1
|
Pharmacological prevention of intimal hyperplasia: A state-of-the-art review. Pharmacol Ther 2022; 235:108157. [PMID: 35183591 DOI: 10.1016/j.pharmthera.2022.108157] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Intimal hyperplasia (IH) occurs in a considerable number of cases of blood vessel reconstruction by stenting or balloon angioplasty, venous bypass grafting, and arteriovenous dialysis accesses. It is triggered by endothelial injury during the vascular intervention and leads to vessel restenosis with life-threatening consequences for patients. To date, the drugs used for IH prevention in clinics-paclitaxel and rapalog drugs-have been focusing primarily on the vascular smooth muscle cell (VSMC) proliferation pathway of IH development. Limitations, such as endothelial toxicity and inappropriate drug administration timing, have spurred the search for new and efficient pharmacological approaches to control IH. In this state-of-the-art review, we present the pathways of IH development, focusing on the key events and actors involved in IH. Subsequently, we discuss different drugs and drug combinations interfering with these pathways based on their effect on peripheral circulation IH models in animal studies, or on clinical reports. The reports were obtained through an extensive search of peer-reviewed publications in Pubmed, Embase, and Google Scholar, with search equations composed based on five concepts around IH and their various combinations. To improve vascular intervention outcomes, rethinking of conventional therapeutic approaches to IH prevention is needed. Exploring local application of drugs and drug combinations acting on different pathophysiological pathways of IH development has the potential to provide effective and safe restenosis prevention.
Collapse
|
2
|
Mansouri A, Reiner Ž, Ruscica M, Tedeschi-Reiner E, Radbakhsh S, Bagheri Ekta M, Sahebkar A. Antioxidant Effects of Statins by Modulating Nrf2 and Nrf2/HO-1 Signaling in Different Diseases. J Clin Med 2022; 11:1313. [PMID: 35268403 PMCID: PMC8911353 DOI: 10.3390/jcm11051313] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Statins are competitive inhibitors of hydroxymethylglutaryl-CoA (HMG-CoA) reductase and have been used to treat elevated low-density lipoprotein cholesterol (LDL-C) for almost four decades. Antioxidant and anti-inflammatory properties which are independent of the lipid-lowering effects of statins, i.e., their pleiotropic effects, might be beneficial in the prevention or treatment of many diseases. This review discusses the antioxidant effects of statins achieved by modulating the nuclear factor erythroid 2 related factor 2/ heme oxygenase-1 (Nrf2/HO-1) pathway in different organs and diseases. Nrf2 and other proteins involved in the Nrf2/HO-1 signaling pathway have a crucial role in cellular responses to oxidative stress, which is a risk factor for ASCVD. Statins can significantly increase the DNA-binding activity of Nrf2 and induce the expression of its target genes, such as HO-1 and glutathione peroxidase) GPx, (thus protecting the cells against oxidative stress. Antioxidant and anti-inflammatory properties of statins, which are independent of their lipid-lowering effects, could be partly explained by the modulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Atena Mansouri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Željko Reiner
- Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, 10000 Zagreb, Croatia;
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy;
| | - Eugenia Tedeschi-Reiner
- University Hospital Center Sestre Milosrdnice, University of Osijek, Vinogradska Cesta 29, 10000 Zagreb, Croatia;
| | - Shabnam Radbakhsh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Mariam Bagheri Ekta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, A.P. Avtsyn Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia;
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
3
|
Sanyour HJ, Rickel AP, Hong Z. The interplay of membrane cholesterol and substrate on vascular smooth muscle biomechanics. CURRENT TOPICS IN MEMBRANES 2020; 86:279-299. [PMID: 33837696 PMCID: PMC8041049 DOI: 10.1016/bs.ctm.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
Cardiovascular disease (CVD) remains the primary cause of death worldwide. Specifically, atherosclerosis is a CVD characterized as a slow progressing chronic inflammatory disease. During atherosclerosis, vascular walls accumulate cholesterol and cause fatty streak formation. The progressive changes in vascular wall stiffness exert alternating mechanical cues on vascular smooth muscle cells (VSMCs). The detachment of VSMCs in the media layer of the vessel and migration toward the intima is a critical step in atherosclerosis. VSMC phenotypic switching is a complicated process that modifies VSMC structure and biomechanical function. These changes affect the expression and function of cell adhesion molecules, thus impacting VSMC migration. Accumulating evidence has shown cholesterol is capable of regulating cellular migration, proliferation, and spreading. However, the interaction and coordinated effects of both cellular cholesterol and the extracellular matrix (ECM) stiffness/composition on VSMC biomechanics remains to be elucidated.
