targeting of the HO system could provide novel therapeutic opportunities for the treatment of several cardiovascular and renal diseases. The goal of this review is to highlight the important role of HO in the regulation of cardiovascular and renal function and protection from disease and to highlight areas in which targeting of the HO system needs to be translated to help benefit patient populations.
Collapse
Affiliation(s)
- Heather A Drummond
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA.
| | - Zachary L Mitchell
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA.
| | - Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, Vahalla, NY 10595, USA.
- Joan C. Edwards School of Medicine, Marshall University, Huntington, VA 25701, USA.
| | - David E Stec
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA.
| |
Collapse
|
47
|
Eltobshy SAG, Hussein AM, Elmileegy AA, Askar MH, Khater Y, Metias EF, Helal GM. Effects of heme oxygenase-1 upregulation on isoproterenol-induced myocardial infarction. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:203-217. [PMID: 31080351 PMCID: PMC6488703 DOI: 10.4196/kjpp.2019.23.3.203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/23/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022]
Abstract
The present study was designed to examine the effect of heme oxygenase-1 (HO-1) induction by cobalt protoporphyrin (CoPP) on the cardiac functions and morphology, electrocardiogram (ECG) changes, myocardial antioxidants (superoxide dismutase [SOD] and glutathione [GSH]), and expression of heat shock protein (Hsp) 70 and connexin 43 (Cx-43) in myocardial muscles in isoproterenol (ISO) induced myocardial infarction (MI). Thirty two adult male Sprague Dawely rats were divided into 4 groups (each 8 rats): normal control (NC) group, ISO group: received ISO at dose of 150 mg/kg body weight intraperitoneally (i.p.) for 2 successive days; ISO + Trizma group: received (ISO) and Trizma (solvent of CoPP) at dose of 5 mg/kg i.p. injection 2 days before injection of ISO, with ISO at day 0 and at day 2 after ISO injections; and ISO + CoPP group: received ISO and CoPP at a dose of 5 mg/kg dissolved in Trizma i.p. injection as Trizma. We found that, administration of ISO caused significant increase in heart rate, corrected QT interval, ST segment, cardiac enzymes (lactate dehydrogenase, creatine kinase-muscle/brain), cardiac HO-1, Hsp70 with significant attenuation in myocardial GSH, SOD, and Cx-43. On the other hand, administration of CoPP caused significant improvement in ECG parameters, cardiac enzymes, cardiac morphology; antioxidants induced by ISO with significant increase in HO-1, Cx-43, and Hsp70 expression in myocardium. In conclusions, we concluded that induction of HO-1 by CoPP ameliorates ISO-induced myocardial injury, which might be due to up-regulation of Hsp70 and gap junction protein (Cx-43).
Collapse
Affiliation(s)
- Somaia A G Eltobshy
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Abdelaziz M Hussein
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Asaad A Elmileegy
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mona H Askar
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Yomna Khater
- Medical Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Emile F Metias
- Department of Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ghada M Helal
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
48
|
Yang P, Zhou Y, Xia Q, Yao L, Chang X. Astragaloside IV Regulates the PI3K/Akt/HO-1 Signaling Pathway and Inhibits H9c2 Cardiomyocyte Injury Induced by Hypoxia-Reoxygenation. Biol Pharm Bull 2019; 42:721-727. [PMID: 30867343 DOI: 10.1248/bpb.b18-00854] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Astragaloside IV (AS-IV) is one of the main pharmacologically active compounds found in Astragalus membranaceus. AS-IV has protective effects against ischemia-reperfusion injury (IRI), but its mechanism of action has not yet been determined. This study aims to investigate the effect of AS-IV on IRI and its effect on the phosphadylinositol 3-kinase (PI3K)/Akt/heme oxygenase (HO-1) signaling pathway through in vitro experiments. Firstly, a cell culture model of myocardiocyte hypoxia-reoxygenation (H/R) injury was replicated. After AS-IV treatment, cell viability, reactive oxygen species (ROS) levels, as well as the content or activity of the cellular factors lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA), interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), were measured to evaluate the effect of treatment with AS-IV. The effect of AS-IV on HO-1 protein expression and nuclear factor E2-related factor 2 (Nrf2) and Bach1 protein expression was determined by Western blotting. Finally, a reversal of the effect of AS-IV treatment was observed following co-incubation with a PI3K inhibitor. Our results show that AS-IV has good protective effect on H/R injury and has anti-oxidative stress and anti-inflammatory effects. It can regulate the expression of Nrf2 and Bach1 proteins in the nucleus and promote the expression of HO-1 protein, while a PI3K inhibitor can partially reverse the above effects. This study suggests that the PI3K/Akt/HO-1 signaling pathway may be a key signaling pathway for the anti-IRI effect of AS-IV.
