1
|
Bouzazi D, Mami W, Mosbah A, Marrakchi N, Ben Ahmed M, Messadi E. Natriuretic-like Peptide Lebetin 2 Mediates M2 Macrophage Polarization in LPS-Activated RAW264.7 Cells in an IL-10-Dependent Manner. Toxins (Basel) 2023; 15:toxins15040298. [PMID: 37104236 PMCID: PMC10142756 DOI: 10.3390/toxins15040298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
Snake natriuretic peptide (NP) Lebetin 2 (L2) has been shown to improve cardiac function and reduce fibrosis as well as inflammation by promoting M2-type macrophages in a reperfused myocardial infarction (MI) model. However, the inflammatory mechanism of L2 remains unclear. Therefore, we investigated the effect of L2 on macrophage polarization in lipopolysaccharide (LPS)-activated RAW264.7 cells in vitro and explored the associated underlying mechanisms. TNF-α, IL-6 and IL-10 levels were assessed using an ELISA assay, and M2 macrophage polarization was determined by flow cytometry. L2 was used at non-cytotoxic concentrations determined by a preliminary MTT cell viability assay, and compared to B-type natriuretic peptide (BNP). In LPS-activated cells, both peptides reduced TNF-α and IL-6 release compared to controls. However, only L2 increased IL-10 release in a sustained manner and promoted downstream M2 macrophage polarization. Pretreatment of LPS-activated RAW264.7 cells with the selective NP receptor (NPR) antagonist isatin abolished both IL-10 and M2-like macrophage potentiation provided by L2. In addition, cell pretreatment with the IL-10 inhibitor suppressed L2-induced M2 macrophage polarization. We conclude that L2 exerts an anti-inflammatory response to LPS by regulating the release of inflammatory cytokines via stimulating of NP receptors and promoting M2 macrophage polarization through activation of IL-10 signaling.
Collapse
Affiliation(s)
- Dorsaf Bouzazi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Wael Mami
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Amor Mosbah
- Laboratory of Biotechnology and Bio-Geo Resources Valorization (LR11ES31), Higher Institute of Biotechnology of Sidi Thabet (ISBST), University of Manouba, Tunis 2010, Tunisia
| | - Naziha Marrakchi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Melika Ben Ahmed
- Laboratoire de Transmission, Department of Clinical Immunology, Contrôle et Immunobiologie des Infections, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| | - Erij Messadi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires (P2C), Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis 1068, Tunisia
| |
Collapse
|
2
|
Bon-Mathier AC, Déglise T, Rignault-Clerc S, Bielmann C, Mazzolai L, Rosenblatt-Velin N. Brain Natriuretic Peptide Protects Cardiomyocytes from Apoptosis and Stimulates Their Cell Cycle Re-Entry in Mouse Infarcted Hearts. Cells 2022; 12:cells12010007. [PMID: 36611800 PMCID: PMC9818267 DOI: 10.3390/cells12010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Brain Natriuretic Peptide (BNP) supplementation after infarction increases heart function and decreases heart remodeling. BNP receptors, NPR-A and NPR-B are expressed on adult cardiomyocytes (CMs). We investigated whether a part of the BNP cardioprotective effect in infarcted and unmanipulated hearts is due to modulation of the CM fate. For this purpose, infarcted adult male mice were intraperitoneally injected every two days during 2 weeks with BNP or saline. Mice were sacrificed 1 and 14 days after surgery. BNP or saline was also injected intraperitoneally every two days into neonatal pups (3 days after birth) for 10 days and in unmanipulated 8-week-old male mice for 2 weeks. At sacrifice, CMs were isolated, counted, measured, and characterized by qRT-PCR. The proportion of mononucleated CMs was determined. Immunostainings aimed to detect CM re-entry in the cell cycle were performed on the different hearts. Finally, the signaling pathway activated by BNP treatment was identified in in vitro BNP-treated adult CMs and in CMs isolated from BNP-treated hearts. An increased number of CMs was detected in the hypoxic area of infarcted hearts, and in unmanipulated neonatal and adult hearts after BNP treatment. Accordingly, Troponin T plasma concentration was significantly reduced 1 and 3 days after infarction in BNP-treated mice, demonstrating less CM death. Furthermore, higher number of small, dedifferentiated and mononucleated CMs were identified in adult BNP-treated hearts when compared to saline-treated hearts. BNP-treated CMs express higher levels of mRNAs coding for hif1 alpha and for the different cyclins than CMs isolated from saline-treated hearts. Higher percentages of CMs undergoing DNA synthesis, expressing Ki67, phospho histone3 and Aurora B were detected in all BNP-treated hearts, demonstrating that CMs re-enter into the cell cycle. BNP effect on adult CMs in vivo is mediated by NPR-A binding and activation of the ERK MAP kinase pathway. Interestingly, an increased number of CMs was also detected in adult infarcted hearts treated with LCZ696, an inhibitor of the natriuretic peptide degradation. Altogether, our results identified BNP and all therapies aimed to increase BNP's bioavailability as new cardioprotective targets as BNP treatment leads to an increased number of CMs in neonatal, adult unmanipulated and infarcted hearts.
Collapse
|
3
|
Tourki B, Dumesnil A, Belaidi E, Ghrir S, Godin-Ribuot D, Marrakchi N, Richard V, Mulder P, Messadi E. Lebetin 2, a Snake Venom-Derived B-Type Natriuretic Peptide, Provides Immediate and Prolonged Protection against Myocardial Ischemia-Reperfusion Injury via Modulation of Post-Ischemic Inflammatory Response. Toxins (Basel) 2019; 11:toxins11090524. [PMID: 31510060 PMCID: PMC6784001 DOI: 10.3390/toxins11090524] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/08/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) followed by left ventricular (LV) remodeling is the most frequent cause of heart failure. Lebetin 2 (L2), a snake venom-derived natriuretic peptide, exerts cardioprotection during acute myocardial ischemia-reperfusion (IR) ex vivo. However, its effects on delayed consequences of IR injury, including post-MI inflammation and fibrosis have not been defined. Here, we determined whether a single L2 injection exerts cardioprotection in IR murine models in vivo, and whether inflammatory response to ischemic injury plays a role in L2-induced effects. We quantified infarct size (IS), fibrosis, inflammation, and both endothelial cell and cardiomyocyte densities in injured myocardium and compared these values with those induced by B-type natriuretic peptide (BNP). Both L2 and BNP reduced IS, fibrosis, and inflammatory response after IR, as evidenced by decreased leukocyte and proinflammatory M1 macrophage infiltrations in the infarcted area compared to untreated animals. However, only L2 increased anti-inflammatory M2-like macrophages. L2 also induced a higher density of endothelial cells and cardiomyocytes. Our data show that L2 has strong, acute, prolonged cardioprotective effects in post-MI that are mediated, at least in part, by the modulation of the post-ischemic inflammatory response and especially, by the enhancement of M2-like macrophages, thus reducing IR-induced necrotic and fibrotic effects.
Collapse
Affiliation(s)
- Bochra Tourki
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, 1068 Tunis, Tunisia.