Collapse
Affiliation(s)
- Hanna J Sanyour
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States
| | - Alex P Rickel
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States
| | - Zhongkui Hong
- Department of Biomedical Engineering, University of South Dakota, Vermillion, SD, United States.
| |
Collapse
|
4
|
Curcumin, a Multifaceted Hormetic Agent, Mediates an Intricate Crosstalk between Mitochondrial Turnover, Autophagy, and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3656419. [PMID: 32765806 PMCID: PMC7387956 DOI: 10.1155/2020/3656419] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/01/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023]
Abstract
Curcumin has extensive therapeutic potential because of its antioxidant, anti-inflammatory, and antiproliferative properties. Multiple preclinical studies in vitro and in vivo have proven curcumin to be effective against various cancers. These potent effects are driven by curcumin's ability to induce G2/M cell cycle arrest, induce autophagy, activate apoptosis, disrupt molecular signaling, inhibit invasion and metastasis, and increase the efficacy of current chemotherapeutics. Here, we focus on the hormetic behavior of curcumin. Frequently, low doses of natural chemical products activate an adaptive stress response, whereas high doses activate acute responses like autophagy and cell death. This phenomenon is often referred to as hormesis. Curcumin causes cell death and primarily initiates an autophagic step (mitophagy). At higher doses, cells undergo mitochondrial destabilization due to calcium release from the endoplasmic reticulum, and die. Herein, we address the complex crosstalk that involves mitochondrial biogenesis, mitochondrial destabilization accompanied by mitophagy, and cell death.
Collapse
|
5
|
Sanyour HJ, Li N, Rickel AP, Torres HM, Anderson RH, Miles MR, Childs JD, Francis KR, Tao J, Hong Z. Statin-mediated cholesterol depletion exerts coordinated effects on the alterations in rat vascular smooth muscle cell biomechanics and migration. J Physiol 2020; 598:1505-1522. [PMID: 32083311 DOI: 10.1113/jp279528] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
KEY POINTS This study demonstrates and evaluates the changes in rat vascular smooth muscle cell biomechanics following statin-mediated cholesterol depletion. Evidence is presented to show correlated changes in migration and adhesion of vascular smooth muscle cells to extracellular matrix proteins fibronectin and collagen. Concurrently, integrin α5 expression was enhanced but not integrin α2. Atomic force microscopy analysis provides compelling evidence of coordinated reduction in vascular smooth muscle cell stiffness and actin cytoskeletal orientation in response to statin-mediated cholesterol depletion. Proof is provided that statin-mediated cholesterol depletion remodels total vascular smooth muscle cell cytoskeletal orientation that may additionally participate in altering ex vivo aortic vessel function. It is concluded that statin-mediated cholesterol depletion may coordinate vascular smooth muscle cell migration and adhesion to different extracellular matrix proteins and regulate cellular stiffness and cytoskeletal orientation, thus impacting the biomechanics of the cell. ABSTRACT Not only does cholesterol induce an inflammatory response and deposits in foam cells at the atherosclerotic plaque, it also regulates cellular mechanics, proliferation and migration in atherosclerosis progression. Statins are HMG-CoA reductase inhibitors that are known to inhibit cellular cholesterol biosynthesis and are clinically prescribed to patients with hypercholesterolemia or related cardiovascular conditions. Nonetheless, the effect of statin-mediated cholesterol management on cellular biomechanics is not fully understood. In this study, we aimed to assess the effect of fluvastatin-mediated cholesterol management on primary rat vascular smooth muscle cell (VSMC) biomechanics. Real-time measurement of cell adhesion, stiffness, and imaging were performed using atomic force microscopy (AFM). Cellular migration on extra cellular matrix (ECM) protein surfaces was studied by time-lapse imaging. The effect of changes in VSMC biomechanics on aortic function was assessed using an ex vivo myograph system. Fluvastatin-mediated cholesterol depletion (-27.8%) lowered VSMC migration distance on a fibronectin (FN)-coated surface (-14.8%) but not on a type 1 collagen (COL1)-coated surface. VSMC adhesion force to FN (+33%) and integrin α5 expression were enhanced but COL1 adhesion and integrin α2 expression were unchanged upon cholesterol depletion. In addition, VSMC stiffness (-46.6%) and ex vivo aortic ring contraction force (-40.1%) were lowered and VSMC actin cytoskeletal orientation was reduced (-24.5%) following statin-mediated cholesterol depletion. Altogether, it is concluded that statin-mediated cholesterol depletion may coordinate VSMC migration and adhesion to different ECM proteins and regulate cellular stiffness and cytoskeletal orientation, thus impacting the biomechanics of the cell and aortic function.
