1
|
Wang K, Zhu Q, Liu W, Wang L, Li X, Zhao C, Wu N, Ma C. Mitochondrial apoptosis in response to cardiac ischemia-reperfusion injury. J Transl Med 2025; 23:125. [PMID: 39875870 PMCID: PMC11773821 DOI: 10.1186/s12967-025-06136-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
In patients with acute myocardial infarction (AMI), thrombolytic therapy and revascularization strategies allow complete recanalization of occluded epicardial coronary arteries. However, approximately 35% of patients still experience myocardial ischemia/reperfusion (I/R) injury, which contributing to increased AMI mortality. Therefore, an accurate understanding of myocardial I/R injury is important for preventing and treating AMI. The death of each cell (cardiomyocytes, endothelial cells, vascular smooth muscle cells, cardiac fibroblasts, and mesenchymal stem cells) after myocardial ischemia/reperfusion is associated with apoptosis due to mitochondrial dysfunction. Abnormal opening of the mitochondrial permeability transition pore, aberrant mitochondrial membrane potential, Ca2+ overload, mitochondrial fission, and mitophagy can lead to mitochondrial dysfunction, thereby inducing mitochondrial apoptosis. The manifestation of mitochondrial apoptosis varies according to cell type. Here, we reviewed the characteristics of mitochondrial apoptosis in cardiomyocytes, endothelial cells, vascular smooth muscle cells, cardiac fibroblasts, and mesenchymal stem cells following myocardial ischemia/reperfusion.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Wen Liu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Linyuan Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China
| | - Nan Wu
- The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China.
- Clinical Medical Research Center of Imaging in Liaoning Province, Shenyang, China.
| |
Collapse
|
2
|
Role of c-Src and reactive oxygen species in cardiovascular diseases. Mol Genet Genomics 2023; 298:315-328. [PMID: 36700976 DOI: 10.1007/s00438-023-01992-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
Oxidative stress, caused by the over production of oxidants or inactivity of antioxidants, can modulate the redox state of several target proteins such as tyrosine kinases, mitogen-activated protein kinases and tyrosine phosphatases. c-Src is one such non-receptor tyrosine kinase which activates NADPH oxidases (Noxs) in response to various growth factors and shear stress. Interaction between c-Src and Noxs is influenced by cell type and primary messengers such as angiotensin II, which binds to G-protein coupled receptor and activates the intracellular signaling cascade. c-Src stimulated activation of Noxs results in elevated release of intracellular and extracellular reactive oxygen species (ROS). These ROS species disturb vascular homeostasis and cause cardiac hypertrophy, coronary artery disease, atherosclerosis and hypertension. Interaction between c-Src and ROS in the pathobiology of cardiac fibrosis is hypothesized to be influenced by cell type and stimuli. c-Src and ROS have a bidirectional relationship, thus increased ROS levels due to c-Src mediated activation of Noxs can further activate c-Src by promoting the oxidation and sulfenylation of critical cysteine residues. This review highlights the role of c-Src and ROS in mediating downstream signaling pathways underlying cardiovascular diseases. Furthermore, due to the central role of c-Src in activation of various signaling proteins involved in differentiation, migration, proliferation, and cytoskeletal reorganization of vascular cells, it is presented as therapeutic target for treating cardiovascular diseases except cardiac fibrosis.
Collapse
|
3
|
Nejat R, Torshizi MF, Najafi DJ. S Protein, ACE2 and Host Cell Proteases in SARS-CoV-2 Cell Entry and Infectivity; Is Soluble ACE2 a Two Blade Sword? A Narrative Review. Vaccines (Basel) 2023; 11:204. [PMID: 36851081 PMCID: PMC9968219 DOI: 10.3390/vaccines11020204] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Since the spread of the deadly virus SARS-CoV-2 in late 2019, researchers have restlessly sought to unravel how the virus enters the host cells. Some proteins on each side of the interaction between the virus and the host cells are involved as the major contributors to this process: (1) the nano-machine spike protein on behalf of the virus, (2) angiotensin converting enzyme II, the mono-carboxypeptidase and the key component of renin angiotensin system on behalf of the host cell, (3) some host proteases and proteins exploited by SARS-CoV-2. In this review, the complex process of SARS-CoV-2 entrance into the host cells with the contribution of the involved host proteins as well as the sequential conformational changes in the spike protein tending to increase the probability of complexification of the latter with angiotensin converting enzyme II, the receptor of the virus on the host cells, are discussed. Moreover, the release of the catalytic ectodomain of angiotensin converting enzyme II as its soluble form in the extracellular space and its positive or negative impact on the infectivity of the virus are considered.
Collapse
Affiliation(s)
- Reza Nejat
- Department of Anesthesiology and Critical Care Medicine, Laleh Hospital, Tehran 1467684595, Iran
| | | | | |
Collapse
|
4
|
Espitia-Corredor JA, Boza P, Espinoza-Pérez C, Lillo JM, Rimassa-Taré C, Machuca V, Osorio-Sandoval JM, Vivar R, Bolivar S, Pardo-Jiménez V, Sánchez-Ferrer CF, Peiró C, Díaz-Araya G. Angiotensin II Triggers NLRP3 Inflammasome Activation by a Ca 2+ Signaling-Dependent Pathway in Rat Cardiac Fibroblast Ang-II by a Ca 2+-Dependent Mechanism Triggers NLRP3 Inflammasome in CF. Inflammation 2022; 45:2498-2512. [PMID: 35867264 DOI: 10.1007/s10753-022-01707-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 06/09/2022] [Indexed: 11/05/2022]
Abstract
Angiotensin II (Ang-II) is a widely studied hypertensive, profibrotic, and pro-inflammatory peptide. In the heart, cardiac fibroblasts (CF) express type 1 angiotensin II receptors (AT1R), Toll-like receptor-4 (TLR4), and the NLRP3 inflammasome complex, which play important roles in pro-inflammatory processes. When activated, the NLRP3 inflammasome triggers proteolytic cleavage of pro-IL-1, resulting in its activation. However, in CF the mechanism by which Ang-II assembles and activates the NLRP3 inflammasome remains not fully known. To elucidate this important point, we stimulated TLR4 receptors in CF and evaluated the signaling pathways by which Ang-II triggers the assembly and activity. In cultured rat CF, pro-IL-1β levels, NLRP3, ASC, and caspase-1 expression levels were determined by Western blot. NLRP3 inflammasome complex assembly was analyzed by immunocytochemistry, whereas by ELISA, we analyzed NLRP3 inflammasome activity and [Formula: see text] release. In CF, Ang-II triggered NLRP3 inflammasome assembly and caspase-1 activity; and in LPS-pretreated CF, Ang-II also triggered [Formula: see text] secretion. These effects were blocked by losartan (AT1R antagonist), U73221 (PLC inhibitor), 2-APB (IP3R antagonist), and BAPTA-AM (Ca2+ chelator) indicating that the AT1R/PLC/IP3R/Ca2+ pathway is involved. Finally, bafilomycin A1 prevented Ang-II-induced [Formula: see text] secretion, indicating that a non-classical protein secretion mechanism is involved. These findings suggest that in CF, Ang-II by a Ca2+-dependent mechanism triggers NLRP3 inflammasome assembly and activation leading to [Formula: see text] secretion through a non-conventional protein secretion mechanism.