- Université Carthage Tunis, 1054 Bizerte, Tunisia.
| | - Anais Dumesnil
- Normandie Univ, UNIROUEN, Inserm U1096, FHU REMOD-VHF, 76000 Rouen, France.
| | - Elise Belaidi
- Université Grenoble Alpes, Inserm U1042, Laboratoire HP2, 38000 Grenoble, France.
| | - Slim Ghrir
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, 1068 Tunis, Tunisia.
| | - Diane Godin-Ribuot
- Université Grenoble Alpes, Inserm U1042, Laboratoire HP2, 38000 Grenoble, France.
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, 1068 Tunis, Tunisia.
| | - Vincent Richard
- Normandie Univ, UNIROUEN, Inserm U1096, FHU REMOD-VHF, 76000 Rouen, France.
| | - Paul Mulder
- Normandie Univ, UNIROUEN, Inserm U1096, FHU REMOD-VHF, 76000 Rouen, France.
| | - Erij Messadi
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, 1068 Tunis, Tunisia.
| |
Collapse
|
4
|
Suppression of Stim1 reduced intracellular calcium concentration and attenuated hypoxia/reoxygenation induced apoptosis in H9C2 cells. Biosci Rep 2017; 37:BSR20171249. [PMID: 29089467 PMCID: PMC5700273 DOI: 10.1042/bsr20171249] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 01/04/2023] Open
Abstract
Objective: Previous studies have demonstrated Stromal interaction molecule 1 (STIM1)-mediated store-operated Ca2+ entry (SOCE) contributes to intracellular Ca2+ accumulation. The present study aimed to investigate the expression of STIM1 and its downstream molecules Orai1/TRPC1 in the context of myocardial ischemia/reperfusion injury (MIRI) and the effect of STIM1 inhibition on Ca2+ accumulation and apoptosis in H9c2 cardiomyocytes subjected to hypoxia/reoxygenation (H/R). Methods: Expression of STIM1/Orai1/TRPC1 was determined by RT-PCR and Western blot in mice subjected to MIRI and H9C2 cardiomyocytes subjected to H/R. To knock-down STIM1, H9C2 cardiomyocytes was transfected with Stealth SiRNA. Apoptosis was analyzed by both flow cytometry and TUNEL assay. Cell viability was measured by MTT assay. Intracellular Ca2+ concentration was detected by laser scanning confocal microscopy using Fluo-3/AM probe. Furthermore, the opening of mitochondrial permeability transition pore (mPTP) was assessed by coloading with calcein AM and CoCl2, while ROS generation was evaluated using the dye DCFH-DA in H9C2 cardiomyocytes. Results: Expression of STIM1/Orai1/TRPC1 significantly increased in transcript and translation level after MIRI in vivo and H/R in vitro. In H9C2 cardiomyocytes subjected to H/R, intracellular Ca2+ accumulation significantly increased compared with control group, along with enhanced mPTP opening and elevated ROS generation. However, suppression of STIM1 by SiRNA significantly decreased apoptosis and intracellular Ca2+ accumulation induced by H/R in H9C2 cardiomyocytes, accompanied by attenuated mPTP opening and decreased ROS generation. In addition, suppression of STIM1 increased the Bcl-2/Bax ratio, decreased Orai1/TRPC1, and cleaved caspase-3 expression. Conclusion: Suppression of STIM1 reduced intracellular calcium level and attenuated hypoxia/reoxygenation induced apoptosis in H9C2 cardiomyocytes. Our findings provide a new perspective in understanding STIM1-mediated calcium overload in the setting of MIRI.
Collapse
|
5
|
Suveren E, Baxter GF, Iskit AB, Turker AU. Cardioprotective effects of Viscum album L. subsp. album (European misletoe) leaf extracts in myocardial ischemia and reperfusion. JOURNAL OF ETHNOPHARMACOLOGY 2017; 209:203-209. [PMID: 28689799 DOI: 10.1016/j.jep.2017.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Viscum album L. (European mistletoe) is a hemiparasitic plant belonging to Loranthaceae family and has been used in Turkish traditional medicine for the treatment of cardiovascular disorders and heart diseases such as hypertension, tachycardia and angina pectoris. AIM OF THE STUDY The present study investigated the cardioprotective effects of V. album leaf extracts in myocardial ischemia and reperfusion injury in rats. MATERIAL AND METHODS Lyophilized aqueous (AVa) and methanolic (MVa) extracts of V. album were prepared from dried leaf. The isolated hearts were perfused with V. album extracts prior to and during 35min of ischemia induced by coronary artery occlusion. After 120min of coronary reperfusion, infarct size was determined by triphenyltetrazolium staining. RESULTS Both AVa and MVa extracts reduced the extent of infarction compared with untreated control hearts, but protective effect of MVa had more potential in low concentration; infarct size as proportion of ischemic risk zone: AVa 17.5±1.5%; Mva 20.3±2.5%, both P<0.01 versus control 38.1±1.4%. This protective effect was comparable to infarct limitation induced by ischemic preconditioning (21.5±2.4%). Inhibition of nitric oxide synthesis with L-NG-nitroarginine methyl ester completely abrogated the protection afforded by both extracts. ATP-sensitive K+ channel blockade by glibenclamide abrogated the protection afforded by MVa while attenuating, but not abolishing, the protective action of Ava. CONCLUSIONS This study provided the first experimental evidence that V. album leaf extracts can mediate nitric oxide-dependent cardioprotection against myocardial injury produced by ischemia/reperfusion insult. With this study, popular usage of V. album extracts in Turkish folk medicine as a remedy for cardiac diseases was justified.
Collapse
Affiliation(s)
- Eylem Suveren
- Department of Nursing, Bolu School of Health, Abant Izzet Baysal University, Bolu, Turkey.
| | - Gary F Baxter
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK.
| | - Alper B Iskit
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| | - Arzu Ucar Turker
- Department of Biology, Faculty of Arts and Sciences, Abant Izzet Baysal University, Bolu, Turkey.