Collapse
Affiliation(s)
- Hanna J Sanyour
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Na Li
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Alex P Rickel
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Haydee M Torres
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, 57007, USA
| | - Ruthellen H Anderson
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Miranda R Miles
- BioSNTR, Sioux Falls, SD, 57107, USA.,Mechanical Engineering Department, South Dakota State University, Brookings, SD, 57007, USA
| | - Josh D Childs
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| | - Kevin R Francis
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Jianning Tao
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD, 57104, USA.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.,Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, 57007, USA
| | - Zhongkui Hong
- Department of Biomedical Engineering, University of South Dakota, Sioux Falls, SD, 57107, USA.,BioSNTR, Sioux Falls, SD, 57107, USA
| |
Collapse
|
6
|
Villalpando DM, Gómez Rivas J, Flynn D, R de Bethencourt F, Ferrer M. Gonadal function protects against organ culture-induced vascular damage. Involvement of prostanoids. Prostaglandins Other Lipid Mediat 2020; 148:106406. [PMID: 31945460 DOI: 10.1016/j.prostaglandins.2019.106406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/25/2019] [Accepted: 12/23/2019] [Indexed: 02/03/2023]
Abstract
Androgen deprivation induces vascular dysfunction in which altered release and action of prostanoids has been extensively studied. On the other hand, the vascular organ-culture system has been reported as a valid model for phenotypic changes that occur in several cardiovascular pathologies. Since there are no studies analyzing the impact of androgenic loss on vascular vulnerability during induced vascular damage, the objective of this study was to analyze the possible preventive role of male sex hormones on the organ culture-induced vascular damage in rat aorta. The link to possible changes in gross structure was also analyzed. For this purpose, fresh and 20 h-cultured aortic arterial segments from intact and orchidectomized rats were used to analyze: (i) the release and vasomotor effect of the thromboxane A2 (TXA2), prostaglandin (PG) E2, PGF2α and PGI2; (ii) the vasodilator response induced by acetylcholine (ACh) as well as the involvement of prostanoids, in particular TXA2, in the ACh-induced response; (iii) the effect of activation of thromboxane/prostaglandin (TP) receptors on the ACh-induced response; and (iv) the vascular structure. The results showed that organ culture: i) increased production of prostanoids; ii) increased prostanoids-induced vasomotor responses; iii) decreased ACh-induced relaxation after incubation with indomethacin, a blocker of cyclooxygenases; iv) increased the ACh-induced relaxation after incubation with the TXA2 synthase inhibitor, furegrelate, more in arteries from orchidectomized rats than in those of intact rats; v) diminished ACh-induced relaxation after U-46619 incubation only in arteries from orchidectomized rats; and vi) preserved the integrity of the different vascular layers. These results showed the protective role of male sex hormones against the induced vascular damage, since a decreased deleterious effect of prostanoids, in particular that of TXA2, was observed in arteries from rats with intact gonadal function.