Collapse
Affiliation(s)
- Jenaro Antonio Espitia-Corredor
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile.,Faculty of Medicine, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.,PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain.,Faculty of Chemical and Pharmaceutical Sciences, Advanced Center of Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Pía Boza
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Claudio Espinoza-Pérez
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - José Miguel Lillo
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Constanza Rimassa-Taré
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Víctor Machuca
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - José Miguel Osorio-Sandoval
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Raúl Vivar
- Molecular and Clinical Pharmacology Program, Faculty of Medicine, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile
| | - Samir Bolivar
- Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla, Colombia
| | - Viviana Pardo-Jiménez
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Carlos Félix Sánchez-Ferrer
- Faculty of Medicine, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Concepción Peiró
- Faculty of Medicine, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Guillermo Díaz-Araya
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile. .,Faculty of Chemical and Pharmaceutical Sciences, Advanced Center of Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.
| |
Collapse
|
5
|
Salama A, Mansour D, Hegazy R. The cardio and renoprotective role of ginseng against epinephrine-induced myocardial infarction in rats: Involvement of angiotensin II type 1 receptor/protein kinase C. Toxicol Rep 2021; 8:908-919. [PMID: 33996504 PMCID: PMC8099916 DOI: 10.1016/j.toxrep.2021.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/26/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022] Open
Abstract
Epinephrine induced MI with renal complication through Nrf2/NF-κB imbalance and PKC/AT1R. Ginseng abolished ECG changes induced by epinephrine and stimulated Nrf2. Ginseng reduced upregulation of PKC, NF-κB, and AT1R induced by epinephrine. Ginseng inhibited iNOS and corrected renal disorder in epinephrine model of MI.
The expression of angiotensin II type 1 receptor (AT1 receptor)/protein kinase C (PKC) in heart tissues has a vital role in myocardial infarction (MI). The current work aimed to clarify the renal complication enhanced by MI following epinephrine injection via AT1 receptor/ PKC expression; in addition, the impact of ginseng extract on epinephrine-induced MI and its renal complication was assessed. Adult male albino Wistar rats were pretreated orally with ginseng extract (200 & 400 mg/kg/day) for 14 days, then two successive doses of epinephrine injection (100 mg/kg, i.p.). Epinephrine evoked electrocardiographic (ECG) and renal changes accompanied with a significant increase in heart and kidney contents of malodialdehyde (MDA), nitric oxide (NO), protein kinase C (PKC), heart contents of nuclear factor-kabba B (NF-κB) and angiotensin 1receptor (AT1R), as well as a decrease in heart and kidney reduced glutathione (GSH) and nuclear factor-erythroid-related factor 2 (Nrf2) contents. In histopathological investigations epinephrine exhibited deleterious heart changes in the form of acute MI with the presence of necrosis of cardiomyocytes with iNOS expression and produced glomerulus and renal tubules degeneration. Pretreatment of rats with ginseng extract in both doses significantly corrected epinephrine-induced heart and renal changes. The current work revealed that epinephrine-induced MI associated with aggravated renal complication and ginseng extract has cardio and reno protective role against this as it reduces infarct size, preserves cardiac and renal tissues and functions through activating Nrf2 and down-regulating NF-κB, PKC, AT1R and iNOS.
Collapse
Affiliation(s)
- Abeer Salama
- Pharmacology Department, Medical Division, National Research Centre, 33 El Bohouth St. (Former El-Tahrir St.), 12622 Dokki, Cairo, Egypt
| | - Dina Mansour
- Pharmacology Department, Medical Division, National Research Centre, 33 El Bohouth St. (Former El-Tahrir St.), 12622 Dokki, Cairo, Egypt
| | - Rehab Hegazy
- Pharmacology Department, Medical Division, National Research Centre, 33 El Bohouth St. (Former El-Tahrir St.), 12622 Dokki, Cairo, Egypt
| |
Collapse
|
6
|
Vivar R, Anfossi R, Humeres C, Catalán M, Reyes C, Cárdenas S, Contreras A, Aránguiz P, González F, Diaz-Araya G. FoxO1 is required for high glucose-dependent cardiac fibroblasts into myofibroblast phenoconversion. Cell Signal 2021; 83:109978. [PMID: 33722671 DOI: 10.1016/j.cellsig.2021.109978] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022]
Abstract
In the normal heart, cardiac fibroblasts (CFs) maintain extracellular matrix (ECM) homeostasis, whereas in pathological conditions, such as diabetes mellitus (DM), CFs converse into cardiac myofibroblasts (CMFs) and this CFs phenoconversion increase the synthesis and secretion of ECM proteins, promoting cardiac fibrosis and heart dysfunction. High glucose (HG) conditions increase TGF-β1 expression and FoxO1 activity, whereas FoxO1 is crucial to CFs phenoconversion induced by TGF-β1. In addition, FoxO1 increases CTGF expression, whereas CTGF plays an active role in the fibrotic process induced by hyperglycemia. However, the role of FoxO1 and CTGF in CFs phenoconversion induced by HG is not clear. In this study, we investigated the effects of FoxO1 pharmacological inhibition on CFs phenoconversion in both in vitro and ex vivo models of DM. Our results demonstrate that HG induces CFs phenoconversion and FoxO1 activation. Moreover, AS1842856, a pharmacological inhibitor of FoxO1 activity, prevents CFs phenoconversion and CTGF expression increase induced by HG, whereas these results were corroborated by FoxO1 silencing. Additionally, K252a, a pharmacological blocker of CTGF receptor, prevents HG-induced CFs phenoconversion, which was corroborated with CTGF expression knockdown. Furthermore, through CFs isolation from heart of diabetic rats, we showed that hyperglycemia induces FoxO1 activation, the increase of CTGF expression and CFs phenoconversion, whereas the FoxO1 activity inhibition reverses the effects induced by hyperglycemia on CFs. Altogether, our results demonstrate that FoxO1 and CTGF are necessary for CFs phenoconversion induced by HG and suggest that both proteins are likely to become a potential targeted drug for fibrotic response induced by hyperglycemic conditions.