| |
Collapse
|
6
|
Tourki B, Matéo P, Morand J, Elayeb M, Godin-Ribuot D, Marrakchi N, Belaidi E, Messadi E. Lebetin 2, a Snake Venom-Derived Natriuretic Peptide, Attenuates Acute Myocardial Ischemic Injury through the Modulation of Mitochondrial Permeability Transition Pore at the Time of Reperfusion. PLoS One 2016; 11:e0162632. [PMID: 27618302 PMCID: PMC5019389 DOI: 10.1371/journal.pone.0162632] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/25/2016] [Indexed: 12/28/2022] Open
Abstract
Cardiac ischemia is one of the leading causes of death worldwide. It is now well established that natriuretic peptides can attenuate the development of irreversible ischemic injury during myocardial infarction. Lebetin 2 (L2) is a new discovered peptide isolated from Macrovipera lebetina venom with structural similarity to B-type natriuretic peptide (BNP). Our objectives were to define the acute cardioprotective actions of L2 in isolated Langendorff-perfused rat hearts after regional or global ischemia-reperfusion (IR). We studied infarct size, left ventricular contractile recovery, survival protein kinases and mitochondrial permeability transition pore (mPTP) opening in injured myocardium. L2 dosage was determined by preliminary experiments at its ability to induce cyclic guanosine monophosphate (cGMP) release without changing hemodynamic effects in normoxic hearts. L2 was found to be as effective as BNP in reducing infarct size after the induction of either regional or global IR. Both peptides equally improved contractile recovery after regional IR, but only L2 increased coronary flow and reduced severe contractile dysfunction after global ischemia. Cardioprotection afforded by L2 was abolished after isatin or 5-hydroxydecanote pretreatment suggesting the involvement of natriuretic peptide receptors and mitochondrial KATP (mitoKATP) channels in the L2-induced effects. L2 also increased survival protein expression in the reperfused myocardium as evidenced by phosphorylation of signaling pathways PKCε/ERK/GSK3β and PI3K/Akt/eNOS. IR induced mitochondrial pore opening, but this effect was markedly prevented by L2 treatment. These data show that L2 has strong cardioprotective effect in acute ischemia through stimulation of natriuretic peptide receptors. These beneficial effects are mediated, at least in part, by mitoKATP channel opening and downstream activated survival kinases, thus delaying mPTP opening and improving IR-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Bochra Tourki
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
- Université Carthage Tunis, Bizerte, Tunisia
| | - Philippe Matéo
- Laboratoire de Signalisation et Physiopathologie Cardiovasculaire, UMR-S 1180, Faculté de Pharmacie, Université Paris Sud, Paris, France
| | - Jessica Morand
- Laboratoire d’Hypoxie et Physiopathologie Cardiaque, Inserm U1042, Faculté de Pharmacie, Université Grenoble Alpes, Grenoble, France
| | - Mohamed Elayeb
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Diane Godin-Ribuot
- Laboratoire d’Hypoxie et Physiopathologie Cardiaque, Inserm U1042, Faculté de Pharmacie, Université Grenoble Alpes, Grenoble, France
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Elise Belaidi
- Laboratoire d’Hypoxie et Physiopathologie Cardiaque, Inserm U1042, Faculté de Pharmacie, Université Grenoble Alpes, Grenoble, France
| | - Erij Messadi
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
- * E-mail:
| |
Collapse
|
7
|
He F, Xu BL, Chen C, Jia HJ, Wu JX, Wang XC, Sheng JL, Huang L, Cheng J. Methylophiopogonanone A suppresses ischemia/reperfusion-induced myocardial apoptosis in mice via activating PI3K/Akt/eNOS signaling pathway. Acta Pharmacol Sin 2016; 37:763-71. [PMID: 27063216 DOI: 10.1038/aps.2016.14] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/06/2016] [Indexed: 12/24/2022]
Abstract
AIM The dried tuber root of Ophiopogon japonicus has been used in the traditional Chinese medicine for treatment of myocardial ischemia and thrombosis. In this study we investigated the effects of methylophiopogonanone A (MO-A), a major homoisoflavonoid in Ophiopogon japonicus, on myocardial ischemia/reperfusion (I/R) injury. METHODS Mice were pretreated with MO-A (10 mg·kg(-1)·d(-1), po) for 2 weeks and then subjected to transient occlusion of the left anterior descending coronary artery. Cardiac function was evaluated, and the infarct size and apoptosis index were assessed. The mechanisms underlying the cardio-protection of MO-A were analyzed in H9C2 rat cardiomyocytes subjected to hypoxia/reoxygenation (H/R). The cell viability and apoptosis were evaluated; apoptotic and relevant signaling proteins were analyzed. NO levels in the culture medium were assessed. RESULTS In I/R mice, pretreatment with MO-A significantly reduced the infarct size (by 60.7%) and myocardial apoptosis (by 56.8%), and improved cardiac function. In H9C2 cells subjected to H/R, pretreatment with MO-A (10 μmol/L) significantly decreased apoptosis and cleaved caspase-3 expression, elevated the Bcl-2/Bax ratio and restored NO production. Furthermore, pretreatment with MO-A markedly increased the activation of PI3K/Akt/eNOS pathway in H9C2 cells subjected to H/R, and the protective effects of MO-A were abolished in the presence of the PI3K inhibitor wortmannin (100 nmol/L). CONCLUSION MO-A attenuates I/R-induced myocardial apoptosis in mice via activating the PI3K/Akt/eNOS signaling pathway.
Collapse
|
8
|
Pharmacological Therapy in the Heart as an Alternative to Cellular Therapy: A Place for the Brain Natriuretic Peptide? Stem Cells Int 2016; 2016:5961342. [PMID: 26880973 PMCID: PMC4735943 DOI: 10.1155/2016/5961342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/08/2015] [Accepted: 10/08/2015] [Indexed: 02/08/2023] Open
Abstract
The discovery that stem cells isolated from different organs have the ability to differentiate into mature beating cardiomyocytes has fostered considerable interest in developing cellular regenerative therapies to treat cardiac diseases associated with the loss of viable myocardium. Clinical studies evaluating the potential of stem cells (from heart, blood, bone marrow, skeletal muscle, and fat) to regenerate the myocardium and improve its functional status indicated that although the method appeared generally safe, its overall efficacy has remained modest. Several issues raised by these studies were notably related to the nature and number of injected cells, as well as the route and timing of their administration, to cite only a few. Besides the direct administration of cardiac precursor cells, a distinct approach to cardiac regeneration could be based upon the stimulation of the heart's natural ability to regenerate, using pharmacological approaches. Indeed, differentiation and/or proliferation of cardiac precursor cells is controlled by various endogenous mediators, such as growth factors and cytokines, which could thus be used as pharmacological agents to promote regeneration. To illustrate such approach, we present recent results showing that the exogenous administration of the natriuretic peptide BNP triggers “endogenous” cardiac regeneration, following experimental myocardial infarction.
