1
|
Kamenshchikov NO, Duong N, Berra L. Nitric Oxide in Cardiac Surgery: A Review Article. Biomedicines 2023; 11:1085. [PMID: 37189703 PMCID: PMC10135597 DOI: 10.3390/biomedicines11041085] [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: 01/24/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
Perioperative organ injury remains a medical, social and economic problem in cardiac surgery. Patients with postoperative organ dysfunction have increases in morbidity, length of stay, long-term mortality, treatment costs and rehabilitation time. Currently, there are no pharmaceutical technologies or non-pharmacological interventions that can mitigate the continuum of multiple organ dysfunction and improve the outcomes of cardiac surgery. It is essential to identify agents that trigger or mediate an organ-protective phenotype during cardiac surgery. The authors highlight nitric oxide (NO) ability to act as an agent for perioperative protection of organs and tissues, especially in the heart-kidney axis. NO has been delivered in clinical practice at an acceptable cost, and the side effects of its use are known, predictable, reversible and relatively rare. This review presents basic data, physiological research and literature on the clinical application of NO in cardiac surgery. Results support the use of NO as a safe and promising approach in perioperative patient management. Further clinical research is required to define the role of NO as an adjunct therapy that can improve outcomes in cardiac surgery. Clinicians also have to identify cohorts of responders for perioperative NO therapy and the optimal modes for this technology.
Collapse
Affiliation(s)
- Nikolay O. Kamenshchikov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Nicolette Duong
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Respiratory Care Service, Patient Care Services, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lorenzo Berra
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Respiratory Care Service, Patient Care Services, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
2
|
Orhan E, Gündüz Ö, Kaya O, Öznur M, Şahin E. Transferring the protective effect of remote ischemic preconditioning on skin flap among rats by blood serum. J Plast Surg Hand Surg 2019; 53:198-203. [DOI: 10.1080/2000656x.2019.1582422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Erkan Orhan
- Department of Plastic Surgery, School of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Özgür Gündüz
- Department of Medical Pharmacology, School of Medicine, Trakya University, Edirne, Turkey
| | - Oktay Kaya
- Department of Physiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Meltem Öznur
- Department of Pathology, School of Medicine, Namik Kemal University, Tekirdağ, Turkey
| | - Ertan Şahin
- Department of Nuclear Medicine, School of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
3
|
Remote Postconditioning Induced by Trauma Protects the Mouse Heart against Ischemia Reperfusion Injury. Involvement of the Neural Pathway and Molecular Mechanisms. Cardiovasc Drugs Ther 2017; 30:271-80. [PMID: 27067902 DOI: 10.1007/s10557-016-6661-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE Abdominal superficial surgical incision elicits cardioprotection against myocardial ischemia reperfusion (I/R) injury in mice. This cardioprotective phenomenon, termed remote preconditioning of trauma (RPCT), results in an 80 to 85 % reduction in cardiac infarct size. We evaluated cardioprotection and the molecular mechanisms of remote postconditioning of trauma (RPostCT) in a murine I/R injury model. METHODS Mice were analyzed using a previously established I/R injury model. An abdominal superficial surgical incision was made 45 min after myocardial ischemia at the end of coronary occlusion, and infarct size was determined 24 h after reperfusion. RESULTS The results indicated that a strong cardioprotective effect occurred during RPostCT (56.94 ± 2.71 % sham vs. 15.58 ± 2.16 % RPostCT; the mean area of the infarct divided by the mean area of the region at risk; p ≤ 0.05; n = 10). Furthermore, pharmacological intervention revealed neurogenic signaling involvement in the beneficial effects of RPostCT via sensory and sympathetic thoracic nerves. Pharmacological experiments in transgenic mice demonstrated that bradykinin receptors, β-adrenergic receptors (AR), and protein kinase C were implicated in the cardioprotective effects of RPostCT. CONCLUSIONS RPostCT significantly decreased myocardial infarction size via neurogenic transmission and various signaling pathways. This study describes a new cardiac I/R injury prevention method that might lead to the development of therapies that are more clinically relevant for myocardial I/R injury.
Collapse
|
4
|
Song Y, Ye YJ, Li PW, Zhao YL, Miao Q, Hou DY, Ren XP. The Cardioprotective Effects of Late-Phase Remote Preconditioning of Trauma Depends on Neurogenic Pathways and the Activation of PKC and NF-κB (But Not iNOS) in Mice. J Cardiovasc Pharmacol Ther 2015; 21:310-9. [PMID: 26450997 DOI: 10.1177/1074248415609435] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/24/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND A superficial abdominal surgical incision elicits cardioprotection against cardiac ischemia-reperfusion (I/R) injury in mice. This process, called remote preconditioning of trauma (RPCT), has both an early and a late phase. Previous investigations have demonstrated that early RPCT reduces cardiac infarct size by 80% to 85%. We evaluated the cardioprotective and molecular mechanisms of late-phase RPCT in a murine I/R injury model. METHODS Wild-type mice, bradykinin (BK) 2 receptor knockout mice, 3M transgenic mice (nuclear factor κB [NF-κb] repressor inhibitor of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha [IκBα((S32A, S36A, Y42F))]), and inducible nitric oxide synthase (iNOS) knockout mice were analyzed using a previously established I/R injury model. A noninvasive abdominal surgical incision was made 24 hours prior to I/R injury and the infarct size was determined at 24 hours post-I/R injury. RESULTS The results indicated that a strong cardioprotective effect occurred during late-phase RPCT (58.42% ± 1.89% sham vs 29.41% ± 4.00% late RPCT, mean area of the infarct divided by the mean area of the risk region; P ≤ .05; n = 10). Furthermore, pharmacological intervention revealed the involvement of neurogenic signaling in the beneficial effects of late RPCT via sensory and sympathetic thoracic nerves. Pharmacological experiments in transgenic mice-implicated BK receptors, β-adrenergic receptors, protein kinase C, and NF-κB but not iNOS signaling in the cardioprotective effects of late RPCT. CONCLUSION Late RPCT significantly decreased myocardial infarct size via neurogenic transmission and various other signaling pathways. This protective mechanism differentiates late and early RPCT. This study describes a new cardiac I/R injury prevention method and refines the concept of RPCT.
Collapse
Affiliation(s)
- Y Song
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Y J Ye
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - P W Li
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Y L Zhao
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Q Miao
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - D Y Hou
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - X P Ren
- Hand and Microsurgery Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China State-Province Key Laboratories of Biomedicine-Pharmaceutics, Harbin Medical University, Harbin, China Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine in Loyola University, Chicago, IL, USA
| |
Collapse
|
5
|
Das A, Durrant D, Salloum FN, Xi L, Kukreja RC. PDE5 inhibitors as therapeutics for heart disease, diabetes and cancer. Pharmacol Ther 2014; 147:12-21. [PMID: 25444755 DOI: 10.1016/j.pharmthera.2014.10.003] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
Abstract
The phosphodiesterase 5 (PDE5) inhibitors, including sildenafil (Viagra™), vardenafil (Levitra™), and tadalafil (Cialis™) have been developed for treatment of erectile dysfunction. Moreover, sildenafil and tadalafil are used for the management of pulmonary arterial hypertension in patients. Since our first report showing the cardioprotective effect of sildenafil in 2002, there has been tremendous growth of preclinical and clinical studies on the use of PDE5 inhibitors for cardiovascular diseases and cancer. Numerous animal studies have demonstrated that PDE5 inhibitors have powerful protective effect against myocardial ischemia/reperfusion (I/R) injury, doxorubicin cardiotoxicity, ischemic and diabetic cardiomyopathy, cardiac hypertrophy, Duchenne muscular dystrophy and the improvement of stem cell efficacy for myocardial repair. Mechanistically, PDE5 inhibitors protect the heart against I/R injury through increased expression of nitric oxide synthases, activation of protein kinase G (PKG), PKG-dependent hydrogen sulfide generation, and phosphorylation of glycogen synthase kinase-3β - a master switch immediately proximal to mitochondrial permeability transition pore and the end effector of cardioprotection. In addition, PDE5 inhibitors enhance the sensitivity of certain types of cancer to standard chemotherapeutic drugs, including doxorubicin. Many clinical trials with PDE5 inhibitors have focused on the potential cardiovascular and anti-cancer benefits. Despite mixed results of these clinical trials, there is a continuing strong interest by basic scientists and clinical investigators in exploring their new clinical uses. It is our hope that future new mechanistic investigations and carefully designed clinical trials would help in reaping additional benefits of PDE5 inhibitors for cardiovascular disease and cancer in patients.
Collapse
Affiliation(s)
- Anindita Das
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - David Durrant
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Fadi N Salloum
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Rakesh C Kukreja
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|
6
|
Krenz M, Baines C, Kalogeris T, Korthuis R. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardioprotection. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00090ed1v01y201309isp044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
7
|
Dacho AK, Dietz A, Mueller K. Histological effect on the adipocutaneous flap in rats after preconditioning with 2-chloro-N(6) -cyclopentyladenosine. Head Neck 2013; 36:1189-99. [PMID: 23893540 DOI: 10.1002/hed.23433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 03/03/2013] [Accepted: 07/04/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND 2-chloro-N(6) -cyclopentyladenosine (CCPA) was proven to be a protective factor in ischemic reperfusion injury. The purpose of this study was to determine how CCPA would affect the single tissue layers of the adipocutaneous flap. METHODS Seventy male Wistar rats were divided into 5 experimental groups. Samples were taken of the area of flap necrosis and the wound margin after classical or pharmacological preconditioning on the fifth postoperative day. All samples were fixed in formaldehyde, embedded in paraplast, and analyzed in 3- to 4-μm sections (hemalaun-eosin stain and light microscopy). RESULTS In general, wound healing was alike and remained unaffected by the experimental design. The most sensitive part of the flap during preconditioning is the subcutis. The number of neutrophils and of plasma cells is reduced significantly (p < .05). CONCLUSION CCPA has an effect on each tissue layer of the flap. Subcutis became apparent as the most sensitive layer. CCPA influences complement pathway and neutrophils directly and indirectly.