Collapse
Affiliation(s)
- Raúl Vivar
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Renatto Anfossi
- Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudio Humeres
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Christopher Reyes
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Simone Cárdenas
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Contreras
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, 2520000 Viña del Mar, Chile
| | - Fabiola González
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Guillermo Diaz-Araya
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.; Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
7
|
Ding J, Tang Q, Luo B, Zhang L, Lin L, Han L, Hao M, Li M, Yu L, Li M. Klotho inhibits angiotensin II-induced cardiac hypertrophy, fibrosis, and dysfunction in mice through suppression of transforming growth factor-β1 signaling pathway. Eur J Pharmacol 2019; 859:172549. [PMID: 31325434 DOI: 10.1016/j.ejphar.2019.172549] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/13/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
Recent studies have revealed critical roles of transforming growth factor-β1 (TGF-β1) and microRNA-132 (miR-132), a downstream mediator of TGF-β1, in the pathogenesis of cardiac remodeling. In this study, we tested whether the antiaging protein klotho modifies angiotensin II (Ang II)-induced cardiac remodeling through regulating TGF-β1-miR-132 axis. We found that both klotho and the TGF-β1 inhibitor LY364947 significantly inhibited cardiac hypertrophy, fibrosis, and dysfunction in Ang II-infused mice, as evidenced by the ratios of heart weight to body weight (HW/BW), heart weight to tibial length (HW/TL), cardiomyocyte cross-sectional area, fibrotic area, and expression of prohypertrophic genes (ANP, β-MHC) and fibrotic marker genes (α-SMA, collagen I), echocardiographic parameters. Meanwhile, klotho also significantly inhibited Ang II-induced protein expression of TGF-β1 and phosphorylated Smad2/3 in the heart tissues and cultured cardiomyocytes and cardiac fibroblasts. In vitro experiments demonstrated that Ang II-induced cardiomyocyte hypertrophy and proliferation and activation of cardiac fibroblasts were markedly inhibited by klotho, LY364947 or the miR-132 inhibitor anti-miR-132. Both klotho and the TGF-β1 inhibitor LY364947 downregulated the miR-132 expression. Additionally, klotho decreased Ang II-induced protein expressions of cardiac fibroblast growth factor (FGF)23 in vivo and in vitro. The decreased protein levels of klotho in serum and renal tissues of Ang II-infused mice were elevated by klotho. Klotho downregulated the protein levels of TGF-β1 in renal tissues of Ang II-infused mice. In conclusion, our results suggest that klotho prevents Ang II-induced cardiac remodeling and dysfunction through modifying the TGF-β1-miR-132 axis, providing an experimental basis for clinical treatment on cardiac remodeling.
Collapse
Affiliation(s)
- Jieqiong Ding
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Qiong Tang
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Binhua Luo
- School of Pharmacy, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Lijun Zhang
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Li Lin
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Lu Han
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Miaomiao Hao
- School of Pharmacy, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Mingyue Li
- School of Pharmacy, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Liangzhu Yu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China.
| | - Mincai Li
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China.
| |
Collapse
|
8
|
Cai W, Zhong S, Zheng F, Zhang Y, Gao F, Xu H, Cai X, Lan J, Huang D, Shi G. Angiotensin II confers resistance to apoptosis in cardiac myofibroblasts through the AT1/ERK1/2/RSK1 pathway. IUBMB Life 2019; 71:261-276. [PMID: 30452117 DOI: 10.1002/iub.1967] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 02/05/2023]
Abstract
Myofibroblast apoptosis is essential for normal resolution of wound repair, including cardiac infarction repair. Impaired cardiac myofibroblast (CMF) apoptosis is associated with excessive extracellular matrix (ECM) deposition, which could be responsible for pathological cardiac fibrosis. Conventionally, angiotensin II (Ang II), a soluble peptide, is implicated in fibrogenesis because it induces cardiac fibroblast (CFb) proliferation, differentiation, and collagen synthesis. However, the role of Ang II in regulation of CMF survival and apoptosis has not been fully clarified. In this report, we cultured neonatal rat CFbs, which transform into CMFs after passage 3 (6-8 days), and investigated the effects of Ang II on CMFs challenged by TNF-α combined with cycloheximide and the underlying mechanisms. Here, we show that Ang II rapidly activates MAPKs but not AKT in CMFs and confers apoptosis resistance, as evidenced by the inhibition of caspase-3 cleavage, early apoptotic cells and late apoptotic cells. This inhibitory effect of Ang II was reversed by blockade of AT1 or inactivation of ERK1/2 or RSK1 but not AT2, indicating that activation of the prosurvival AT1/ERK1/2/RSK1 signaling pathway mediates apoptosis resistance. TGF-β, a latent fibrotic factor, was found to have no relation to Ang II-induced apoptosis resistance in our study. Furthermore, Ang II-mediated apoptosis resistance, which was conferred by activation of the AT1/ERK1/2/RSK1 signaling pathway, was also confirmed in human adult ventricular cardiac myofibroblasts. Collectively, our findings suggest a novel profibrotic mechanism of Ang II in which it promotes myofibroblast resistance to apoptosis in addition to classical mechanisms, providing a potential novel therapeutic approach by targeting prosurvival signaling pathways. © 2018 IUBMB Life, 71(1):261-276, 2019.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensin II Type 2 Receptor Blockers/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Butadienes/pharmacology
- Cell Survival/drug effects
- Cycloheximide/pharmacology
- Flavonoids/pharmacology
- Gene Expression Regulation
- Humans
- Imidazoles/pharmacology
- Losartan/pharmacology
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myofibroblasts/cytology
- Myofibroblasts/drug effects
- Myofibroblasts/metabolism
- Nitriles/pharmacology
- Primary Cell Culture
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Signal Transduction
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Wenfeng Cai
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Shuping Zhong
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Fuchun Zheng
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Hongquan Xu
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiangna Cai
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Junhong Lan
- Department of Plastic Surgery, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
- Pharmaceutical Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
- Department of Cardiovascular Diseases, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
9
|
Bian J, Zhang S, Yi M, Yue M, Liu H. The mechanisms behind decreased internalization of angiotensin II type 1 receptor. Vascul Pharmacol 2018; 103-105:1-7. [DOI: 10.1016/j.vph.2018.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/19/2018] [Accepted: 01/24/2018] [Indexed: 01/05/2023]
|
10
|
Phuong HTA, Yu L, Park BM, Kim SH. Comparative effects of angiotensin II and angiotensin-(4-8) on blood pressure and ANP secretion in rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:667-674. [PMID: 29200910 PMCID: PMC5709484 DOI: 10.4196/kjpp.2017.21.6.667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/26/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022]
Abstract
Angiotensin II (Ang II) is metabolized from N-terminal by aminopeptidases and from C-terminal by Ang converting enzyme (ACE) to generate several truncated angiotensin peptides (Angs). The truncated Angs have different biological effects but it remains unknown whether Ang-(4-8) is an active peptide. The present study was to investigate the effects of Ang-(4-8) on hemodynamics and atrial natriuretic peptide (ANP) secretion using isolated beating rat atria. Atrial stretch caused increases in atrial contractility by 60% and in ANP secretion by 70%. Ang-(4-8) (0.01, 0.1, and 1 µM) suppressed high stretch-induced ANP secretion in a dose-dependent manner. Ang-(4-8) (0.1 µM)-induced suppression of ANP secretion was attenuated by the pretreatment with an antagonist of Ang type 1 receptor (AT1R) but not by an antagonist of AT2R or AT4R. Ang-(4-8)-induced suppression of ANP secretion was attenuated by the pretreatment with inhibitor of phospholipase (PLC), inositol triphosphate (IP3) receptor, or nonspecific protein kinase C (PKC). The potency of Ang-(4-8) to inhibit ANP secretion was similar to Ang II. However, Ang-(4-8) 10 µM caused an increased mean arterial pressure which was similar to that by 1 nM Ang II. Therefore, we suggest that Ang-(4-8) suppresses high stretch-induced ANP secretion through the AT1R and PLC/IP3/PKC pathway. Ang-(4-8) is a biologically active peptide which functions as an inhibition mechanism of ANP secretion and an increment of blood pressure.