Collapse
|
9
|
Breivik L, Jensen A, Guvåg S, Aarnes EK, Aspevik A, Helgeland E, Hovland S, Brattelid T, Jonassen AK. B-type natriuretic peptide expression and cardioprotection is regulated by Akt dependent signaling at early reperfusion. Peptides 2015; 66:43-50. [PMID: 25698234 DOI: 10.1016/j.peptides.2015.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 01/16/2023]
Abstract
Exogenously administered B-type natriuretic peptide (BNP) has been shown to offer cardioprotection through activation of particulate guanylyl cyclase (pGC), protein kinase G (PKG) and KATP channel opening. The current study explores if cardioprotection afforded by short intermittent BNP administration involves PI3K/Akt/p70s6k dependent signaling, and whether this signaling pathway may participate in regulation of BNP mRNA expression at early reperfusion. Isolated Langendorff perfused rat hearts were subjected to 30min of regional ischemia and 120min of reperfusion (IR). Applying intermittent 3×30s infusion of BNP peptide in a postconditioning like manner (BNPPost) reduced infarct size by >50% compared to controls (BNPPost 17±2% vs. control 42±4%, p<0.001). Co-treatment with inhibitors of the PI3K/Akt/p70s6k pathway (wortmannin, SH-6 and rapamycin) completely abolished the infarct-limiting effect of BNP postconditioning (BNPPost+Wi 36±5%, BNPPost+SH-6 41±4%, BNPPost+Rap 37±6% vs. BNPPost 17±2%, p<0.001). Inhibition of natriuretic peptide receptors (NPR) by isatin also abrogated BNPPost cardioprotection (BNPPost+isatin 46±2% vs. BNPPost 17±2%, p<0.001). BNPPost also significantly phosphorylated Akt and p70s6k at early reperfusion, and Akt phosphorylation was inhibited by SH-6 and isatin. Myocardial BNP mRNA levels in the area at risk (AA) were significantly elevated at early reperfusion as compared to the non-ischemic area (ANA) (Ctr(AA) 2.7±0.5 vs. Ctr(ANA) 1.2±0.2, p<0.05) and the ischemic control tissue (Ctr(AA) 2.7±0.5 vs. ischemia 1.0±0.1, p<0.05). Additional experiments also revealed a significant higher BNP mRNA level in ischemic postconditioned (IPost) hearts as compared to ischemic controls (IPost 6.7±1.3 vs. ischemia 1.0±0.2, p<0.05), but showed no difference from controls run in parallel (Ctr 5.4±0.8). Akt inhibition by SH-6 completely abrogated this elevation (IPost 6.7±1.3 vs. IPost+SH-6 1.8±0.7, p<0.05) (Ctr 5.4±0.8 vs. SH-6 1.5±0.9, p<0.05). In conclusion, Akt dependent signaling is involved in mediating the cardioprotection afforded by intermittent BNP infusion at early reperfusion, and may also participate in regulation of reperfusion induced BNP expression.
Collapse
Affiliation(s)
- L Breivik
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway.
| | - A Jensen
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - S Guvåg
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - E K Aarnes
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - A Aspevik
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - E Helgeland
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - S Hovland
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - T Brattelid
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - A K Jonassen
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| |
Collapse
|
10
|
Inserte J, Garcia-Dorado D. The cGMP/PKG pathway as a common mediator of cardioprotection: translatability and mechanism. Br J Pharmacol 2015; 172:1996-2009. [PMID: 25297462 DOI: 10.1111/bph.12959] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/16/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022] Open
Abstract
Cardiomyocyte cell death occurring during myocardial reperfusion (reperfusion injury) contributes to final infarct size after transient coronary occlusion. Different interrelated mechanisms of reperfusion injury have been identified, including alterations in cytosolic Ca(2+) handling, sarcoplasmic reticulum-mediated Ca(2+) oscillations and hypercontracture, proteolysis secondary to calpain activation and mitochondrial permeability transition. All these mechanisms occur during the initial minutes of reperfusion and are inhibited by intracellular acidosis. The cGMP/PKG pathway modulates the rate of recovery of intracellular pH, but has also direct effect on Ca(2+) oscillations and mitochondrial permeability transition. The cGMP/PKG pathway is depressed in cardiomyocytes by ischaemia/reperfusion and preserved by ischaemic postconditioning, which importantly contributes to postconditioning protection. The present article reviews the mechanisms and consequences of the effect of ischaemic postconditioning on the cGMP/PKG pathway, the different pharmacological strategies aimed to stimulate it during myocardial reperfusion and the evidence, limitations and promise of translation of these strategies to the clinical practice. Overall, the preclinical and clinical evidence suggests that modulation of the cGMP/PKG pathway may be a therapeutic target in the context of myocardial infarction.
Collapse
Affiliation(s)
- Javier Inserte
- Cardiology Department, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | |
Collapse
|
11
|
Bielmann C, Rignault-Clerc S, Liaudet L, Li F, Kunieda T, Sogawa C, Zehnder T, Waeber B, Feihl F, Rosenblatt-Velin N. Brain natriuretic peptide is able to stimulate cardiac progenitor cell proliferation and differentiation in murine hearts after birth. Basic Res Cardiol 2014; 110:455. [DOI: 10.1007/s00395-014-0455-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/06/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022]
|
12
|
Abstract
In the 30 years since the identification of the natriuretic peptides, their involvement in regulating fluid and blood pressure has become firmly established. Data indicating a role for these hormones in lifestyle-related metabolic and cardiovascular disorders have also accumulated over the past decade. Dysregulation of the natriuretic peptide system has been associated with obesity, glucose intolerance, type 2 diabetes mellitus, and essential hypertension. Moreover, the natriuretic peptides have been implicated in the protection against atherosclerosis, thrombosis, and myocardial ischaemia. All these conditions can coexist and potentially lead to heart failure, a syndrome associated with a functional natriuretic peptide deficiency despite high circulating concentrations of immunoreactive peptides. Therefore, dysregulation of the natriuretic peptide system, a 'natriuretic handicap', might be an important factor in the initiation and progression of metabolic dysfunction and its accompanying cardiovascular complications. This Review provides a summary of the natriuretic peptide system and its involvement in these cardiometabolic conditions. We propose that these peptides might have an integrating role in lifestyle-related metabolic and cardiovascular disorders.
Collapse
|
13
|
Bice JS, Burley DS, Baxter GF. Novel approaches and opportunities for cardioprotective signaling through 3',5'-cyclic guanosine monophosphate manipulation. J Cardiovasc Pharmacol Ther 2014; 19:269-82. [PMID: 24572031 DOI: 10.1177/1074248413518971] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Limiting the injurious effects of myocardial ischemia-reperfusion is a desirable therapeutic target, which has been investigated extensively over the last three decades. Here we provide an up to date review of the literature documenting the experimental and clinical research demonstrating the effects of manipulating cGMP for the therapeutic targeting of the injurious effects of ischemic heart disease. Augmentation of the cyclic nucleotide cGMP plays a crucial role in many cardioprotective signaling pathways. There is an extensive body of literature which supports pharmacological targeting of cGMP or upstream activators in models of ischemia-reperfusion to limit injury. NO donors have long been utilised to manipulate cGMP, and more recently non-NO synthase derived NOx species have been investigated, resulting in their evaluation in clinical trials for the treatment of ischemic heart disease. Encouraging results demonstrate that natriuretic peptides are worthy candidates in manipulating cGMP and its downstream effectors to afford cytoprotection. Synthetic ligands have been designed which co-activate natriuretic peptide receptors to improve targeting this pathway. Advances have been made in targeting the soluble guanylyl cyclase which catalyzes the production of cGMP independently of the endogenous ligand NO using NO-independent stimulators and activators of sGC. These novel compounds show promise as a new class of drugs that target this signaling cascade specifically under pathological conditions when endogenous NO production may be compromised. Regulating the degradation of cGMP via phosphodiesterase inhibition also shows therapeutic potential. It is clear that production and regulation of cGMP is complex, indeed its spatial production and cellular distribution are only just emerging.