Collapse
Affiliation(s)
- Andreas K Dacho
- Department of Plastic Surgery, St. Josef Medical Center, Essen, Germany
| | | | | |
Collapse
|
8
|
Comparison of various methods of vessel ligation: what is the safest method? Surg Endosc 2013; 27:3129-38. [DOI: 10.1007/s00464-013-2866-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 02/06/2013] [Indexed: 10/27/2022]
|
9
|
Time course analysis of cardiac pacing-induced gene expression changes in the canine heart. Mol Cell Biochem 2012; 372:257-66. [PMID: 23014934 DOI: 10.1007/s11010-012-1467-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
Abstract
Rapid right ventricular pacing in anesthetized dogs results in marked protection against ischemia and reperfusion-induced ventricular arrhythmias, 24 h later. We have previous evidence that this protection associates with altered expression of genes, encoding proteins involved in the delayed cardioprotection. However, the sequence of transcriptional changes occurring between the pacing stimulus and the test ischemia has not yet been elucidated. Thus, we designed studies in which the expression of 29 genes was examined by real-time PCR at various time intervals, i.e., immediately (0 h), 6, 12, and 24 h after short periods (4 times 5 min) of rapid (240 beats min(-1)) right ventricular pacing in the canine. Sham-operated dogs (the pacing electrode was introduced but the dogs were not paced) served as controls. Compared with these dogs, pacing induced an early up-regulation of genes which encode, for example, HSP90, MnSOD, ERK1, PKCε, Bcl2, and sGC; all these somehow relate to the early phase of the protection. These genes remained either up-regulated or, after a transient lower expression (around 6 h), were up-regulated again, suggesting their involvement in the delayed protection. There were also some genes which down-regulated soon after the pacing stimulus (e.g., Bax, Casp3, Casp9, MMP9, GSK3β), and showed also low expression 24 h later. Genes encoding eNOS and iNOS, as well as Cx43 were only up-regulated 12 h after pacing. We conclude that cardiac pacing induces time-dependent changes in gene expression, and the sequence of these changes is important in the development of the delayed protection.
Collapse
|
10
|
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: 3.4] [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
|
11
|
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: 2.2] [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
|
12
|
Murillo D, Kamga C, Mo L, Shiva S. Nitrite as a mediator of ischemic preconditioning and cytoprotection. Nitric Oxide 2011; 25:70-80. [PMID: 21277988 DOI: 10.1016/j.niox.2011.01.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 01/04/2011] [Accepted: 01/13/2011] [Indexed: 10/24/2022]
Abstract
Ischemia/reperfusion (IR) injury is a central component in the pathogenesis of several diseases and is a leading cause of morbidity and mortality in the western world. Subcellularly, mitochondrial dysfunction, characterized by depletion of ATP, calcium-induced opening of the mitochondrial permeability transition pore, and exacerbated reactive oxygen species (ROS) formation, plays an integral role in the progression of IR injury. Nitric oxide (NO) and more recently nitrite (NO(2)(-)) are known to modulate mitochondrial function, mediate cytoprotection after IR and have been implicated in the signaling of the highly protective ischemic preconditioning (IPC) program. Here, we review what is known about the role of NO and nitrite in cytoprotection after IR and consider the putative role of nitrite in IPC. Focus is placed on the potential cytoprotective mechanisms involving NO and nitrite-dependent modulation of mitochondrial function.
Collapse
Affiliation(s)
- Daniel Murillo
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
13
|
Abstract
A standard ischemic preconditioning (IPC) stimulus of one or more brief episodes of non-lethal myocardial ischemia and reperfusion elicits a bi-phasic pattern of cardioprotection. The first phase manifests almost immediately following the IPC stimulus and lasts for 1-2 h, after which its effect disappears (termed classical or early IPC). The second phase of cardioprotection appears 12-24 h later and lasts for 48-72 h (termed the Second Window of Protection [SWOP] or delayed or late IPC). The cardioprotection conferred by delayed IPC is robust and ubiquitous but is not as powerful as early IPC. Although there are some similarities in the mechanisms underlying early and delayed IPC, one of the major distinctions between the two is the latter's requirement for de novo protein synthesis of distal mediators such as iNOS and COX-2 which mediate the cardioprotection 24 h after the IPC stimulus. The phenomenon of delayed IPC has been demonstrated in man using a variety of experimental models. However, its clinical application has been limited by the same factors which affect early IPC- i.e. the need to intervene before the onset of myocardial ischemia, thereby restricting its potential clinical utility to planned settings of acute myocardial ischemia-reperfusion injury such as coronary artery bypass graft surgery, cardiac transplantation and percutaneous coronary intervention. In this article, the focus will be on the origins of delayed IPC, the mechanisms underlying its delayed cardioprotective effect, and the potential areas for its clinical application.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | | |
Collapse
|
14
|
Tranter M, Ren X, Forde T, Wilhide ME, Chen J, Sartor MA, Medvedovic M, Jones WK. NF-kappaB driven cardioprotective gene programs; Hsp70.3 and cardioprotection after late ischemic preconditioning. J Mol Cell Cardiol 2010; 49:664-72. [PMID: 20643136 DOI: 10.1016/j.yjmcc.2010.07.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 06/30/2010] [Accepted: 07/01/2010] [Indexed: 12/14/2022]
Abstract
It has been shown that the transcription factor NF-kappaB is necessary for late phase cardioprotection after ischemic preconditioning (IPC) in the heart, and yet is injurious after ischemia/reperfusion (I/R). However the downstream gene expression programs that underlie the contribution of NF-kappaB to cardioprotection after late IPC are incompletely understood. The objective of this study was to delineate the specific genes that are regulated by NF-kappaB immediately after a late IPC stimulus and validate the methodology for the identification of NF-kappaB-dependent genes that contribute to cardioprotection. A directed microarray analysis identified 238 genes as up or downregulated in an NF-kappaB-dependent manner 3.5h after late IPC. Among these are several genes previously implicated in late IPC. Gene ontological analysis showed that the most significant group of NF-kappaB-dependent genes are heat shock response genes, including the genes encoding Hsp70.1 and Hsp70.3. Though an Hsp70.1/70.3 double knockout failed to exhibit cardioprotection, late IPC was intact in the Hsp70.1 single knockout. After I/R, the Hsp70.1/70.3 double knockout and the Hsp70.1 single knockout had significantly increased and reduced infarct size, respectively. These results delineate the immediate NF-kappaB-dependent transcriptome after late IPC. One of the major categories of NF-kappaB-dependent genes induced by late IPC is the heat shock response. The results of infarct studies confirm that Hsp70.3 is protective after IPC. However, though Hsp70.1 and Hsp70.3 are coordinately regulated, their functions are opposing after I/R injury.
Collapse
Affiliation(s)
- Michael Tranter
- Department of Pharmacology & Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Effects of folic acid on cardiac myocyte apoptosis in rats with streptozotocin-induced diabetes mellitus. Cardiovasc Drugs Ther 2008; 22:299-304. [PMID: 18470605 DOI: 10.1007/s10557-008-6109-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 03/25/2008] [Indexed: 02/05/2023]
Abstract
BACKGROUND The effect of folic acid on cardiac myocyte apoptosis secondary to diabetes is unknown. METHODS Diabetic rats were divided into diabetic control (DC, n = 11), low-dose (LDF, 0.4 mg/kg/day, n = 12) and high-dose (HDF, 1.2 mg/kg/day, n = 12) folic acid groups. Non-diabetic rats (n = 11) were used as the normal control (NC). RESULTS After 11 weeks of treatment, compared with the NC group, the DC group showed a reduced blood levels of reactive oxygen species (ROS, P < 0.01). The rate of cardiac myocyte apoptosis in the diabetic control group was also greater than in the non-diabetic control group (P < 0.01). In folic acid-treated rats, the blood levels of ROS was higher than in the diabetic control group (P < 0.05). There was a dose-dependent reduction in the rate of cardiac myocyte apoptosis in the folic acid groups (P < 0.01), and this was accompanied by an increased level of anti-apoptotic protein Bcl-2 and decreased level of pro-apoptotic protein Bax and Fas (P < 0.01). CONCLUSIONS Dietary folic acid supplementation diminishes the cardiac myocyte apoptosis in streptozotocin-induced diabetes. The apoptosis suppression is accompanied by an increase in the expression of Bcl-2 and a decrease in Bax and Fas.
Collapse
|
16
|
Yang J, Wu LJ, Tashiro SI, Onodera S, Ikejima T. Nitric oxide activated by p38 and NF-kappaB facilitates apoptosis and cell cycle arrest under oxidative stress in evodiamine-treated human melanoma A375-S2 cells. Free Radic Res 2008; 42:1-11. [PMID: 18324518 DOI: 10.1080/10715760701762407] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nitric oxide (NO) has been identified as a fundamental molecule that interplays with reactive oxygen species (ROS) in determining cell fate. As a previous study indicated that ROS was stimulated in evodiamine-induced human melanoma A375-S2 cell apoptosis, the goal of this study was to investigate the role of NO in the cells. In this study, it was found that evodiamine has a strong inductive effect on NO production synthesized by inducible NOS (iNOS) enzyme in a positive-feedback manner. The generated NO was further showed to induce apoptosis and cell cycle arrest and linked to the activation of p53 and p21. After interruption of p38 and nuclear factor-kappaB (NF-kappaB) by pre-treatment with SB203580 and PDTC, iNOS expression, NO synthesis and cell damage were all significantly blocked. It was concluded that p38 and NF-kappaB were critical to the NO producing system, which contributed greatly to the apoptosis and cell cycle arrest in evodiamine-incubated cells.