Collapse
Affiliation(s)
- Hoang Thi Ai Phuong
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju 54907, Korea
| | - Lamei Yu
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju 54907, Korea
| | - Byung Mun Park
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju 54907, Korea
| | - Suhn Hee Kim
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju 54907, Korea
| |
Collapse
|
11
|
A peptide vaccine targeting angiotensin II attenuates the cardiac dysfunction induced by myocardial infarction. Sci Rep 2017; 7:43920. [PMID: 28266578 PMCID: PMC5339733 DOI: 10.1038/srep43920] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 02/01/2017] [Indexed: 12/24/2022] Open
Abstract
A peptide vaccine targeting angiotensin II (Ang II) was recently developed as a novel treatment for hypertension to resolve the problem of noncompliance with pharmacotherapy. Ang II plays a crucial role in the pathogenesis of cardiac remodeling after myocardial infarction (MI), which causes heart failure. In the present study, we examined whether the Ang II vaccine is effective in preventing heart failure. The injection of the Ang II vaccine in a rat model of MI attenuated cardiac dysfunction in association with an elevation in the serum anti-Ang II antibody titer. Furthermore, any detrimental effects of the Ang II vaccine were not observed in the rats that underwent sham operations. Treatment with immunized serum from Ang II vaccine-injected rats significantly suppressed post-MI cardiac dysfunction in MI rats and Ang II-induced remodeling-associated signaling in cardiac fibroblasts. Thus, our present study demonstrates that the Ang II vaccine may provide a promising novel therapeutic strategy for preventing heart failure.
Collapse
|
12
|
Losartan attenuates aortic endothelial apoptosis induced by chronic intermittent hypoxia partly via the phospholipase C pathway. Sleep Breath 2017; 21:679-689. [PMID: 28190165 DOI: 10.1007/s11325-017-1479-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/31/2016] [Accepted: 02/03/2017] [Indexed: 10/20/2022]
Abstract
PURPOSE Endoplasmic reticulum (ER) stress is known to play key roles in the development of endothelial cell apoptosis induced by chronic intermittent hypoxia (CIH), and the angiotensin II-phospholipase C-inositol-1,4,5-triphosphate (AngII-PLC-IP3) pathway has been demonstrated to induce ER stress. To explore whether the AngII-PLC-IP3 pathway is involved in the vascular damage induced by CIH, we examined whether the AngII-PLC-IP3 pathway is involved in ER stress induced by CIH and whether losartan, a selective angiotensin II type 1 receptor (AT1R) blocker, could suppress endothelial cell apoptosis induced by CIH. METHODS Adult male Sprague Dawley rats were subjected to 8 h/day of intermittent hypoxia/normoxia, with or without losartan, a selective AT1R blocker, and/or U73122, a selective PLC inhibitor, for 8 weeks. Endothelial cell apoptosis, ER stress markers, and levels of PLC-γ1 and IP3R expression were determined. RESULTS Losartan prevented increases in PLC-γ1 and IP3R protein levels and inhibited ER stress markers induced by CIH. Addition of U73122 reproduced all the protective effects of losartan. Losartan administration before CIH significantly ameliorated CIH-induced endothelial cell apoptosis. CONCLUSIONS This study showed that the AngII-PLC-IP3 pathway is involved in ER stress induced by CIH and that pre-losartan administration ameliorates endothelial cell apoptosis following CIH partly via inhibition of the AngII-PLC-IP3 pathway and ER stress.
Collapse
|
13
|
Díaz-Araya G, Vivar R, Humeres C, Boza P, Bolivar S, Muñoz C. Cardiac fibroblasts as sentinel cells in cardiac tissue: Receptors, signaling pathways and cellular functions. Pharmacol Res 2015; 101:30-40. [PMID: 26151416 DOI: 10.1016/j.phrs.2015.07.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 12/24/2022]
Abstract
Cardiac fibroblasts (CF) not only modulate extracellular matrix (ECM) proteins homeostasis, but also respond to chemical and mechanical signals. CF express a variety of receptors through which they modulate the proliferation/cell death, autophagy, adhesion, migration, turnover of ECM, expression of cytokines, chemokines, growth factors and differentiation into cardiac myofibroblasts (CMF). Differentiation of CF to CMF involves changes in the expression levels of various receptors, as well as, changes in cell phenotype and their associated functions. CF and CMF express the β2-adrenergic receptor, and its stimulation activates PKA and EPAC proteins, which differentially modulate the CF and CMF functions mentioned above. CF and CMF also express different levels of Angiotensin II receptors, in particular, AT1R activation increases collagen synthesis and cell proliferation, but its overexpression activates apoptosis. CF and CMF express different levels of B1 and B2 kinin receptors, whose stimulation by their respective agonists activates common signaling transduction pathways that decrease the synthesis and secretion of collagen through nitric oxide and prostacyclin I2 secretion. Besides these classical functions, CF can also participate in the inflammatory response of cardiac repair, through the expression of receptors commonly associated to immune cells such as Toll like receptor 4, NLRP3 and interferon receptor. The activation by their respective agonists modulates the cellular functions already described and the release of cytokines and chemokines. Thus, CF and CMF act as sentinel cells responding to a plethora of stimulus, modifying their own behavior, and that of neighboring cells.
Collapse
Affiliation(s)
- G Díaz-Araya
- Laboratory of Molecular Pharmacology, Chemical Pharmacological and Toxicological Department, Faculty of Chemical and Pharmaceutical Sciences, FONDAP Advanced Center for Chronic diseases ACCDiS, University of Chile, Santiago, Chile.
