1
|
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
|
2
|
Wang H, Siren J, Perttunen S, Immonen K, Chen Y, Narumanchi S, Kosonen R, Paavola J, Laine M, Tikkanen I, Lakkisto P. Deficiency of heme oxygenase 1a causes detrimental effects on cardiac function. J Cell Mol Med 2024; 28:e18243. [PMID: 38509740 PMCID: PMC10955162 DOI: 10.1111/jcmm.18243] [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: 09/25/2023] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
Humans lacking heme oxygenase 1 (HMOX1) display growth retardation, haemolytic anaemia, and vulnerability to stress; however, cardiac function remains unclear. We aimed to explore the cardiac function of zebrafish lacking hmox1a at baseline and in response to stress. We generated zebrafish hmox1a mutants using CRISPR/Cas9 genome editing technology. Deletion of hmox1a increases cardiac output and further induces hypertrophy in adults. Adults lacking hmox1a develop myocardial interstitial fibrosis, restrain cardiomyocyte proliferation and downregulate renal haemoglobin and cardiac antioxidative genes. Larvae lacking hmox1a fail to respond to hypoxia, whereas adults are insensitive to isoproterenol stimulation in the heart, suggesting that hmox1a is necessary for cardiac response to stress. Haplodeficiency of hmox1a stimulates non-mitochondrial respiration and cardiac cell proliferation, increases cardiac output in larvae in response to hypoxia, and deteriorates cardiac function and structure in adults upon isoproterenol treatment. Intriguingly, haplodeficiency of hmox1a upregulates cardiac hmox1a and hmox1b in response to isoproterenol. Collectively, deletion of hmox1a results in cardiac remodelling and abrogates cardiac response to hypoxia and isoproterenol. Haplodeficiency of hmox1a aggravates cardiac response to the stress, which could be associated with the upregulation of hmox1a and hmox1b. Our data suggests that HMOX1 homeostasis is essential for maintaining cardiac function and promoting cardioprotective effects.
Collapse
Affiliation(s)
- Hong Wang
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Juuso Siren
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Sanni Perttunen
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | | | - Yu‐Chia Chen
- Department of AnatomyUniversity of HelsinkiHelsinkiFinland
| | | | - Riikka Kosonen
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Jere Paavola
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
- Heart and Lung CentreUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Mika Laine
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
- Heart and Lung CentreUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Ilkka Tikkanen
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
- Abdominal Centre NephrologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Päivi Lakkisto
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
- Department of Clinical ChemistryUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| |
Collapse
|
3
|
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
|
4
|
Vilahur G, Sutelman P, Ben-Aicha S, Mendieta G, Radiké M, Schoch L, Casaní L, Borrell-Pagés M, Padro T, Badimon L. Supplementation With Spirulina Reduces Infarct Size and Ameliorates Cardiac Function in a Pig Model of STEMI. Front Pharmacol 2022; 13:891801. [PMID: 35592428 PMCID: PMC9113432 DOI: 10.3389/fphar.2022.891801] [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: 03/08/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Aims: Myocardial infarction (MI) is the clinical manifestation of atherosclerotic coronary artery disease. Spirulina is an algae known to ameliorate cardiometabolic disorders and with proven anti-inflammatory and anti-oxidant effects. We investigated, in a highly translatable animal model, whether oral supplementation with spirulina protects against the deleterious effects triggered by ST-elevation MI (STEMI). Methods: Pigs were fed a regular diet supplemented with spirulina (1 g/animal/bid) or placebo-control for 10 days. Thereafter, animals were subjected to 1.5 h percutaneous balloon-induced coronary occlusion (STEMI) followed by 2.5 h reperfusion and then sacrificed. We assessed infarct size and cardiac function. Blood samples and infarcted and remote myocardial tissue were obtained. Results: Spirulina supplementation reduced infarct size by 64%, increased myocardial salvage by 18%, and improved cardiac function by 30% vs. controls (p < 0.05). These benefits were associated with attenuation in DNA-oxidative damage and apoptotic markers and increased iNOS in the infarcted myocardium, higher AMPK activation in the remote myocardium, and lower myocardial MCP-1 expression. Systemically, spirulina attenuated Cox-2 expression in STEMI-activated peripheral blood mononuclear cells and enhanced TNF-α release acutely post-STEMI. Additionally, spirulina decreased weight gain progression over time (p < 0.05) without changes in lipids, glucose, liver or kidney parameters. Conclusion: A 10-day supplementation with spirulina exerts cardioprotection in a preclinical setting of STEMI by limiting cardiac damage and improving ventricular contractility through anti-oxidative, anti-inflammatory, and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Gemma Vilahur
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Pablo Sutelman
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Soumaya Ben-Aicha
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Guiomar Mendieta
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Department of Cardiology, Clinic Hospital, Barcelona, Spain
| | - Monika Radiké
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Leonie Schoch
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Laura Casaní
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - María Borrell-Pagés
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,CiberCV, Institute Carlos III, Madrid, Spain.,Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Abouleisa RRE, Salama ABM, Ou Q, Tang XL, Solanki M, Guo Y, Nong Y, McNally L, Lorkiewicz PK, Kassem KM, Ahern BM, Choudhary K, Thomas R, Huang Y, Juhardeen HR, Siddique A, Ifthikar Z, Hammad SK, Elbaz AS, Ivey KN, Conklin DJ, Satin J, Hill BG, Srivastava D, Bolli R, Mohamed TMA. Transient Cell Cycle Induction in Cardiomyocytes to Treat Subacute Ischemic Heart Failure. Circulation 2022; 145:1339-1355. [PMID: 35061545 PMCID: PMC9038650 DOI: 10.1161/circulationaha.121.057641] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND The regenerative capacity of the heart after myocardial infarction is limited. Our previous study showed that ectopic introduction of 4 cell cycle factors (4F; CDK1 [cyclin-dependent kinase 1], CDK4 [cyclin-dependent kinase 4], CCNB [cyclin B1], and CCND [cyclin D1]) promotes cardiomyocyte proliferation in 15% to 20% of infected cardiomyocytes in vitro and in vivo and improves cardiac function after myocardial infarction in mice. METHODS Using temporal single-cell RNA sequencing, we aimed to identify the necessary reprogramming stages during the forced cardiomyocyte proliferation with 4F on a single cell basis. Using rat and pig models of ischemic heart failure, we aimed to start the first preclinical testing to introduce 4F gene therapy as a candidate for the treatment of ischemia-induced heart failure. RESULTS Temporal bulk and single-cell RNA sequencing and further biochemical validations of mature human induced pluripotent stem cell-derived cardiomyocytes treated with either LacZ or 4F adenoviruses revealed full cell cycle reprogramming in 15% of the cardiomyocyte population at 48 hours after infection with 4F, which was associated mainly with sarcomere disassembly and metabolic reprogramming (n=3/time point/group). Transient overexpression of 4F, specifically in cardiomyocytes, was achieved using a polycistronic nonintegrating lentivirus (NIL) encoding 4F; each is driven by a TNNT2 (cardiac troponin T isoform 2) promoter (TNNT2-4Fpolycistronic-NIL). TNNT2-4Fpolycistronic-NIL or control virus was injected intramyocardially 1 week after myocardial infarction in rats (n=10/group) or pigs (n=6-7/group). Four weeks after injection, TNNT2-4Fpolycistronic-NIL-treated animals showed significant improvement in left ventricular ejection fraction and scar size compared with the control virus-treated animals. At 4 months after treatment, rats that received TNNT2-4Fpolycistronic-NIL still showed a sustained improvement in cardiac function and no obvious development of cardiac arrhythmias or systemic tumorigenesis (n=10/group). CONCLUSIONS This study provides mechanistic insights into the process of forced cardiomyocyte proliferation and advances the clinical feasibility of this approach by minimizing the oncogenic potential of the cell cycle factors owing to the use of a novel transient and cardiomyocyte-specific viral construct.