Collapse
Affiliation(s)
- Jia Yang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, PR, China
| | | | | | | | | |
Collapse
|
17
|
Liao X, Liu JM, Du L, Tang A, Shang Y, Wang SQ, Chen LY, Chen Q. Nitric oxide signaling in stretch‐induced apoptosis of neonatal rat cardiomyocytes. FASEB J 2006; 20:1883-5. [PMID: 16877524 DOI: 10.1096/fj.06-5717fje] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Pressure overload associated with hypertension is an important pathological factor leading to heart remodeling and ultimately heart failure partially due to cardiomyocyte apoptosis. Here we show that endogenous NO signaling plays a critical role in mechanical stretch-induced cardiomyocyte apoptosis. Mechanical stretch induced elevated expression of both eNOS and inducible NO synthase (iNOS) and increased synthesis of NO. A sustained increase in iNOS expression was also found in hearts of hypertensive rats in vivo. Blockade of NO signaling by inhibitors of NOS (L-NAME and AMT) or downstream guanylyl cyclase (ODQ) strongly inhibited stretch-induced apoptosis, mitochondria depolarization, and cytochrome c release, suggesting that NO is required in stretch-induced cardiomyocyte apoptosis. The expression of iNOS, but not eNOS, was blocked by L-NAME and ODQ, indicating that the iNOS induction is NO dependent. The initial elevation of NO is likely due to Ca(2+)-dependent activation of eNOS because elimination of intracellular calcium by EGTA-AM inhibited both iNOS induction and NO elevation. Other calcium signaling inhibitors (nifedipine, ryanodine, thapsigargin, and ionic gadolinium) also attenuated the initial NO elevation. These data indicate that mechanical signals initiate Ca(2+)-dependent NO synthesis, which is further amplified by activation of NO-induced iNOS expression, to regulate cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Xudong Liao
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Wakeno-Takahashi M, Otani H, Nakao S, Imamura H, Shingu K. Isoflurane induces second window of preconditioning through upregulation of inducible nitric oxide synthase in rat heart. Am J Physiol Heart Circ Physiol 2005; 289:H2585-91. [PMID: 16006547 DOI: 10.1152/ajpheart.00400.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The second window of preconditioning (SWOP) induced by inhalation of volatile anesthetics has been documented in the rat heart and is triggered by nitric oxide synthase (NOS), but involvement of NOS in the mediator phase of isoflurane-induced SWOP has not been demonstrated. We tested the hypothesis that isoflurane-induced SWOP is mediated through upregulation of inducible NOS (iNOS). Rats inhaled 0.75 minimum alveolar concentration (MAC) isoflurane, 1.5 MAC isoflurane, or O2 for 2 h. After 24, 48, 72, and 96 h, the isolated heart was perfused with buffer and subjected to 30 min of ischemia followed by 2 h of reperfusion. Inhalation of 0.75 and 1.5 MAC isoflurane significantly limited infarct size after ischemia-reperfusion 24–72 h after isoflurane inhalation. The maximum effect was obtained 48 h after inhalation of 1.5 MAC isoflurane. Postischemic left ventricular function was improved only 48 h after inhalation of 1.5 MAC isoflurane. iNOS expression and activity in the heart were increased 24–72 h after inhalation of 1.5 MAC isoflurane; this increase was less pronounced after inhalation of 0.75 MAC isoflurane. A selective iNOS inhibitor, 1400W (10 μM), abolished iNOS activation and cardioprotection induced 48 h after inhalation of 1.5 MAC isoflurane. These results suggest that isoflurane inhalation induces SWOP after 24–72 h through overexpression and activation of iNOS in the rat heart.
Collapse
|
19
|
Lasley RD, Keith BJ, Kristo G, Yoshimura Y, Mentzer RM. Delayed adenosine A1 receptor preconditioning in rat myocardium is MAPK dependent but iNOS independent. Am J Physiol Heart Circ Physiol 2005; 289:H785-91. [PMID: 15833799 DOI: 10.1152/ajpheart.01008.2004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adenosine A1 receptor delayed preconditioning (PC) against myocardial infarction has been well described; however, there have been limited investigations of the signaling mechanisms that mediate this phenomenon. In addition, there are multiple conflicting reports on the role of inducible nitric oxide synthase (iNOS) in mediating A1 late-phase PC. The purpose of this study was to determine the roles of the p38 and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinases (MAPKs) in in vivo delayed A1 receptor PC and whether this protection at the myocyte level is due to upregulation of iNOS. Myocardial infarct size was measured in open-chest anesthetized rats 24 h after treatment with vehicle or the adenosine A1 agonist 2-chloro-N6-cyclopentyladenosine (CCPA; 100 microg/kg ip). Additional rats receiving CCPA were pretreated with the p38 inhibitor SB-203580 (1 mg/kg ip) or the MAPK/ERK kinase (MEK) inhibitor PD-098059 (0.5 mg/kg ip). At 24 h after CCPA administration, a group of animals was given the iNOS inhibitor 1400 W 10 min before ischemia. Treatment with CCPA reduced infarct size from 48 +/- 2 to 28 +/- 2% of the area at risk, an effect that was blocked by both SB-203580 and PD-098059 but not 1400 W. Ventricular myocytes isolated 24 h after CCPA injection exhibited significantly reduced oxidative stress during H2O2 exposure compared with myocytes from vehicle-injected animals, and this effect was not blocked by the iNOS inhibitor 1400 W. Western blot analysis of whole heart and cardiac myocyte protein samples revealed no expression of iNOS 6 or 24 h after CCPA treatment. These results indicate that adenosine A1 receptor delayed PC in rats is mediated by MAPK-dependent mechanisms, but this phenomenon is not associated with the early or late expression of iNOS.
Collapse
Affiliation(s)
- Robert D Lasley
- Department of Surgery, Univ. of Kentucky College of Medicine, MN276, Chandler Medical Center, 800 Rose St., Lexington, KY 40536-0298, USA.
| | | | | | | | | |
Collapse
|
20
|
Abstract
Despite significant advances in myocardial revascularization and reperfusion, coronary artery disease and subsequently myocardial infarction, are the leading causes of morbidity and mortality in the United States. Strategies which improve the myocardial substrate during and following a myocardial infarction-such as the regrowth of functional blood vessels to the ischemic myocardium would be of great clinical importance. This review article attempts to address this important clinical issue through identifying potential signalling mechanisms by various mode of preconditioning that cause angiogenesis.
Collapse
Affiliation(s)
- Nilanjana Maulik
- Molecular Cardiology Laboratory, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06030-1110, USA.
| |
Collapse
|
21
|
Abstract
Ischemic preconditioning (IP) is defined as a brief period of ischemia ("preclamping") followed by tissue reperfusion, thereby increasing ischemic tolerance for a subsequent longer ischemic period. Several studies showed the effectiveness of classic local IP by preclamping the flap pedicle. There are two temporally and mechanically different types of IP: acute preconditioning, which is induced by preclamping the flap pedicle briefly before flap ischemia, and late preconditioning, induced by a preclamping procedure 24-48 h before flap ischemia. However, both types of local ischemic preconditioning are rarely used clinically, most likely since they can be applied only by invasive means, significantly increase operation time, or even require a second surgical procedure. Several studies from our laboratory showed, in different experimental models, that acute IP, enhancement of flap survival, and improvement of reperfusion microcirculation can be achieved not only by preclamping the flap pedicle, but also by induction of an ischemia/reperfusion event in a body area distant from the flap prior to elevation. This new acute remote IP procedure can be applied without invasive means, using limb tourniquet ischemia briefly before flap ischemia. The effectiveness of acute remote IP was confirmed by other authors in large animal models. Another of our studies showed that late remote IP using a limb tourniquet 24 h before flap ischemia attenuates ischemia/reperfusion in muscle flaps, whereas it was ineffective in adipocutaneous flaps. The exact mechanism of "classic" as well as remote IP is not yet finally determined, although several studies demonstrated that endogenous nitric oxide plays an important role. In summary, the use of a tourniquet to induce limb ischemia before flap ischemia could provide a new, alternative, noninvasive remote IP protocol, although late remote IP might be effective only in muscle flaps. However, the possible future clinical application for late IP is elective flap surgery, whereas acute remote IP could even be used in emergency flaps.
Collapse
Affiliation(s)
- Markus V Küntscher
- BG, Department of Hand, Plastic, and Reconstructive Surgery, Trauma Center Ludwigshafen, and Department of Plastic and Hand Surgery, University of Heidelberg, Heidelberg, Germany.
| | | | | |
Collapse
|
22
|
D'Urso G, Frascarelli S, Balzan S, Zucchi R, Montali U. Production of ouabain-like factor in normal and ischemic rat heart. J Cardiovasc Pharmacol 2004; 43:657-62. [PMID: 15071352 DOI: 10.1097/00005344-200405000-00007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Endogenous ouabain-like factor (OLF) has been detected in mammalian plasma, adrenal gland, and hypothalamus. We investigate whether cardiac tissue may also produce OLF. HPLC chromatographic separation of cardiac extracts showed that RIA-determined OLF activity coincided with the elution profile of exogenous ouabain and with the ability to inhibit 86Rb uptake in human erythrocytes. OLF activity was remarkably higher in excised hearts (3.94 +/- 0.84 pmol/g wet weight by RIA) than in rat blood (0.05 +/- 0.02 pmol/ml). Similar values were obtained in perfused working hearts, without significant changes over time from 5 to 30 minutes of aerobic perfusion. Significant OLF release in the perfusion buffer was also observed (0.54 +/- 0.05 pmoles over 30 minutes). In hearts subjected to 15 minutes of aerobic perfusion followed by 15 minutes of global myocardial ischemia OLF concentration was remarkably increased (8.59 +/- 1.13 versus 4.58 +/- 0.57 pmol/g wet weight by RIA, P < 0.01; an increase after ischemia was confirmed by the assay of 86Rb uptake). Our findings suggest that the rat heart is able to produce OLF, and that its concentration increases during ischemia. Myocardial OLF might modulate the Na/K-ATPase, producing relevant effects on ionic homeostasis and/or gene transcription.
Collapse
Affiliation(s)
- Giuseppina D'Urso
- Dipartimento di Scienze dell'Uomo e dell'Ambiente, University of Pisa, Pisa, Italy
| | | | | | | | | |
Collapse
|
23
|
Zhang F, Oswald T, Holt J, Gerzenshtein J, Lei MP, Lineaweaver WC. Regulation of inducible nitric oxide synthase in ischemic preconditioning of muscle flap in a rat model. Ann Plast Surg 2004; 52:609-13. [PMID: 15166998 DOI: 10.1097/01.sap.0000099713.64498.9e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Ischemic preconditioning has been shown to influence flap tolerance to prolonged ischemia. Nitric oxide (NO) synthesis is one of the proposed mechanisms involved in ischemic preconditioning. In this study, the molecular marker of NO is examined in correlation with ischemic preconditioning on improving muscle flap survival. METHODS Fifty male Sprague-Dawley rats were randomized into experimental and control groups. The gracilis muscle flap with femoral vascular pedicle was used as a flap model. Ischemic preconditioning consisted of 3 sequences of clamping the pedicle for 10 minutes followed by 10 minutes of reperfusion for a total of 1 hour. In part I, the experimental group (n = 10) underwent ischemic preconditioning for 1 hour. In the control group (n = 10), the flaps were dissected without clamping of the pedicle. Both groups were then subjected to 4 hours of global ischemia by continuous pedicle clamping, after which the flaps were sutured to their beds. On postoperative day 3, flap survival was determined by gross and histologic examinations. The evaluators were blinded to the treatment. In part II, the experimental group (n = 12) underwent ischemic preconditioning, while the control group (n = 12) did not. The flaps from each group were harvested for inducible nitric oxide synthase (iNOS) gene expression using reverse transcriptase-polymerase chain reaction at the end of 1 hour after reperfusion and at 4 hours of global ischemia. RESULTS The results indicated a significantly higher survival rate in the experimental group than in the control group (90 versus 50%, P < 0.05). iNOS gene expression was significantly higher in the experimental group than in the control group at 1 hour after ischemic preconditioning (0.73+/-0.18 versus 0.26+/-0.11, P < 0.01). However, after 4 hours of global ischemia, iNOS expression in the control group was statistically higher than in the experimental group (0.83+/-0.16 versus 0.26+/-0.07, P < 0.01). CONCLUSIONS We conclude that ischemic preconditioning can enhance flap tolerance to ischemia-reperfusion injury and improve flap viability rate. This study provides evidence that the regulation of NOS may play a role in ischemic preconditioning phenomenon and warrants further investigation.