| | - R Vivar
- Laboratory of Molecular Pharmacology, Chemical Pharmacological and Toxicological Department, Faculty of Chemical and Pharmaceutical Sciences, FONDAP Advanced Center for Chronic diseases ACCDiS, University of Chile, Santiago, Chile
| | - C Humeres
- Laboratory of Molecular Pharmacology, Chemical Pharmacological and Toxicological Department, Faculty of Chemical and Pharmaceutical Sciences, FONDAP Advanced Center for Chronic diseases ACCDiS, University of Chile, Santiago, Chile
| | - P Boza
- Laboratory of Molecular Pharmacology, Chemical Pharmacological and Toxicological Department, Faculty of Chemical and Pharmaceutical Sciences, FONDAP Advanced Center for Chronic diseases ACCDiS, University of Chile, Santiago, Chile
| | - S Bolivar
- Laboratory of Molecular Pharmacology, Chemical Pharmacological and Toxicological Department, Faculty of Chemical and Pharmaceutical Sciences, FONDAP Advanced Center for Chronic diseases ACCDiS, University of Chile, Santiago, Chile
| | - C Muñoz
- Laboratory of Molecular Pharmacology, Chemical Pharmacological and Toxicological Department, Faculty of Chemical and Pharmaceutical Sciences, FONDAP Advanced Center for Chronic diseases ACCDiS, University of Chile, Santiago, Chile
| |
Collapse
|
14
|
Zhao L, Xu C, Xu J. Autoantibodies against β1 Receptor and AT1 Receptor in Type 2 Diabetes Patients with Left Ventricular Dilatation. Cardiology 2014; 129:191-6. [DOI: 10.1159/000365782] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/04/2014] [Indexed: 11/19/2022]
|
15
|
Vivar R, Humeres C, Ayala P, Olmedo I, Catalán M, García L, Lavandero S, Díaz-Araya G. TGF-β1 prevents simulated ischemia/reperfusion-induced cardiac fibroblast apoptosis by activation of both canonical and non-canonical signaling pathways. Biochim Biophys Acta Mol Basis Dis 2013; 1832:754-62. [PMID: 23416528 DOI: 10.1016/j.bbadis.2013.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 12/30/2012] [Accepted: 02/07/2013] [Indexed: 12/01/2022]
Abstract
Ischemia/reperfusion injury is a major cause of myocardial death. In the heart, cardiac fibroblasts play a critical role in healing post myocardial infarction. TGF-β1 has shown cardioprotective effects in cardiac damage; however, if TGF-β1 can prevent cardiac fibroblast death triggered by ischemia/reperfusion is unknown. Therefore, we test this hypothesis, and whether the canonical and/or non-canonical TGF-β1 signaling pathways are involved in this protective effect. Cultured rat cardiac fibroblasts were subjected to simulated ischemia/reperfusion. Cell viability was analyzed by trypan blue exclusion and propidium iodide by flow cytometry. The processing of procaspases 8, 9 and 3 to their active forms was assessed by Western blot, whereas subG1 population was evaluated by flow cytometry. Levels of total and phosphorylated forms of ERK1/2, Akt and Smad2/3 were determined by Western blot. The role of these signaling pathways on the protective effect of TGF-β1 was studied using specific chemical inhibitors. Simulated ischemia over 8h triggers a significant cardiac fibroblast death, which increased by reperfusion, with apoptosis actively involved. These effects were only prevented by the addition of TGF-β1 during reperfusion. TGF-β1 pretreatment increased the levels of phosphorylated forms of ERK1/2, Akt and Smad2/3. The inhibition of ERK1/2, Akt and Smad3 also blocked the preventive effects of TGF-β1 on cardiac fibroblast apoptosis induced by simulated ischemia/reperfusion. Overall, our data suggest that TGF-β1 prevents cardiac fibroblast apoptosis induced by simulated ischemia-reperfusion through the canonical (Smad3) and non canonical (ERK1/2 and Akt) signaling pathways.
Collapse
Affiliation(s)
- Raúl Vivar
- Centro Estudios Moleculares de la Célula, Facultad de Ciencias Químicas y Farmacéuticas/Facultad de Medicina, Universidad de Chile, Chile
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Inhibition of anti-apoptotic signals by Wortmannin induces apoptosis in the remote myocardium after LAD ligation: evidence for a protein kinase C-δ-dependent pathway. Mol Cell Biochem 2012; 372:275-83. [PMID: 23010893 DOI: 10.1007/s11010-012-1469-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 09/14/2012] [Indexed: 01/04/2023]
Abstract
It has been shown that, in the remote myocardium after infarction (MI), protein kinase C (PKC) inhibition reduces apoptosis both by blocking proapoptotic pathways and by activating antiapoptotic signals including the Akt pathway. However, it was open if vice versa, blockade of antiapoptotic pathways may influence proapoptotic signals. To clarify this, the present study tested the effects of the PI3-kinase blocker Wortmannin on proapoptotic signals and on apoptosis execution in the remote myocardium after infarction. Rats were subjected to MI by LAD ligation in situ. Some were pre-treated with Wortmannin alone or in combination with the PKC inhibitor Chelerythrine. After 24 h, pro- and anti-apoptotic signals (caspase-3, PKC isoforms, p38-MAPK, p42/44-MAPK, Akt, Bad), and marker of apoptosis execution (TUNEL) were quantified in the myocardium remote from the infarction. Wortmannin treatment increased apoptosis in the remote myocardium both at baseline and after MI, together with an activation of the PKC-δ/p38-MAPK-pathway. PKC-ε and p42/44-MAPK were unaffected. Combined treatment with Wortmannin and Chelerythrine fully reversed the pro-apoptotic effects of Wortmannin both at baseline and after MI. The PKC-δ-p38-MAPK-pathway as a strong signal for apoptosis in the non-infarcted myocardium can be influenced by targeting the anti-apoptotic PI3-kinase pathway. This gives evidence of a bi-directional crosstalk of pro- and anti-apoptotic signals after infarction.
Collapse
|
17
|
Vivar R, Humeres C, Varela M, Ayala P, Guzmán N, Olmedo I, Catalán M, Boza P, Muñoz C, Díaz Araya G. Cardiac fibroblast death by ischemia/reperfusion is partially inhibited by IGF-1 through both PI3K/Akt and MEK-ERK pathways. Exp Mol Pathol 2012; 93:1-7. [PMID: 22537549 DOI: 10.1016/j.yexmp.2012.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 12/21/2011] [Accepted: 01/31/2012] [Indexed: 11/18/2022]
Abstract
UNLABELLED Cardiac fibroblast (CF) death by ischemia/reperfusion (I/R) has major implications for cardiac wound healing. Although IGF-1 has well-known cytoprotective effects, no study has been done on CF subjected to simulated I/R. Simulated ischemia of neonate rat CF was performed in a free oxygen chamber in an ischemic medium; reperfusion was done in normal culture conditions. Cell viability was evaluated by trypan blue assay, and apoptosis by a FACS flow cytometer; p-ERK-1/2 and p-Akt levels were determined by western blot. We showed that simulated I/R triggers CF death by necrosis and apoptosis. IGF-1 partially inhibits I/R-induced apoptosis. PD98059 and LY294002 neutralize the preventive effects of IGF-1. CONCLUSION IGF-1 partially inhibits CF apoptosis induced by simulated I/R by PI3K/Akt- and MEK/ERK1/2-dependent signaling pathways.