Collapse
Affiliation(s)
- Riham R. E. Abouleisa
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Abou Bakr M. Salama
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Faculty of Medicine, Zagazig University, Egypt
| | - Qinghui Ou
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Xian-Liang Tang
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Mitesh Solanki
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Yiru Guo
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Yibing Nong
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Lindsey McNally
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | - Pawel K. Lorkiewicz
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | - Kamal M. Kassem
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | | | | | | | - Yu Huang
- Gladstone Institute, San Francisco, CA, U.S.A
| | | | - Aisha Siddique
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Zainab Ifthikar
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sally K. Hammad
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Department of Biochemistry Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ayman S. Elbaz
- Department of Bioengineering, Speed School of Engineering, University of Louisville, KY, U.S.A
| | | | - Daniel J Conklin
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, KY, U.S.A
| | - Bradford G. Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
| | | | - Roberto Bolli
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
| | - Tamer M A Mohamed
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, U.S.A
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, U.S.A
- Department of Bioengineering, Speed School of Engineering, University of Louisville, KY, U.S.A
- Department of Pharmacology and Toxicology, University of Louisville, KY, U.S.A
- Institute of Cardiovascular Sciences, University of Manchester, U.K
| |
Collapse
|
6
|
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
|
7
|
Dunn LL, Kong SMY, Tumanov S, Chen W, Cantley J, Ayer A, Maghzal GJ, Midwinter RG, Chan KH, Ng MKC, Stocker R. Hmox1 (Heme Oxygenase-1) Protects Against Ischemia-Mediated Injury via Stabilization of HIF-1α (Hypoxia-Inducible Factor-1α). Arterioscler Thromb Vasc Biol 2021; 41:317-330. [PMID: 33207934 DOI: 10.1161/atvbaha.120.315393] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Hmox1 (heme oxygenase-1) is a stress-induced enzyme that catalyzes the degradation of heme to carbon monoxide, iron, and biliverdin. Induction of Hmox1 and its products protect against cardiovascular disease, including ischemic injury. Hmox1 is also a downstream target of the transcription factor HIF-1α (hypoxia-inducible factor-1α), a key regulator of the body's response to hypoxia. However, the mechanisms by which Hmox1 confers protection against ischemia-mediated injury remain to be fully understood. Approach and Results: Hmox1 deficient (Hmox1-/-) mice had impaired blood flow recovery with severe tissue necrosis and autoamputation following unilateral hindlimb ischemia. Autoamputation preceded the return of blood flow, and bone marrow transfer from littermate wild-type mice failed to prevent tissue injury and autoamputation. In wild-type mice, ischemia-induced expression of Hmox1 in skeletal muscle occurred before stabilization of HIF-1α. Moreover, HIF-1α stabilization and glucose utilization were impaired in Hmox1-/- mice compared with wild-type mice. Experiments exposing dermal fibroblasts to hypoxia (1% O2) recapitulated these key findings. Metabolomics analyses indicated a failure of Hmox1-/- mice to adapt cellular energy reprogramming in response to ischemia. Prolyl-4-hydroxylase inhibition stabilized HIF-1α in Hmox1-/- fibroblasts and ischemic skeletal muscle, decreased tissue necrosis and autoamputation, and restored cellular metabolism to that of wild-type mice. Mechanistic studies showed that carbon monoxide stabilized HIF-1α in Hmox1-/- fibroblasts in response to hypoxia. CONCLUSIONS Our findings suggest that Hmox1 acts both downstream and upstream of HIF-1α, and that stabilization of HIF-1α contributes to Hmox1's protection against ischemic injury independent of neovascularization.
Collapse
Affiliation(s)
- Louise L Dunn
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia (L.L.D., W.C., A.A., G.J.M., R.S.)
| | - Stephanie M Y Kong
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
| | - Sergey Tumanov
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
- Heart Research Institute, Newtown, NSW, Australia (S.T., W.C., A.A., K.H.C., M.K.C.N., R.S.)
| | - Weiyu Chen
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia (L.L.D., W.C., A.A., G.J.M., R.S.)
- Heart Research Institute, Newtown, NSW, Australia (S.T., W.C., A.A., K.H.C., M.K.C.N., R.S.)
| | | | - Anita Ayer
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia (L.L.D., W.C., A.A., G.J.M., R.S.)
- Heart Research Institute, Newtown, NSW, Australia (S.T., W.C., A.A., K.H.C., M.K.C.N., R.S.)
| | - Ghassan J Maghzal
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia (L.L.D., W.C., A.A., G.J.M., R.S.)
| | - Robyn G Midwinter
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia (L.L.D., W.C., A.A., G.J.M., R.S.)
- Centre for Vascular Research, School of Medical Sciences (Pathology), and Bosch Institute, Sydney Medical School, The University of Sydney, Australia (R.G.M., R.S.)
| | - Kim H Chan
- Heart Research Institute, Newtown, NSW, Australia (S.T., W.C., A.A., K.H.C., M.K.C.N., R.S.)
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia (K.H.C., M.K.C.N.)
| | - Martin K C Ng
- Heart Research Institute, Newtown, NSW, Australia (S.T., W.C., A.A., K.H.C., M.K.C.N., R.S.)
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia (K.H.C., M.K.C.N.)
| | - Roland Stocker
- The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia (L.L.D., S.M.Y.K., S.T., W.C., A.A., G.J.M., R.S.)
