1
|
Zhao J, Yoshizumi M. A Comprehensive Retrospective Study on the Mechanisms of Cyclic Mechanical Stretch-Induced Vascular Smooth Muscle Cell Death Underlying Aortic Dissection and Potential Therapeutics for Preventing Acute Aortic Aneurysm and Associated Ruptures. Int J Mol Sci 2024; 25:2544. [PMID: 38473793 DOI: 10.3390/ijms25052544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Acute aortic dissection (AAD) and associated ruptures are the leading causes of death in cardiovascular diseases (CVDs). Hypertension is a prime risk factor for AAD. However, the molecular mechanisms underlying AAD remain poorly understood. We previously reported that cyclic mechanical stretch (CMS) leads to the death of rat aortic smooth muscle cells (RASMCs). This review focuses on the mechanisms of CMS-induced vascular smooth muscle cell (VSMC) death. Moreover, we have also discussed the potential therapeutics for preventing AAD and aneurysm ruptures.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan
| | - Masanori Yoshizumi
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan
| |
Collapse
|
2
|
Luo L, Cai Y, Zhang Y, Hsu CG, Korshunov VA, Long X, Knight PA, Berk BC, Yan C. Role of PDE10A in vascular smooth muscle cell hyperplasia and pathological vascular remodelling. Cardiovasc Res 2022; 118:2703-2717. [PMID: 34550322 PMCID: PMC9890476 DOI: 10.1093/cvr/cvab304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/17/2021] [Indexed: 02/05/2023] Open
Abstract
AIMS Intimal hyperplasia is a common feature of vascular remodelling disorders. Accumulation of synthetic smooth muscle cell (SMC)-like cells is the main underlying cause. Current therapeutic approaches including drug-eluting stents are not perfect due to the toxicity on endothelial cells and novel therapeutic strategies are needed. Our preliminary screening for dysregulated cyclic nucleotide phosphodiesterases (PDEs) in growing SMCs revealed the alteration of PDE10A expression. Herein, we investigated the function of PDE10A in SMC proliferation and intimal hyperplasia both in vitro and in vivo. METHODS AND RESULTS RT-qPCR, immunoblot, and in situ proximity ligation assay were performed to determine PDE10A expression in synthetic SMCs and injured vessels. We found that PDE10A mRNA and/or protein levels are up-regulated in cultured SMCs upon growth stimulation, as well as in intimal cells in injured mouse femoral arteries. To determine the cellular functions of PDE10A, we focused on its role in SMC proliferation. The anti-mitogenic effects of PDE10A on SMCs were evaluated via cell counting, BrdU incorporation, and flow cytometry. We found that PDE10A deficiency or inhibition arrested the SMC cell cycle at G1-phase with a reduction of cyclin D1. The anti-mitotic effect of PDE10A inhibition was dependent on cGMP-dependent protein kinase Iα (PKGIα), involving C-natriuretic peptide (CNP) and particulate guanylate cyclase natriuretic peptide receptor 2 (NPR2). In addition, the effects of genetic depletion and pharmacological inhibition of PDE10A on neointimal formation were examined in a mouse model of femoral artery wire injury. Both PDE10A knockout and inhibition decreased injury-induced intimal thickening in femoral arteries by at least 50%. Moreover, PDE10A inhibition decreased ex vivo remodelling of cultured human saphenous vein segments. CONCLUSIONS Our findings indicate that PDE10A contributes to SMC proliferation and intimal hyperplasia at least partially via antagonizing CNP/NPR2/cGMP/PKG1α signalling and suggest that PDE10A may be a novel drug target for treating vascular occlusive disease.
Collapse
Affiliation(s)
- Lingfeng Luo
- Department of Biochemistry and Biophysics, University of Rochester School
of Medicine and Dentistry, Rochester, NY,
USA
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Yujun Cai
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Yishuai Zhang
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Chia G Hsu
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Vyacheslav A Korshunov
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Xiaochun Long
- Department of Vascular Biology Center and Medicine, Medical College of
Georgia, Augusta, GA, USA
| | - Peter A Knight
- Department of Surgery, University of Rochester School of Medicine and
Dentistry, Rochester, NY, USA
| | - Bradford C Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| | - Chen Yan
- Department of Medicine, Aab Cardiovascular Research Institute, University
of Rochester School of Medicine and Dentistry, Rochester,
NY, USA
| |
Collapse
|
3
|
Xu H, Xiao J. ACE2 Promotes the Synthesis of Pulmonary Surfactant to Improve AT II Cell Injury via SIRT1/eNOS Pathway. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:7710129. [PMID: 34471421 PMCID: PMC8405332 DOI: 10.1155/2021/7710129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/19/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE We aimed to explore the level of PS, cell viability, inflammatory factors, and apoptosis in neonatal respiratory distress syndrome (ARDS). Besides, we explored the potential relationship between ACE2, SIRT1/eNOS pathway, and hypoxia-induced AT II cell damage. METHODS The hUC-MSC-derived AT II cells were verified by IF and ICC, whereas qRT-PCR was used for PS and AT II cell marker (CK-8 and KGF). The AT II cell damage model was established by hypoxia exposure. The enhanced expression of ACE2 was tested after transfection with pcDNA3.1-ACE2 by western blot. The effects of hypoxia and ACE2 on AT II cells were evaluated by MTT, western blot, ELISA, and flow cytometry. The involvement of the SIRT1/eNOS pathway in AT II cell's protective functions against NRDS was verified with the addition of SIRT1 inhibitor EX527. RESULTS Based on the successful differentiation of AT II cells from hUC-MSCs and the buildup of AT II cell damage model, the overexpressed ACE2 impeded the hypoxia-induced cellular damage of AT II cells. It also counteracted the inhibitory effects of hypoxia on the secretion of PS. Overexpression of ACE2 rescued the cell viability and suppressed the secretion of inflammatory cytokines and the apoptosis of AT II cells triggered by hypoxia. And ACE2 activated the SIRT1/eNOS pathway to play its cell-protective and anti-inflammatory roles. CONCLUSION Our findings provided information that ACE2 prevented AT II cells from inflammatory damage through activating the SIRT1/eNOS pathway, which suggested that ACE2 might become a novel protective agent applied in the protection and treatment of NRDS.
Collapse
Affiliation(s)
- Hailing Xu
- Department of Respiratory Medicine, Laizhou People's Hospital of Shandong Province, China
| | - Jianguang Xiao
- Department of Thoracic Surgery, Laizhou People's Hospital of Shandong Province, China
| |
Collapse
|
4
|
Boraldi F, Lofaro FD, Quaglino D. Apoptosis in the Extraosseous Calcification Process. Cells 2021; 10:cells10010131. [PMID: 33445441 PMCID: PMC7827519 DOI: 10.3390/cells10010131] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Extraosseous calcification is a pathologic mineralization process occurring in soft connective tissues (e.g., skin, vessels, tendons, and cartilage). It can take place on a genetic basis or as a consequence of acquired chronic diseases. In this last case, the etiology is multifactorial, including both extra- and intracellular mechanisms, such as the formation of membrane vesicles (e.g., matrix vesicles and apoptotic bodies), mitochondrial alterations, and oxidative stress. This review is an overview of extraosseous calcification mechanisms focusing on the relationships between apoptosis and mineralization in cartilage and vascular tissues, as these are the two tissues mostly affected by a number of age-related diseases having a progressively increased impact in Western Countries.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
- Correspondence:
| | - Francesco Demetrio Lofaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
- Interuniversity Consortium for Biotechnologies (CIB), Italy
| |
Collapse
|
5
|
Zhao J, Nakahira K, Kimura A, Kyotani Y, Yoshizumi M. Upregulation of iNOS Protects Cyclic Mechanical Stretch-Induced Cell Death in Rat Aorta Smooth Muscle Cells. Int J Mol Sci 2020; 21:E8660. [PMID: 33212839 PMCID: PMC7698365 DOI: 10.3390/ijms21228660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/06/2020] [Accepted: 11/14/2020] [Indexed: 01/09/2023] Open
Abstract
Aortic dissection and aneurysm are associated with abnormal hemodynamic loads originating from hypertension. Our previous study demonstrated that cyclic mechanical stretch (CMS, mimicked hypertension) caused the death of rat aortic smooth muscle cells (RASMCs) in a mitogen activated-protein kinases (MAPKs)-dependent manner. The current study investigated the effects of inducible nitric oxide synthase (iNOS) on CMS-induced RASMC death. cDNA microarrays for CMS-treated RASMCs showed that iNOS expression levels were increased in response to CMS. Real-time polymerase chain reaction (PCR) analysis demonstrated that this increase was p38 MAPK (p38)-dependent. NO production was also increased. This increase could be inhibited by p38 and iNOS inhibitors. Thus, CMS-induced iNOS synthesized NO. CMS-induced cell death in RASMCs was increased by the iNOS inhibitor but abrogated by the long-acting NO donor DETA-NONOate. Increased iNOS expression was confirmed in the abdominal aortic constriction mouse model. Signal transducers and activators of transcription 1 (STAT1) was activated in stretched RASMCs, and iNOS expression and NO production were inhibited by the STAT1 inhibitor nifuroxazide. Our findings suggest that RASMCs were protected by iNOS from CMS-stimulated cell death through the STAT1 and p38 signal pathways independently.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/enzymology
- Gene Expression Regulation, Enzymologic
- Male
- Mechanotransduction, Cellular
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/enzymology
- Nitric Oxide Synthase Type II/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Stress, Mechanical
- Up-Regulation
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan;
| | - Yoji Kyotani
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| | - Masanori Yoshizumi
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| |
Collapse
|
6
|
Unal O, Ulukan MO, Bakuy V, Kaytaz B, Artan S, Aral E, Oztas DM, Beyaz MO, Ugurlucan M, Sevin B. Comparison of the Apoptotic Effects of Topically Applied Papaverine, Diltiazem, and Nitroprusside to Internal Thoracic Artery. Braz J Cardiovasc Surg 2020; 35:626-633. [PMID: 33118726 PMCID: PMC7598950 DOI: 10.21470/1678-9741-2019-0251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Objective To detect and to compare the apoptotic effects of intraoperatively topically applied diltiazem, papaverine, and nitroprusside. Methods Internal thoracic artery segments of ten patients were obtained during coronary bypass grafting surgery. Each internal thoracic artery segment was divided into four pieces and immersed into four different solutions containing separately saline (Group S), diltiazem (Group D), papaverine (Group P), and nitroprusside (Group N). Each segment was examined with both hematoxylin-eosin and the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) method in order to determine and quantify apoptosis. Results Apoptotic cells were counted in 50 microscopic areas of each segment. No significant difference was observed among the four groups according to hematoxylin-eosin staining. However, the TUNEL method revealed a significant increase in mean apoptotic cells in the diltiazem group when compared with the other three groups (Group S=4.25±1.4; Group D=13.31±2.8; Group N=9.48±2.09; Group P=10.75±2.37). The differences between groups were significant (P=0.0001). No difference was observed between the samples of the diabetic and non-diabetic patients in any of the study groups. Conclusion The benefit of topically applied vasodilator drugs must outweigh the potential adverse effects. In terms of apoptosis, diltiazem was found to have the most deleterious effects on internal thoracic artery graft segments. Of the analyzed medical agents, nitroprusside was found to have the least apoptotic activity, followed by papaverine. Diabetes did not have significant effect on the occurrence of apoptosis in left internal thoracic artery grafts.