Collapse
Affiliation(s)
| | - Juan Gómez Rivas
- Servicio de Urología, Hospital Universitario La Paz, Madrid, Spain; Instituto de Investigación Hospital Universitario La Paz (IdiPAZ) Madrid, Spain
| | - Daniel Flynn
- Departamento de Fisiología, Facultad de Medicina, UAM, Spain
| | - Fermín R de Bethencourt
- Servicio de Urología, Hospital Universitario La Paz, Madrid, Spain; Instituto de Investigación Hospital Universitario La Paz (IdiPAZ) Madrid, Spain
| | - Mercedes Ferrer
- Departamento de Fisiología, Facultad de Medicina, UAM, Spain; Instituto de Investigación Hospital Universitario La Paz (IdiPAZ) Madrid, Spain.
| |
Collapse
|
7
|
Li B, Wang Z, Hu Z, Zhang M, Ren Z, Zhou Z, Huang J, Hu X. P38 MAPK Signaling Pathway Mediates Angiotensin II-Induced miR143/145 Gene Cluster Downregulation during Aortic Dissection Formation. Ann Vasc Surg 2017; 40:262-273. [PMID: 28167124 DOI: 10.1016/j.avsg.2016.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/18/2016] [Accepted: 09/09/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND We endeavored to prove that angiotensin II (Ang II) regulates both the expression of micro-RNA143/145 (miR143/145) and differentiation of vascular smooth muscle cells (VSMCs) during the formation of aortic dissection (AD). We also studied the contribution of p38 mitogen-activated protein kinase (MAPK) signaling pathway toward this process. METHODS Ascending aortic tissues were harvested from the patients with AD and organ donors. Tissues were immunostained with labeled antibodies targeting p38 MAPK, phospho-p38 MAPK, alpha-smooth muscle actin (α-SMA), and osteopontin (OPN). Next, we treated mouse aortic VSMCs with different regimens of Ang II (duration and dosages) in vitro and determined expression levels of miR143/145 and VSMC phenotype marker proteins (α-SMA and OPN) by quantitative polymerase chain reaction and/or western blotting. SB203580 was used to inhibit the p38 MAPK signaling pathway. Finally, the VSMC phenotype was validated by immunofluorescence microscopy. RESULTS Expression of phospho-p38 MAPK was significantly greater in the AD tissue. Ang II induced the phenotypic switching of VSMCs along with the downregulation of an miR143/145 gene cluster. Expression of OPN and phospho-p38 was significantly increased in VSMCs treated with 0.1 μM Ang II for 12 hr. Furthermore, the expression of miR143 and miR145 was downregulated by Ang II treatment. When the p38 MAPK signaling pathway was blocked by pretreatment with an SB203580 inhibitor, the expression of miR143, miR145, and VSMC phenotypic markers was not affected by Ang II. Immunohistochemical staining of aortic tissues donated by AD patients and healthy donors showed that the expression of α-SMA decreased in pathological tissue, while the OPN increased and the arrangement of the smooth muscle cells of the media was dysregulated, which we verified in vitro. CONCLUSIONS Ang II could regulate the expression of miR143/145 gene cluster and the phenotypic switching of VSMCs via the p38 MAPK signaling pathway. This may play an important role in the pathogenesis of AD.
Collapse
MESH Headings
- Actins/metabolism
- Adult
- Aortic Dissection/enzymology
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Angiotensin II/pharmacology
- Animals
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Case-Control Studies
- Cells, Cultured
- Dose-Response Relationship, Drug
- Female
- Humans
- Male
- Mice, Inbred C57BL
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Multigene Family
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Phosphorylation
- Signal Transduction/drug effects
- Time Factors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Bowen Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China.
| | - Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Zhen Zhou
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Jizhen Huang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People's Republic of China
| |
Collapse
|
8
|
Xie Q, Zhang D. Effects of Statins and Xuezhikang on the Expression of Secretory Phospholipase A2, Group IIA in Rat Vascular Smooth Muscle Cells. Int Heart J 2017; 58:115-124. [PMID: 28123160 DOI: 10.1536/ihj.16-163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Secretory phospholipase A2, group IIA (sPLA2-IIA) is involved in this process and plays a critical role. However, the exact role of sPLA2-IIA in cardiovascular inflammation is more complicated and remains unclear. Furthermore, both statins and Xuezhikang (XZK) are widely used in the prevention and treatment of cardiovascular disease risk because of their pleiotropic effects on the cardiovascular system. However, their effects on sPLA2-IIA are still controversial. We investigated the regulation of sPLA2-IIA by rat thoracic aorta smooth muscle cells (VSMCs) in culture. Cells were first incubated with IL-1β alone to induce expression of sPLA2-IIA and then treated with several concentrations of statins or XZK for different times in the absence or presence of IL-1β. We tested the expression of sPLA2-IIA, including sPLA2-IIA mRNA, protein, as well as activity. We found that statins or IL-1β increase the expression of sPLA2-IIA in VSMCs and the effect is based on a synergetic relationship between them. However, for the first time, we observed that XZK effectively reduces sPLA2-IIA expression in IL-1β-treated VSMCs. Our findings may shine a new light on the clinical use of XZK and statins in the prevention and treatment of atherosclerosis-related thrombosis.
Collapse
Affiliation(s)
- Qiang Xie
- Department of Cardiology, The First Hospital of Xiamen University
| | | |
Collapse
|
9
|
Deng CY, Yang H, Kuang SJ, Rao F, Xue YM, Zhou ZL, Liu XY, Shan ZX, Li XH, Lin QX, Wu SL, Yu XY. Upregulation of 5-hydroxytryptamine receptor signaling in coronary arteries after organ culture. PLoS One 2014; 9:e107128. [PMID: 25202989 PMCID: PMC4159325 DOI: 10.1371/journal.pone.0107128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/13/2014] [Indexed: 02/07/2023] Open
Abstract
Background 5-Hydroxytryptamine (5-HT) is a powerful constrictor of coronary arteries and is considered to be involved in the pathophysiological mechanisms of coronary-artery spasm. However, the mechanism of enhancement of coronary-artery constriction to 5-HT during the development of coronary artery disease remains to be elucidated. Organ culture of intact blood-vessel segments has been suggested as a model for the phenotypic changes of smooth muscle cells in cardiovascular disease. Methodology/Principal Findings We wished to characterize 5-HT receptor-induced vasoconstriction and quantify expression of 5-HT receptor signaling in cultured rat coronary arteries. Cumulative application of 5-HT produced a concentration-dependent vasoconstriction in fresh and 24 h-cultured rat coronary arteries without endothelia. 5-HT induced greater constriction in cultured coronary arteries than in fresh coronary arteries. U46619- and CaCl2-induced constriction in the two groups was comparable. 5-HT stimulates the 5-HT2A receptor and cascade of phospholipase C to induce coronary vasoconstriction. Calcium influx through L-type calcium channels and non-L-type calcium channels contributed to the coronary-artery constrictions induced by 5-HT. The contractions mediated by non-L-type calcium channels were significantly enhanced in cultured coronary arteries compared with fresh coronary arteries. The vasoconstriction induced by thapsigargin was also augmented in cultured coronary arteries. The decrease in Orai1 expression significantly inhibited 5-HT-evoked entry of Ca2+ in coronary artery cells. Expression of the 5-HT2A receptor, Orai1 and STIM1 were augmented in cultured coronary arteries compared with fresh coronary arteries. Conclusions An increased contraction in response to 5-HT was mediated by the upregulation of 5-HT2A receptors and downstream signaling in cultured coronary arteries.