Collapse
Affiliation(s)
- Justin S Bice
- 1School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | | | | |
Collapse
|
14
|
Lyu T, Zhao Y, Zhang T, Zhou W, Yang F, Ge H, Ding S, Pu J, He B. Natriuretic Peptides as an Adjunctive Treatment for Acute Myocardial Infarction. Int Heart J 2014; 55:8-16. [DOI: 10.1536/ihj.13-109] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ting Lyu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Tuo Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Wen Zhou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Fan Yang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Heng Ge
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Song Ding
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| | - Ben He
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University
| |
Collapse
|
15
|
Bice JS, Keim Y, Stasch JP, Baxter GF. NO-independent stimulation or activation of soluble guanylyl cyclase during early reperfusion limits infarct size. Cardiovasc Res 2013; 101:220-8. [PMID: 24259501 PMCID: PMC3896250 DOI: 10.1093/cvr/cvt257] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aims Guanylyl cyclase-cyclic guanosine monophosphate signalling plays an important role in endogenous cardioprotective signalling. The aim was to assess the potential of direct pharmacological activation and stimulation of soluble guanylyl cyclase, targeting different redox states of the enzyme, to limit myocardial necrosis during early reperfusion. Methods and results Rat isolated hearts were subjected to reversible left coronary artery occlusion (ischaemia-reperfusion) and infarct size was assessed by the tetrazolium staining technique. Administration during early reperfusion of BAY 41-2272, an NO-independent, haem-dependent stimulator of soluble guanylyl cyclase targeting the reduced state, or BAY 60-2770, an NO-independent, haem-independent activator targeting the oxidized state, significantly limited infarct size. Inhibition of NO synthesis did not abrogate this protection, but exogenous perfusion of NO with BAY 41-2272 produced a synergistic effect. The haem site oxidiser, ODQ abrogated the protection afforded by BAY 41-2272 but potentiated the protection afforded by BAY 60-2770. Targeting both the reduced and oxidized forms of sGC together did not afford additive protection. Conclusions Targeting either reduced or oxidized forms of sGC during early reperfusion affords cardioprotection, providing support for the concept that direct sGC manipulation at reperfusion has therapeutic potential for the management of acute myocardial infarction.
Collapse
Affiliation(s)
- Justin S Bice
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | | | | | | |
Collapse
|
16
|
Chang R, Li Y, Yang X, Yue Y, Dou L, Wang Y, Zhang W, Li X. Protective role of deoxyschizandrin and schisantherin A against myocardial ischemia-reperfusion injury in rats. PLoS One 2013; 8:e61590. [PMID: 23620773 PMCID: PMC3631228 DOI: 10.1371/journal.pone.0061590] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/12/2013] [Indexed: 12/23/2022] Open
Abstract
Background Our previous studies suggested that deoxyschizandrin (DSD) and schisantherin A (STA) may have cardioprotective effects, but information in this regard is lacking. Therefore, we explored the protective role of DSD and STA in myocardial ischemia–reperfusion (I/R) injury. Methodology/Principal Findings Anesthetized male rats were treated once with DSD and STA (each 40 µmol/kg) through the tail vein after 45 min of ischemia, followed by 2-h reperfusion. Cardiac function, infarct size, biochemical markers, histopathology and apoptosis were measured and mRNA expression of gp91phox in myocardial tissue assessed by RT-PCR. Neonatal rat cardiomyocytes were pretreated with DSD and STA and then damaged by H2O2. Cell apoptosis was tested by a flow cytometric assay. Compared with the I/R group: (i) DSD and STA could significantly reduce the abnormalities of LVSP, LVEDP, ±dp/dtmax and arrhythmias, thereby showing their protective roles in cardiac function; (ii) DSD and STA could significantly attenuate the infarct size and MDA release while increasing SOD activity, suggesting a role in reducing myocardial injury; (iii) tissue morphology and myocardial textual analysis revealed that DSD and STA mitigated changes in myocardial histopathology; (iv) DSD and STA decreased apoptosis (33.56±2.58% to 10.28±2.80% and 10.98±1.99%, respectively) and caspase-3 activity in the myocardium (0.62±0.02 OD/mg to 0.38±0.02 OD/mg and 0.32±0.02 OD/mg, respectively), showing their protective effects upon cardiomyocytes; and (v) DSD and STA had similar protective effects on I/R injury as those seen with the positive control metoprolol. In vitro, DSD and STA could significantly decrease the apoptosis of neonatal cardiomyocytes. Conclusions/Significance These data suggest that DSD and STA can protect against myocardial I/R injury. The underlining mechanism may be related to their role in inhibiting cardiomyocyte apoptosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Arrhythmias, Cardiac/blood
- Arrhythmias, Cardiac/complications
- Arrhythmias, Cardiac/drug therapy
- Arrhythmias, Cardiac/physiopathology
- Cardiotonic Agents/pharmacology
- Cardiotonic Agents/therapeutic use
- Caspase 3/metabolism
- Cyclooctanes/administration & dosage
- Cyclooctanes/chemistry
- Cyclooctanes/pharmacology
- Cyclooctanes/therapeutic use
- Cytoprotection/drug effects
- Dioxoles/administration & dosage
- Dioxoles/chemistry
- Dioxoles/pharmacology
- Dioxoles/therapeutic use
- Disease Models, Animal
- Gene Expression Regulation/drug effects
- Hemodynamics/drug effects
- Hydrogen Peroxide/pharmacology
- Lignans/administration & dosage
- Lignans/chemistry
- Lignans/pharmacology
- Lignans/therapeutic use
- Male
- Malondialdehyde/blood
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Myocardial Infarction/complications
- Myocardial Infarction/drug therapy
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardial Reperfusion Injury/blood
- Myocardial Reperfusion Injury/complications
- Myocardial Reperfusion Injury/drug therapy
- Myocardial Reperfusion Injury/physiopathology
- Myocardium/enzymology
- Myocardium/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- NADPH Oxidase 2
- NADPH Oxidases/genetics
- NADPH Oxidases/metabolism
- Polycyclic Compounds/administration & dosage
- Polycyclic Compounds/chemistry
- Polycyclic Compounds/pharmacology
- Polycyclic Compounds/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Superoxide Dismutase/metabolism
Collapse
Affiliation(s)
- Ruimiao Chang
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yong Li
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xingxin Yang
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yuan Yue
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Lili Dou
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yanwei Wang