Collapse
Affiliation(s)
| | - Yuping Zhou
- The Affiliated Hospital of Medical School of Ningbo University
| | - Qing Xia
- Ningbo College of Health Sciences
| | | | | |
Collapse
|
49
|
Abstract
Nonapoptotic cell death-induced tissue damage has been implicated in a variety of diseases, including neurodegenerative disorder, inflammation, and stroke. In this study, we demonstrate that ferroptosis, a newly defined iron-dependent cell death, mediates both chemotherapy- and ischemia/reperfusion-induced cardiomyopathy. RNA-sequencing analysis revealed up-regulation of heme oxygenase 1 by doxorubicin as a major mechanism of ferroptotic cardiomyopathy. As a result, heme oxygenase 1 degrades heme and releases free iron in cardiomyocytes, which in turn leads to generation of oxidized lipids in the mitochondria membrane. Most importantly, both iron chelation therapy and pharmacologically blocking ferroptosis could significantly alleviate cardiomyopathy in mice. These findings suggest targeting ferroptosis as a strategy for treating deadly heart disease. Heart disease is the leading cause of death worldwide. A key pathogenic factor in the development of lethal heart failure is loss of terminally differentiated cardiomyocytes. However, mechanisms of cardiomyocyte death remain unclear. Here, we discovered and demonstrated that ferroptosis, a programmed iron-dependent cell death, as a mechanism in murine models of doxorubicin (DOX)- and ischemia/reperfusion (I/R)-induced cardiomyopathy. In canonical apoptosis and/or necroptosis-defective Ripk3−/−, Mlkl−/−, or Fadd−/−Mlkl−/− mice, DOX-treated cardiomyocytes showed features of typical ferroptotic cell death. Consistently, compared with dexrazoxane, the only FDA-approved drug for treating DOX-induced cardiotoxicity, inhibition of ferroptosis by ferrostatin-1 significantly reduced DOX cardiomyopathy. RNA-sequencing results revealed that heme oxygenase-1 (Hmox1) was significantly up-regulated in DOX-treated murine hearts. Administering DOX to mice induced cardiomyopathy with a rapid, systemic accumulation of nonheme iron via heme degradation by Nrf2-mediated up-regulation of Hmox1, which effect was abolished in Nrf2-deficent mice. Conversely, zinc protoporphyrin IX, an Hmox1 antagonist, protected the DOX-treated mice, suggesting free iron released on heme degradation is necessary and sufficient to induce cardiac injury. Given that ferroptosis is driven by damage to lipid membranes, we further investigated and found that excess free iron accumulated in mitochondria and caused lipid peroxidation on its membrane. Mitochondria-targeted antioxidant MitoTEMPO significantly rescued DOX cardiomyopathy, supporting oxidative damage of mitochondria as a major mechanism in ferroptosis-induced heart damage. Importantly, ferrostatin-1 and iron chelation also ameliorated heart failure induced by both acute and chronic I/R in mice. These findings highlight that targeting ferroptosis serves as a cardioprotective strategy for cardiomyopathy prevention.
Collapse
|
50
|
An W, Yu Y, Zhang Y, Zhang Z, Yu Y, Zhao X. Exogenous IL-19 attenuates acute ischaemic injury and improves survival in male mice with myocardial infarction. Br J Pharmacol 2019; 176:699-710. [PMID: 30460984 DOI: 10.1111/bph.14549] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Myocardial infarction (MI) is one of the leading causes of death in China and often results in the development of heart failure. In this work, we tested the therapeutic role of Interleukin-19 (IL-19) in mice with MI and investigated the underlying molecular mechanism. EXPERIMENTAL APPROACH Mice were subjected to MI by ligation of left anterior descending coronary artery (LAD) and treated with IL-19 (10ng g-1 ; i.p.). KEY RESULTS Protein expression of IL-19 and its receptor in myocardium were upregulated 24 hrs post-MI in male mice. IL-19 treatment decreased infarct and apoptosis in myocardium, accompanied by enhanced haem oxygenase-1 (HO-1) activities and reduced malondialdehyde (MDA) formation. Pretreatment with IL-19 upregulated HO-1 expression in cultured neonatal mouse ventricular myocytes and attenuated oxygen-glucose deprivation (OGD)-induced injuries in vitro. Furthermore, IL-19 preserved cardiac function and improved survival of mice with MI. IL-19 reduced inflammatory infiltrates and suppressed formation of TNF-α, IL-1β, and IL-6. More importantly, IL-19 inhibited polarization toward proinflammatory M1 macrophages and stimulated M2 macrophage polarization in myocardium of mice with MI. IL-19 enhanced protein levels of vascular endothelial growth factor (VEGF) and promoted angiogenesis in myocardium of mice with MI. In addition, IL-19 treatment increased DNA-binding of the transcription factor STAT3 in myocardium of mice with MI. CONCLUSIONS AND IMPLICATIONS Treatment with exogenous IL-19 attenuated acute ischemic injury and improved survival of mice with MI. The mechanisms underlying these effects involved induction of HO-1, M2 macrophage polarization, angiogenesis, and STAT3 activation.
Collapse
Affiliation(s)
- Weishuai An
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yongsheng Yu
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuefan Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhigang Zhang
- Department of Cardiology, Putuo Center Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunhua Yu
- Department of Geriatrics, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Xianxian Zhao
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|