- Heart Research Institute, Newtown, NSW, Australia (S.T., W.C., A.A., K.H.C., M.K.C.N., R.S.)
- Centre for Vascular Research, School of Medical Sciences (Pathology), and Bosch Institute, Sydney Medical School, The University of Sydney, Australia (R.G.M., R.S.)
| |
Collapse
|
8
|
Arad M, Waldman M, Abraham NG, Hochhauser E. Therapeutic approaches to diabetic cardiomyopathy: Targeting the antioxidant pathway. Prostaglandins Other Lipid Mediat 2020; 150:106454. [PMID: 32413571 DOI: 10.1016/j.prostaglandins.2020.106454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/23/2020] [Accepted: 05/06/2020] [Indexed: 12/25/2022]
Abstract
The global epidemic of cardiovascular disease continues unabated and remains the leading cause of death both in the US and worldwide. We hereby summarize the available therapies for diabetes and cardiovascular disease in diabetics. Clearly, the current approaches to diabetic heart disease often target the manifestations and certain mediators but not the specific pathways leading to myocardial injury, remodeling and dysfunction. Better understanding of the molecular events determining the evolution of diabetic cardiomyopathy will provide insight into the development of specific and targeted therapies. Recent studies largely increased our understanding of the role of enhanced inflammatory response, ROS production, as well as the contribution of Cyp-P450-epoxygenase-derived epoxyeicosatrienoic acid (EET), Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1α (PGC-1α), Heme Oxygenase (HO)-1 and 20-HETE in pathophysiology and therapy of cardiovascular disease. PGC-1α increases production of the HO-1 which has a major role in protecting the heart against oxidative stress, microcirculation and mitochondrial dysfunction. This review describes the potential drugs and their downstream targets, PGC-1α and HO-1, as major loci for developing therapeutic approaches beside diet and lifestyle modification for the treatment and prevention of heart disease associated with obesity and diabetes.
Collapse
Affiliation(s)
- Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Maayan Waldman
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Tel Aviv, Israel
| | - Nader G Abraham
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
9
|
Drummond GS, Baum J, Greenberg M, Lewis D, Abraham NG. HO-1 overexpression and underexpression: Clinical implications. Arch Biochem Biophys 2019; 673:108073. [PMID: 31425676 DOI: 10.1016/j.abb.2019.108073] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/23/2019] [Accepted: 08/10/2019] [Indexed: 12/11/2022]
Abstract
In this review we examine the effects of both over- and under-production of heme oxygenase-1 (HO-1) and HO activity on a broad spectrum of biological systems and on vascular disease. In a few instances e.g., neonatal jaundice, overproduction of HO-1 and increased HO activity results in elevated levels of bilirubin requiring clinical intervention with inhibitors of HO activity. In contrast HO-1 levels and HO activity are low in obesity and the HO system responds to mitigate the deleterious effects of oxidative stress through increased levels of bilirubin (anti-inflammatory) and CO (anti-apoptotic) and decreased levels of heme (pro-oxidant). Site specific HO-1 overexpression diminishes adipocyte terminal differentiation and lipid accumulation of obesity mediated release of inflammatory molecules. A series of diverse strategies have been implemented that focus on increasing HO-1 and HO activity that are central to reversing the clinical complications associated with diseases including, obesity, metabolic syndrome and vascular disease.
Collapse
Affiliation(s)
- George S Drummond
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Jeffrey Baum
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Menachem Greenberg
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - David Lewis
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA; Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25701, USA.
| |
Collapse
|
10
|
Inducible cardiac-specific overexpression of cyclooxygenase-2 (COX-2) confers resistance to ischemia/reperfusion injury. Basic Res Cardiol 2019; 114:32. [PMID: 31278427 DOI: 10.1007/s00395-019-0741-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/26/2019] [Indexed: 10/26/2022]
Abstract
The role of cyclooxygenase-2 (COX-2) in cardiovascular biology remains controversial. Although COX-2 has been reported to mediate the protective actions of late preconditioning, other studies show that it is also an important mediator of inflammation, toxic shock, and apoptosis, resulting in significant dysfunction and injury in several tissues. To determine whether increased myocardial COX-2, in itself, is protective, cardiac-specific, inducible (Tet-off) COX-2 transgenic (iCOX-2 TG) mice were generated by crossbreeding α-MyHC-tTA transgenic mice (tetracycline transactivator [tTA]) with CMV/TRE-COX-2 transgenic mice. Three months after COX-2 induction, mice were subjected to a 30-min coronary occlusion and 24 h of reperfusion. Three different lines (L5, L7, and L8) of iCOX-2 TG mice were studied; in all three lines, infarct size was markedly reduced compared with WT mice: L5 TG/TG 23.4 ± 5.8 vs. WT/WT 48.5 ± 6.1% of risk region; L7 TG/TG 23.2 ± 6.2 vs. WT/WT 53.3 ± 3.6%; and L8 TG/TG 23.5 ± 2.8 vs. WT/WT 52.7 ± 4.6% (P < 0.05 for each). COX-2 inhibition with NS-398 completely abolished the cardioprotection provided by COX-2 overexpression. This study for the first time utilizes an inducible cardiac-specific COX-2 overexpression system to examine the role of this enzyme in ischemia/reperfusion injury in vivo. We demonstrate that induced cardiac-specific overexpression of COX-2 exerts a potent cardioprotective effect against myocardial infarction in mice, and that chronic COX-2 overexpression is not associated with any apparent deleterious effects. We also show that PGE2 levels are upregulated in COX-2 overexpressing cardiac tissue, confirming increased enzyme activity. Finally, we have developed a valuable genetic tool to further our understanding of the role of COX-2 in ischemia/reperfusion injury and other settings. The concept that COX-2 is chronically protective has important therapeutic implications for studies of long-term gene therapy aimed at increasing myocardial COX-2 content as well as other COX-2- based strategies.
Collapse
|
11
|
Modulation of the monocyte/macrophage system in heart failure by targeting heme oxygenase-1. Vascul Pharmacol 2018; 112:79-90. [PMID: 30213580 DOI: 10.1016/j.vph.2018.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/26/2018] [Accepted: 08/29/2018] [Indexed: 01/14/2023]
Abstract
Upon myocardial infarction (MI) immune system becomes activated by extensive necrosis of cardiomyocytes releasing intracellular molecules called damage-associated molecular patterns. Overactive and prolonged immune responses are likely to be responsible for heart failure development and progression in patients surviving the ischemic episode. Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. This stress-inducible enzyme is induced by various oxidative and inflammatory signals. Consequently, biological actions of HO-1 are not limited to degradation of a toxic heme released from hemoproteins, but also provide an adaptive cellular response against chronic inflammation and oxidative injury. Indeed, the immunomodulatory and anti-inflammatory properties of HO-1 were demonstrated in several experimental studies, as well as in human cases of genetic HO-1 deficiency. HO-1 was shown to suppress the production, myocardial infiltration and inflammatory properties of monocytes and macrophages what resulted in limitation of post-MI cardiac damage. This review specifically addresses the role of HO-1, heme and its degradation products in macrophage biology and post-ischemic cardiac repair. A more complete understanding of these mechanisms is essential to develop new therapeutic approaches.