Collapse
MESH Headings
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
- Animals
- Calcium/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- ORAI1 Protein
- Organ Culture Techniques/methods
- Rats
- Rats, Sprague-Dawley
- Receptor, Serotonin, 5-HT2A/genetics
- Receptor, Serotonin, 5-HT2A/metabolism
- Serotonin/genetics
- Serotonin/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Stromal Interaction Molecule 1
- Thapsigargin/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/genetics
- Vasoconstriction/drug effects
- Vasoconstriction/genetics
Collapse
Affiliation(s)
- Chun-Yu Deng
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Hui Yang
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Su-Juan Kuang
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Fang Rao
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Yu-Mei Xue
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhi-Ling Zhou
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xiao-Ying Liu
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Zhi-Xin Shan
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Xiao-Hong Li
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Qiu-Xiong Lin
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Shu-Lin Wu
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
- * E-mail: (SLW); (XYY)
| | - Xi-Yong Yu
- Medical Research Center of Guangdong General Hospital, Guangzhou, P.R. China
- Guangdong Provincial Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
- * E-mail: (SLW); (XYY)
| |
Collapse
|
10
|
Yin Y, Wu X, Yang Z, Zhao J, Wang X, Zhang Q, Yuan M, Xie L, Liu H, He Q. The potential efficacy of R8-modified paclitaxel-loaded liposomes on pulmonary arterial hypertension. Pharm Res 2013; 30:2050-62. [PMID: 23756757 DOI: 10.1007/s11095-013-1058-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 04/10/2013] [Indexed: 11/26/2022]
Abstract
PURPOSE In this paper, a novel liposomal formulation of paclitaxel modified with octaarginine (R8) was fabricated and the therapeutic efficacy of it on pulmonary arterial hypertension was evaluated. METHODS Octaarginine-modified stealth liposomes loaded with PTX (R8-PTX-LIP) were prepared and characterized. Vector cytoxicity and anti-proliferation ability of different formulations on primary cultured VSMCs were determined with MTT assay. The uptake capacity of VSMCs on different formulations were evaluated by flow cytometry, and the influences on cytoskeletons of liposomes were investigated by cytoskeleton staining with rhodamine-phalloidin. The biodistribution of liposomes were imaged by a CCD camera using a near-infrared fluorophore DiD. The therapeutic efficacy of different PTX-formulations of PAH was evaluated by hemodynamic measurement, right ventricular hypertrophic parameters and vessel diameters. RESULTS The cellular uptake of R8 modified liposomes (R8-LIP) was improved noticeably compared with other groups. All liposomes did not exert cytotoxicity on VSMCs in 24 h. R8-PTX-LIP exhibited the strongest inhibitory effect on the proliferation of VSMCs among all the formulations (p < 0.001). R8-PTX-LIP could reverse the phenotype transformation, and inhibit cell migration. mPAP, (RV/LV+S) and the wall thickness of small distal pulmonary arteries of rats treated with R8-PTX-LIP were significantly lower than those from other groups (p < 0.001). CONCLUSIONS In conclusion, the drug delivery system of R8-modified paclitaxel-loaded liposomes we established showed pronounced inhibitory effect over VSMCs proliferation and cytoskeleton formation in vitro, a stronger pulmonary delivery ability in vivo, and was effective on PAH, showing the potential for pulmonary drug delivery system for PAH treatment.
Collapse
Affiliation(s)
- Yujia Yin
- The Pulmonary Vascular Remodeling Research Unit Department of Pediatric, West China Second University Hospital, Sichuan University, No. 20, Section 3, RenminNanLu Road, Chengdu, Sichuan 610041, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Addition of adult serum improves endothelium-dependent relaxation of organ-cultured rat mesenteric artery via inhibiting mitochondrial reactive oxygen species. Vascul Pharmacol 2013; 58:105-11. [DOI: 10.1016/j.vph.2012.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/31/2012] [Accepted: 08/26/2012] [Indexed: 11/24/2022]
|
12
|
Long X, Cowan SL, Miano JM. Mitogen-activated protein kinase 14 is a novel negative regulatory switch for the vascular smooth muscle cell contractile gene program. Arterioscler Thromb Vasc Biol 2012; 33:378-86. [PMID: 23175675 DOI: 10.1161/atvbaha.112.300645] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Several studies have shown through chemical inhibitors that p38 mitogen-activated protein kinase (MAPK) promotes vascular smooth muscle cell (VSMC) differentiation. Here, we evaluate the effects of knocking down a dominant p38MAPK isoform on VSMC differentiation. METHODS AND RESULTS Knockdown of p38MAPKα (MAPK14) in human coronary artery SMCs unexpectedly increases VSMC differentiation genes, such as miR145, ACTA2, CNN1, LMOD1, and TAGLN, with little change in the expression of serum response factor (SRF) and 2 SRF cofactors, myocardin (MYOCD) and myocardin-related transcription factor A (MKL1). A variety of chemical and biological inhibitors demonstrate a critical role for a RhoA-MKL1-SRF-dependent pathway in mediating these effects. MAPK14 knockdown promotes MKL1 nuclear localization and VSMC marker expression, an effect partially reversed with Y27632; in contrast, MAP2K6 (MKK6) blocks MKL1 nuclear import and VSMC marker expression. Immunostaining and Western blotting of injured mouse carotid arteries reveal elevated MAPK14 (both total and phosphorylated) and reduced VSMC marker expression. CONCLUSIONS Reduced MAPK14 expression evokes unanticipated increases in VSMC contractile genes, suggesting an unrecognized negative regulatory role for MAPK14 signaling in VSMC differentiation.