Collapse
Affiliation(s)
- Raúl Vivar
- Laboratorio de Farmacoquímica y Centro FONDAP CEMC, Centro de Estudios Moleculares de Célula, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Velez Rueda JO, Palomeque J, Mattiazzi A. Early apoptosis in different models of cardiac hypertrophy induced by high renin-angiotensin system activity involves CaMKII. J Appl Physiol (1985) 2012; 112:2110-20. [PMID: 22492934 DOI: 10.1152/japplphysiol.01383.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The objective of this study was to establish whether 1) hyperactivity of renin-angiotensin-aldosterone system (RAAS) produces apoptosis in early stages of cardiac disease; and 2) Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) is involved in these apoptotic events. Two models of hypertrophy were used at an early stage of cardiac disease: spontaneously hypertensive rats (SHR) and isoproterenol-treated rats (Iso-rats). At 4 mo, SHR showed blood pressure, aldosterone serum levels, used as RAAS activity index, and left ventricular mass index, used as hypertrophy index, above control values by 84.2 ± 2.6 mmHg, 211.2 ± 25.8%, and 8.6 ± 1.1 mg/mm, respectively. There was also an increase in apoptotis (Bax-to-Bcl-2 ratio and terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling positive cells) associated with an enhancement of CaMKII activity with respect to age-matched controls (phosphorylated-CaMKII, 98.7 ± 14.1 above control). Similar results were observed in 4-mo-old Iso-rats. Cardiac function studied by echocardiography remained unaltered in all groups. Enalapril treatment significantly prevented hypertrophy, apoptosis, and CaMKII activity. Moreover, intracellular Ca(2+) handling in isolated myocytes was similar between SHR, Iso-rats, and their aged-matched controls. However, SHR and Iso-rats showed a significant increase in superoxide anion generation (lucigenin) and lipid peroxidation (thiobarbituric acid reactive substance). In transgenic mice with targeted cardiomyocyte expression of a CaMKII inhibitory peptide (AC3-I) or a scrambled control peptide (AC3-C), Iso treatment increased thiobarbituric acid reactive substance in both strains, whereas it increased CaMKII activity and apoptosis only in AC3-C mice. Endogenous increases in RAAS activity induce ROS and CaMKII-dependent apoptosis in vivo. CaMKII activation could not be associated with intracellular Ca(2+) increments and was directly related to the increase in oxidative stress.
Collapse
Affiliation(s)
- J Omar Velez Rueda
- Centro de Investigaciones Cardiovasculares, Consejo Nacional de Investigaciones Científicas y Técnicas-La Plata, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, Argentina
| | | | | |
Collapse
|
19
|
Reina S, Sterin-Borda L, Borda E. Anti-M(3) peptide IgG from Sjögren's syndrome triggers apoptosis in A253 cells. Cell Immunol 2012; 275:33-41. [PMID: 22513175 DOI: 10.1016/j.cellimm.2012.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 03/06/2012] [Accepted: 03/26/2012] [Indexed: 01/16/2023]
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune disease that targets salivary and lachrymal glands, characterized by anti-cholinergic autoantibodies directed against the M(3) muscarinic acetylcholine receptor (mAChR). The aim of this work was to evaluate if cholinergic autoantibodies contained in IgG purified from Sjögren sera could trigger apoptosis of A253 cell line. We also determined if caspase-3 and matrix metalloproteinase-3 (MMP-3) are involved in the induction of A253 cell death. Our results demonstrated that anti-cholinergic autoantibodies stimulate apoptosis and inositol phosphate (InsP) accumulation accompanied by caspase-3 activation and MMP-3 production. All of these effects were blunted by atropine and J104794, indicating that M(3) mAChRs are impacted by the anti-cholinergic autoantibodies. The intracellular pathway leading to autoantibody-induced biological effects involves phospholipase C (PLC), calcium/calmodulin (CaM) and extracellular calcium as demonstrated by treatment with U-73122, W-7, verapamil, BAPTA and BAPTA-AM, all of which blocked the effects of the anti-cholinergic autoantibodies. In conclusion, anti-cholinergic autoantibodies in IgG purified from pSS patient's sera mediates apoptosis of the A253 cell line in an InsP, caspase-3 and MMP-3 dependent manner.
Collapse
Affiliation(s)
- Silvia Reina
- Pharmacology Unit, School of Dentistry, Buenos Aires University and Argentine National Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | | | | |
Collapse
|
20
|
Copaja M, Venegas D, Aranguiz P, Canales J, Vivar R, Avalos Y, Garcia L, Chiong M, Olmedo I, Catalán M, Leyton L, Lavandero S, Díaz-Araya G. Simvastatin disrupts cytoskeleton and decreases cardiac fibroblast adhesion, migration and viability. Toxicology 2012; 294:42-9. [PMID: 22306966 DOI: 10.1016/j.tox.2012.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 01/16/2012] [Accepted: 01/20/2012] [Indexed: 12/31/2022]
Abstract
Statins reduce the isoprenoids farnesyl and geranylgeranyl pyrophosphate, essential intermediates, which control a diversity of cellular events such as cytoskeleton integrity, adhesion, migration and viability. Cardiac fibroblasts are the major non-myocyte cell constituent in the normal heart, and play a key role in the maintenance of extracellular matrix. The effects of simvastatin on cardiac fibroblast processes previously mentioned remain unknown. Our aims were to investigate the effects of simvastatin on cytoskeleton structure and focal adhesion complex assembly and their relationships with cell adhesion, migration and viability in cultured cardiac fibroblasts. To this end, cells were treated with simvastatin for 24 h and changes in actin cytoskeleton, levels of vimentin and paxillin as well as their subcellular localization were analyzed by Western blot and immunocytochemistry, respectively. Cell adhesion to plastic or collagen coated dishes, migration in Transwell chambers, and cell viability were analyzed after simvastatin treatment. Our results show that simvastatin disrupts actin cytoskeleton and focal adhesion complex evaluated by phalloidin stain and immunocytochemistry for paxillin and vinculin. All these effects occurred by a cholesterol synthesis-independent mechanism. Simvastatin decreased cell adhesion, migration and viability in a concentration-dependent manner. Finally, simvastatin decreased angiotensin II-induced phospho-paxillin levels and cell adhesion. We concluded that simvastatin disrupts cytoskeleton integrity and focal adhesion complex assembly in cultured cardiac fibroblasts by a cholesterol-independent mechanism and consequently decreases cell migration, adhesion and viability.
Collapse
Affiliation(s)
- Miguel Copaja
- Centro Estudios Moleculares de la Célula, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The renin-angiotensin system (RAS) plays an important role in regulating blood pressure, water-salt balance and the pathogenesis of cardiovascular diseases. Angiotensin II (Ang II) is the physiologically active mediator and mediates the main pathophysiological actions in RAS. Ang II exerts the effects by activating its receptors, primarily type 1 (AT1R) and type 2 (AT2R). Most of the known pathophysiological effects of Ang II are mediated by AT1R activation. The precise physiological function of AT2R is still not clear. Generally, AT2R is considered to oppose the effects of AT1R. Lectin-like oxidized low-density lipoprotein scavenger receptor-1 (LOX-1) is one of the major receptors responsible for binding, internalizing and degrading ox-LDL. The activation of LOX-1 has been known to be related to many pathophysiological events, including endothelial dysfunction and injury, fibroblast growth, and vascular smooth muscle cell hypertrophy. Many of these alterations are present in atherosclerosis, hypertension, and myocardial ischemia and remodeling. A growing body of evidence suggests the existence of a cross-talk between LOX-1 and Ang II receptors. Their interplays are embodied in the reciprocal regulation of their expression and activity. Their interplays are involved in a series of signals. Recent studies suggests that reactive oxygen species (ROS), nitric oxide (NO), protein kinase C (PKC) and mitogen activated protein kinases (MAPKs) are important signals responsible for their cross-talk. This paper reviews these aspects of dyslipidemia and RAS activation.