Collapse
Affiliation(s)
- Feng Zhang
- Division of Plastic Surgery, University of Mississippi Medical Center, Jackson, 39216, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Fisher SG, Marber MS. An in vivo model of ischaemia-reperfusion injury and ischaemic preconditioning in the mouse heart. J Pharmacol Toxicol Methods 2004; 48:161-9. [PMID: 14986865 DOI: 10.1016/s1056-8719(03)00046-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Our aim was to develop a robust protocol for inducing ischaemia-reperfusion injury and preconditioning in the in vivo mouse heart. METHODS Animals were prepared by standard techniques adapted for the mouse. RESULTS Thirty minutes of ischaemia appeared to strike the most appropriate balance between large infarct volumes with the risk of heart failure and excessively small infarcts. With this duration of ischaemia, a protocol of three sets of 5 min of ischaemia and 5 min of reperfusion was found to precondition, leading to significantly smaller infarct sizes (infarct volume: risk zone volume reduced to 10.1 +/- 2.7% from 41.39% +/- 3.0; P < .05, n = 6 and 7, respectively). Late preconditioning also occurred (infarct volume: risk zone volume = 57.9 +/- 10.1% vs. 35.4 +/- 4.2%, sham compared to late preconditioning, respectively; P = .001, n = 7). DISCUSSION Whilst further refinements may indeed be possible, we feel this article details a valuable, robust protocol for in vivo studies of ischaemia-reperfusion injury and preconditioning in the mouse heart.
Collapse
Affiliation(s)
- Simon G Fisher
- Department of Cardiology, Division of Cardiovascular Science, GKT School of Medicine, King's College London, The Rayne Institute, St. Thomas' Hospital, Lambeth Palace Road, London SE1 7EH, UK.
| | | |
Collapse
|
25
|
Wang X, Yin C, Xi L, Kukreja RC. Opening of Ca2+-activated K+ channels triggers early and delayed preconditioning against I/R injury independent of NOS in mice. Am J Physiol Heart Circ Physiol 2004; 287:H2070-7. [PMID: 15217801 DOI: 10.1152/ajpheart.00431.2004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Opening of Ca2+-activated K+ (KCa) channels has been shown to confer early cardioprotection. It is unknown whether the opening of these channels also induces delayed cardioprotection. In addition, we determined the involvement of nitric oxide synthases (NOSs), which have been implicated in cardioprotection induced by opening of mitochondrial ATP-sensitive K+ (KATP) channels. Adult male ICR mice were pretreated with the KCa-channel opener NS-1619 either 10 min or 24 h before 30 min of global ischemia and 60 min of reperfusion (I/R) in Langendorff mode. Infusion of NS-1619 (10 microM) for 10 min before I/R led to smaller infarct sizes as compared with the vehicle (DMSO)-treated group (P <0.05). This infarct-limiting effect of NS-1619 was associated with improvement in ventricular functional recovery after I/R. The NS-1619-induced protection was abolished by coadministration with the KCa-channel blocker paxilline (1 microM). Similarly, pretreatment with NS-1619 (1 mg/kg ip) induced delayed protection 24 h later (P <0.05). Interestingly, the NS-1619-induced late protection was not blocked by the NOS inhibitor Nomega-nitro-L-arginine methyl ester (15 mg/kg ip). Unlike diazoxide (the opener of mitochondrial KATP channels), NS-1619 did not increase the expression of inducible or endothelial NOS. Western blot analysis demonstrated the existence of alpha- and beta-subunits of KCa channels in mouse heart tissue. We conclude that opening of KCa channels leads to both early and delayed preconditioning effects through a mechanism that is independent of nitric oxide.
Collapse
Affiliation(s)
- Xiaoyin Wang
- Division of Cardiology, Box 980281, Virginia Commonwealth Univ., Richmond, VA 23298-0281, USA
| | | | | | | |
Collapse
|
26
|
Xuan YT, Guo Y, Zhu Y, Han H, Langenbach R, Dawn B, Bolli R. Mechanism of cyclooxygenase-2 upregulation in late preconditioning. J Mol Cell Cardiol 2003; 35:525-37. [PMID: 12738234 PMCID: PMC3210730 DOI: 10.1016/s0022-2828(03)00076-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although the cardioprotection of late preconditioning (PC) is known to be mediated by both inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), the signaling mechanism responsible for COX-2 upregulation and the interaction between iNOS and COX-2 remain unknown. A total of 122 mice were used to address this issue. In wild-type mice preconditioned with six cycles of 4-min coronary occlusion-4-min reperfusion, ischemic PC resulted in rapid activation of nuclear STAT1/3 through tyrosine phosphorylation (STAT1: 339 +/- 48% of control; STAT3: 389 +/- 46% of control) and increased STAT1/3-DNA binding activity (687 +/- 58% of control) at 30 min after PC, with subsequent upregulation of COX-2 protein (373 +/- 60% of control) and activity(increased myocardial levels of PGE2, PGF(2alpha), and 6-keto-PGF(1alpha)) at 24 h. However, COX-1 protein was not changed 24 h after ischemic PC. Pretreatment with the Janus tyrosine kinase (JAK) inhibitor AG-490 before the six occlusion-reperfusion cycles blocked both the tyrosine phosphorylation of STAT1/3 and the subsequent upregulation of COX-2 protein, demonstrating a necessary role of the JAK-STAT pathway in the induction of COX-2. Targeted disruption of the iNOS gene (iNOS-/-) did not block the increased expression of COX-2 protein 24 h after ischemic PC but completely blocked the increase in COX-2 activity, whereas targeted disruption of the COX-2 gene (COX-2-/-) did not alter ischemic PC-induced iNOS induction. Immunoprecipitation of preconditioned heart tissues with anti-COX-2 antibodies followed by immunoblotting with anti-iNOS antibodies revealed that the increased iNOS protein co-precipitated with COX-2. We conclude that (i) the upregulation of COX-2 protein expression after ischemic PC is mediated by a JAK1/2-STAT1/3-signaling cascade; (ii) COX-2 activity requires upregulated iNOS and iNOS-derived NO; and (iii) COX-2 forms complexes with iNOS, supporting a direct interaction between these two proteins. To our knowledge, this is the first evidence that myocardial COX-2 is upregulated via a JAK1/2-STAT1/3 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Roberto Bolli
- Corresponding author. Tel.: +1-502-852-1837; fax: +1-502-852-6474. (R. Bolli)
| |
Collapse
|
27
|
Ferdinandy P, Schulz R. Nitric oxide, superoxide, and peroxynitrite in myocardial ischaemia-reperfusion injury and preconditioning. Br J Pharmacol 2003; 138:532-43. [PMID: 12598407 PMCID: PMC1573696 DOI: 10.1038/sj.bjp.0705080] [Citation(s) in RCA: 327] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
There appears to be a controversy in the study of myocardial ischaemia-reperfusion injury and preconditioning whether nitric oxide (NO) plays a protective or detrimental role. A number of findings and the interpretation of the results to date do not support such a controversy. An understanding of the latest developments in NO, superoxide (O(2)(-)*) and peroxynitrite (ONOO(-)) biology, as well as the various ischaemic animal models utilized is necessary to resolve the apparent controversy. NO is an important cardioprotective molecule via its vasodilator, antioxidant, antiplatelet, and antineutrophil actions and it is essential for normal heart function. However, NO is detrimental if it combines with O(2)(-)* to form ONOO(-) which rapidly decomposes to highly reactive oxidant species. There is a critical balance between cellular concentrations of NO, O(2)(-)*, and superoxide dismutase which physiologically favour NO production but in pathological conditions such as ischaemia and reperfusion result in ONOO(-) formation. In contrast, exposure of the heart to brief episode(s) of ischaemia markedly enhances its ability to withstand a subsequent ischaemic injury. The triggering of this endogenous cardioprotective mechanism known as preconditioning requires both NO and O(2)(-)* synthesis. However, preconditioning in turn attenuates the overproduction of NO, O(2)(-)* and ONOO(-) during a subsequent episode of ischaemia and reperfusion, thereby protecting the heart. Here we review the roles of NO, O(2)(-)*, and ONOO(-) in both ischaemia-reperfusion injury and preconditioning.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720 Hungary.
| | | |
Collapse
|
28
|
Küntscher MV, Kastell T, Altmann J, Menke H, Gebhard MM, Germann G. Acute remote ischemic preconditioning II: the role of nitric oxide. Microsurgery 2003; 22:227-31. [PMID: 12375287 DOI: 10.1002/micr.10042] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The purpose of this study was to determine whether nitric oxide (NO) plays a role in the mechanism of acute "classic" as well as acute remote ischemic preconditioning (IP). Thirty-two male Wistar rats were divided into five experimental groups. The rat cremaster flap in vivo microscopy model was used for assessment of ischemia/reperfusion injury. In the control group (CG, n = 8), a 2-hr flap ischemia was induced after preparation of the cremaster muscle. The animals of group NO (n = 6) received 500 nmol/kg of the NO-donor spermine/nitric oxide complex (Sper/NO) intravenously 30 min prior to ischemia. The group LN + P (L-NAME + preclamping, n = 6) received 10 mg/kg Nomega-nitro-L-arginine methyl ester (L-NAME) intravenously before preclamping of the flap pedicle (10-min cycle length, 30-min reperfusion). L-NAME (10 mg/kg) was administered in group LN + T (L-NAME + tourniquet, n = 6) before ischemia of the right hindlimb was induced, using a tourniquet for 10 min after flap elevation. The limb was then reperfused for 30 min. Thereafter, flap ischemia was induced in each group as in group CG. In vivo microscopy was performed after 1 hr of flap reperfusion in each animal. Group NO demonstrated a significantly higher red blood cell velocity (RBV) in the first-order arterioles and capillaries, a higher capillary flow, and a decreased number of leukocytes adhering to the endothelium (stickers) of the postcapillary venules by comparison to all other groups (P < 0.05). The average capillary RBV and capillary flow were still higher in the CG than in the groups receiving L-NAME (P < 0.05). The data show that NO plays an important role in the mechanism of both acute "classic" as well as acute remote IP, since the administration of a NO-donor previous to ischemia simulates the effect of IP, whereas the nonspecific blocking of NO synthesis by L-NAME abolishes the protective effect of flap preconditioning.