Collapse
|
12
|
Kirby RJ, Divlianska DB, Whig K, Bryan N, Morfa CJ, Koo A, Nguyen KH, Maloney P, Peddibhotla S, Sessions EH, Hershberger PM, Smith LH, Malany S. Discovery of Novel Small-Molecule Inducers of Heme Oxygenase-1 That Protect Human iPSC-Derived Cardiomyocytes from Oxidative Stress. J Pharmacol Exp Ther 2018; 364:87-96. [PMID: 29101218 DOI: 10.1124/jpet.117.243717] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/31/2017] [Indexed: 01/09/2023] Open
Abstract
Oxidative injury to cardiomyocytes plays a critical role in cardiac pathogenesis following myocardial infarction. Transplantation of stem cell-derived cardiomyocytes has recently progressed as a novel treatment to repair damaged cardiac tissue but its efficacy has been limited by poor survival of transplanted cells owing to oxidative stress in the post-transplantation environment. Identification of small molecules that activate cardioprotective pathways to prevent oxidative damage and increase survival of stem cells post-transplantation is therefore of great interest for improving the efficacy of stem cell therapies. This report describes a chemical biology phenotypic screening approach to identify and validate small molecules that protect human-induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) from oxidative stress. A luminescence-based high-throughput assay for cell viability was used to screen a diverse collection of 48,640 small molecules for protection of hiPSC-CMs from peroxide-induced cell death. Cardioprotective activity of "hit" compounds was confirmed using impedance-based detection of cardiomyocyte monolayer integrity and contractile function. Structure-activity relationship studies led to the identification of a potent class of compounds with 4-(pyridine-2-yl)thiazole scaffold. Examination of gene expression in hiPSC-CMs revealed that the hit compound, designated cardioprotectant 312 (CP-312), induces robust upregulation of heme oxygenase-1, a marker of the antioxidant response network that has been strongly correlated with protection of cardiomyocytes from oxidative stress. CP-312 therefore represents a novel chemical scaffold identified by phenotypic high-throughput screening using hiPSC-CMs that activates the antioxidant defense response and may lead to improved pharmacological cardioprotective therapies.
Collapse
Affiliation(s)
- R Jason Kirby
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Daniela B Divlianska
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Kanupriya Whig
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Nadezda Bryan
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Camilo J Morfa
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Ada Koo
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Kevin H Nguyen
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Patrick Maloney
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Satayamaheshwar Peddibhotla
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - E Hampton Sessions
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Paul M Hershberger
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Layton H Smith
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| | - Siobhan Malany
- Sanford Burham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, Orlando, Florida
| |
Collapse
|
13
|
Otterbein LE, Foresti R, Motterlini R. Heme Oxygenase-1 and Carbon Monoxide in the Heart: The Balancing Act Between Danger Signaling and Pro-Survival. Circ Res 2017; 118:1940-1959. [PMID: 27283533 DOI: 10.1161/circresaha.116.306588] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Understanding the processes governing the ability of the heart to repair and regenerate after injury is crucial for developing translational medical solutions. New avenues of exploration include cardiac cell therapy and cellular reprogramming targeting cell death and regeneration. An attractive possibility is the exploitation of cytoprotective genes that exist solely for self-preservation processes and serve to promote and support cell survival. Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. The remarkable cardioprotective effects ascribed to heme oxygenase-1 are best evidenced by its ability to regulate inflammatory processes, cellular signaling, and mitochondrial function ultimately mitigating myocardial tissue injury and the progression of vascular-proliferative disease. We discuss here new insights into the role of heme oxygenase-1 and heme on cardiovascular health, and importantly, how they might be leveraged to promote heart repair after injury.
Collapse
Affiliation(s)
- Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Roberta Foresti
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| | - Roberto Motterlini
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| |
Collapse
|
14
|
Abstract
Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.
Collapse
Affiliation(s)
- Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Abolfazl Zarjou
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Anupam Agarwal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
15
|
Miranda B, Barrabés JA, Figueras J, Pineda V, Rodríguez-Palomares J, Lidón RM, Sambola A, Bañeras J, Otaegui I, García-Dorado D. Plasma bilirubin values on admission and ventricular remodeling after a first anterior ST-segment elevation acute myocardial infarction. Ann Med 2016; 48:1-9. [PMID: 26631587 DOI: 10.3109/07853890.2015.1112027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Bilirubin may elicit cardiovascular protection and heme oxygenase-1 overexpression attenuated post-infarction ventricular remodeling in experimental animals, but the association between bilirubin levels and post-infarction remodeling is unknown. MATERIALS AND METHODS In 145 patients with a first anterior ST-segment elevation acute myocardial infarction (STEMI), we assessed whether plasma bilirubin on admission predicted adverse remodeling (left ventricular end-diastolic volume [LVEDV] increase ≥20% between discharge and 6 months, estimated by magnetic resonance imaging). RESULTS Patients' baseline characteristics and management were comparable among bilirubin tertiles. LVEDV increased at 6 months (P < 0.001) with respect to the initial exam, but the magnitude of this increase was similar across increasing bilirubin tertiles (10.8 [30.2], 10.1 [22.9], and 12.7 [24.3]%, P = 0.500). Median (25-75 percentile) bilirubin values in patients with and without adverse remodeling were 0.75 (0.60-0.93) and 0.73 (0.60-0.92) mg/dL (P = 0.693). Absence of final TIMI flow grade 3 (odds ratio 3.92, 95% CI 1.12-13.66) and a history of hypertension (2.04, 0.93-4.50), but not admission bilirubin, were independently associated with adverse remodeling. Bilirubin also did not predict the increase in ejection fraction at 6 months. CONCLUSIONS Admission bilirubin values are not related to LVEDV or ejection fraction progression after a first anterior STEMI and do not predict adverse ventricular remodeling. Key messages Bilirubin levels are inversely related to cardiovascular disease, and overexpression of heme oxygenase-1 (the enzyme that determines bilirubin production) has prevented post-infarction ventricular remodeling in experimental animals, but the association between bilirubin levels and the progression of ventricular volumes and function in patients with acute myocardial infarction remained unexplored. In this cohort of patients with a first acute anterior ST-segment elevation myocardial infarction receiving contemporary management, bilirubin levels on admission were not predictive of the changes in left ventricular volumes or ejection fraction at 6 months measured by serial cardiac magnetic resonance imaging. The data are contrary to a significant protective effect of bilirubin against post-infarction ventricular remodeling.