Collapse
Affiliation(s)
- Xianwei Wang
- Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
22
|
Copaja M, Venegas D, Aránguiz P, Canales J, Vivar R, Catalán M, Olmedo I, Rodríguez AE, Chiong M, Leyton L, Lavandero S, Díaz-Araya G. Simvastatin induces apoptosis by a Rho-dependent mechanism in cultured cardiac fibroblasts and myofibroblasts. Toxicol Appl Pharmacol 2011; 255:57-64. [PMID: 21651924 DOI: 10.1016/j.taap.2011.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 05/19/2011] [Accepted: 05/23/2011] [Indexed: 11/17/2022]
Abstract
UNLABELLED Several clinical trials have shown the beneficial effects of statins in the prevention of coronary heart disease. Additionally, statins promote apoptosis in vascular smooth muscle cells, in renal tubular epithelial cells and also in a variety of cell lines; yet, the effects of statins on cardiac fibroblast and myofibroblast, primarily responsible for cardiac tissue healing are almost unknown. Here, we investigated the effects of simvastatin on cardiac fibroblast and myofibroblast viability and studied the molecular cell death mechanism triggered by simvastatin in both cell types. METHODS Rat neonatal cardiac fibroblasts and myofibroblasts were treated with simvastatin (0.1-10μM) up to 72h. Cell viability and apoptosis were evaluated by trypan blue exclusion method and by flow cytometry, respectively. Caspase-3 activation and Rho protein levels and activity were also determined by Western blot and pull-down assay, respectively. RESULTS Simvastatin induces caspase-dependent apoptosis of cardiac fibroblasts and myofibroblasts in a concentration- and time-dependent manner, with greater effects on fibroblasts than myofibroblasts. These effects were prevented by mevalonate, farnesylpyrophosphate and geranylgeranylpyrophosphate, but not squalene. These last results suggest that apoptosis was dependent on small GTPases of the Rho family rather than Ras. CONCLUSION Simvastatin triggered apoptosis of cardiac fibroblasts and myofibroblasts by a mechanism independent of cholesterol synthesis, but dependent of isoprenilation of Rho protein. Additionally, cardiac fibroblasts were more susceptible to simvastatin-induced apoptosis than cardiac myofibroblasts. Thus simvastatin could avoid adverse cardiac remodeling leading to a less fibrotic repair of the damaged tissues.
Collapse
Affiliation(s)
- Miguel Copaja
- Centro FONDAP Estudios Moleculares de la Célula, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pojoga LH, Romero JR, Yao TM, Loutraris P, Ricchiuti V, Coutinho P, Guo C, Lapointe N, Stone JR, Adler GK, Williams GH. Caveolin-1 ablation reduces the adverse cardiovascular effects of N-omega-nitro-L-arginine methyl ester and angiotensin II. Endocrinology 2010; 151:1236-46. [PMID: 20097717 PMCID: PMC2840694 DOI: 10.1210/en.2009-0514] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Caveolae are the major cellular membrane structure through which extracellular mediators transmit information to intracellular signaling pathways. In vascular tissue (but not ventricular myocardium), caveolin-1 (cav-1) is the main component of caveolae; cav-1 modulates enzymes and receptors, such as the endothelial nitric oxide synthase and the angiotensin II (AngII) type 1 receptor. Evidence suggests that AngII and aldosterone (ALDO) are important mediators of ventricular injury. We have described a model of biventricular damage in rodents that relies on treatment with N-omega-nitro-l-arginine methyl ester (L-NAME (nitric oxide synthase inhibitor)) and AngII. This damage initiated at the vascular level and was observed only in the presence of ALDO and an activated mineralocorticoid receptor (MR). We hypothesize that cav-1 modulates the adverse cardiac effects mediated by ALDO in this animal model. To test this hypothesis, we assessed the ventricular damage and measures of inflammation, in wild-type (WT) and cav-1 knockout (KO) mice randomized to either placebo or L-NAME/AngII treatment. Despite displaying cardiac hypertrophy at baseline and higher blood pressure responses to L-NAME/AngII, cav-1 KO mice displayed, as compared with WT, decreased treatment-induced biventricular damage as well as decreased transcript levels of the proinflammatory marker plasminogen activator inhibitor-1. Additionally, L-NAME/AngII induced an increase in cardiac MR levels in WT but not cav-1-ablated mice. Moreover and despite similar circulating ALDO levels in both genotypes, the myocardial damage (as determined histologically and by plasminogen activator inhibitor-1 mRNA levels) was less sensitive to ALDO levels in cav-1 KO vs. WT mice, consistent with decreased MR signaling in the cav-1 KO. Thus, we conclude that the L-NAME/AngII-induced biventricular damage is mediated by a mechanism partially dependent on cav-1 and signaling via MR/ALDO.
Collapse
Affiliation(s)
- Luminita H Pojoga
- Brigham and Women's Hospital/Harvard Medical School, Department of Endocrinology, Diabetes, and Hypertension, 221 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Reina S, Sterin-Borda L, Passafaro D, Borda E. Muscarinic cholinoceptor activation by pilocarpine triggers apoptosis in human skin fibroblast cells. J Cell Physiol 2010; 222:640-7. [PMID: 19927300 DOI: 10.1002/jcp.21981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The aim of the present work was to examine the role of muscarinic acetylcholine receptors (mAChRs) on apoptosis in human skin fibroblast cells. Neonatal human skin fibroblast cultures were stimulated with pilocarpine in the presence or absence of specific antagonists. Pilocarpine stimulates apoptosis, total inositol phosphates (InsP) accumulation and nitric oxide synthase (NOS) activity. All these effects were inhibited by atropine, mustard hydrochloride (4-DAMP) and pirenzepine, indicating that M(1) and M(3) mAChRs are implicated in pilocarpine action. Pilocarpine apoptotic action is accompanied by caspase-3 and JNK activation. The intracellular pathway leading to pilocarpine-induced biological effects involved phospholipase C, calcium/calmodulin and extracellular calcium as U-73122, W-7, verapamil, BAPTA and BAPTA-AM blocked pilocarpine effects. L-NMMA, a NOS inhibitor, had no effect, indicating that the enzyme does not participate in the apoptosis phenomenon. These results may contribute to a better understanding of the modulatory role of the parasympathetic muscarinic system on the apoptotic human skin fibroblast process.