Collapse
Affiliation(s)
- Orcun Unal
- Eskisehir Osmangazi University Medical Faculty Department of Cardiovascular Surgery Eskisehir Turkey Department of Cardiovascular Surgery, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey.,Yedikule Hospital of Pulmonary Diseases and Thoracic Surgery Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Yedikule Hospital of Pulmonary Diseases and Thoracic Surgery, Istanbul, Turkey
| | - Mustafa Ozer Ulukan
- Eskisehir Osmangazi University Medical Faculty Department of Cardiovascular Surgery Eskisehir Turkey Department of Cardiovascular Surgery, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey.,Istanbul Medipol University Medical Faculty Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Medipol University Medical Faculty, Istanbul, Turkey
| | - Vedat Bakuy
- Baskent University Medical Faculty Department of Cardiovascular Surgery Ankara Turkey Department of Cardiovascular Surgery, Baskent University Medical Faculty, Ankara, Turkey
| | - Behiye Kaytaz
- Eskisehir Osmangazi University Medical Faculty Department of Genetics Eskisehir Turkey Department of Genetics, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey
| | - Sevilhan Artan
- Eskisehir Osmangazi University Medical Faculty Department of Genetics Eskisehir Turkey Department of Genetics, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey
| | - Erinc Aral
- Eskisehir Osmangazi University Medical Faculty Department of Histology and Embryology Eskisehir Turkey Department of Histology and Embryology, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey
| | - Didem Melis Oztas
- Bagcilar Education and Research Hospital Cardiovascular Surgery Clinic Istanbul Turkey Cardiovascular Surgery Clinic, Bagcilar Education and Research Hospital, Istanbul, Turkey
| | - Metin Onur Beyaz
- Istanbul Medipol University Medical Faculty Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Medipol University Medical Faculty, Istanbul, Turkey
| | - Murat Ugurlucan
- Istanbul Medipol University Medical Faculty Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Medipol University Medical Faculty, Istanbul, Turkey
| | - Behcet Sevin
- Eskisehir Osmangazi University Medical Faculty Department of Cardiovascular Surgery Eskisehir Turkey Department of Cardiovascular Surgery, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey
| |
Collapse
|
7
|
Angiotensin II inhibits apoptosis of mouse aortic smooth muscle cells through regulating the circNRG-1/miR-193b-5p/NRG-1 axis. Cell Death Dis 2019; 10:362. [PMID: 31043588 PMCID: PMC6494886 DOI: 10.1038/s41419-019-1590-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Abstract
Angiotensin II (Ang II) is known to promote proliferation of vascular smooth muscle cells (VSMCs) in vascular remodeling, but whether it has an anti-apoptotic effect needs to be explored. Neuregulin-1 (NRG-1) as a member of the epidermal growth factor family was reported to suppress the proliferation of VSMCs by activating ErbB receptors, and therefore we hypothesized that there might be a cross talk between the anti-apoptotic effect of Ang II and the anti-proliferative effect of NRG-1 in VSMCs. The aim of the present study was to observe the expression and role of NRG-1 underlying the inhibitory effect of Ang II on apoptosis of mouse aortic smooth muscle cells (MASMCs). It was found that NRG-1 expression was down-regulated via the circNRG-1/miR-193b-5p-mediated post-transcriptional mechanism in response to Ang II. In addition, NRG-1 overexpression reversed the inhibitory effect of Ang II on apoptosis in MASMCs. Our data may provide a molecular basis for further understanding the mechanism of Ang II in suppressing the apoptosis of MASMCs by decreasing NRG-1 expression at circular RNA and micro RNA levels. The circNRG-1/miR-193b-5p/NRG-1 axis may prove to be a potential target for Ang II to inhibit the apoptosis of VSMCs and lead to vascular remodeling.
Collapse
|
8
|
Yoo YM, Jung EM, Ahn C, Jeung EB. Nitric oxide prevents H 2O 2-induced apoptosis in SK-N-MC human neuroblastoma cells. Int J Biol Sci 2018; 14:1974-1984. [PMID: 30585261 PMCID: PMC6299366 DOI: 10.7150/ijbs.28050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2022] Open
Abstract
Nitric oxide (NO) is a cellular signaling molecule in many physiological and pathological processes including neuroprotector. Here we examined the antiapoptotic effect of NO in SK-N-MC cells. H2O2 treatment (10-200 μM) induced cell death in a dose-dependent manner and pretreatment of cells with 100 μM S-nitroso-N-acetylpenicillamine (SNAP), an NO donor, attenuated the occurrence of H2O2-induced cell death. DAPI staining showed H2O2-induced nuclear fragmentation and NO treatment suppressed it. NO inhibited the proteolytic activation of caspase-3 and mitochondrial cytochrome c release. Treatment of soluble guanylyl cyclase inhibitor ODQ decreased the protective effect of SNAP on H2O2-treated cells and increased caspase 3-like enzyme activity and activation, cytochrome c release, PARP cleavage, and DNA fragmentation, indicating that cGMP is a key mediator in NO-mediated antiapoptosis. The cGMP analog 8-Br-cGMP blocked H2O2-induced apoptotic cell death; reduction of caspase-3 enzyme, cytochrome c release, and caspase-8 and -9. These preventive effects of SNAP and 8-Br-cGMP were suppressed by PKG inhibitor KT5823. Levels of PKGI, PKGII, and p-VASP proteins were increased by SNAP and 8-Br-cGMP and suppressed by KT5823 treatment. These results indicate that PKG is a downstream signal mediator in the suppression of apoptosis by NO and cGMP. Akt activation was inhibited the PI3K inhibitors LY294002 and Wortmannin, resulting in the inhibition of cell viability and increase of cytochrome c release. SNAP induced phosphorylation of Akt and Bad and then increased the interactions between 14-3-3β and p-Bad. These data suggest that the NO suppresses H2O2-induced SK-N-MC cell apoptosis by suppressing apoptosis signal mediating the interaction between 14-3-3β and Bad phosphorylation via PKG/PI3K/Akt.
Collapse
Affiliation(s)
| | | | | | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| |
Collapse
|
9
|
Pandey KN. Molecular and genetic aspects of guanylyl cyclase natriuretic peptide receptor-A in regulation of blood pressure and renal function. Physiol Genomics 2018; 50:913-928. [PMID: 30169131 DOI: 10.1152/physiolgenomics.00083.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Natriuretic peptides (NPs) exert diverse effects on several biological and physiological systems, such as kidney function, neural and endocrine signaling, energy metabolism, and cardiovascular function, playing pivotal roles in the regulation of blood pressure (BP) and cardiac and vascular homeostasis. NPs are collectively known as anti-hypertensive hormones and their main functions are directed toward eliciting natriuretic/diuretic, vasorelaxant, anti-proliferative, anti-inflammatory, and anti-hypertrophic effects, thereby, regulating the fluid volume, BP, and renal and cardiovascular conditions. Interactions of NPs with their cognate receptors display a central role in all aspects of cellular, biochemical, and molecular mechanisms that govern physiology and pathophysiology of BP and cardiovascular events. Among the NPs atrial and brain natriuretic peptides (ANP and BNP) activate guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA) and initiate intracellular signaling. The genetic disruption of Npr1 (encoding GC-A/NPRA) in mice exhibits high BP and hypertensive heart disease that is seen in untreated hypertensive subjects, including high BP and heart failure. There has been a surge of interest in the NPs and their receptors and a wealth of information have emerged in the last four decades, including molecular structure, signaling mechanisms, altered phenotypic characterization of transgenic and gene-targeted animal models, and genetic analyses in humans. The major goal of the present review is to emphasize and summarize the critical findings and recent discoveries regarding the molecular and genetic regulation of NPs, physiological metabolic functions, and the signaling of receptor GC-A/NPRA with emphasis on the BP regulation and renal and cardiovascular disorders.