Collapse
Affiliation(s)
- Berta Miranda
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - José A Barrabés
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Jaume Figueras
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Victor Pineda
- b Servicio de Radiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - José Rodríguez-Palomares
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Rosa-Maria Lidón
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Antonia Sambola
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Jordi Bañeras
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - Imanol Otaegui
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| | - David García-Dorado
- a Servicio de Cardiología, Hospital Universitari Vall d'Hebron, VHIR, Universitat Autònoma de Barcelona , Barcelona , Spain
| |
Collapse
|
16
|
Abraham NG, Junge JM, Drummond GS. Translational Significance of Heme Oxygenase in Obesity and Metabolic Syndrome. Trends Pharmacol Sci 2015; 37:17-36. [PMID: 26515032 DOI: 10.1016/j.tips.2015.09.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/10/2015] [Accepted: 09/17/2015] [Indexed: 01/04/2023]
Abstract
The global epidemic of obesity continues unabated with sequelae of diabetes and metabolic syndrome. This review reflects the dramatic increase in research on the role of increased expression of heme oxygenase (HO)-1/HO-2, biliverdin reductase, and HO activity on vascular disease. The HO system engages with other systems to mitigate the deleterious effects of oxidative stress in obesity and cardiovascular disease (CVD). Recent reports indicate that HO-1/HO-2 protein expression and HO activity have several important roles in hemostasis and reactive oxygen species (ROS)-dependent perturbations associated with metabolic syndrome. HO-1 protects tissue during inflammatory stress in obesity through the degradation of pro-oxidant heme and the production of carbon monoxide (CO) and bilirubin, both of which have anti-inflammatory and anti-apoptotic properties. By contrast, repression of HO-1 is associated with increases of cellular heme and inflammatory conditions including hypertension, stroke, and atherosclerosis. HO-1 is a major focus in the development of potential therapeutic strategies to reverse the clinical complications of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA; Marshall University, Joan C. Edwards School of Medicine, Huntington, WV 25701, USA.
| | - Joshua M Junge
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA
| | - George S Drummond
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA
| |
Collapse
|
17
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
18
|
Czibik G, Derumeaux G, Sawaki D, Valen G, Motterlini R. Heme oxygenase-1: an emerging therapeutic target to curb cardiac pathology. Basic Res Cardiol 2014; 109:450. [PMID: 25344086 DOI: 10.1007/s00395-014-0450-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/05/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022]
Abstract
Activation of heme oxygenase-1 (HO-1), a heme-degrading enzyme responsive to a wide range of cellular stress, is traditionally considered to convey adaptive responses to oxidative stress, inflammation and vasoconstriction. These diversified effects are achieved through the degradation of heme to carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin by biliverdin reductase) and ferric iron. Recent findings have added antiproliferative and angiogenic effects to the list of HO-1/CO actions. HO-1 along with its reaction products bilirubin and CO are protective against ischemia-induced injury (myocardial infarction, ischemia-reperfusion (IR)-injury and post-infarct structural remodelling). Moreover, HO-1, and CO in particular, possess acute antihypertensive effects. As opposed to these curative potentials, the long-believed protective effect of HO-1 in cardiac remodelling in response to pressure overload and type 2 diabetes mellitus (DM) has been questioned by recent work. These challenges, coupled with emerging regulatory mechanisms, motivate further in-depth studies to help understand untapped layers of HO-1 regulation and action. The outcomes of these efforts may shed new light on critical mechanisms that could be used to harness the protective potential of this enzyme for the therapeutic benefit of patients suffering from such highly prevalent cardiovascular disorders.
Collapse
Affiliation(s)
- Gabor Czibik
- INSERM U955, Equipe 8, Faculty of Medicine, DHU A-TVB, Hôpital Henri Mondor, APHP, Creteil, University of Paris-Est, 3rd Floor, room 3006, Paris, France,
| | | | | | | | | |
Collapse
|
19
|
Zhou C, Li H, Yao Y, Li L. Delayed remote ischemic preconditioning produces an additive cardioprotection to sevoflurane postconditioning through an enhanced heme oxygenase 1 level partly via nuclear factor erythroid 2-related factor 2 nuclear translocation. J Cardiovasc Pharmacol Ther 2014; 19:558-66. [PMID: 24651515 DOI: 10.1177/1074248414524479] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although both sevoflurane postconditioning (SPoC) and delayed remote ischemic preconditioning (DRIPC) have been proved effective in various animal and human studies, the combined effect of these 2 strategies remains unclear. Therefore, this study was designed to investigate this effect and elucidate the related signal mechanisms in a Langendorff perfused rat heart model. After 30-minute balanced perfusion, isolated hearts were subjected to 30-minute ischemia followed by 60-minute reperfusion except 90-minute perfusion for control. A synergic cardioprotective effect of SPoC (3% v/v) and DRIPC (4 cycles 5-minute occlusion/5-minute reflow at the unilateral hindlimb once per day for 3 days before heart isolation) was observed with facilitated cardiac functional recovery and decreased cardiac enzyme release. The infarct size-limiting effect was more pronounced in the combined group (6.76% ± 2.18%) than in the SPoC group (16.50% ± 4.55%, P < .001) or in the DRIPC group (10.22% ± 2.57%, P = .047). Subsequent analysis revealed that an enhanced heme oxygenase 1 (HO-1) expression, but not protein kinase B/AKt or extracellular signal-regulated kinase 1 and 2 activation, was involved in the synergic cardioprotective effect, which was further confirmed in the messenger RNA level of HO-1. Such trend was also observed in the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, an upstream regulation of HO-1. In addition, correlation analysis showed a significantly positive relationship between HO-1 expression and Nrf2 translocation (r = 0.729, P < .001). Hence, we conclude that DRIPC may produce an additive cardioprotection to SPoC through an enhanced HO-1 expression partly via Nrf2 translocation.