Collapse
Affiliation(s)
- Silvia Reina
- Argentine National Research Council (CONICET), Buenos Aires, Argentina
| | | | | | | |
Collapse
|
25
|
Palomeque J, Rueda OV, Sapia L, Valverde CA, Salas M, Petroff MV, Mattiazzi A. Angiotensin II-induced oxidative stress resets the Ca2+ dependence of Ca2+-calmodulin protein kinase II and promotes a death pathway conserved across different species. Circ Res 2009; 105:1204-12. [PMID: 19850941 DOI: 10.1161/circresaha.109.204172] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RATIONALE Angiotensin (Ang) II-induced apoptosis was reported to be mediated by different signaling molecules. Whether these molecules are either interconnected in a single pathway or constitute different and alternative cascades by which Ang II exerts its apoptotic action, is not known. OBJECTIVE To investigate in cultured myocytes from adult cat and rat, 2 species in which Ang II has opposite inotropic effects, the signaling cascade involved in Ang II-induced apoptosis. METHODS AND RESULTS Ang II (1 micromol/L) reduced cat/rat myocytes viability by approximately 40%, in part, because of apoptosis (TUNEL/caspase-3 activity). In both species, apoptosis was associated with reactive oxygen species (ROS) production, Ca(2+)/calmodulin-dependent protein kinase (CaMK)II, and p38 mitogen-activated protein kinase (p38MAPK) activation and was prevented by the ROS scavenger MPG (2-mercaptopropionylglycine) or the NADPH oxidase inhibitor DPI (diphenyleneiodonium) by CaMKII inhibitors (KN-93 and AIP [autocamtide 2-related inhibitory peptide]) or in transgenic mice expressing a CaMKII inhibitory peptide and by the p38MAPK inhibitor, SB202190. Furthermore, p38MAPK overexpression exacerbated Ang II-induced cell mortality. Moreover, although KN-93 did not affect Ang II-induced ROS production, it prevented p38MAPK activation. Results further show that CaMKII can be activated by Ang II or H(2)O(2), even in the presence of the Ca(2+) chelator BAPTA-AM, in myocytes and in EGTA-Ca(2+)-free solutions in the presence of the calmodulin inhibitor W-7 in in vitro experiments. CONCLUSIONS (1) The Ang II-induced apoptotic cascade converges in both species, in a common pathway mediated by ROS-dependent CaMKII activation which results in p38MAPK activation and apoptosis. (2) In the presence of Ang II or ROS, CaMKII may be activated at subdiastolic Ca(2+) concentrations, suggesting a new mechanism by which ROS reset the Ca(2+) dependence of CaMKII to extremely low Ca(2+) levels.
Collapse
Affiliation(s)
- Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, Facultad de Medicina, Universidad Nacional de La Plata, Centro Científico Tecnológico CONICET, La Plata, Argentina
| | | | | | | | | | | | | |
Collapse
|
26
|
Chen S, Li G, Zhang W, Wang J, Sigmund CD, Olson JE, Chen Y. Ischemia-induced brain damage is enhanced in human renin and angiotensinogen double-transgenic mice. Am J Physiol Regul Integr Comp Physiol 2009; 297:R1526-31. [PMID: 19759335 DOI: 10.1152/ajpregu.91040.2008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate the role of brain angiotensin II (ANG II) in the pathogenesis of injury following ischemic stroke, mice overexpressing renin and angiotensinogen (R+A+) and their wild-type control animals (R-A-) were used for experimental ischemia studies. Focal brain ischemia was induced by middle cerebral artery occlusion (MCAO). The severity of ischemic injury was determined by measuring neurological deficits and histological damage at 24 and 48 h after MCAO, respectively. To exclude the influence of blood pressure and local collateral blood flow, brain slices were used for oxygen and glucose deprivation (OGD) studies. The severity of OGD-induced damage was determined by measuring indicators of tissue swelling and cell death, the intensity of the intrinsic optical signal (IOS), and the number of propidium iodide (PI) staining cells, respectively. Results showed 1) R+A+ mice showed higher neurological deficit score (3.8 +/- 0.5 and 2.5 +/- 0.3 for R+A+ and R-A-, respectively, P < 0.01) and larger infarct volume (22.2 +/- 1.6% and 14.1 +/- 1.2% for R+A+ and R-A-, respectively, P < 0.01); 2) The R+A+ brain slices showed more severe tissue swelling and cell death in the cortex (IOS: 140 +/- 6% and 114 +/- 10%; PI: 139 +/- 20 cells/field and 39 +/- 9 cells/field for R+A+ and R-A-, respectively, P < 0.01); 3) treatment with losartan (20 micromol/l) abolished OGD-induced exaggeration of cell injury seen in R+A+ mice. The data indicate that activation of ANG II/AT(1) signaling is harmful to brain exposed to ischemia.
Collapse
Affiliation(s)
- Shuzhen Chen
- Departments of Pharmacology and Toxicology, Wright State University, Dayton, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Aránguiz-Urroz P, Soto D, Contreras A, Troncoso R, Chiong M, Montenegro J, Venegas D, Smolic C, Ayala P, Thomas WG, Lavandero S, Díaz-Araya G. Differential participation of angiotensin II type 1 and 2 receptors in the regulation of cardiac cell death triggered by angiotensin II. Am J Hypertens 2009; 22:569-76. [PMID: 19300422 DOI: 10.1038/ajh.2009.32] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The Angiotensin II (Ang II) type 1 (AT(1)R) and type 2 (AT(2)R) receptors are increased in the heart following myocardial infarction and dilated cardiomyopathy, yet their contribution at a cellular level to compensation and/or failure remains controversial. METHODS We ectopically expressed AT(1)R and AT(2)R in cultured adult rat cardiomyocytes and cardiac fibroblasts to investigate Ang II-mediated cardiomyocyte hypertrophy and cardiac cell viability. RESULTS In adult rat cardiomyocytes, Ang II did not induce hypertrophy via the AT(1)R, and no effect of Ang II on cell viability was observed following AT(1)R or AT(2)R expression. In adult rat cardiac fibroblasts, Ang II stimulated cell death by apoptosis via the AT(1)R (but not the AT(2)R), which required the presence of extracellular calcium, and induced a rapid dissipation of mitochondrial membrane potential, which was significant from 8 h. CONCLUSIONS We conclude that Ang II/AT(1)R triggers apoptosis in adult rat cardiac fibroblasts, which is dependent on Ca2+ influx.
Collapse
|
28
|
Abstract
Increased activity of the renin angiotensin system with enhanced levels of angiotensin II leads to oxidative stress with endothelial dysfunction, hypertension, and atherosclerosis. Epidemiologic studies revealed a higher cancer mortality and an increased kidney cancer incidence in hypertensive patients. Because elevated angiotensin II levels might contribute to carcinogenesis, we tested whether angiotensin II induces DNA damage in the kidney. In isolated perfused mouse kidneys, as little as 1 nmol/L angiotensin II caused a significant increase in DNA strand breaks, measured with the comet assay. This damage was independent of the hemodynamic effect of angiotensin II and mediated by the angiotensin II type 1 receptor. Angiotensin II also caused double-strand breaks in the cells of the isolated perfused kidney, detected with an antibody against the double-strand break marker gamma-H2AX. Studies in cell culture allowed further characterization of the DNA damage induced by angiotensin II. Single- and double-strand breaks, abasic sites, and 7,8-dihydro-8-oxo-guanine, all types of oxidative DNA lesions, were detected in angiotensin II-treated renal cells. The majority of detected strand breaks was repaired within 1 hour, but double-strand breaks increased and persisted for at least 24 hours.
Collapse
Affiliation(s)
- Ursula Schmid
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | | | | | | | | |
Collapse
|