Collapse
Affiliation(s)
- Markus V Küntscher
- Department of Plastic and Hand Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Strasse 33, 67071 Ludwigshafen, Germany.
| | | | | | | | | | | |
Collapse
|
29
|
Huwiler A, Pfeilschifter J. Nitric Oxide Signalling with a Special Focus on Lipid-Derived Mediators. Biol Chem 2003; 384:1379-89. [PMID: 14669981 DOI: 10.1515/bc.2003.154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The ways in which cells communicate among each other concerns all aspects of biology, from developmental processes to diseases. Nitric oxide (NO) is one of the most remarkable and unusual regulatory molecules. It is a labile free radical gas that is not stored but generated on demand, and has been implicated in an extraordinarily diverse range of physiological and pathophysiological functions. The modulation of cell signalling by free radicals is an emerging area of research that provides insight into the orchestration of cell adaptation to a changing microenvironment. In a multicellular organism this serves to coordinate complex physiological responses, such as inflammation. Cell signalling is also accompanied by rapid remodelling of membrane lipids by activated lipases. The discovery that NO, which does not reversibly interact with membrane receptors like conventional hormones and growth factors, targets enzymes such as phospholipase A2, sphingomyelinases or ceramidases, has stimulated growing interest in the crosstalk between redox and lipid signalling.
Collapse
Affiliation(s)
- Andrea Huwiler
- Pharmazentrum Frankfurt, Klinikum der Johann-Wolfgang-Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | | |
Collapse
|
30
|
Loubani M, Galiñanes M. Pharmacological and ischemic preconditioning of the human myocardium: mitoK(ATP) channels are upstream and p38MAPK is downstream of PKC. BMC PHYSIOLOGY 2002; 2:10. [PMID: 12123527 PMCID: PMC117790 DOI: 10.1186/1472-6793-2-10] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2002] [Accepted: 07/18/2002] [Indexed: 11/10/2022]
Abstract
BACKGROUND These studies investigate the role of mitoK(ATP) channels, protein kinase C (PKC) and Mitogen activated protein kinase (p38MAPK) on the cardioprotection of ischemic (IP) and pharmacological preconditioning (PP) of the human myocardium and their sequence of activation. RESULTS Right atrial appendages from patients undergoing elective cardiac surgery were equilibrated for 30 min and then subjected to 90 min of simulated ischemia followed by 120 min reoxygenation. At the end of each protocol creatinine kinase leakage (CK U/g wet wt) and the reduction of MTT to formazan dye (mM/g wet wt) were measured. Similar protection was obtained with alpha1 agonist phenylephrine, adenosine and IP and their combination did not afford additional cardioprotection. Blockade of mitoK(ATP) channels with 5-hydroxydecanoate, PKC with chelerythrine, or p38MAPK with SB203580 abolished the protection of IP and of PP. In additional studies, the stimulation of mitoK(ATP) channels with diazoxide or activation of PKC with PMA or p38MAPK with anisomycin induced identical protection to that of IP and PP. The protection induced by diazoxide was abolished by blockade of PKC and by blockade of p38MAPK. Furthermore, the protection induced by PMA was abolished by SB203580 but not by 5-hydroxydecanoate, whereas the protection induced by anisomycin was unaffected by either 5-hydroxydecanoate or chelerythrine. CONCLUSIONS Opening of mitoK(ATP) channels and activation of PKC and p38MAPK are obligatory steps in the signal transduction cascade of IP and PP of the human myocardium with PKC activation being downstream of the opening of mitoK(ATP) channels and upstream of p38MAPK activation.
Collapse
Affiliation(s)
- Mahmoud Loubani
- Department of Integrative Human Cardiovascular Physiology and Functional Genomics, Division of Cardiac Surgery, University of Leicester, Glenfield Hospital, Groby Road, Leicester
| | - Manuel Galiñanes
- Department of Integrative Human Cardiovascular Physiology and Functional Genomics, Division of Cardiac Surgery, University of Leicester, Glenfield Hospital, Groby Road, Leicester
| |
Collapse
|
31
|
Küntscher MV, Juran S, Menke H, Gebhard MM, Erdmann D, Germann G. The role of pre-ischaemic application of the nitric oxide donor spermine/nitric oxide complex in enhancing flap survival in a rat model. BRITISH JOURNAL OF PLASTIC SURGERY 2002; 55:430-3. [PMID: 12372374 DOI: 10.1054/bjps.2002.3871] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Spermine/nitric oxide complex (Sper/NO) is a new nitric oxide (NO) donor with a long half-life providing controlled biological release of NO in vivo. The purpose of this study was to determine whether flap survival could be improved by pre-ischaemic or post-ischaemic intravenous administration of Sper/NO. We divided 37 male Wistar rats into four experimental groups. An extended epigastric adipocutaneous flap was raised in each animal. The mean area of flap necrosis was assessed for all groups on the fifth postoperative day, using planimetry software. The average area of flap necrosis was mean +/- s.d. = 68.2%+/-18.1% in the control group, and 29.7% +/- 13.3% in the non-ischaemic controls. The group with pre-ischaemic application of Sper/NO demonstrated an average flap necrosis of mean+/-s.d. = 11.2%+/-5.9%, whereas this increased to 59.2%+/-14.4% in the group receiving Sper/NO 5 min prior to reperfusion. The group with pre-ischaemic application of Sper/NO showed a significantly lower area of flap necrosis than either of the control groups or the group receiving Sper/NO just prior to reperfusion (P < 0.05). The group receiving Sper/NO just prior to reperfusion demonstrated a significantly higher mean area of flap necrosis than the non-ischaemic controls (P < 0.05), but did not differ significantly from the control group. Our data show that pharmacological preconditioning and enhancement of flap survival can be achieved by intravenous administration of Sper/NO. The application of Sper/NO at the end of the ischaemia period or in the early reperfusion period provides no protection against ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- M V Küntscher
- Department of Plastic and Hand Surgery, Burn Center, BG Trauma Center Ludwigshafen, Ludwigshafen, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Bishopric NH. A thousand times NO. J Mol Cell Cardiol 2002; 34:601-6. [PMID: 12054846 DOI: 10.1006/jmcc.2002.2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Abstract
Ischemic preconditioning (PC) is a polygenic defensive cellular adaptive phenomenon whereby brief ischemic stimuli render the heart resistant to subsequent similar stress. The late phase of ischemic PC lasts for three to four days, protects against both myocardial stunning and infarction, and thus has considerable clinical relevance. Diverse signaling molecules released by a sublethal ischemic stress initiate a complex signal transduction cascade that modulates the expression of cardioprotective genes. Nitric oxide (NO), generated by the endothelial NO synthase (NOS) and acting via the formation of reactive oxygen species, activates the epsilon isoform of protein kinase C (PKC), which activates the Src family of protein tyrosine kinases (Src and Lck) and transcription factors (nuclear factor-kappaB, and possibly others), with resultant upregulation of the inducible NOS (iNOS) gene and protein expression. iNOS, and other cardioprotective proteins, including cyclooxygenase-2 and aldose reductase, confer resistance to subsequent ischemic stress. This delayed protection can also be mimicked, in the absence of ischemia, by administering NO-releasing agents, a situation that can be potentially exploited for cardioprotection in clinical situations. Identification of this novel bifunctional (trigger and mediator) role played by NO in cardiac protection, not only advances the knowledge regarding its signaling functions, but also offers a potential therapeutic strategy for patients with coronary artery disease. The purpose of this review is to summarize the current evidence in support of this critical role played by NO in ischemic PC.
Collapse
Affiliation(s)
- Buddhadeb Dawn
- Experimental Research Laboratory, Division of Cardiology, University of Louisville, Louisville, Kentucky 40292, USA
| | | |
Collapse
|
34
|
Shinmura K, Xuan YT, Tang XL, Kodani E, Han H, Zhu Y, Bolli R. Inducible nitric oxide synthase modulates cyclooxygenase-2 activity in the heart of conscious rabbits during the late phase of ischemic preconditioning. Circ Res 2002; 90:602-8. [PMID: 11909825 DOI: 10.1161/01.res.0000012202.52809.40] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cyclooxygenase-2 (COX-2) is known to mediate the cardioprotective effects of the late phase of ischemic preconditioning (PC); however, the signaling pathways involved in COX-2 induction following ischemic PC are unknown. In addition, although inducible nitric oxide synthase (iNOS) has been identified as a co-mediator of late PC together with COX-2, the interaction between iNOS and COX-2 in the heart is unknown. Using conscious rabbits, we found that the induction of COX-2 expression 24 hours after ischemic PC was blocked by pretreatment with inhibitors of protein kinase C (PKC), Src protein tyrosine kinases (PTKs), and nuclear factor-kappaB (NF-kappaB) but not by inhibitors of NOS or scavengers of reactive oxygen species (ROS). The selective iNOS inhibitors SMT and 1400W, given 24 hours after PC, abrogated the increase in myocardial prostaglandin E2 (PGE2) and 6-keto-PGF1alpha, whereas the selective soluble guanylate cyclase inhibitor ODQ had no effect. COX-2 selective inhibitors (celecoxib and NS-398) did not affect iNOS activity. These results demonstrate that (i) ischemic PC upregulates cardiac COX-2 via PKC-, Src PTK-, and NF-kappaB-dependent signaling pathways, whereas generation of NO and ROS is not necessary, and (ii) the activity of newly synthesized COX-2 following PC requires iNOS-derived NO whereas iNOS activity is independent of COX-2-derived prostanoids, indicating that COX-2 is located downstream of iNOS in the protective pathway of late PC. The data also indicate that iNOS modulates COX-2 activity via cGMP-independent mechanisms. To our knowledge, this is the first demonstration that iNOS-derived NO drives prostanoid synthesis by COX-2 in the heart. NO-mediated activation of COX-2 may be a heretofore unrecognized mechanism by which NO exerts its salubrious effects in the late phase of PC.
Collapse
Affiliation(s)
- Ken Shinmura
- Experimental Research Laboratory, Division of Cardiology, University of Louisville and Jewish Hospital Heart and Lung Institute, Louisville, Ky 40292, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Pfeilschifter J, Beck KF, Eberhardt W, Huwiler A. Changing gears in the course of glomerulonephritis by shifting superoxide to nitric oxide-dominated chemistry. Kidney Int 2002; 61:809-15. [PMID: 11849430 DOI: 10.1046/j.1523-1755.2002.00225.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The glomerular response to injury displays astonishingly uniform features that include infiltration with professional immune cells, activation and proliferation of resident glomerular cells and matrix expansion. Cross-communication of intrinsic mesangial cells with invading immune cells is crucial for the fate of glomerular injury: progression to glomerulosclerosis or resolution and repair. The formation of free radicals, particularly of nitric oxide and superoxide, are key events that initiate redox-based signal transduction and gene expression. The balance between these radicals constitutes redox-operated genetic switches that ensure self-limited inflammatory responses to tissue injury. The aberrant production of the mediators, however, may sustain matrix accumulation and result in irreversible alteration of glomerular structure and function.