Collapse
Affiliation(s)
- Chenghui Zhou
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huatong Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuntai Yao
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihuan Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Mahabadi AA, Lehmann N, Möhlenkamp S, Kälsch H, Bauer M, Schulz R, Moebus S, Jöckel KH, Erbel R, Heusch G. Association of bilirubin with coronary artery calcification and cardiovascular events in the general population without known liver disease: the Heinz Nixdorf Recall study. Clin Res Cardiol 2014; 103:647-53. [PMID: 24633678 DOI: 10.1007/s00392-014-0697-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/27/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Bilirubin is a product of heme oxygenase and an antioxidant per se. In selected cohorts and patient studies, bilirubin was associated with decreased risk for cardiovascular events. We aimed to determine the association of serum bilirubin with coronary artery calcification (CAC) and incident cardiovascular disease in the unselected general population. METHODS Participants without prior coronary heart or liver disease were drawn from the population-based Heinz Nixdorf Recall study. CAC was measured from electron beam computed tomography and quantified using the Agatston method. Incident major cardiovascular events (coronary events, stroke, cardiovascular death) were assessed during follow-up. The association of bilirubin with CAC score and incident events was calculated using regression analysis. RESULTS A total of 3,553 subjects (mean age 59.4 years, 44 % male) were included. Bilirubin was inversely correlated with CAC in men [estimated % change in (CAC + 1) per each gender-specific standard deviation of bilirubin [95 % confidence interval (CI): -6.1 (-11.6; -1.7) %, p = 0.032] but less so in women [-3.6 (-7.7; 0.2) %, p = 0.065], when adjusting for age only. With adjustment for traditional cardiovascular risk factors, the association was less pronounced [men: -2.5 (-7.5; 2.6) %, p = 0.35, women: -0.2 (-4.1; 3.5) %, p = 0.9]. Likewise, higher bilirubin by one standard deviation was associated with 19 % lower frequency of incident cardiovascular events in age- and gender-adjusted Cox regression analysis [hazard ratio (95 % CI): 0.81 (0.68; 0.96), p = 0.014]; this effect was slightly attenuated after adjustment for traditional cardiovascular risk factors [0.87 (0.73; 1.04), p = 0.13]. CONCLUSION Our data from a general population suggest that a potential protective role of bilirubin in the atherosclerosis process is largely secondary to a more beneficial risk factor profile.
Collapse
Affiliation(s)
- Amir A Mahabadi
- Department of Cardiology, West German Heart Center, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Tongguan Capsule Protects against Myocardial Ischemia and Reperfusion Injury in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:159237. [PMID: 24073004 PMCID: PMC3774060 DOI: 10.1155/2013/159237] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/19/2013] [Accepted: 08/01/2013] [Indexed: 12/28/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) can induce lethal ventricular arrhythmia and myocardial infarction. One of the clinical strategies for managing patients with high risk of myocardial I/R is to prevent the occurrence of arrhythmias and limit the size of infarction following a coronary episode. Tongguan Capsule (TGC) is one of the popular herbal remedies in treating coronary artery disease in the clinics of Chinese medicine. However, the potential roles and mechanisms of TGC in reducing I/R injury are still unclear. The present study statistically assessed the effectiveness of TGC in reducing I/R injury by comparing the infarct size (IS), risk region (RR), and arrhythmia (in electrocardiogram) among four groups of surgically created mice models of myocardial I/R: SHAM, I/R, VER (I/R with verapamil 20 mg/kg pretreatment), and TGC (I/R with TGC 5 g/kg/d pretreatment). We found that IS was significantly smaller in the TGC and VER groups than I/R group, and the incidence of arrhythmias was reduced in the TGC group compared with I/R group, although there were no differences in RR among the four groups. We conclude that TGC is effective in reducing I/R injury in mice. These results provided an experimental basis for clinical application of TGC in reducing I/R injury.
Collapse
|
22
|
Therapeutic approaches to limit hemolysis-driven endothelial dysfunction: scavenging free heme to preserve vasculature homeostasis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:396527. [PMID: 23781294 PMCID: PMC3678425 DOI: 10.1155/2013/396527] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/29/2013] [Accepted: 05/14/2013] [Indexed: 12/11/2022]
Abstract
Hemolysis results in the release of hemoglobin and heme into the bloodstream and is associated with the development of several pathologic conditions of different etiology, including hemoglobinopathies, hemolytic anemias, bacterial infections, malaria, and trauma. In addition, hemolysis is associated with surgical procedures, hemodialysis, blood transfusion, and other conditions in which mechanical forces can lead to red blood cell rupture. Free plasma hemoglobin and heme are toxic for the vascular endothelium since heme iron promotes oxidative stress that causes endothelial activation responsible for vasoocclusive events and thrombus formation. Moreover, free hemoglobin scavenges nitric oxide, reducing its bioavailability, and heme favours ROS production, thus causing oxidative nitric oxide consumption. This results in the dysregulation of the endothelium vasodilator:vasoconstrictor balance, leading to severe vasoconstriction and hypertension. Thus, endothelial dysfunction and impairment of cardiovascular function represent a common feature of pathologic conditions associated with hemolysis. In this review, we discuss how hemoglobin/heme released following hemolysis may affect vascular function and summarise the therapeutic approaches available to limit hemolysis-driven endothelial dysfunction. Particular emphasis is put on recent data showing the beneficial effects obtained through the use of the plasma heme scavenger hemopexin in counteracting heme-mediated endothelial damage in mouse models of hemolytic diseases.
Collapse
|
23
|
Current world literature. Curr Opin Pediatr 2012; 24:770-9. [PMID: 23146873 DOI: 10.1097/mop.0b013e32835af8de] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Gene transfer of heme oxygenase-1 using an adeno-associated virus serotype 6 vector prolongs cardiac allograft survival. J Transplant 2012; 2012:740653. [PMID: 23125915 PMCID: PMC3480008 DOI: 10.1155/2012/740653] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 09/07/2012] [Accepted: 09/07/2012] [Indexed: 11/18/2022] Open
Abstract
Introduction. Allograft survival can be prolonged by overexpression of cytoprotective genes such as heme oxygenase-1 (HO-1). Modifications in vector design and delivery have provided new opportunities to safely and effectively administer HO-1 into the heart prior to transplantation to improve long-term graft outcome. Methods. HO-1 was delivered to the donor heart using an adeno-associated virus vector (AAV) with a pseudotype 6 capsid and vascular endothelial growth factor (VEGF) to enhance myocardial tropism and microvascular permeability. Survival of mouse cardiac allografts, fully or partially mismatched at the MHC, was determined with and without cyclosporine A. Intragraft cytokine gene expression was examined by PCR.
Results. The use of AAV6 to deliver HO-1 to the donor heart, combined with immunosuppression, prolonged allograft survival by 55.3% when donor and recipient were completely mismatched at the MHC and by 94.6% if partially mismatched. The combination of gene therapy and immunosuppression was more beneficial than treatment with either AAV6-HO-1 or CsA alone. IL-17a, b, e and f were induced in the heart at rejection.