Collapse
Affiliation(s)
- Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine , New Orleans, Louisiana
| |
Collapse
|
10
|
Wilstein Z, Alligood DM, McLure VL, Miller AC. Mathematical model of hypertension-induced arterial remodeling: A chemo-mechanical approach. Math Biosci 2018; 303:10-25. [PMID: 29758218 DOI: 10.1016/j.mbs.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/31/2018] [Accepted: 05/04/2018] [Indexed: 01/22/2023]
Abstract
The development of chronic hypertension is a poorly described process involving many chemical and structural changes to the artery. Typically, mathematical models of this disease focus primarily on the mechanical aspects such as arterial geometry, elasticity, and tissue content, or alternatively on the chemical drivers of vasoactivity such as nitric oxide and reactive oxygen species. This paper presents a model that considers the powerful interaction between mechanical and biochemical drivers of hypertension and arterial remodeling. Based on biological processes thought to be involved in the development of hypertension, we have built a system of algebraic, differential, and integral equations. Endothelial dysfunction, which is known to limit vasodilation, is explicitly considered in the model and plays a vital role in the development of chronic hypertension. Numerical solutions to the system are consistent with available experimental data for normal and spontaneously-hypertensive rats.
Collapse
Affiliation(s)
- Zahava Wilstein
- Department of Mathematics & Computer Science, Berry College, Mount Berry, GA 30149, United States.
| | - Daniel M Alligood
- Department of Mathematics & Computer Science, Berry College, Mount Berry, GA 30149, United States.
| | - Valerie L McLure
- Department of Mathematics & Computer Science, Berry College, Mount Berry, GA 30149, United States.
| | - Austinn C Miller
- Mercer University School of Medicine, Macon, GA 31207, United States.
| |
Collapse
|
11
|
Yue Y, Ma K, Li Z, Wang Z. Angiotensin II type 1 receptor-associated protein regulates carotid intimal hyperplasia through controlling apoptosis of vascular smooth muscle cells. Biochem Biophys Res Commun 2018; 495:2030-2037. [DOI: 10.1016/j.bbrc.2017.12.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 11/30/2022]
|
12
|
Li X, Shen F, Wang K, Lin S, Zhou L, Chen S, Wang J, Huang N. Endothelial mimetic multifunctional surfaces fabricated via polydopamine mediated copper immobilization. J Mater Chem B 2018; 6:7582-7593. [DOI: 10.1039/c8tb01976b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nitric oxide (NO), which is continuously released from the normal healthy endodermis cell layer of the vascular system, plays a crucial role in the stability and health maintenance of blood vessels.
Collapse
Affiliation(s)
- Xin Li
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | | | - Kebing Wang
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Shuang Lin
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Lei Zhou
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Si Chen
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Jin Wang
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| | - Nan Huang
- Key Laboratories of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- China
| |
Collapse
|
13
|
Chemokines protect vascular smooth muscle cells from cell death induced by cyclic mechanical stretch. Sci Rep 2017; 7:16128. [PMID: 29170451 PMCID: PMC5701048 DOI: 10.1038/s41598-017-15867-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/03/2017] [Indexed: 01/08/2023] Open
Abstract
The pulsatile nature of blood flow exposes vascular smooth muscle cells (VSMCs) in the vessel wall to cyclic mechanical stretch (CMS), which evokes VSMC proliferation, cell death, phenotypic switching, and migration, leading to vascular remodeling. These responses have been observed in many cardiovascular diseases; however, the underlying mechanisms remain unclear. We have revealed that CMS of rat aortic smooth muscle cells (RASMCs) causes JNK- and p38-dependent cell death and that a calcium channel blocker and angiotensin II receptor antagonist decreased the phosphorylation of JNK and p38 and subsequently decreased cell death by CMS. In the present study, we showed that the expression of Cxcl1 and Cx3cl1 was induced by CMS in a JNK-dependent manner. The expression of Cxcl1 was also induced in VSMCs by hypertension produced by abdominal aortic constriction (AAC). In addition, antagonists against the receptors for CXCL1 and CX3CL1 increased cell death, indicating that CXCL1 and CX3CL1 protect RASMCs from CMS-induced cell death. We also revealed that STAT1 is activated in RASMCs subjected to CMS. Taken together, these results indicate that CMS of VSMCs induces inflammation-related gene expression, including that of CXCL1 and CX3CL1, which may play important roles in the stress response against CMS caused by hypertension.
Collapse
|
14
|
Trittmann JK, Jin Y, Chicoine LG, Liu Y, Chen B, Nelin LD. An arginase-1 SNP that protects against the development of pulmonary hypertension in bronchopulmonary dysplasia enhances NO-mediated apoptosis in lymphocytes. Physiol Rep 2017; 4:4/22/e13041. [PMID: 27895230 PMCID: PMC5358007 DOI: 10.14814/phy2.13041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/20/2016] [Accepted: 10/26/2016] [Indexed: 11/24/2022] Open
Abstract
Arginase and nitric oxide synthase (NOS) share a common substrate, l‐arginine, and have opposing effects on vascular remodeling. Arginase is the first step in polyamine and proline synthesis necessary for cellular proliferation, while NO produced from NOS promotes apoptosis. Previously, we identified a single nucleotide polymorphism (SNP) in the arginase‐1 (ARG1) gene, rs2781666 (T‐allele) that was associated with a decreased risk for developing pulmonary hypertension (PH) in a cohort of infants with bronchopulmonary dysplasia (BPD). In this study, we utilized lymphocytes from neonates (the only readily available cells from these patients expressing the two genotypes of interest) with either the rs2781666 SNP (TT) or wild type (GG) to test the hypothesis that the protection of the ARG1 SNP against the development of PH in BPD would involve augmented NO production leading to more apoptosis. Lymphocytes were stimulated with IL‐4, IL‐13, and phorbol myristate acetate (PMA). We found that TT lymphocytes had similar levels of arginase I and arginase II expression, but there was a tendency for lower urea production (a surrogate marker of arginase activity), than in the GG lymphocytes. The TT lymphocytes also had significantly greater NO production than did GG lymphocytes despite no differences in iNOS expression between genotypes. Furthermore, the TT lymphocytes had lower numbers of viable cells, and higher levels of cleaved caspase‐3 than did GG lymphocytes. Inhibiting NOS activity using Nω‐Nitro‐l‐arginine methyl ester hydrochloride (l‐NAME) significantly decreased cleaved caspase‐3 levels in the TT lymphocytes. These data demonstrate that the TT genotype results in greater levels of NO production leading to more apoptosis, which is consistent with the concept that BPD patients with the TT genotype are protected against the development of PH by producing greater basal levels of endogenous NO.
Collapse
Affiliation(s)
- Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio .,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio
| | - Louis G Chicoine
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
15
|
Ngo JP, Ow CP, Gardiner BS, Kar S, Pearson JT, Smith DW, Evans RG. Diffusive shunting of gases and other molecules in the renal vasculature: physiological and evolutionary significance. Am J Physiol Regul Integr Comp Physiol 2016; 311:R797-R810. [DOI: 10.1152/ajpregu.00246.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/27/2016] [Indexed: 01/22/2023]
Abstract
Countercurrent systems have evolved in a variety of biological systems that allow transfer of heat, gases, and solutes. For example, in the renal medulla, the countercurrent arrangement of vascular and tubular elements facilitates the trapping of urea and other solutes in the inner medulla, which in turn enables the formation of concentrated urine. Arteries and veins in the cortex are also arranged in a countercurrent fashion, as are descending and ascending vasa recta in the medulla. For countercurrent diffusion to occur, barriers to diffusion must be small. This appears to be characteristic of larger vessels in the renal cortex. There must also be gradients in the concentration of molecules between afferent and efferent vessels, with the transport of molecules possible in either direction. Such gradients exist for oxygen in both the cortex and medulla, but there is little evidence that large gradients exist for other molecules such as carbon dioxide, nitric oxide, superoxide, hydrogen sulfide, and ammonia. There is some experimental evidence for arterial-to-venous (AV) oxygen shunting. Mathematical models also provide evidence for oxygen shunting in both the cortex and medulla. However, the quantitative significance of AV oxygen shunting remains a matter of controversy. Thus, whereas the countercurrent arrangement of vasa recta in the medulla appears to have evolved as a consequence of the evolution of Henle’s loop, the evolutionary significance of the intimate countercurrent arrangement of blood vessels in the renal cortex remains an enigma.
Collapse
Affiliation(s)
- Jennifer P. Ngo
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
| | - Connie P.C. Ow
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
| | - Bruce S. Gardiner
- School of Engineering and Information Technology, Murdoch University, Perth, Western Australia
| | - Saptarshi Kar
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | - James T. Pearson
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
- Monash Biomedical Imaging Facility, Monash University, Melbourne, Australia
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - David W. Smith
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | - Roger G. Evans
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
| |
Collapse
|
16
|
Gonzalez-Lopez L, Cardona-Muñoz EG, Celis A, García-de la Torre I, Orozco-Barocio G, Salazar-Paramo M, Garcia-Gonzalez C, Garcia-Gonzalez A, Sanchez-Ortiz A, Trujillo-Hernandez B, Gamez-Nava JI. Therapy with intermittent pulse cyclophosphamide for pulmonary hypertension associated with systemic lupus erythematosus. Lupus 2016; 13:105-12. [PMID: 14995003 DOI: 10.1191/0961203304lu509oa] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The aim of this study was to compare the efficacy of intravenouscyclophosphamide(IVCYC) versus oral enalapril in mild or moderate pulmonary hypertension (PH) in systemic lupus erythematosus (SLE). Thirty-four patients with SLE who had systolic pulmonary artery pressure (SPAP) > 30mmHg by Doppler echocardiography were randomized to receive IVCYC (0.5g/mt2 body surface area, monthly), or oral enalapril (10mg/day) for six months. The primary outcome was the significant decrease in SPAP. An additional outcome measure included the improvement in the heart functional class (NYHA). Sixteen patients received cyclophosphamide and 18 enalapril. IVCYC decreased the median values of SPAP from 41 to 28mmHg (P < 0.001), and enalapril from 35 to 27mmHg (P 0.02). IVCYC reduced more than twice as much SPAP than enalapril (P 0.04). In those patients with SPAP ≥35mmHg, cyclophosphamidedecreased from 43 to 27mmHg (P 0.003), but enalapril was not effective (P 0.14). The NYHA functional class improved only in those with cyclophosphamide (P 0.021). Also IVCYC had a higher frequency of side effects including infections (RR 1.6; 95% CI, 1.001-2.47), and gastrointestinal side effects (RR 14.6; 95% CI,2.15-99.68). We concluded that IVCYC was effective in mild and moderate PH associated with SLE. Further research is needed to evaluate its long-term efficacy.