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Weifang Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xiaoni Li
- College of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi Province, China
- * E-mail:
| |
Collapse
|
17
|
Matheeussen V, Jungraithmayr W, De Meester I. Dipeptidyl peptidase 4 as a therapeutic target in ischemia/reperfusion injury. Pharmacol Ther 2012; 136:267-82. [DOI: 10.1016/j.pharmthera.2012.07.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 07/16/2012] [Indexed: 01/21/2023]
|
18
|
New aspects of the interactions between the cardiovascular nitric oxide system and natriuretic peptides. Biochem Biophys Res Commun 2011; 406:161-4. [DOI: 10.1016/j.bbrc.2011.02.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 02/11/2011] [Indexed: 11/20/2022]
|
19
|
Ren B, Wu H, Yin R, Xu L, Jing H, Li M, Jiang F, Wang Z. B-Type Natriuretic Peptide Pretreatment Attenuates Heart Ischemia-Reperfusion Injury in Rats. Transplant Proc 2010; 42:4496-8. [DOI: 10.1016/j.transproceed.2010.09.163] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 09/28/2010] [Indexed: 11/26/2022]
|
20
|
Kim T, Harman PK, Lyons R, Gaskins RB, Hobson CE, Evans SM, Tribble CG, Ejaz AA, Beaver TM. Brain Natriuretic Peptide is Not Reno-Protective during Renal Ischemia-Reperfusion Injury in the Rat. J Surg Res 2010; 164:e13-9. [DOI: 10.1016/j.jss.2010.06.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 06/11/2010] [Accepted: 06/19/2010] [Indexed: 11/29/2022]
|
21
|
Recombinant human erythropoietin pretreatment attenuates heart ischemia-reperfusion injury in rats by suppressing the systemic inflammatory response. Transplant Proc 2010; 42:1595-7. [PMID: 20620481 DOI: 10.1016/j.transproceed.2009.11.050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Accepted: 11/24/2009] [Indexed: 11/24/2022]
Abstract
BACKGROUND Ischemia-reperfusion (I/R) injury may influence graft function after transplantation. Erythropoietin (EPO) attenuates I/R injury in various animal organs such as intestine, brain, and kidney. OBJECTIVE To evaluate the effects of pretreatment with recombinant human EPO (rhEPO) on I/R-induced heart injury. MATERIALS AND METHODS A rat model of I/R injury was established by ligating the left descending coronary artery for 30 minutes, followed by reperfusion for 4 hours. Fifty Sprague-Dawley rats were divided into 5 groups: sham operation; I/R; I/R+rhEPO, 100 U/kg; I/R+rhEPO, 1000 U/kg; and I/R+rhEPO, 5000 U/kg. Electrocardiograms were assessed continuously to note arrhythmia caused by reperfusion. Serum concentrations of interleukin (IL)-6 and IL-8, and tumor necrosis factor-alpha were measured at 2 and 4 hours after reperfusion. RESULTS The rhEPO-treated animals exhibited dosage-dependent significant reduction in the incidence of ventricular arrhythmia caused by reperfusion, and markedly decreased serum concentrations of IL-6, IL-8, and tumor necrosis factor-alpha (P < .05) compared with the I/R group (P < .05). CONCLUSION The rhEPO attenuates myocardial I/R injury in rats, at least in part related to inhibition of the system inflammatory response.
Collapse
|
22
|
Long-term effects of B-type natriuretic peptide infusion after acute myocardial infarction in a rat model. J Cardiovasc Pharmacol 2010; 55:14-20. [PMID: 19858735 DOI: 10.1097/fjc.0b013e3181c5e743] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The effects of exogenous B-type natriuretic peptide (BNP) on postmyocardial infarction (MI) are not known. We tested the hypothesis that in vivo infusion of BNP would improve cardiac function and affect left ventricular (LV) remodeling in an experimental model of MI. METHODS MI was induced by coronary ligation in rats and confirmed by echocardiography. 19 rats were randomized to 1 of 3 groups: sham (n = 7), MI + saline (n = 5), MI + BNP (400 ng.kg(-1).minute(-1)) (n = 7). Infusions were delivered for 7 days via venous catheters tunneled to an infusion pump. Rats were followed for 8 weeks. Echocardiography, hemodynamics, histology, and in vivo and ex vivo pressure-volume relationships were examined. RESULTS LV systolic pressure, LV dP/dtmax, and infarct size improved with BNP treatment versus control MI group (132 +/- 4 vs.110 +/- 2 mm Hg, 8097 +/- 317 vs. 5816 +/- 378 mm Hg/s, 19.3% +/- 1.6% vs. 23.3% +/- 1.9%, respectively; all P < 0.05). Ex vivo end-diastolic pressure-volume relationship demonstrated reduced diastolic dysfunction after BNP therapy (P < 0.05 vs. control MI). Serum BNP levels confirmed delivery of BNP. CONCLUSIONS We demonstrate beneficial effects on LV function and decreased LV remodeling with BNP infusion in an experimental model of acute MI.
Collapse
|
23
|
He JG, Chen YL, Chen BL, Huang YY, Yao FJ, Chen SL, Dong YG. B-type natriuretic peptide attenuates cardiac hypertrophy via the transforming growth factor-ß1/smad7 pathway in vivo and in vitro. Clin Exp Pharmacol Physiol 2009; 37:283-9. [PMID: 19719752 DOI: 10.1111/j.1440-1681.2009.05281.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. Previously, we showed that long-term treatment of rats after myocardial infarction (MI) with B-type natriuretic peptide (BNP) prevented ventricular remodelling. However, it is unclear whether long-term BNP treatment affects cardiac hypertrophy and, if so, its mechanism of action. In the present study, we investigated the effects of long-term BNP treatment on cardiac hypertrophy and the molecular mechanisms involved. 2. Cardiac hypertrophy was established in rats by ligation of the left anterior descending coronary artery. After treatment with BNP (5 or 15 microg/kg per day) for 8 weeks, indices of cardiac hypertrophy were determined. In separate in vitro experiments, cardiomyocyte hypertrophy was induced by treatment of cardiomyocytes with 10(-6) mol/L angiotensin (Ang) II for 48 h and cell surface area and [(3)H] incorporation were measured. Transforming growth factor (TGF)-beta1 and smad7 mRNA and protein expression in vivo and in vitro were detected using reverse transcription-polymerase chain reaction and western blotting. 3. Long-term BNP treatment dose-dependently attenuated cardiac hypertrophy and improved cardiac function in rats after MI. Furthermore, BNP attenuated the upregulation of TGF-beta1 and downregulation of smad7 mRNA and protein expression. The in vitro experiments further proved that BNP inhibited cardiac hypertrophy and changes in the TGF-beta1/smad7 pathway, which were completely blocked by the cyclic GMP-dependent protein kinase (PKG) inhibitor, KT5823 (cells were treated with 10(-6) mol/L KT5823 for 48 h). 4. The results of the present study demonstrate that long-term treatment of rats with BNP dose-dependently attenuates cardiac hypertrophy and that this is associated with downregulation of TGF-beta1 and upregulation of smad7 via PKG signalling. Long-term BNP treatment may be a new therapeutic strategy to prevent cardiac hypertrophy and progression to heart failure.