Collapse
Affiliation(s)
- Josef Pfeilschifter
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany.
| | | | | | | |
Collapse
|
36
|
Wang Y, Guo Y, Zhang SX, Wu WJ, Wang J, Bao W, Bolli R. Ischemic preconditioning upregulates inducible nitric oxide synthase in cardiac myocyte. J Mol Cell Cardiol 2002; 34:5-15. [PMID: 11812160 DOI: 10.1006/jmcc.2001.1482] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent evidence has shown that the cardioprotection afforded by the late phase of ischemic preconditioning (PC) is mediated by upregulation of inducible nitric oxide synthase (iNOS). However, the specific cardiac cell type(s) that express(es) iNOS in response to ischemic PC remains unknown. Thus, mice underwent a sequence of six cycles of 4-min coronary occlusion/4-min reperfusion, which induces late PC, and tissue samples were collected at serial times for measurement of mRNA (Northern) and protein levels (Western). In addition, whole heart samples were cryosectioned for in situ hybridization and immunohistochemistry. The steady-state levels of iNOS mRNA in the ischemic regions started to increase at 1 h after ischemic PC, peaked at 3 h (201+/-31% of sham, n=5 P<0.01) and remained elevated at 24 h (177+/-22% of sham, n=5 P<0.01). In accordance with these data, iNOS protein expression was increased at 24 h (219+/-41% of sham, n=5 P<0.01). In contrast, neither endothelial nitric oxide synthase (eNOS) mRNA levels nor its protein expression changed at any time-point. The magnitude of iNOS upregulation after ischemic PC was mild compared with that noted 66 h after permanent coronary occlusion (360+/-53% of sham) or 8 h after endotoxin (3117+/-61% of control). After ischemic PC, diffuse iNOS signals were detected with in situ hybridization and immunohistochemistry in the cytoplasmic space of cardiac myocytes and, to a lesser degree, in the wall of large vessels, but were absent in smooth muscle and endothelium of small vessels and in fibroblasts. This pattern contrasted with that observed in mouse hearts subjected to permanent coronary occlusion where strong iNOS signals were concentrated in inflammatory cells but absent in cardiac myocytes. Thus, not only the degree of iNOS expression but also its cellular distribution were profoundly different in reversibly injured (preconditioned) v infarcted myocardium. We conclude that iNOS is rapidly upregulated after ischemic PC and that cardiac myocytes are the main source of ischemic PC-induced iNOS expression. This study demonstrates, for the first time, a differential pattern of iNOS expression in sublethal (PC) v lethal ischemia, which may have important implication for the role of iNOS in these two settings.
Collapse
Affiliation(s)
- Yang Wang
- Experimental Research Laboratory, Division of Cardiology, Louisville, KY 40292, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Bolli R. Cardioprotective function of inducible nitric oxide synthase and role of nitric oxide in myocardial ischemia and preconditioning: an overview of a decade of research. J Mol Cell Cardiol 2001; 33:1897-918. [PMID: 11708836 DOI: 10.1006/jmcc.2001.1462] [Citation(s) in RCA: 423] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Over the past decade, an enormous number of studies (>100) have focused on the role of nitric oxide (NO) in myocardial ischemia. It is important to distinguish the function of NO in unstressed (non-preconditioned) myocardium from its function in preconditioned myocardium (i.e. myocardium that has shifted to a defensive phenotype in response to stress). Of the 92 studies that have examined the role of NO in modulating the severity of ischemia/reperfusion injury in non-preconditioned myocardium, the vast majority [67 (73%)] have concluded that NO (either endogenous or exogenous) has a protective effect and only 11 (12%) found a detrimental effect. The proportion of studies supporting a cytoprotective role of NO is similar in vivo[35 (71%) out of 49] and in vitro[32 (74%) out of 43]. With regard to the delayed acquisition of tolerance to ischemia [late preconditioning (PC)], overwhelming evidence indicates a critical role of NO in this phenomenon. Specifically, enhanced biosynthesis of NO by eNOS is essential to trigger the late phase of ischemia-induced and exercise-induced PC, and enhanced NO production by iNOS is obligatorily required to mediate the anti-stunning and anti-infarct actions of late PC elicited by five different stimuli (ischemia, adenosine A1 agonists, opioid delta1 agonists, endotoxin derivatives and exercise). Thus, NO plays a dual role in the pathophysiology of the late phase of PC, acting initially as the trigger and subsequently as the mediator of this adaptive response ("NO hypothesis of late PC"). The diversity of the PC stimuli that converge on iNOS implies that the upregulation of this enzyme is a central mechanism whereby the myocardium protects itself from ischemia. The NO hypothesis of late PC has thus revealed a cytoprotective function of iNOS in the heart, a novel paradigm which has recently been extended to other tissues, including kidney and intestine. Other corollaries of this hypothesis are that the heart responds to stress in a biphasic manner, utilizing eNOS as an immediate but short-term response and iNOS as a delayed but long-term defense, and that the fundamental difference between non-preconditioned and late preconditioned myocardium is the tissue level of iNOS-derived NO, which is tonically higher in the latter compared with the former. Hence, late PC can be viewed as a state of enhanced NO synthesis. The NO hypothesis of late PC has important therapeutic implications. In experimental animals, administration of NO donors in lieu of ischemia can faithfully reproduce the molecular and functional aspects of ischemia-induced late PC, indicating that NO is not only necessary but also sufficient to induce late PC. The recent demonstration that nitroglycerin also induces late PC in patients provides proof-of-principle for the concept that nitrates could be used as a PC-mimetic therapy for the prophylaxis of ischemic injury in the clinical arena. This novel application of nitrates could be as important as, or perhaps even more important than, their current use as antianginal and preload-reducing agents. In addition, gene transfer of either eNOS or iNOS has been shown to replicate the infarct-sparing actions of ischemic PC, suggesting that NOS gene therapy could be an effective strategy for alleviating ischemia/reperfusion injury. Ten years of research have demonstrated that NO plays a fundamental biological role in protecting the heart against ischemia/reperfusion injury. The time has come to translate this enormous body of experimental evidence into clinically useful therapies by harnessing the cytoprotective properties of NO.
Collapse
Affiliation(s)
- R Bolli
- Division of Cardiology, University of Louisville and the Jewish Hospital Heart and Lung Institute, Louisville, KY 40292, USA
| |
Collapse
|
38
|
Xiao L, Lu R, Hu CP, Deng HW, Li YJ. Delayed cardioprotection by intestinal preconditioning is mediated by calcitonin gene-related peptide. Eur J Pharmacol 2001; 427:131-5. [PMID: 11557265 DOI: 10.1016/s0014-2999(01)01231-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Previous studies have shown that nitric oxide and calcitonin gene-related peptide (CGRP) are involved in mediation of the delayed cardioprotection of ischemic or pharmacological preconditioning, and nitric oxide can evoke the release of CGRP. In the present study, we examined the role of CGRP in nitric oxide-mediated delayed cardioprotection by brief intestinal ischemia in rats. The serum concentration of creatine kinase and infarct size were measured after 45-min coronary artery occlusion and 180-min reperfusion. Ischemic preconditioning was induced by six cycles of 4-min ischemia and 4-min reperfusion of the small intestine. Pretreatment with intestinal ischemic preconditioning for 24, 48, or 72 h significantly reduced infarct size and creatine kinase release, and the effects of ischemic preconditioning were completely abolished by L-nitroarginine methyl ester (L-NAME, 10 mg/kg, i.p.), an inhibitor of nitric oxide synthase, or by pretreatment with capsaicin (50 mg/kg, s.c.), which selectively depletes transmitters in capsaicin-sensitive sensory nerves. Intestinal preconditioning caused a significant increase in plasma concentrations of CGRP, and the effect was also abolished by L-NAME or capsaicin. These results suggest that the delayed cardioprotection afforded by intestinal ischemic preconditioning is mediated by endogenous CGRP via the nitric oxide pathway.
Collapse
Affiliation(s)
- L Xiao
- Department of Pharmacology, Hunan Medical University, Changsha, Hunan 410078, China
| | | | | | | | | |
Collapse
|
39
|
Dick JMC, Van Molle W, Libert C, Lefebvre RA. Antisense knockdown of inducible nitric oxide synthase inhibits the relaxant effect of VIP in isolated smooth muscle cells of the mouse gastric fundus. Br J Pharmacol 2001; 134:425-33. [PMID: 11564662 PMCID: PMC1572958 DOI: 10.1038/sj.bjp.0704262] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
1. Our previous results showed that the non-selective nitric oxide synthase (NOS) inhibitor L-N(G)-nitroarginine (L-NOARG) and the selective inducible NOS (iNOS) inhibitor N-(3-(acetaminomethyl)-benzyl)acetamidine (1400W) inhibited the relaxant effect of vasoactive intestinal polypeptide (VIP) in isolated smooth muscle cells of the mouse gastric fundus, suggesting the involvement of iNOS. The identity of the NOS isoform involved in the VIP-induced relaxation in isolated smooth muscle cells of the mouse gastric fundus was now further investigated by use of antisense oligodeoxynucleotides (aODNs) to iNOS. 2. Incubation of isolated smooth muscle cells with fluorescein isothiocyanate (FITC)-labelled aODNs showed that nuclear accumulation occurs quickly and reaches saturation after 60 min. The in vivo intravenous administration of aODNs to iNOS, 24 and 12 h before murine tumour necrosis factor alpha (mTNFalpha) challenge, significantly reduced the nitrite levels induced by the mTNFalpha challenge. 3. Intravenous administration of aODNs to iNOS in mice, 24 and 12 h before isolation of the gastric smooth muscle cells, decreased the inhibitory effect of the NOS inhibitors L-NOARG and 1400W on the relaxant effect of VIP, whereas neither saline nor sODNs had any influence. 4. Preincubation of the isolated smooth muscle cells with aODNs almost abolished the inhibitory effect of L-NOARG and 1400W on the VIP-induced relaxation, whereas sODNs failed. 5. These results illustrate that the inhibitory effect of NOS inhibitors in isolated smooth muscle cells of the mouse gastric fundus is due to inactivation of iNOS. iNOS, probably induced by the isolation procedure of the smooth muscle cells, seems involved in the relaxant effect of VIP in isolated gastric smooth muscle cells.