Collapse
Affiliation(s)
- L Gonzalez-Lopez
- Department of Internal Medicine-Rheumatology, Hospital General Regional 110-Instituto Mexicano del Seguro Social, Guadalajara, México.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Slim RM, Song Y, Albassam M, Dethloff LA. Apoptosis and Nitrative Stress Associated with Phosphodiesterase Inhibitor-Induced Mesenteric Vasculitis in Rats. Toxicol Pathol 2016; 31:638-45. [PMID: 14585732 DOI: 10.1080/01926230390241972] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Nitric oxide may play a role in phosphodiesterase (PDE) inhibitor-induced rat mesenteric vasculitis. The present study was conducted to identify cellular sources of iNOS, determine the distribution of nitrotyrosine (NT) residues as a footprint of peroxynitrite (ONOO-) production, and evaluate their association with vascular apoptosis. To dissociate primary events from secondary changes associated with the inflammatory response, rats were given the PDE IV inhibitor CI-1018 orally at 750 mg/kg alone or concurrently with dexamethasone (DEX) intraperitoneally at 1 mg/kg for 4—5 days. Neutrophil (PMN) involvement in apoptosis was investigated in CI-1018 treated rats dosed with rabbit anti-rat PMN serum (APS). iNOS expression, NT residues, and caspase-3 were detected by immuno-histochemistry. Apoptosis was evaluated by TUNEL assay. CI-1018 induced vascular lesions were associated with iNOS expression in endothelial cells and inflammatory infiltrates; NT was evident only in the latter. Caspase-3 and TUNEL-positive staining were prominent only in medial smooth muscle cells (SMC) from CI-1018-treated rats and only when associated with active inflammation. iNOS- and NT-positive inflammatory cells were present in close proximity to SMC with caspase-3 staining. Inflammatory infiltrates were absent in rats given DEX with minimal SMC necrosis and hemorrhage remained. DEX eliminated apoptosis and immunoreactivity associated with caspase-3, iNOS, and NT. APS depletion of PMNs decreased the incidence and severity of vasculitis but failed to abolish completely caspase-3 immunoreactivity. Expression patterns for caspase-3, iNOS, and NT demonstrated that nitrative stress is a prominent feature of PDE inhibitor-induced vasculitis, with a possible role in medial SMC apoptosis. Further, medial SMC apoptosis may not be a primary event, but instead may be secondary to the inflammatory response.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antilymphocyte Serum/immunology
- Antilymphocyte Serum/pharmacology
- Apoptosis/drug effects
- Caspase 3
- Caspases/metabolism
- Dexamethasone/administration & dosage
- Dexamethasone/pharmacology
- Drug Therapy, Combination
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Female
- Immunoenzyme Techniques
- In Situ Nick-End Labeling
- Injections, Intraperitoneal
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/pathology
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type II
- Oxidative Stress/drug effects
- Peroxynitrous Acid/metabolism
- Phosphodiesterase Inhibitors/administration & dosage
- Phosphodiesterase Inhibitors/toxicity
- Rats
- Rats, Wistar
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
- Vasculitis/chemically induced
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- Rabih M Slim
- Departments of Drug Safety Evaluation, Pfizer Global Research and Development, Ann Arbor Laboratories, Ann Arbor, Michigan 48105, USA
| | | | | | | |
Collapse
|
18
|
Morales RC, Bahnson ESM, Havelka GE, Cantu-Medellin N, Kelley EE, Kibbe MR. Sex-based differential regulation of oxidative stress in the vasculature by nitric oxide. Redox Biol 2015; 4:226-33. [PMID: 25617803 PMCID: PMC4803798 DOI: 10.1016/j.redox.2015.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/08/2015] [Accepted: 01/12/2015] [Indexed: 11/28/2022] Open
Abstract
Background Nitric oxide (•NO) is more effective at inhibiting neointimal hyperplasia following arterial injury in male versus female rodents, though the etiology is unclear. Given that superoxide (O2•−) regulates cellular proliferation, and •NO regulates superoxide dismutase-1 (SOD-1) in the vasculature, we hypothesized that •NO differentially regulates SOD-1 based on sex. Materials and methods Male and female vascular smooth muscle cells (VSMC) were harvested from the aortae of Sprague-Dawley rats. O2•− levels were quantified by electron paramagnetic resonance (EPR) and HPLC. sod-1 gene expression was assayed by qPCR. SOD-1, SOD-2, and catalase protein levels were detected by Western blot. SOD-1 activity was measured via colorimetric assay. The rat carotid artery injury model was performed on Sprague-Dawley rats ±•NO treatment and SOD-1 protein levels were examined by Western blot. Results In vitro, male VSMC have higher O2•− levels and lower SOD − 1 activity at baseline compared to female VSMC (P < 0.05). •NO decreased O2•− levels and increased SOD − 1 activity in male (P<0.05) but not female VSMC. •NO also increased sod− 1 gene expression and SOD − 1 protein levels in male (P<0.05) but not female VSMC. In vivo, SOD-1 levels were 3.7-fold higher in female versus male carotid arteries at baseline. After injury, SOD-1 levels decreased in both sexes, but •NO increased SOD-1 levels 3-fold above controls in males, but returned to baseline in females. Conclusions Our results provide evidence that regulation of the redox environment at baseline and following exposure to •NO is sex-dependent in the vasculature. These data suggest that sex-based differential redox regulation may be one mechanism by which •NO is more effective at inhibiting neointimal hyperplasia in male versus female rodents.
The baseline redox environment in the vascular is sex-dependent. Nitric oxide differentially affects the vascular redox environment between the sexes. Nitric oxide decreases superoxide (O2.) levels, by increasing SOD-1 activity, sod1 gene expression and SOD-1 protein levels in male vascular smooth muscle cells, but not in females. Sex-based differential redox regulation may be one mechanism by which is more effective at inhibiting neointimal hyperplasia in male versus female rodents.
Collapse
Affiliation(s)
- Rommel C Morales
- Division of Vascular Surgery, Northwestern University, Chicago, IL, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - Edward S M Bahnson
- Division of Vascular Surgery, Northwestern University, Chicago, IL, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | - George E Havelka
- Division of Vascular Surgery, Northwestern University, Chicago, IL, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
| | | | - Eric E Kelley
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melina R Kibbe
- Division of Vascular Surgery, Northwestern University, Chicago, IL, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
19
|
Izawa K, Okada M, Sumitomo K, Nakagawa N, Aizawa Y, Kawabe J, Kikuchi K, Hasebe N. Impaired glutathione redox system paradoxically suppresses angiotensin II-induced vascular remodeling. PLoS One 2014; 9:e108115. [PMID: 25343455 PMCID: PMC4208744 DOI: 10.1371/journal.pone.0108115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 08/18/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Angiotensin II (AII) plays a central role in vascular remodeling via oxidative stress. However, the interaction between AII and reduced glutathione (GSH) redox status in cardiovascular remodeling remains unknown. METHODS In vivo: The cuff-induced vascular injury model was applied to Sprague Dawley rats. Then we administered saline or a GSH inhibitor, buthionine sulfoximine (BSO, 30 mmol/L in drinking water) for a week, subsequently administered 4 more weeks by osmotic pump with saline or AII (200 ng/kg/minute) to the rats. In vitro: Incorporation of bromodeoxyuridine (BrdU) was measured to determine DNA synthesis in cultured rat vascular smooth muscle cells (VSMCs). RESULTS BSO reduced whole blood GSH levels. Systolic blood pressure was increased up to 215 ± 4 mmHg by AII at 4 weeks (p<0.01), which was not affected by BSO. Superoxide production in vascular wall was increased by AII and BSO alone, and was markedly enhanced by AII+BSO. The left ventricular weight to body weight ratio was significantly increased in AII and AII+BSO as compared to controls (2.52 ± 0.08, 2.50 ± 0.09 and 2.10 ± 0.07 mg/g respectively, p<0.05). Surprisingly, the co-treatment of BSO totally abolished these morphological changes. Although the vascular circumferential wall stress was well compensated in AII, significantly increased in AII+BSO. The anti-single-stranded DNA staining revealed increasing apoptotic cells in the neointima of injured arteries in BSO groups. BrdU incorporation in cultured VSMCs with AII was increased dose-dependently. Furthermore it was totally abolished by BSO and was reversed by GSH monoethyl ester. CONCLUSIONS We demonstrated that a vast oxidative stress in impaired GSH redox system totally abolished AII-induced vascular, not cardiac remodeling via enhancement of apoptosis in the neointima and suppression of cell growth in the media. The drastic suppression of remodeling may result in fragile vasculature intolerable to mechanical stress by AII.