Collapse
Affiliation(s)
- Jian-Gui He
- Department of Cardiology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
24
|
George I, Morrow B, Xu K, Yi GH, Holmes J, Wu EX, Li Z, Protter AA, Oz MC, Wang J. Prolonged effects of B-type natriuretic peptide infusion on cardiac remodeling after sustained myocardial injury. Am J Physiol Heart Circ Physiol 2009; 297:H708-17. [PMID: 19525373 DOI: 10.1152/ajpheart.00661.2008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
B-type natriuretic peptide (BNP) is an established first-line therapy for acute decompensated heart failure (HF), but its efficacy in preventing left ventricular (LV) remodeling after myocardial injury is unknown. The goal of this study was to evaluate the effects of BNP therapy on remodeling after ischemic injury in an awake canine model. Dogs were chronically instrumented for hemodynamics. Ischemia was created by daily coronary embolization (Embo; 3.1 x 10(4) beads/day) for 3 wk; 60 min after the first embolization, BNP (100 ng x kg(-1) x min(-1); n = 6) or saline (control; n = 6) was continuously infused via a left atrial catheter for 3 wk. Hemodynamics and echocardiography were performed in an awake state at baseline, 3 wk after Embo + BNP infusion, and 4 wk after stopping Embo + BNP infusion. End-systolic elastance (E(es)) and LV change in pressure over time (dP/dt) were preserved throughout Embo + BNP therapy versus control therapy (E(es): 3.76 +/- 1.01 vs. 1.41 +/- 0.16 mmHg/ml; LV dP/dt: 2,417 +/- 96 vs. 2,068 +/- 95 mmHg/s; both P < 0.05 vs. control). LV end-diastolic dimension was significantly smaller in BNP-treated dogs compared with control dogs (4.29 +/- 0.10 vs. 4.77 +/- 0.17 cm), and ejection fraction was maintained in treated dogs vs. control dogs (53 +/- 1% vs. 46 +/- 2%) (both P < 0.05 vs. control). Cyclooxygenase (COX)-2 expression in terminal LV tissue was significantly reduced after BNP therapy. Treatment with continuous infusion of BNP preserved LV geometry, improved systolic function, and prevented the progression of systolic HF after persistent ischemic injury.
Collapse
Affiliation(s)
- Isaac George
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fox AA, Collard CD, Shernan SK, Seidman CE, Seidman JG, Liu KY, Muehlschlegel JD, Perry TE, Aranki SF, Lange C, Herman DS, Meitinger T, Lichtner P, Body SC. Natriuretic peptide system gene variants are associated with ventricular dysfunction after coronary artery bypass grafting. Anesthesiology 2009; 110:738-47. [PMID: 19326473 PMCID: PMC2735337 DOI: 10.1097/aln.0b013e31819c7496] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ventricular dysfunction (VnD) after primary coronary artery bypass grafting is associated with increased hospital stay and mortality. Natriuretic peptides have compensatory vasodilatory, natriuretic, and paracrine influences on myocardial failure and ischemia. The authors hypothesized that natriuretic peptide system gene variants independently predict risk of VnD after primary coronary artery bypass grafting. METHODS A total of 1,164 patients undergoing primary coronary artery bypass grafting with cardiopulmonary bypass at two institutions were prospectively enrolled. After prospectively defined exclusions, 697 patients of European descent (76 with VnD) were analyzed. VnD was defined as need for at least 2 new inotropes and/or new mechanical ventricular support after coronary artery bypass grafting. A total of 139 haplotype-tagging single nucleotide polymorphisms (SNPs) within 7 genes (NPPA, NPPB, NPPC, NPR1, NPR2, NPR3, CORIN) were genotyped. SNPs univariately associated with VnD were entered into logistic regression models adjusting for clinical covariates predictive of VnD. To control for multiple comparisons, permutation analyses were conducted for all SNP associations. RESULTS After adjusting for clinical covariates and multiple comparisons within each gene, seven NPPA/NPPB SNPs (rs632793, rs6668352, rs549596, rs198388, rs198389, rs6676300, rs1009592) were associated with decreased risk of postoperative VnD (additive model; odds ratios 0.44-0.55; P = 0.010- 0.036) and four NPR3 SNPs (rs700923, rs16890196, rs765199, rs700926) were associated with increased risk of postoperative VnD (recessive model; odds ratios 3.89-4.28; P = 0.007-0.034). CONCLUSIONS Genetic variation within the NPPA/NPPB and NPR3 genes is associated with risk of VnD after primary coronary artery bypass grafting. Knowledge of such genotypic predictors may result in better understanding of the molecular mechanisms underlying postoperative VnD.
Collapse
Affiliation(s)
- Amanda A Fox
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Impact of Nesiritide on Renal Function and Mortality in Patients Suffering from Heart Failure. Cardiovasc Drugs Ther 2009; 23:221-33. [DOI: 10.1007/s10557-009-6167-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Wu B, Jiang H, Lin R, Cui B, Wen H, Lu Z. Pretreatment with B-type Natriuretic Peptide Protects the Heart From Ischemia-Reperfusion Injury by Inhibiting Myocardial Apoptosis. TOHOKU J EXP MED 2009; 219:107-14. [DOI: 10.1620/tjem.219.107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Bing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University
- Department of Cardiology, First Hospital of Quanzhou Affiliated to Fujian Medical University
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University
| | - Rong Lin
- Department of Cardiology, First Hospital of Quanzhou Affiliated to Fujian Medical University
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University
| | - Huazhi Wen
- Department of Cardiology, Renmin Hospital of Wuhan University
| | - Zhibing Lu
- Department of Cardiology, Renmin Hospital of Wuhan University
| |
Collapse
|
28
|
Ishibashi Y, Takahashi N, Tokumaru A, Karino K, Sugamori T, Sakane T, Kodani N, Kunizawa Y, Yoshitomi H, Sato H, Oyake N, Murakami Y, Shimada T. Activation of inducible NOS in peripheral vessels and outcomes in heart failure patients. J Card Fail 2008; 14:724-31. [PMID: 18995176 DOI: 10.1016/j.cardfail.2008.06.450] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Revised: 06/25/2008] [Accepted: 06/30/2008] [Indexed: 11/13/2022]
Abstract
BACKGROUND Activation of inducible nitric oxide synthase (iNOS) has been reported in congestive heart failure (CHF) conditions. However, it is unknown whether activation of iNOS affects prognosis of CHF patients. We prospectively studied the influence of activation of iNOS in the forearm on the outcome of CHF patients. METHODS AND RESULTS Forearm blood flow (FBF) responses to 3 doses of acetylcholine (ACh) and nitroglycerin (NTG), and 4 doses of a selective iNOS inhibitor (aminoguanidine: Amn) and a nonselective NOS inhibitor (L-NMMA) were examined using plethysmography in 68 patients with CHF from idiopathic dilated cardiomyopathy. Plasma brain natriuretic peptide (BNP) and tumor necrosis factor-alpha (TNF-alpha) were also measured in all patients. During the mean follow-up period of 3.8 years, 25 patients were hospitalized for worsening heart failure and 9 of these patients died. Patients with adverse events had a diminished vasodilator response to ACh (P < .001) compared to patients without adverse events. Amn significantly decreased FBF (P < .001) in patients with adverse events, but not in patients without adverse events. FBF responses to NTG and L-NMMA were not significantly different between the 2 groups. When grouped by maximum FBF responses to each drug above and below the median value, multivariate Cox proportional hazards model analyses for cardiac event showed a significance in the FBF response to Amn (adjusted hazard ratio 5.89, P < .001). FBF responses to maximum dose of Amn significantly correlated with BNP and TNF-alpha levels (both P < .001). CONCLUSIONS CHF patients with vascular iNOS activation, as demonstrated by a greater vasoconstrictor response to Amn, had poor outcomes. Activation of iNOS in peripheral vessels, associated with proinflammatory cytokines in accordance to the severity of heart failure, is a marker for, or contributes to, adverse events in patients with CHF.