Collapse
Affiliation(s)
- J M C Dick
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, B-9000 Ghent, Belgium
| | - W Van Molle
- Department of Molecular Biology, Flanders Interuniversity Institute for Biotechnology and Ghent University, K.L. Ledeganckstraat 35, B-9000 Ghent, Belgium
| | - C Libert
- Department of Molecular Biology, Flanders Interuniversity Institute for Biotechnology and Ghent University, K.L. Ledeganckstraat 35, B-9000 Ghent, Belgium
| | - R A Lefebvre
- Heymans Institute of Pharmacology, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, B-9000 Ghent, Belgium
- Author for correspondence:
| |
Collapse
|
40
|
Ricciardi R, Schaffer BK, Kim RD, Shah SA, Donohue SE, Wheeler SM, Quarfordt SH, Callery MP, Meyers WC, Chari RS. Protective effects of ischemic preconditioning on the cold-preserved liver are tyrosine kinase dependent. Transplantation 2001; 72:406-12. [PMID: 11502967 DOI: 10.1097/00007890-200108150-00008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Little data exist regarding the use of ischemic preconditioning before sustained hepatic cold storage. We hypothesized that ischemic preconditioning protects hepatic grafts via a tyrosine kinase-dependent pathway. METHODS Six porcine livers underwent routine harvest (control). Five other livers underwent 15 min of in situ ischemia followed by 15 min of reflow before harvest (ischemic preconditioning). Another five livers were pretreated with a tyrosine kinase inhibitor (genistein) before preconditioning. Upon reperfusion and after 2 hours of cold storage, graft function, graft circulatory impairment, and markers of cellular damage were analyzed. Tissue cytoplasmic extracts were analyzed for tyrosine phosphorylation with Western blot. Significance was determined with t tests. RESULTS Ischemic-preconditioned grafts demonstrated enhanced bile production, augmented responses to a bile acid challenge, and elevated O2 consumption (P<0.05) compared to controls. Also, preconditioned grafts demonstrated improved hepatic tissue blood flow and decreased hepatic vascular resistance (P<0.005) compared to controls. Endothelial cell preservation (factor VIII immunostain) was improved in preconditioned graft biopsies compared to controls. With genistein pretreatment, all observed improvements returned to control levels. Analysis of cytoplasmic extracts demonstrated an increase in tyrosine phosphorylation before cold ischemia in preconditioned grafts only, but not in control or genistein-pretreated grafts. CONCLUSIONS The data indicate that ischemic preconditioning protects the liver from sustained cold ischemia and that tyrosine kinases are involved in preconditioning responses.
Collapse
Affiliation(s)
- R Ricciardi
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Müllenheim J, Rulands R, Wietschorke T, Frässdorf J, Preckel B, Schlack W. Late preconditioning is blocked by racemic ketamine, but not by S(+)-ketamine. Anesth Analg 2001; 93:265-70, 1st contents page. [PMID: 11473841 DOI: 10.1097/00000539-200108000-00005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
UNLABELLED Racemic ketamine blocks K(ATP) channels in isolated cells and abolishes short-term cardioprotection against prolonged ischemia. We investigated the effects of racemic ketamine and S(+)-ketamine on ischemic late preconditioning (LPC) in the rabbit heart in vivo. A coronary occluder was chronically implanted in 36 rabbits. After recovery, the rabbits divided into four groups (each n = 9). LPC was induced in conscious rabbits by a 5-min coronary occlusion. Twenty-four hours later, the animals were instrumented for measurement of left ventricular systolic pressure (LVSP, tip manometer), cardiac output (CO, ultrasonic flowprobe) and myocardial infarct size (triphenyltetrazolium staining). All rabbits were then subjected to 30-min coronary occlusion and 2 h reperfusion. Controls underwent the ischemia-reperfusion program without LPC. To test whether racemic ketamine or S(+)-ketamine blocks the cardioprotection induced by LPC, the drugs (10 mg/kg) were given 10 min before the 30-min ischemia. Hemodynamic values were not significantly different between groups during the experiments (baseline: LVSP, 94 +/- 3 mm Hg [mean +/- SEM] and CO, 243 +/- 9 mL/min; coronary occlusion: LVSP, 93% +/- 4% of baseline and CO, 84% +/- 4%; after 2 h of reperfusion: LVSP, 85% +/- 4% and CO, 83% +/- 4%). LPC reduced infarct size from 44% +/- 3% of the area at risk in controls to 22% +/- 3% (P = 0.002). Administration of racemic ketamine abolished the cardioprotective effects of LPC (44 +/- 4%, P = 0.002). S(+)-ketamine did not affect the infarct size reduction induced by LPC (26 +/- 6%, P = 0.88). IMPLICATIONS Racemic ketamine, but not S(+)-ketamine, blocks the cardioprotection induced by ischemic late preconditioning in rabbit hearts in vivo. Thus, the influence of ketamine on ischemic late preconditioning is most likely enantiomer specific, and the use of S(+)-ketamine may be preferable in patients with coronary artery disease.
Collapse
Affiliation(s)
- J Müllenheim
- Institut für Klinische Anaesthesiologie, Heinrich-Heine-Universität, Postfach 10 10 07, D-40001 Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Dana A, Baxter GF, Yellon DM. Delayed or second window preconditioning induced by adenosine A1 receptor activation is independent of early generation of nitric oxide or late induction of inducible nitric oxide synthase. J Cardiovasc Pharmacol 2001; 38:278-87. [PMID: 11483878 DOI: 10.1097/00005344-200108000-00014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Transient adenosine A1 receptor (A1R) activation induces a second window or delayed preconditioning against myocardial infarction 24-72 h later. Early generation of nitric oxide and delayed induction of nitric oxide synthase have been implicated in mediating delayed cardioprotection after ischemic preconditioning in rabbits. Recent evidence indicates that some of the regulatory roles of adenosine in cardiac tissue may be mediated by A1R-induced generation of nitric oxide. This study examined the role of nitric oxide in the mediation of A1R-induced delayed preconditioning against infarction. Pharmacologic preconditioning of rabbits with the selective A1R agonist 2-chloro-N6-cyclopentyladenosine 100 microg/kg (CCPA) significantly reduced myocardial infarct size compared with control animals, after 30 min regional ischemia and 2 h reperfusion in vivo 24 h later (27.3+/-4.7 vs. 46.0+/-3.7%, respectively; p = 0.001). Nonselective inhibition of nitric oxide synthase with N(G)-nitro-L-arginine methyl ester (10 mg/kg) before administration of CCPA did not affect this infarct limitation at 24 h. Selective inhibition of inducible nitric oxide synthase before the prolonged ischemic insult on day 2, with two structurally independent inducible nitric oxide synthase inhibitors, L-N(6)-(1-iminoethyl)-lysine (10 mg/kg) or aminoguanidine (300 mg/kg), did not abrogate the reduction in infarction observed by pharmacologic preconditioning with CCPA 24 h earlier. These results suggest that the second window or delayed protection against myocardial infarction observed 24 h after pharmacologic preconditioning with an adenosine A1 agonist occurs independently of either early generation of nitric oxide or subacute induction of inducible nitric oxide synthase.
Collapse
Affiliation(s)
- A Dana
- The Hatter Institute for Cardiovascular Studies, University College London Hospital and Medical School, UK
| | | | | |
Collapse
|
43
|
|
44
|
Patel HH, Hsu A, Moore J, Gross GJ. BW373U86, a delta opioid agonist, partially mediates delayed cardioprotection via a free radical mechanism that is independent of opioid receptor stimulation. J Mol Cell Cardiol 2001; 33:1455-65. [PMID: 11448134 DOI: 10.1006/jmcc.2001.1408] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Opioids have been shown to produce both an early and delayed phase of cardioprotection; however, the signaling pathways involved, particularly in the delayed response, have not been well defined. Therefore, we investigated the potential of BW373U86 (BW), a potent delta opioid agonist, to produce delayed cardioprotection and characterized the role of opioid receptors and oxygen-derived free radicals (OFRs) in this delayed response. All rats underwent 30 min of ischemia followed by 2 h of reperfusion. The rats were divided into four groups. First, rats were pretreated with selective opioid receptor antagonists or the antioxidant, 2-mercaptopropionyl glycine (2-MPG), in the presence of BW and allowed to recover for 24 h before the ischemia-reperfusion protocol. Second, rats were pretreated with BW, allowed to recover for 24 h, and subsequently treated with either opioid antagonists or 2-MPG, 10 min prior to the ischemia-reperfusion protocol. Third, rats underwent ischemic preconditioning (IPC) (1x5 min occlusion) both with and without 2-MPG to determine the role of OFRs in acute cardioprotection. Fourth, rats were pretreated with TAN-67, an opioid agonist known to signal through the delta1 opioid receptor in the presence and absence of 2-MPG. Control rats were injected with saline and allowed to recover for 24 h. BW produced a bell-shaped dose-related reduction in infarct size with a maximal reduction observed at 0.1 mg/kg v control (16+/-3%v 60+/-3%, P<0.001). Surprisingly, the delayed protection induced by BW was only partially blocked by pretreatment with the delta1-selective antagonist, BNTX; however, it was completely blocked by pretreatment with 2-MPG (47+/-5%, P<0.001). Only naloxone given acutely inhibited the protective effects of BW; however, at the dose used, 2-MPG partially reduced the protective effect of acute IPC. TAN-67 (0.1 mg/kg) also produced a significant reduction in infarct size compared to control (18+/-4%v 60+/-3%, P<0.001). This protection was blocked by pretreatment with 2-MPG (42+/-4%, P<0.001). These data suggest that BW and TAN-67 mediate delayed cardioprotection via a free radical mechanism that appears to be only partially dependent on delta opioid receptor stimulation. Furthermore, it is the early burst in OFRs that is crucial to initiating the protective effect.