Collapse
Affiliation(s)
- Kazuma Izawa
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Motoi Okada
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kazuhiro Sumitomo
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Naoki Nakagawa
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yoshiaki Aizawa
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Junichi Kawabe
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kenjiro Kikuchi
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Naoyuki Hasebe
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
20
|
Pandey KN. Guanylyl cyclase/natriuretic peptide receptor-A signaling antagonizes phosphoinositide hydrolysis, Ca(2+) release, and activation of protein kinase C. Front Mol Neurosci 2014; 7:75. [PMID: 25202235 PMCID: PMC4141235 DOI: 10.3389/fnmol.2014.00075] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/05/2014] [Indexed: 11/25/2022] Open
Abstract
Thus far, three related natriuretic peptides (NPs) and three distinct sub-types of cognate NP receptors have been identified and characterized based on the specific ligand binding affinities, guanylyl cyclase activity, and generation of intracellular cGMP. Atrial and brain natriuretic peptides (ANP and BNP) specifically bind and activate guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA), and C-type natriuretic peptide (CNP) shows specificity to activate guanylyl cyclase/natriuretic peptide receptor-B (GC-B/NPRB). All three NPs bind to natriuretic peptide receptor-C (NPRC), which is also known as clearance or silent receptor. The NPRA is considered the principal biologically active receptor of NP family; however, the molecular signaling mechanisms of NP receptors are not well understood. The activation of NPRA and NPRB produces the intracellular second messenger cGMP, which serves as the major signaling molecule of all three NPs. The activation of NPRB in response to CNP also produces the intracellular cGMP; however, at lower magnitude than that of NPRA, which is activated by ANP and BNP. In addition to enhanced accumulation of intracellular cGMP in response to all three NPs, the levels of cAMP, Ca2+ and inositol triphosphate (IP3) have also been reported to be altered in different cells and tissue types. Interestingly, ANP has been found to lower the concentrations of cAMP, Ca2+, and IP3; however, NPRC has been proposed to increase the levels of these metabolic signaling molecules. The mechanistic studies of decreased and/or increased levels of cAMP, Ca2+, and IP3 in response to NPs and their receptors have not yet been clearly established. This review focuses on the signaling mechanisms of ANP/NPRA and their biological effects involving an increased level of intracellular accumulation of cGMP and a decreased level of cAMP, Ca2+, and IP3 in different cells and tissue systems.
Collapse
Affiliation(s)
- Kailash N Pandey
- Department of Physiology, School of Medicine, Tulane University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
21
|
The Renin-Angiotensin-aldosterone system in vascular inflammation and remodeling. Int J Inflam 2014; 2014:689360. [PMID: 24804145 PMCID: PMC3997861 DOI: 10.1155/2014/689360] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/28/2014] [Accepted: 03/03/2014] [Indexed: 12/31/2022] Open
Abstract
The RAAS through its physiological effectors plays a key role in promoting and maintaining inflammation. Inflammation is an important mechanism in the development and progression of CVD such as hypertension and atherosclerosis. In addition to its main role in regulating blood pressure and its role in hypertension, RAAS has proinflammatory and profibrotic effects at cellular and molecular levels. Blocking RAAS provides beneficial effects for the treatment of cardiovascular and renal diseases. Evidence shows that inhibition of RAAS positively influences vascular remodeling thus improving CVD outcomes. The beneficial vascular effects of RAAS inhibition are likely due to decreasing vascular inflammation, oxidative stress, endothelial dysfunction, and positive effects on regeneration of endothelial progenitor cells. Inflammatory factors such as ICAM-1, VCAM-1, TNFα, IL-6, and CRP have key roles in mediating vascular inflammation and blocking RAAS negatively modulates the levels of these inflammatory molecules. Some of these inflammatory markers are clinically associated with CVD events. More studies are required to establish long-term effects of RAAS inhibition on vascular inflammation, vascular cells regeneration, and CVD clinical outcomes. This review presents important information on RAAS's role on vascular inflammation, vascular cells responses to RAAS, and inhibition of RAAS signaling in the context of vascular inflammation, vascular remodeling, and vascular inflammation-associated CVD. Nevertheless, the review also equates the need to rethink and rediscover new RAAS inhibitors.
Collapse
|
22
|
Abstract
The ubiquitous gaseous signaling molecule nitric oxide participates in the regulation of a variety of physiological and pathological processes, including adult neurogenesis. Adult neurogenesis, or the generation of new neurons in the adult brain, is a restricted event confined to areas with neurogenic capability. Although nitric oxide has been shown to mediate conflicting effects on adult neurogenesis, which may be partly explained by its unique characteristics, more studies are required in order to fully comprehend and appreciate the mechanisms involved. Neuropeptide Y, a neurotransmitter shown to be an important regulator of adult hippocampal neurogenesis, acts through intracellular nitric oxide to induce an increase in neural progenitor cell proliferation.
Collapse
|
23
|
Emond ZM, Kibbe MR. Clinical science review article: understanding the implications of diabetes on the vascular system. Vasc Endovascular Surg 2011; 45:481-9. [PMID: 21571777 DOI: 10.1177/1538574411408354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Patients with diabetes comprise an extremely complex subset of patients for the vascular surgeon. Often, they have numerous comorbidities that can further complicate matters. The diabetic environment is highly complex and the interplay of various diseases makes this an extremely challenging condition to manage. Knowing the mechanisms by which diabetes inflicts adverse microscopic changes in the vasculature allows the clinician to anticipate problems and minimize the heightened risks observed in diabetic patients undergoing surgery. In this review, we will illustrate how diabetes affects the vasculature and how the molecular and cellular derangements that occur in diabetic environments lead to these pathophysiologic consequences.
Collapse
Affiliation(s)
- Zachary M Emond
- Department of Surgery, University of Illinois at Chicago, IL, USA
| | | |
Collapse
|
24
|
Abstract
Significant advances have been made in understanding the pathogenesis and clinical physiology of primary hypertension. This article presents an overview of the physiology of normal blood pressure control and the pathophysiologic mechanisms that predispose individuals and populations to primary hypertension. The role of genetics, environment, and the gene-environment interaction is discussed. The spectrum of changes in physiologic states that result in chronic increases of arterial blood pressure are reviewed. The nature and characteristics of feedback loops and the primary modulating systems, the central and peripheral nervous systems, and circulating and tissue hormones are reviewed. The role of the endothelium of the artery and its production of endothelin, nitric oxide, angiotensin II, as well as other vasoactive substances in response to various stimuli, is also discussed. A unifying pathway for the development of hypertension and the practical implications for the prevention and control of hypertension are discussed.
Collapse
Affiliation(s)
- Mukesh Singh
- University of Chicago Pritzker School of Medicine, IL 60637, USA
| | | | | |
Collapse
|
25
|
Cerrito MG, Scagliarini A, Froio A, Liloia A, Busnelli M, Giovannoni R, Otterbein LE, Mainetti L, Villa M, Bach FH, Leone BE, Biasi GM, Lavitrano M. Heme Oxygenase-1 Inhibition Prevents Intimal Hyperplasia Enhancing Nitric Oxide-Dependent Apoptosis of Vascular Smooth Muscle Cells. Biol Pharm Bull 2011; 34:1204-14. [DOI: 10.1248/bpb.34.1204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Alberto Froio
- Vascular Surgery Unit, Department of Surgical Sciences, University of Milano-Bicocca
| | - Angela Liloia
- Vascular Surgery Unit, Department of Surgical Sciences, University of Milano-Bicocca
| | - Marco Busnelli
- Molecular Medicine Laboratory, University of Milano-Bicocca
| | | | | | - Lara Mainetti
- Molecular Medicine Laboratory, University of Milano-Bicocca
| | - Matteo Villa
- Molecular Medicine Laboratory, University of Milano-Bicocca
| | - Fritz Heintz Bach
- Immunobiology Research Center, Department of Surgery, Harvard Medical School
| | | | - Giorgio Maria Biasi
- Vascular Surgery Unit, Department of Surgical Sciences, University of Milano-Bicocca
| | | |
Collapse
|
26
|
Beyazit Y, Purnak T, Guven GS, Haznedaroglu IC. Local bone marrow Renin-Angiotensin system and atherosclerosis. Cardiol Res Pract 2010; 2011:714515. [PMID: 21234405 PMCID: PMC3014698 DOI: 10.4061/2011/714515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 10/14/2010] [Accepted: 10/23/2010] [Indexed: 12/13/2022] Open
Abstract
Local hematopoietic bone marrow (BM) renin-angiotensin system (RAS) affects the growth, production, proliferation differentiation, and function of hematopoietic cells. Angiotensin II (Ang II), the dominant effector peptide of the RAS, regulates cellular growth in a wide variety of tissues in pathobiological states. RAS, especially Ang II and Ang II type 1 receptor (AT1R), has considerable proinflammatory and proatherogenic effects on the vessel wall, causing progression of atherosclerosis. Recent investigations, by analyzing several BM chimeric mice whose BM cells were positive or negative for AT1R, disclosed that AT1R in BM cells participates in the pathogenesis of atherosclerosis. Therefore, AT1R blocking not only in vascular cells but also in the BM could be an important therapeutic approach to prevent atherosclerosis. The aim of this paper is to review the function of local BM RAS in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Yavuz Beyazit
- Department of Gastroenterology, Turkiye Yuksek Ihtisas Teaching and Research Hospital, 06100 Ankara, Turkey
| | | | | | | |
Collapse
|
27
|
Protein kinase G activity prevents pathological-level nitric oxide-induced apoptosis and promotes DNA synthesis/cell proliferation in vascular smooth muscle cells. Cardiovasc Pathol 2010; 19:e221-31. [DOI: 10.1016/j.carpath.2009.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2009] [Revised: 08/26/2009] [Accepted: 11/02/2009] [Indexed: 11/19/2022] Open
|
28
|
Chen C, Hu JT, Tu YJ, Wu JC, Liang J, Gao LL, Wang ZG, Yang BF, Dong DL. Effects of isosorbide mononitrate on the restoration of injured artery in mice in vivo. Eur J Pharmacol 2010; 640:150-156. [PMID: 20483352 DOI: 10.1016/j.ejphar.2010.04.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 03/29/2010] [Accepted: 04/28/2010] [Indexed: 10/19/2022]
Abstract
The pharmacological basis of isosorbide mononitrate (ISMN), a widely used drug for cardiovascular diseases, is that it is metabolized to nitric oxide (NO). However, NO is a double-edged sword that results in either beneficial or detrimental effect. Vascular injury is the common consequence of many cardiovascular diseases, but it is not determined whether ISMN influences the restoration of injured artery in vivo. Carotid artery injury was induced by electric stimulation in mice. Vasoconstriction and endothelium-dependent and -independent relaxation were recorded by a multichannel acquisition and analysis system. ISMN (10 mg/kg, p.o.) treatment for 1 week and 1 month had no effect on reendothelialization, histology and function of carotid artery injured by electric stimulation. L-arginine (500 mg/kg, p.o.) and Nomega-nitro-L-arginine methyl ester (L-NAME) (50 mg/kg, p.o.) treatment for 1 week did not affect the reendothelialization process, but L-NAME treatment induced neointimal hyperplasia and inhibited endothelium-dependent relaxation in electrically injured artery. These results suggest that supplement of exogenous or endogenous NO has no effect on the restoration of injured artery, but inhibition of endogenous NO induces neointimal hyperplasia in injured artery. ISMN treatment does not affect the restoration of injured artery.