Conclusions. Pretreatment of cardiac allografts with AAV6-HO-1 plus cyclosporine A prolonged graft survival. HO-1 gene therapy represents a beneficial adjunct to immunosuppressive therapy in cardiac transplantation.
Collapse
|
25
|
Mahan VL, Zurakowski D, Otterbein LE, Pigula FA. Inhaled carbon monoxide provides cerebral cytoprotection in pigs. PLoS One 2012; 7:e41982. [PMID: 22879904 PMCID: PMC3413707 DOI: 10.1371/journal.pone.0041982] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/27/2012] [Indexed: 11/18/2022] Open
Abstract
Carbon monoxide (CO) at low concentrations imparts protective effects in numerous preclinical small animal models of brain injury. Evidence of protection in large animal models of cerebral injury, however, has not been tested. Neurologic deficits following open heart surgery are likely related in part to ischemia reperfusion injury that occurs during cardiopulmonary bypass surgery. Using a model of deep hypothermic circulatory arrest (DHCA) in piglets, we evaluated the effects of CO to reduce cerebral injury. DHCA and cardiopulmonary bypass (CPB) induced significant alterations in metabolic demands, including a decrease in the oxygen/glucose index (OGI), an increase in lactate/glucose index (LGI) and a rise in cerebral blood pressure that ultimately resulted in increased cell death in the neocortex and hippocampus that was completely abrogated in piglets preconditioned with a low, safe dose of CO. Moreover CO-treated animals maintained normal, pre-CPB OGI and LGI and corresponding cerebral sinus pressures with no change in systemic hemodynamics or metabolic intermediates. Collectively, our data demonstrate that inhaled CO may be beneficial in preventing cerebral injury resulting from DHCA and offer important therapeutic options in newborns undergoing DHCA for open heart surgery.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.
| | | | | | | |
Collapse
|
26
|
Wojakowski W, Tendera M, Cybulski W, Zuba-Surma EK, Szade K, Florczyk U, Kozakowska M, Szymula A, Krzych L, Paslawska U, Paslawski R, Milewski K, Buszman PP, Nabialek E, Kuczmik W, Janiszewski A, Dziegiel P, Buszman PE, Józkowicz A, Dulak J. Effects of intracoronary delivery of allogenic bone marrow-derived stem cells expressing heme oxygenase-1 on myocardial reperfusion injury. Thromb Haemost 2012; 108:464-75. [PMID: 22872040 DOI: 10.1160/th12-05-0303] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/10/2012] [Indexed: 12/22/2022]
Abstract
Heme oxygenase-1 (HO-1) decreases apoptosis, inflammation and oxidative stress. The aim of the study was to investigate the effects of intracoronary infusion of allogenic bone marrow cells (BMC) overexpressing HO-1 in the porcine model of myocardial infarction (MI). MI was produced by balloon occlusion of a coronary artery. BMC were transduced with adenoviruses encoding for HO-1 (HO-1 BMC) or GFP (GFP-BMC) genes. Prior to reperfusion animals received HO-1 BMC, control BMC (unmodified or GFP-BMC) or placebo. Left ventricular (LV) ejection fraction (EF), shortening fraction (SF), end-systolic and end-diastolic diameters (EDD, ESD) were assessed by echocardiography before, 30 minutes (min) and 14 days after reperfusion. BMC significantly improved LVEF and SF early (30 min) after reperfusion as well as after 14 days. Early after reperfusion HO-1 BMC were significantly more effective than control BMC, but after 14 days, there were no differences. There were no effect of cells on LV remodelling and diastolic function. Both HO-1 BMC and control BMC significantly reduced the infarct size vs. placebo (17.2 ± 2.7 and 18.8 ± 2.5, respectively, vs. 27.5 ± 5.1, p= 0.02) in histomorphometry. HO-1-positive donor BMC were detected in the infarct border area in pigs receiving HO-1-cells. No significant differences in expression of inflammatory genes (SDF-1, TNF-α, IL-6, miR21, miR29a and miR133a) in the myocardium were found. In conclusion, intracoronary delivery of allogeneic BMC immediately prior to reperfusion improved the LVEF and reduced the infarct size. HO-1 BMC were not superior to control cells after 14 days, however, produced faster recovery of LVEF. Transplanted cells survived in the peri-infarct zone.
Collapse
Affiliation(s)
- Wojciech Wojakowski
- Third Division of Cardiology, Medical University of Silesia, Katowice, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Guo Y, Flaherty MP, Wu WJ, Tan W, Zhu X, Li Q, Bolli R. Genetic background, gender, age, body temperature, and arterial blood pH have a major impact on myocardial infarct size in the mouse and need to be carefully measured and/or taken into account: results of a comprehensive analysis of determinants of infarct size in 1,074 mice. Basic Res Cardiol 2012; 107:288. [PMID: 22864681 DOI: 10.1007/s00395-012-0288-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 12/11/2022]
Abstract
In order to determine whether the myocardial response to ischemia/reperfusion (I/R) injury varies depending on genetic background, gender, age, body temperature, and arterial blood pH, we studied 1,074 mice from 19 strains (including 129S6/SvEvTac (129S6), B6/129P2-Ptgs2(tm1Unc), B6/129SvF(2)/J, B6/129/D2, B6/CBAF1, B6/DBA/1JNcr, BALB/c, BPH2/J, C57BL/6/J (B6/J), C3H/DBA, C3H/FB/FF, C3H/HeJ-Pde6b(rd1), FVB/N/J [FVB/N], FVB/B6, FVB/ICR and Crl:ICR/H [ICR]) and distributed them into 69 groups depending on strain and: (1) two phases of ischemic preconditioning (PC); (2) coronary artery occlusion (O) time; (3) gender; (4) age; (5) blood transfusion; (6) core body temperature; and (7) arterial blood pH. Mice underwent O either without (non-preconditioned [naive]) or with prior cyclic O/reperfusion (R) (PC stimulus) consisting of six 4-min O/4-min R cycles 10 min (early PC, EPC) or 24 h (late PC, LPC) prior to 30 or 45-min O and 24 h R. In B6/J and B6/129/D2 mice, almost the entire risk region was infarcted after a 60-min O. Of the naive mouse hearts, B6/ecSOD(WT) and FVB/N mice had infarct sizes significantly smaller than those of the other mice. All strains except FVB/N benefited from the cardioprotection afforded by the early phase of PC; in contrast, development of LPC was inconsistent amongst groups and was strain-dependent. Female gender (1) was associated with reduced infarct size in ICR mice, (2) determined whether LPC developed in ICR mice, and (3) limited the protection afforded by EPC in 129S6 mice. Importantly, mild hypothermia (1 °C decrease in core temperature) and mild acidosis (0.18 decrease in blood pH) resulted in a striking cardioprotective effect in ICR mice: 67.5 and 43.0 % decrease in infarct size, respectively. Replacing blood losses with crystalloid fluids (instead of blood) during surgery also reduced infarct size. To our knowledge, this is the largest analysis of the determinants of infarct size in mice ever published. The results demonstrate that genetic background, gender, age (but not in ICR), body temperature and arterial blood pH have a major impact on infarct size, and thus need to be carefully measured and/or taken into account when designing a study of myocardial infarction in mice; failure to do so makes results uninterpretable. For example, core temperature and blood pH need to be measured, respiratory acidosis (or alkalosis) and hypothermia (or hyperthermia) must be avoided, and comparisons cannot be made between mouse strains or genders that exhibit different susceptibility to I/R injury (e.g., FVB/N male mice and ICR female mice are inherently protected against I/R injury).