Collapse
Affiliation(s)
- Yutaka Ishibashi
- Divisions of Cardiovascular Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, Shimane, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Affiliation(s)
- Richard Bogle
- Experimental Medicine and Toxicology, Imperial College London, Hammersmith Hospital, London W12 0HS, UK
| | | |
Collapse
|
30
|
Burley DS, Baxter GF. B-type natriuretic peptide at early reperfusion limits infarct size in the rat isolated heart. Basic Res Cardiol 2007; 102:529-41. [PMID: 17896117 DOI: 10.1007/s00395-007-0672-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 07/10/2007] [Accepted: 07/26/2007] [Indexed: 10/22/2022]
Abstract
Natriuretic peptides are regulatory autacoids in the mammalian myocardium whose functions, mediated via particulate guanylyl cyclase/cGMP, may include cytoprotection against ischaemia-reperfusion injury. Previous work has identified that B-type natriuretic peptide (BNP) limits infarct size when administered prior to and during coronary occlusion through a K(ATP) channel-dependent mechanism. The present study examined the hypothesis that the protection afforded by BNP is mediated specifically at reperfusion in a postconditioning-like manner. Langendorff-perfused rat hearts were subjected to 35 min coronary artery occlusion and 120 min reperfusion, and infarct size was determined by tetrazolium staining. Postconditioning was effected by applying six 10-second periods of global ischaemia at the onset of reperfusion.Treatment with either BNP 10 nM or the NO donor S-nitroso-N-acetylpenicillamine (SNAP) 1-10 microM was commenced 5 min prior to reperfusion and continued until 10 min after reperfusion. Control infarct size (% of ischaemic risk zone) was 40.8 +/- 3.7%.BNP at reperfusion induced a significant limitation of infarct size (BNP 22.9 +/- 4.1% P<0.05 vs. control). Co-treatment at reperfusion with BNP and the K(ATP) channel blockers 5-hydroxydecanote (5HD, 100 microM), glibenclamide (Glib; 10 microM) or HMR1098 (10 microM) abolished the infarct-limiting effect of BNP (BNP + 5HD 41.0 +/- 3.9%, BNP + Glib 39.8 +/- 5.6%, BNP + HMR 1098 46.0 +/- 7.1%,P < 0.05 vs. BNP). BNP given together with L-NAME (100 microM) at reperfusion resulted in a marked loss of protection (BNP + L-NAME 53.1 +/- 3.8% P < 0.001 vs. BNP). In a second series of experiments, SNAP (1-10 microM) given at reperfusion was found not to be protective (SNAP 1 microM 30.2 +/- 4.9%, SNAP 2 microM 27.5 +/- 9.5%, SNAP 5 microM 39.2 +/- 5.7%, SNAP 10 microM 33.7 +/- 6.4%, not significant vs. control). In a third series of experiments, postconditioning significantly limited infarct size (14.9 +/- 3.6 % vs. control 34.5 +/- 4.9%, P < 0.01) and this effect of postconditioning was abolished in the presence of isatin (100 microM), a non-specific blocker of particulate guanylyl cyclases (35.1 +/- 6%, P < 0.05 vs. postconditioning). In conclusion, pharmacological activation of pGC by BNP can effectively induce protection against reperfusion injury, by mechanisms involving K(ATP) channel opening and endogenous NO synthase activation. Furthermore, endogenous activation of pGC could play a role in the mechanism of postconditioning.
Collapse
Affiliation(s)
- Dwaine S Burley
- The Royal Veterinary College, University of London, London, UK
| | | |
Collapse
|
31
|
Burley DS, Ferdinandy P, Baxter GF. Cyclic GMP and protein kinase-G in myocardial ischaemia-reperfusion: opportunities and obstacles for survival signaling. Br J Pharmacol 2007; 152:855-69. [PMID: 17700722 PMCID: PMC2078226 DOI: 10.1038/sj.bjp.0707409] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is clear that multiple signalling pathways regulate the critical balance between cell death and survival in myocardial ischaemia-reperfusion. Recent attention has focused on the activation of survival or salvage kinases, particularly during reperfusion, as a common mechanism of many cardioprotective interventions. The phosphatidyl inositol 3'-hydroxy kinase/Akt complex (PI3K/Akt) and p42/p44 mitogen-activated protein kinase cascades have been widely promoted in this respect but the cyclic guanosine 3',5'-monophosphate/cGMP-dependent protein kinase (cGMP/PKG) signal transduction cassette has been less systematically investigated as a survival cascade. We propose that activation of the cGMP/PKG signalling pathway, following activation of soluble or particulate guanylate cyclases, may play a pivotal role in survival signalling in ischaemia-reperfusion, especially in the classical preconditioning, delayed preconditioning and postconditioning paradigms. The resurgence of interest in reperfusion injury, largely as a result of postconditioning-related research, has confirmed that the cGMP/PKG pathway is a pivotal salvage mechanism in reperfusion. Numerous studies suggest that the infarct-limiting effects of preconditioning and postconditioning, exogenously donated nitric oxide (NO), natriuretic peptides, phosphodiesterase inhibitors, and other diverse drugs and mediators such as HMG co-A reductase inhibitors (statins), Rho-kinase inhibitors and adrenomedullin, whether given before and during ischaemia, or specifically at the onset of reperfusion, may be mediated by activation or enhancement of the cGMP pathway, either directly or indirectly via endogenous NO generation downstream of PI3K/Akt. Putative mechanisms of protection include PKG regulation of Ca(2+) homeostasis through the modification of sarcoplasmic reticulum Ca(2+) uptake mechanisms, and PKG-induced opening of ATP-sensitive K(+) channels during ischaemia and/or reperfusion. At present, significant technical obstacles in defining the precise roles played by cGMP/PKG signalling include the heavy reliance on pharmacological PKG inhibitors of uncertain selectivity, difficulties in determining PKG activity in intact tissue, and the growing recognition that intracellular compartmentalisation of the cGMP pool may contribute markedly to the nucleotide's biological actions and biochemical determination. Overall, the body of experimental evidence suggests that cGMP/PKG survival signalling ameliorates irreversible injury associated with ischaemia-reperfusion and may be a tractable therapeutic target.
Collapse
Affiliation(s)
- D S Burley
- Division of Pharmacology, Welsh School of Pharmacy, Cardiff University Cardiff, UK
| | - P Ferdinandy
- Cardiovascular Research Group, University of Szeged Szeged, Hungary
| | - G F Baxter
- Division of Pharmacology, Welsh School of Pharmacy, Cardiff University Cardiff, UK
- Author for correspondence:
| |
Collapse
|