Collapse
Affiliation(s)
- Chang Chen
- Department of Pharmacology, Harbin Medical University, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin 150081, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cabrales P, Han G, Roche C, Nacharaju P, Friedman AJ, Friedman JM. Sustained release nitric oxide from long-lived circulating nanoparticles. Free Radic Biol Med 2010; 49:530-8. [PMID: 20460149 PMCID: PMC2903640 DOI: 10.1016/j.freeradbiomed.2010.04.034] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 04/25/2010] [Accepted: 04/29/2010] [Indexed: 10/19/2022]
Abstract
The current limitations of nitric oxide (NO) delivery systems have stimulated an extraordinary interest in the development of compounds that generate NO in a controlled and sustained manner with a heavy emphasis on the treatment of cardiovascular disease states. This work describes the positive physiological response to the infusion of NO-releasing nanoparticles prepared using a new platform based on hydrogel/glass hybrid nanoparticles. When exposed to moisture, these nanoparticles slowly release therapeutic levels of NO, previously generated through thermal reduction of nitrite to NO trapped within the dry particles. The controlled and sustained release of NO observed from these nanoparticles (NO-np) is regulated by its hydration over extended periods of time. In a dose-dependent manner, circulating NO-np both decreased mean arterial blood pressure and increased exhaled concentrations of NO over a period of several hours. Circulating NO-np induced vasodilatation and increased microvascular perfusion during their several hour circulation lifetime. Control nanoparticles (control-np; without nitrite) did not induce changes in arterial pressure, although a decrease in the number of capillaries perfused and an increase in leukocyte rolling and immobilization in the microcirculation were observed. The NO released by the NO-np prevents the inflammatory response observed after infusion of control-np. These data suggest that NO release from NO-np is advantageous relative to other NO-releasing compounds, because it does not depend on chemical decomposition or enzymatic catalysis; it is only determined by the rate of hydration. Based on the observed physiological properties, NO-np has clear potential as a therapeutic agent and as a research tool to increase our understanding of NO signaling mechanisms within the vasculature.
Collapse
Affiliation(s)
- Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Forte A, Finicelli M, Grossi M, Vicchio M, Alessio N, Santé P, De Feo M, Cotrufo M, Berrino L, Rossi F, Galderisi U, Cipollaro M. DNA damage and repair in a model of rat vascular injury. Clin Sci (Lond) 2010; 118:473-485. [PMID: 19804370 DOI: 10.1042/cs20090416] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 09/18/2009] [Accepted: 10/05/2009] [Indexed: 12/21/2022]
Abstract
Restenosis rate following vascular interventions still limits their long-term success. Oxidative stress plays a relevant role in this pathophysiological phenomenon, but less attention has been devoted to its effects on DNA damage and to the subsequent mechanisms of repair. We analysed in a model of arteriotomy-induced stenosis in rat carotids the time-dependent expression of DNA damage markers and of DNA repair genes, together with the assessment of proliferation and apoptosis indexes. The expression of the oxidative DNA damage marker 7,8-dihydro-8-oxo-2'-deoxyguanosine was increased at 3 and 7 days after arteriotomy, with immunostaining distributed in the injured vascular wall and in perivascular tissue. The expression of the DNA damage marker phospho-H2A.X was less relevant but increasing from 4 hrs to 7 days after arteriotomy, with immunostaining prevalently present in the adventitia and, to a lesser extent, in medial smooth muscle cells at the injury site. RT-PCR indicated a decrease of 8 out of 12 genes of the DNA repair machinery we selected from 4 hrs to 7 days after arteriotomy with the exception of increased Muyth and Slk genes (p<0.05). Western Blot revealed a decrease of p53 and catalase at 3 days after arteriotomy (p<0.05). A maximal 7% of BrdU-positive cells in endothelium and media occurred at 7 days after arteriotomy, while the apoptotic index peaked at 3 days after injury (p<0.05). Our results highlight a persistent DNA damage presumably related to a temporary decreased expression of the DNA repair machinery and of the antioxidant enzyme catalase, playing a role in stenosis progression.
Collapse
Affiliation(s)
- Amalia Forte
- Department of Experimental Medicine, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Arribas SM, Hermida C, González MC, Wang Y, Hinek A. Enhanced survival of vascular smooth muscle cells accounts for heightened elastin deposition in arteries of neonatal spontaneously hypertensive rats. Exp Physiol 2010; 95:550-60. [DOI: 10.1113/expphysiol.2009.050971] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Protection of retinal vasculature by losartan against apoptosis and vasculopathy in rats with spontaneous hypertension. J Hypertens 2010; 28:510-9. [DOI: 10.1097/hjh.0b013e328333663f] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Martel G, Hamet P, Tremblay J. Central role of guanylyl cyclase in natriuretic peptide signaling in hypertension and metabolic syndrome. Mol Cell Biochem 2009; 334:53-65. [PMID: 19937369 DOI: 10.1007/s11010-009-0326-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 11/04/2009] [Indexed: 01/05/2023]
Abstract
Studied for nearly 30 years for its ability to control many parameters, such as vascular smooth muscle cell relaxation, heart fibrosis, and kidney function, the natriuretic peptide (NP) system is now considered to be a key element in several other major metabolic pathways. After stimulation by NPs, natriuretic peptide receptors (NPR) convert GTP to the second messenger cGMP. In addition to its vasodilatory effects and natriuretic and diuretic functions, cGMP has been positively associated with fat cell function, apoptosis, and NPR expression/activity modulation. The NP system is also closely linked to metabolic syndrome (MetS) progression and obesity control. A new era is now on its way targeting the NP system to not only treat high blood pressure, but to also assist in the fight against the obesity pandemic. Here, we summarize recent data on the role of NPs in hypertension and MetS.
Collapse
Affiliation(s)
- G Martel
- Laboratory of Cellular Biology of Hypertension, Centre for Ecogenomic Models of Human Diseases, Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Technopôle Angus, 2901 rue Rachel est, bureau 314, Montreal, QC H1W 4A4, Canada
| | | | | |
Collapse
|
34
|
Recombinant adeno-associated virus-mediated human kallikrein gene therapy protects against hypertensive target organ injuries through inhibiting cell apoptosis. Acta Pharmacol Sin 2009; 30:1253-61. [PMID: 19684610 DOI: 10.1038/aps.2009.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIM Overexpression of human tissue kallikrein (HK), mediated by recombinant adeno-associated virus (rAAV), decreased blood pressure in spontaneous hypertensive rats (SHRs) and reduced injury to the heart, aorta and kidney. In this study, we used both an in vivo animal model and in vitro cell culture system to investigate whether rAAV-mediated HK gene therapy protects against organ damage by inhibiting cell apoptosis. METHODS rAAV encoding HK (rAAV-HK) or LacZ (rAAV-lacZ) were delivered as a control to spontaneously hypertensive rats (SHRs) and cultured human embryonic kidney (HEK) 293 cells. RESULTS Treatment with rAAV-HK decreased cell apoptosis in the target organs of SHRs and also inhibited lipopolysaccharide (LPS)-induced HEK 293 apoptosis. The rAAV-HK delivery system also increased the levels of apoptosis-inhibiting proteins bcl-2 and bcl-x(L), and decreased the level of Bax and the activity of caspase 3, two promoters of apoptosis. In addition to its role in the inhibition of apoptosis, rAAV-HK also activated the cell survival and proliferation signaling pathways ERK1/2 and PI3K/AKT. CONCLUSION rAAV-mediated HK gene delivery has multiple therapeutic possibilities for treating hypertension, not only by decreasing blood pressure, but also by directly inhibiting end-organ damage.
Collapse
|
35
|
Li Z, Wang ZG, Bian C, Chen XD, Li JW, Chen X, Han B, Hou GF, Chu J, Cui Q. Interferon regulatory factor-1 exerts inhibitory effect on neointimal formation after vascular injury. ACTA ACUST UNITED AC 2009; 24:91-6. [PMID: 19618605 DOI: 10.1016/s1001-9294(09)60068-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To investigate the effect of interferon regulatory factors (IRFs) on neointimal formation after vascular injury in the mouse, and its possible mechanism. METHODS Vascular injury was induced by polyethylene cuff placement around the left femoral artery of IRF-1-deficient mice and C57BL/6J mice. The mRNA expressions of IRF-1, IRF-2, angiotensin II type 2 (AT2) receptor, interleukin-1 beta converting enzyme (ICE), inducible nitric oxide synthase (iNOS) were detected by RT-PCR and immunohistochemical staining. RESULTS Neointimal formation after vascular injury was significantly greater in IRF-1-deficient mice than that in C57BL/6J mice (P<0.05). In contrast, TUNEL-positive nuclei to total nuclei in the neointima and media in vascular smooth muscle cell (VSMC) in the injured artery significantly attenuated in IRF-1-deficient mice compared to C57BL/6J mice (P<0.05). The expressions of AT2 receptor as well as pro-apoptotic genes such as ICE and iNOS in C57BL/6J mice were up-regulated in response to vascular injury, but this upregulation was attenuated in IRF-1-deficient mice. CONCLUSIONS Our results suggest that IRF-1 induces VSMC apoptosis and inhibits neointimal formation after vascular injury at least partly due to the upregulation of AT2 receptor, ICE and iNOS expressions. These results indicate that IRF-1 exerts an inhibitory effect on neointimal formation through the induction of apoptosis in VSMCs.