Collapse
Affiliation(s)
- H H Patel
- Medical College of Wisconsin, Department of Pharmacology and Toxicology, Milwaukee, WI 53226, USA
| | | | | | | |
Collapse
|
45
|
Feng J, Li H, Rosenkranz ER. Bradykinin protects the rabbit heart after cardioplegic ischemia via NO-dependent pathways. Ann Thorac Surg 2000; 70:2119-24. [PMID: 11156131 DOI: 10.1016/s0003-4975(00)02148-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Depressed myocardial performance is an important clinical problem after open heart surgery. We hypothesized pretreating with bradykinin would pharmacologically precondition the heart and improve post-ischemic performance, and induce myocardial preconditioning by activating nitric oxide synthase. METHODS Thirty-three rabbit hearts underwent retrograde perfusion with Krebs-Henseleit buffer (KHB) followed by 50 minutes of 37 degrees C cardioplegic ischemia with St. Thomas' cardioplegia solution (StTCP). Ten control hearts received no pretreatment. Ten bradykinin-pretreated hearts received a 10-minute infusion of 0.1 microMol/L bradykinin-enriched KHB and cardioplegic arrest with 0.1 microMol/L bradykinin-enriched StTCP. Six other hearts received 0.1 microMol/L HOE 140, a selective B2 receptor antagonist, added to both the 0.1 microMol/L bradykinin-enriched KHB and 0.1 microMol/L bradykinin-enriched StTCP solutions. Finally, six other hearts received 100 microMol/L of N-omega-nitro-L-arginine methyl ester (L-NAME), an inhibitor of nitric oxide synthase, added to both the 0.1 microMol/L bradykinin-enriched KHB and 0.1 microMol/L bradykinin-enriched StTCP solutions. RESULTS Bradykinin pretreatment significantly improved postischemic performance and coronary flow (CF) compared with control (LVDP: 53 +/- 5* vs 27 +/- 4 mm Hg; +dP/dtmax: 1,025 +/- 93* vs 507 +/- 85 mm Hg/s; CF: 31 +/- 3* vs 22 +/- 2 mL/min; *p < 0.05). Both HOE 140 and L-NAME abolished bradykinin-induced protection, resulting in recovery equivalent to untreated controls. CONCLUSIONS Bradykinin pretreatment improves recovery of ventricular and coronary vascular function via nitric oxide-dependent mechanisms. Pharmacologic preconditioning by bradykinin pretreatment may be an important new strategy for improving myocardial protection during heart surgery.
Collapse
Affiliation(s)
- J Feng
- Daughtry Family Department of Surgery, University of Miami School of Medicine, Florida, USA
| | | | | |
Collapse
|
46
|
Abstract
Unlike the early phase of preconditioning (PC), which lasts 2 to 3 hours and protects against infarction but not against stunning, the late phase of PC lasts 3 to 4 days and protects against both infarction and stunning, suggesting that it may have greater clinical relevance. It is now clear that late PC is a polygenic phenomenon that requires the simultaneous activation of multiple stress-responsive genes. Chemical signals released by a sublethal ischemic stress (such as NO, reactive oxygen species, and adenosine) trigger a complex cascade of signaling events that includes the activation of protein kinase C, Src protein tyrosine kinases, and nuclear factor kappaB and culminates in increased synthesis of inducible NO synthase, cyclooxygenase-2, aldose reductase, Mn superoxide dismutase, and probably other cardioprotective proteins. An analogous sequence of events can be triggered by a variety of stimuli, such as heat stress, exercise, and cytokines. Thus, late PC appears to be a universal response of the heart to stress in general. Importantly, the cardioprotective effects of late PC can be reproduced pharmacologically with clinically relevant agents (eg, NO donors, adenosine receptor agonists, endotoxin derivatives, or opioid receptor agonists), suggesting that this phenomenon might be exploited for therapeutic purposes. The purpose of this review is to summarize current information regarding the pathophysiology and mechanism of late PC.
Collapse
Affiliation(s)
- R Bolli
- Division of Cardiology, University of Louisville and Jewish Hospital Heart and Lung Institute, Louisville, KY 40292, USA.
| |
Collapse
|
47
|
Takano H, Tang XL, Kodani E, Bolli R. Late preconditioning enhances recovery of myocardial function after infarction in conscious rabbits. Am J Physiol Heart Circ Physiol 2000; 279:H2372-81. [PMID: 11045974 DOI: 10.1152/ajpheart.2000.279.5.h2372] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is unknown whether late preconditioning (PC) enhances the recovery of left ventricular (LV) function after a myocardial infarction. Thus 25 conscious rabbits were subjected to a 30-min coronary occlusion followed by 28 days of reperfusion after PC 24 h earlier with either ischemia or nitric oxide donor administration [S-nitroso-N-acetylpenicillamine (SNAP)]. The recovery of wall thickening (WTh) after reperfusion was significantly improved in the ischemic PC and SNAP PC groups compared with controls, both at rest and during dobutamine stress. Interestingly, neither ischemia- nor SNAP-induced late PC attenuated myocardial stunning from day 1 through day 14. Infarct size was smaller in the ischemic PC and SNAP PC groups compared with controls. In all groups, WTh at 28 days was positively and linearly related to the percentage of viable tissue in the region underlying the ultrasonic crystal (r = 0.90), indicating that the improvement in LV function after both ischemia-induced and NO donor-induced late PC can be fully explained by the reduction in infarct size; a separate effect of late PC on LV remodeling or LV contractility need not be invoked. In conclusion, in conscious rabbits late PC, induced either by ischemia or pharmacologically, not only limits infarct size but also enhances the recovery of LV function after myocardial infarction. This finding has important clinical implications and provides triphenyltetrazolium chloride-independent evidence that late PC limits myocellular death after sustained ischemia.
Collapse
Affiliation(s)
- H Takano
- Experimental Research Laboratory, Division of Cardiology, University of Louisville, Louisville, Kentucky 40292, USA
| | | | | | | |
Collapse
|
48
|
Takano H, Tang XL, Bolli R. Differential role of K(ATP) channels in late preconditioning against myocardial stunning and infarction in rabbits. Am J Physiol Heart Circ Physiol 2000; 279:H2350-9. [PMID: 11045972 DOI: 10.1152/ajpheart.2000.279.5.h2350] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of ATP-sensitive potassium (K(ATP)) channels in the late phase of ischemic preconditioning (PC) remains unclear. Furthermore, it is unknown whether K(ATP) channels serve as end effectors both for late PC against infarction and against stunning. Thus, in phase I of this study, conscious rabbits underwent a 30-min coronary occlusion (O) followed by 72 h of reperfusion (R) with or without ischemic PC (6 4-min O/4-min R cycles) 24 h earlier. Late PC reduced infarct size approximately 46% versus controls. The K(ATP) channel blocker 5-hydroxydecanoic acid (5-HD), given 5 min before the 30-min O, abrogated the infarct-sparing effect of late PC but did not alter infarct size in non-PC rabbits. In phase II, rabbits underwent six 4-min O/4-min R cycles for 3 consecutive days (days 1, 2, and 3). In controls, the total deficit of systolic wall thickening (WTh) after the sixth reperfusion was reduced by 46% on day 2 and 54% on day 3 compared with day 1, indicating a late PC effect against myocardial stunning. Neither 5-HD nor glibenclamide, given on day 2, abrogated late PC. The K(ATP) channel opener diazoxide, given on day 1, attenuated stunning, and this effect was completely blocked by 5-HD. Thus the same dose of 5-HD that blocked the antistunning effect of diazoxide failed to block the antistunning effects of late PC. Furthermore, when diazoxide was administered in PC rabbits on day 2, myocardial stunning was further attenuated, indicating that diazoxide and late PC have additive anti-stunning effects. We conclude that K(ATP) channels play an essential role in late PC against infarction but not in late PC against stunning, revealing an important pathogenetic difference between these two forms of cardioprotection.
Collapse
Affiliation(s)
- H Takano
- Experimental Research Laboratory, Division of Cardiology, University of Louisville, Louisville, Kentucky 40292, USA
| | | | | |
Collapse
|
49
|
Shi Y, Pritchard KA, Holman P, Rafiee P, Griffith OW, Kalyanaraman B, Baker JE. Chronic myocardial hypoxia increases nitric oxide synthase and decreases caveolin-3. Free Radic Biol Med 2000; 29:695-703. [PMID: 11053770 DOI: 10.1016/s0891-5849(00)00364-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nitric oxide synthase (NOS) is believed to play an important role in protecting the myocardium against ischemia. Chronic hypoxia from birth increases NOS activity in the myocardium resulting in enhanced nitric oxide production and increased resistance to ischemia. We examined the effects of chronic hypoxia on NOS gene and protein expression and on NOS protein association with caveolin-3. Rabbits were raised from birth in a normoxic (F(I)O(2) = 0.21) or a hypoxic (F(I)O(2) = 0.12) environment for 9 d, and then the hearts were isolated. Ribonuclease protection assays revealed that chronic hypoxia did not alter NOS transcript levels for NOS1, NOS2, or NOS3. The most abundant transcript was NOS3. Western analysis revealed NOS3 was the only isoform detected. Immunoblots of NOS3 immunoprecipitates showed that chronic hypoxia increases NOS3 protein by 2.0 +/- 0.4-fold and decreases the amount of caveolin-3 that can be coprecipitated with NOS3 by 5.5 +/- 0.9-fold. Immunoblots of normoxic and hypoxic hearts showed that chronic hypoxia decreases the amount of caveolin-3 in heart homogenates by 2. 2 +/- 0.5-fold. These data suggest that a decrease in caveolin-3 plays a role in the mechanisms by which chronic hypoxia increases NOS3 activity in the myocardium.
Collapse
Affiliation(s)
- Y Shi
- Division of Pediatric Surgery, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Li RC, Ping P, Zhang J, Wead WB, Cao X, Gao J, Zheng Y, Huang S, Han J, Bolli R. PKCepsilon modulates NF-kappaB and AP-1 via mitogen-activated protein kinases in adult rabbit cardiomyocytes. Am J Physiol Heart Circ Physiol 2000; 279:H1679-89. [PMID: 11009455 DOI: 10.1152/ajpheart.2000.279.4.h1679] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that protein kinase C (PKC)-epsilon, nuclear factor (NF)-kappaB, and mitogen-activated protein kinases (MAPKs) are essential signaling elements in ischemic preconditioning. In the present study, we examined whether activation of PKCepsilon affects the activation of NF-kappaB in cardiac myocytes and whether MAPKs are mediators of this signaling event. Activation of PKCepsilon (+108% above control) in adult rabbit cardiomyocytes to a degree that has been previously shown to protect myocytes against hypoxic injury increased the DNA-binding activity of NF-kappaB (+164%) and activator protein (AP)-1 (+127%) but not that of Elk-1. Activation of PKCeta did not have an effect on these transcription factors. Activation of PKCepsilon also enhanced the phosphorylation activities of the p44/p42 MAPKs and the p54/p46 c-Jun NH(2)-terminal kinases (JNKs). PKCepsilon-induced activation of NF-kappaB and AP-1 was completely abolished by inhibition of the p44/p42 MAPK pathway with PD98059 and by inhibition of the p54/p46 JNK pathway with a dominant negative mutant of MAPK kinase-4, indicating that both signaling pathways are necessary. Taken together, these data identify NF-kappaB and AP-1 as downstream targets of PKCepsilon, thereby establishing a molecular link between activation of PKCepsilon and activation of NF-kappaB and AP-1 in cardiomyocytes. The results further demonstrate that both the p44/p42 MAPK and the p54/p46 JNK signaling pathways are essential mediators of this event.
Collapse
Affiliation(s)
- R C Li
- Experimental Research Laboratory, Division of Cardiology, University of Louisville and the Jewish Hospital Heart and Lung Research Institute, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|