Collapse
Affiliation(s)
- Yiru Guo
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, 550 S. Jackson St., Louisville, KY 40292, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Guo Y, Tukaye DN, Wu WJ, Zhu X, Book M, Tan W, Jones SP, Rokosh G, Narumiya S, Li Q, Bolli R. The COX-2/PGI2 receptor axis plays an obligatory role in mediating the cardioprotection conferred by the late phase of ischemic preconditioning. PLoS One 2012; 7:e41178. [PMID: 22844439 PMCID: PMC3402528 DOI: 10.1371/journal.pone.0041178] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/18/2012] [Indexed: 01/15/2023] Open
Abstract
Background Pharmacologic studies with cyclooxygenase-2 (COX-2) inhibitors suggest that the late phase of ischemic preconditioning (PC) is mediated by COX-2. However, nonspecific effects of COX-2 inhibitors cannot be ruled out, and the selectivity of these inhibitors for COX-2 vs. COX-1 is only relative. Furthermore, the specific prostaglandin (PG) receptors responsible for the salubrious actions of COX-2-derived prostanoids remain unclear. Objective To determine the role of COX-2 and prostacyclin receptor (IP) in late PC by gene deletion. Methods COX-2 knockout (KO) mice (COX-2−/−), prostacyclin receptor KO (IP−/−) mice, and respective wildtype (WT, COX-2+/+ and IP+/+) mice underwent sham surgery or PC with six 4-min coronary occlusion (O)/4-min R cycles 24 h before a 30-min O/24 h R. Results There were no significant differences in infarct size (IS) between non-preconditioned (non-PC) COX-2+/+, COX-2−/−, IP+/+, and IP−/− mice, indicating that neither COX-2 nor IP modulates IS in the absence of PC. When COX-2−/− or IP−/− mice were preconditioned, IS was not reduced, indicating that the protection of late PC was completely abrogated by deletion of either the COX-2 or the IP gene. Administration of the IP selective antagonist, RO3244794 to C57BL6/J (B6) mice 30 min prior to the 30-min O had no effect on IS. When B6 mice were preconditioned 24 h prior to the 30-min O, IS was markedly reduced; however, the protection of late PC was completely abrogated by pretreatment of RO3244794. Conclusions This is the first study to demonstrate that targeted disruption of the COX-2 gene completely abrogates the infarct-sparing effect of late PC, and that the IP, downstream of the COX-2/prostanoid pathway, is a key mediator of the late PC. These results provide unequivocal molecular genetic evidence for an essential role of the COX-2/PGI2 receptor axis in the cardioprotection afforded by the late PC.
Collapse
Affiliation(s)
- Yiru Guo
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Deepali Nivas Tukaye
- Department of Internal Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Xiaoping Zhu
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Michael Book
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Wei Tan
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Steven P. Jones
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Gregg Rokosh
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Shuh Narumiya
- Department of Pharmacology, Kyoto University Faculty of Medicine, Kyoto, Japan
| | - Qianhong Li
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
29
|
Gene transfer as a strategy to achieve permanent cardioprotection I: rAAV-mediated gene therapy with inducible nitric oxide synthase limits infarct size 1 year later without adverse functional consequences. Basic Res Cardiol 2011; 106:1355-66. [PMID: 21779912 DOI: 10.1007/s00395-011-0207-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/04/2011] [Accepted: 07/08/2011] [Indexed: 12/22/2022]
Abstract
The ultimate goal of prophylactic gene therapy is to confer permanent protection against ischemia. Although gene therapy with inducible nitric oxide synthase (iNOS) is known to protect against myocardial infarction at 3 days and up to 2 months, the long-term effects on myocardial ischemic injury and function are unknown. To address this issue, we created a recombinant adeno-associated viral vector carrying the iNOS gene (rAAV/iNOS), which enables long-lasting transgene expression. The ability of rAAV/iNOS to direct the expression of functional iNOS protein was confirmed in COS-7 cells before in vivo gene transfer. Mice received injections in the anterior LV wall of rAAV/LacZ or rAAV/iNOS; 1 year later, they underwent a 30-min coronary occlusion (O) and 4 h of reperfusion (R). iNOS gene transfer resulted in elevated iNOS protein expression (+3-fold vs. the LacZ group, n = 6; P < 0.05) and iNOS activity (+4.4-fold vs. the LacZ group, n = 6; P < 0.05) 1 year later. Infarct size (% of risk region) was dramatically reduced at 1 year after iNOS gene transfer (13.5 ± 2.2%, n = 12, vs. 41.7 ± 2.9%, n = 10, in the LacZ group; P < 0.05). The infarct-sparing effect of iNOS gene therapy at 1 year was as powerful as that observed 24 h after ischemic preconditioning (six 4-min O/4-min R cycles) (19.3 ± 2.3%, n = 11; P < 0.05). Importantly, compared with the LacZ group (n = 11), iNOS gene transfer (n = 10) had no effect on LV dimensions or function for up to 1 year (at 1 year: FS 34.5 ± 2.0 vs. 34.6 ± 2.6%, EF 57.0 ± 2.0 vs. 59.7 ± 2.9%, LVEDD 4.3 ± 0.1 vs. 4.2 ± 0.2 mm, LVESD 2.8 ± 0.1 vs. 2.9 ± 0.2 mm) (echocardiography). These data demonstrate, for the first time, that rAAV-mediated iNOS gene transfer affords long-term, probably permanent (1 year), cardioprotection without adverse functional consequences, providing a strong rationale for further preclinical testing of prophylactic gene therapy.
Collapse
|