Collapse
Affiliation(s)
- Zhen Li
- Vascular Surgery, the Second Artillery General Hospital, Beijing 100088, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lemay J, Hale TM, deBlois D. Neointimal-specific induction of apoptosis by losartan results in regression of vascular lesion in rat aorta. Eur J Pharmacol 2009; 618:45-51. [PMID: 19619526 DOI: 10.1016/j.ejphar.2009.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 06/24/2009] [Accepted: 07/09/2009] [Indexed: 10/20/2022]
Abstract
We previously reported that initiating treatment with the angiotensin II receptor antagonist losartan, prior to and immediately after balloon injury, attenuates neointimal hyperplasia via induction of smooth muscle cell (SMC) apoptosis in the aorta of spontaneously hypertensive rats (SHR). The present study examines whether losartan can induce regression of an established neointima. Balloon angioplasty was performed in the aorta of 1 1 week-old SHR. Five weeks were allowed for neointima formation before rats received placebo or losartan (30 mg/kg/day) for 1 to 4 weeks. Blood pressure was measured by tail cuff plethysmography. Losartan significantly reduced blood pressure (16%) versus placebo within 2 weeks of treatment. In situ labeling with terminal deoxynucleotidyl transferase among neointimal SMC was transiently increased with losartan (10-fold at 2 weeks; P=0.004) in correlation with internucleosomal fragmentation of vascular DNA. Accordingly, losartan reversed neointimal hyperplasia by 43% (P=0.002) and 61% (P=0.007) at weeks 2 and 4, respectively, and neointimal mass by 63% (P<0.001) and 75% (P<0.001) at weeks 2 and 4, respectively, as compared to pre-treatment values. No change in aortic medial hyperplasia or mass was observed during losartan treatment. Taken together, endothelial denudation rendered the underlying media resistant to drug-induced remodeling, while losartan treatment induced vascular lesion regression by inducing apoptosis selectively in neointimal SMC, an effect that may contribute to the reduction of cardiovascular complications in hypertension.
Collapse
Affiliation(s)
- Jacinthe Lemay
- Department of Pharmacology, University of Montreal, Canada
| | | | | |
Collapse
|
37
|
Duran X, Vilahur G, Badimon L. Exogenous in vivo NO-donor treatment preserves p53 levels and protects vascular cells from apoptosis. Atherosclerosis 2009; 205:101-6. [DOI: 10.1016/j.atherosclerosis.2008.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 08/27/2008] [Accepted: 11/18/2008] [Indexed: 10/21/2022]
|
38
|
D'Amora P, Maciel TT, Tambellini R, Mori MA, Pesquero JB, Sato H, Girão MJBC, Guerreiro da Silva IDC, Schor E. Disrupted cell cycle control in cultured endometrial cells from patients with endometriosis harboring the progesterone receptor polymorphism PROGINS. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:215-24. [PMID: 19497994 DOI: 10.2353/ajpath.2009.080966] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Presently, little is understood about how endometriosis is established or maintained, or how genetic factors can predispose women to the disease. Because of the crucial role that the progesterone receptor polymorphism PROGINS plays in predisposing women to the development of endometriosis, we hypothesized that this variant may influence critical steps during endometrial cell metabolism that are involved in the pathogenesis of endometriosis. Eutopic endometria were collected from three sources: women with endometriosis who had a single PROGINS allele (from the progesterone receptor gene); women with endometriosis who had the wild-type progesterone receptor allele; and women without endometriosis who had the wild-type allele. Cells prepared from the eutopic endometria of these women were stimulated with both estradiol and progesterone, and then examined for cell proliferation, viability, and apoptosis. The cells from women with endometriosis that carried the PROGINS allele demonstrated increased proliferation, greater viability, and decreased apoptosis following progesterone treatment. In general, these parameters were very different as compared with those of women with endometriosis but without the PROGINS allele and women in the control group. This result indicates there is a reduced level of progesterone responsiveness in women who carry the PROGINS polymorphism. Because progesterone responsiveness is known to be an important characteristic of women with endometriosis, these data support the contention that the PROGINS polymorphism enhances the endometriosis phenotype.
Collapse
Affiliation(s)
- Paulo D'Amora
- Pelvic Pain and Endometriosis Unit, Gynecology Department, Molecular Gynecology and Proteomics Laboratory, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, Brasil.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dey NB, Busch JL, Francis SH, Corbin JD, Lincoln TM. Cyclic GMP specifically suppresses Type-Ialpha cGMP-dependent protein kinase expression by ubiquitination. Cell Signal 2009; 21:859-66. [PMID: 19168131 PMCID: PMC2673574 DOI: 10.1016/j.cellsig.2009.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 01/03/2009] [Indexed: 01/31/2023]
Abstract
Type I cGMP-dependent protein kinase (PKG-I) mediates nitric oxide (NO) and hormone dependent smooth muscle relaxation and stimulates smooth muscle cell-specific gene expression. Expression of PKG-I in cultured smooth muscle cells depends on culture conditions and is inhibited by inflammatory cytokines such as interleukin-I and tumor necrosis factor-alpha, which are known to stimulate Type II NO synthase (iNOS) expression. We report here that the suppression of PKG-I protein levels in smooth muscle cells is triggered by the ubiquitin/26S proteasome pathway. Incubation of vascular smooth muscle cells with phosphodiesterase-resistant cyclic GMP analogs (e.g., 8-bromo-cGMP) decreases PKG-I protein level in a time- and concentration-dependent manner. To study this process, we tested the effects of 8-Br-cGMP on PKG-I protein level in Cos7 cells, which do not express endogenous type I PKG mRNA. 8-Br-cGMP induced the ubiquitination and down-regulation of PKG-Ialpha, but not PKG-Ibeta. Treatment of cells with the 26S proteasome inhibitor, MG-132, increased ubiquitination of PKG. Blocking PKG-I catalytic activity using the cell-permeant specific PKG-I inhibitor, DT-2, inhibited cGMP-induced PKG-I ubiquitination and down-regulation, suggesting that PKG catalytic activity and autophosphorylation were required for suppression of PKG-I level. Mutation of the known autophosphorylation sites of PKG-Ialpha to alanine uncovered a specific role for autophosphorylation of serine-64 in cGMP-dependent ubiquitination and suppression of PKG-I level. The results suggest that chronic elevation of cGMP, as seen in inflammatory conditions, triggers ubiquitination and degradation of PKG-Ialpha in smooth muscle.
Collapse
Affiliation(s)
- Nupur B. Dey
- The Department of Physiology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Jennifer L. Busch
- The Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Sharron H. Francis
- The Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Jackie D. Corbin
- The Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Thomas M. Lincoln
- The Department of Physiology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
40
|
Tsai EJ, Kass DA. Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics. Pharmacol Ther 2009; 122:216-38. [PMID: 19306895 PMCID: PMC2709600 DOI: 10.1016/j.pharmthera.2009.02.009] [Citation(s) in RCA: 314] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) mediates a wide spectrum of physiologic processes in multiple cell types within the cardiovascular system. Dysfunctional signaling at any step of the cascade - cGMP synthesis, effector activation, or catabolism - have been implicated in numerous cardiovascular diseases, ranging from hypertension to atherosclerosis to cardiac hypertrophy and heart failure. In this review, we outline each step of the cGMP signaling cascade and discuss its regulation and physiologic effects within the cardiovascular system. In addition, we illustrate how cGMP signaling becomes dysregulated in specific cardiovascular disease states. The ubiquitous role cGMP plays in cardiac physiology and pathophysiology presents great opportunities for pharmacologic modulation of the cGMP signal in the treatment of cardiovascular diseases. We detail the various therapeutic interventional strategies that have been developed or are in development, summarizing relevant preclinical and clinical studies.
Collapse
Affiliation(s)
- Emily J Tsai
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
41
|
Fogo AS, Antonioli E, Calixto JB, Campos AH. Tormentic acid reduces vascular smooth muscle cell proliferation and survival. Eur J Pharmacol 2009; 615:50-4. [PMID: 19464287 DOI: 10.1016/j.ejphar.2009.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 04/30/2009] [Accepted: 05/14/2009] [Indexed: 10/20/2022]
Abstract
The triterpene tormentic acid (TA) has been reported to exhibit anticancer, anti-inflammatory and anti-atherogenic properties, and minimal toxicity has been detected in in vivo. Vascular smooth muscle cell (VSMC) proliferation and apoptosis resistance are hallmarks of vasculoproliferative diseases, such as post-angioplasty restenosis. The present study was designed to assess the effects of TA on the phenotype of cultured VSMC. The exposure of VSMC to TA (30 muM) significantly increased apoptosis of serum-deprived A7r5 cells, whereas cell survival in the presence of 10% fetal bovine serum was less affected by the drug. On the other hand, even in the presence of serum, A7r5 cell proliferation was significantly inhibited by TA, an effect that persisted for at least 8 days of daily administration of TA. As preservation of endothelial integrity is critical to normal vascular function, we also evaluated the effects of TA on human umbilical cord endothelial cells (HUVEC). Interestingly, TA did not produce significant changes in the levels of apoptosis and proliferation of HUVEC. Our data indicate that TA is a VSMC apoptosis inducer and proliferation inhibitor. The anti-growth action in VSMC in the presence of serum, and the absence of significant effects in endothelial cells suggest that TA may control VSMC abnormal proliferation and cell death resistance without affecting the normal vasculature. We conclude that TA should be investigated further as a potential tool for the prevention and treatment of proliferative vascular diseases, particularly in the setting of post-angioplasty restenosis.
Collapse
Affiliation(s)
- Anelize S Fogo
- Nephrology Division, Federal University of São Paulo, Brazil
| | | | | | | |
Collapse
|
42
|
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
|
43
|
Ahn S, Kim J, Hara MR, Ren XR, Lefkowitz RJ. {beta}-Arrestin-2 Mediates Anti-apoptotic Signaling through Regulation of BAD Phosphorylation. J Biol Chem 2009; 284:8855-65. [PMID: 19171933 PMCID: PMC2659243 DOI: 10.1074/jbc.m808463200] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
β-Arrestins, originally discovered as terminators of G protein-coupled
receptor signaling, have more recently been appreciated to also function as
signal transducers in their own right, although the consequences for cellular
physiology have not been well understood. Here we demonstrate that
β-arrestin-2 mediates anti-apoptotic cytoprotective signaling stimulated
by a typical 7-transmembrane receptor the angiotensin ATII 1A receptor,
expressed endogenously in rat vascular smooth muscle cells or by transfection
in HEK-293 cells. Receptor stimulation leads to concerted activation of two
pathways, ERK/p90RSK and PI3K/AKT, which converge to phosphorylate and
inactivate the pro-apoptotic protein BAD. Anti-apoptotic effects as well as
pathway activities can be stimulated by an angiotensin analog (SII), which has
been previously shown to activate β-arrestin but not G protein-dependent
signaling, and are abrogated by β-arrestin-2 small interfering RNA. These
findings establish a key role for β-arrestin-2 in mediating cellular
cytoprotective functions by a 7-transmembrane receptor and define the
biochemical pathways involved.
Collapse
Affiliation(s)
- Seungkirl Ahn
- Departments of Medicine and Biochemistry and Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
44
|
Zemse SM, Hilgers RHP, Simkins GB, Rudic RD, Webb RC. Restoration of endothelin-1-induced impairment in endothelium-dependent relaxation by interleukin-10 in murine aortic rings. Can J Physiol Pharmacol 2008; 86:557-65. [PMID: 18758504 DOI: 10.1139/y08-049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelin-1 (ET-1) is implicated in the development of endothelial dysfunction through the generation of reactive oxygen species by NADPH oxidase activation. Interleukin-10 (IL-10) is an antiinflammatory cytokine that stimulates nitric oxide production, decreases superoxide production, and restores endothelial integrity after vascular injury. In this study, we tested whether IL-10 attenuates ET-1-induced endothelial dysfunction by improving acetylcholine (ACh)-induced relaxation of cultured murine aortic rings. Aortic rings (2 mm long) of C57BL/6 mice were incubated in 2 mL DMEM containing 120 U/mL penicillin and 120 microg/mL streptomycin in the presence of one of 4 treatments: vehicle (deionized water), ET-1 (100 nmol/L), recombinant mouse IL-10 (300 ng/mL), or a combination of both ET-1 and IL-10. After incubation at 37 degrees C for either 1 or 6 h (short-term exposure) or 22 h (overnight exposure), rings were mounted in a wire myograph and stretched to a passive force of 5 mN. Endothelium-dependent vasorelaxation was assessed by constructing cumulative concentration-response curves to ACh (0.001-10 micromol/L) during 10 mumol/L phenylephrine (PE)-induced contraction. Short-term exposure of ET-1 did not result in an impairment of ACh-induced relaxation. Overnight exposure of aortic rings to ET-1 resulted in a statistically significant endothelial dysfunction characterized by a reduced maximal relaxation response to ACh compared with that of untreated rings (Emax 57% +/- 3% versus 82% +/- 4%). IL-10 treatment restored ACh-induced relaxation (Emax 77% +/- 3%). Western blotting showed decreased eNOS expression in response to ET-1, whereas vessels treated with a combination of ET-1 and IL-10 showed increased expression of eNOS. Immunohistochemical analysis showed decreased eNOS expression in ET-1-treated vessels compared with those treated with both ET-1 and IL-10. We conclude that, in murine aorta, the antiinflammatory cytokine IL-10 prevents impairment in endothelium-dependent relaxation induced in response to long-term incubation with ET-1 via normalization of eNOS expression.
Collapse
Affiliation(s)
- Saiprasad M Zemse
- Department of Physiology, Medical College of Georgia, Augusta, GA 30912-3000, USA.
| | | | | | | | | |
Collapse
|
45
|
Homocysteine-induced enhanced expression of tissue factor in human vascular smooth muscle cells. ACTA ACUST UNITED AC 2008; 28:520-4. [DOI: 10.1007/s11596-008-0507-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Indexed: 11/24/2022]
|
46
|
Natriuretic Peptides and Cardiovascular Regulation. Cardiovasc Endocrinol 2008. [DOI: 10.1007/978-1-59745-141-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
47
|
Weinmeister P, Lukowski R, Linder S, Traidl-Hoffmann C, Hengst L, Hofmann F, Feil R. Cyclic guanosine monophosphate-dependent protein kinase I promotes adhesion of primary vascular smooth muscle cells. Mol Biol Cell 2008; 19:4434-41. [PMID: 18685080 DOI: 10.1091/mbc.e08-04-0370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The cyclic guanosine monophosphate (cGMP)/cGMP-dependent protein kinase type I (cGKI) pathway regulates many cellular functions. The current study shows that 8-Br-cGMP stimulates the number of attached primary but not that of subcultured murine vascular smooth muscle cells (VSMCs). These effects of 8-Br-cGMP require the presence of cGKI. In agreement with previous studies, cGKI inhibited the number of cells in repeatedly passaged murine VSMCs. Activation of the cGMP/cGKI pathway in freshly isolated primary VSMCs slightly decreased apoptosis and strongly increased cell adhesion. The stimulation of cell adhesion by cGKI involves an inhibition of the RhoA/Rho kinase pathway and increased exposure of beta(1) and beta(3) integrins on the cell surface. Together, these results identify a novel proadhesive function of cGMP/cGKI signaling in primary VSMCs and suggest that the opposing effects of this pathway on VSMC number depend on the phenotypic context of the cells.
Collapse
Affiliation(s)
- Pascal Weinmeister
- Institut für Pharmakologie und Toxikologie, Technischen Universiät München, D-80802 München, Germany.
| | | | | | | | | | | | | |
Collapse
|
48
|
de Resende MM, Greene AS. Effect of ANG II on endothelial cell apoptosis and survival and its impact on skeletal muscle angiogenesis after electrical stimulation. Am J Physiol Heart Circ Physiol 2008; 294:H2814-21. [PMID: 18441208 DOI: 10.1152/ajpheart.00095.2008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that skeletal muscle angiogenesis induced by electrical stimulation is significantly attenuated when SS-13BN/Mcwi rats are fed a high-salt diet. This effect was associated with a large increase in endothelial cell (EC) apoptosis. We hypothesized that the low levels of ANG II during high-salt diet would increase EC apoptosis and consequently diminish the angiogenic response. To test this hypothesis, a series of in vitro and in vivo studies was performed. EC apoptosis and viability were evaluated after incubation with ANG II under serum-free conditions. After 24 h of incubation, ANG II increased EC viability and Bcl-2-to-Bax ratio along with a dose-dependent decrease in EC apoptosis. This effect was blocked by the ANG II type 1 receptor antagonist losartan. To confirm our in vitro results, ANG II (3 ng.kg(-1).min(-1)) was chronically infused in rats fed a high-salt diet (4% NaCl). ANG II decreased EC apoptosis and produced a significant increase (40%) in skeletal muscle angiogenesis after electrical stimulation. These in vivo results were in agreement with our in vitro results and demonstrate that the attenuation of ANG II levels during a high-salt diet may induce EC apoptosis and consequently block the angiogenic response induced by electrical stimulation. Furthermore, under normal conditions, ANG II increases EC viability and protects EC from apoptosis possibly by inactivation of the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Micheline M de Resende
- Biotechnology and Bioengineering Center, Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | |
Collapse
|
49
|
Angiotensin II Signaling in Vascular Physiology and Pathophysiology. SIGNAL TRANSDUCTION IN THE CARDIOVASCULAR SYSTEM IN HEALTH AND DISEASE 2008. [PMCID: PMC7121295 DOI: 10.1007/978-0-387-09552-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Initially recognized as a physiologic regulator of blood pressure and body fluid homeostasis, angiotensin (Ang) II has now been shown in innumerable experiments and clinical studies to contribute to the development and maintenance of cardiovascular disease. Dissection of its signaling mechanisms over the past decades has led to the discovery of several novel concepts, such as tissue-specific metabolism of Ang peptides. Identification and cloning of the various receptors through which Ang II acts on almost all tissues has led to the development of specific pharmacologic inhibitors with proven clinical benefit in patients with cardiovascular disorders. Work on the G-protein-coupled Ang II Type 1 receptor has demonstrated that different receptors interact through oligomerization, compartmentalization, and transactivation, and may explain how Ang II can activate G-protein-independent pathways. Unraveling the downstream effects of Ang II in specific cell types corroborates the importance of the cellular redox state on certain signaling pathways. Finally, the effects of Ang II on cell function and phenotype, such as the expression of inflammatory cytokines and receptors promoting the recruitment of inflammatory cells into vascular tissues, have indicated its role in local inflammation as a general pathogenetic basis of cardiovascular disease. The recognition of Ang II as a contributor to such fundamental pathophysiologic mechanisms, which are believed to be a common pathway for diverse cardiovascular risk factors like hypertension and diabetes, has greatly advanced our knowledge of pathologic signaling in vascular tissues and may help to eventually define novel targets for pharmacologic interventions.
Collapse
|
50
|
McCrann DJ, Nguyen HG, Jones MR, Ravid K. Vascular smooth muscle cell polyploidy: An adaptive or maladaptive response? J Cell Physiol 2008; 215:588-92. [DOI: 10.1002/jcp.21363] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|