Collapse
Affiliation(s)
- Xiaochun Long
- Department of Medicine, Aab Cardiovascular Research Institute, Box CVRI, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
13
|
Morita T, Okada M, Hara Y, Yamawaki H. Mechanisms underlying impairment of endothelium-dependent relaxation by fetal bovine serum in organ-cultured rat mesenteric artery. Eur J Pharmacol 2011; 668:401-6. [DOI: 10.1016/j.ejphar.2011.07.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/21/2011] [Accepted: 07/30/2011] [Indexed: 11/29/2022]
|
14
|
Chuang YH, Chuang WL, Huang SP, Liu CK, Huang CH. Atorvastatin ameliorates tissue damage of obstructed ureter in rats. Life Sci 2011; 89:795-805. [PMID: 21971118 DOI: 10.1016/j.lfs.2011.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 08/18/2011] [Accepted: 09/13/2011] [Indexed: 10/17/2022]
Abstract
AIMS To investigate the effects of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor on the tissue damage and fibrosis of obstructed ureters, 80 rats were studied. MAIN METHODS Atorvastatin, a HMG-CoA reductase inhibitor, was administered to 40 rats at the dose of 20 mg/kg per day 1day before unilateral ligation of ureters and every day thereafter. The other rats served as controls. Eight rats from each group were sacrificed for examination on days 7, 14, 21, 28 and 42 after ligation, respectively. The expressions of transforming growth factor-β1 (TGF-β1), Interleukine-1β (IL-1β), Interleukine-6 (IL-6), tumor necrosis factor-alpha (TNF-α), proliferation cell nuclear antigen (PCNA), and the apoptotic cells in the ureteric smooth muscle were examined. KEY FINDINGS Hydroureter and fibrosis of the muscle layer became progressively aggravated in the ligated ureters of the atorvastatin-treated group and control group. The severities of hydroureter and muscle layer fibrosis in the ligated ureters of the treated group were significantly less than in the control group. The atorvastatin administration also decreased the expression of TGF-β1, IL-1β, IL-6, TNF-α, PCNA and the labeling index of apoptotic cells in the smooth muscle layer of ligated ureters in the treated group. SIGNIFICANCE We concluded that atorvastatin might ameliorate the tissue damage of obstructed ureters, at least partially, via the inhibition on TGF-β1) expression and by diminishing the effects of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Yen-Hwang Chuang
- Department of Anatomy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
15
|
Kaneda T, Tsuruoka S, Fujimura A. Statins inhibited erythropoietin-induced proliferation of rat vascular smooth muscle cells. Eur J Pharmacol 2010; 649:38-43. [DOI: 10.1016/j.ejphar.2010.08.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 08/28/2010] [Accepted: 08/31/2010] [Indexed: 12/25/2022]
|
16
|
Chronic treatment with PDGF-BB and endothelin-1 synergistically induces vascular hyperplasia and loss of contractility in organ-cultured rat tail artery. Atherosclerosis 2010; 214:288-94. [PMID: 21129745 DOI: 10.1016/j.atherosclerosis.2010.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2009] [Revised: 10/23/2010] [Accepted: 11/04/2010] [Indexed: 01/21/2023]
Abstract
OBJECTIVE In this study, we examined the synergistic effects of the two potent pathogenic factors, platelet-derived growth factor-BB (PDGF-BB) and endothelin-1 (ET-1) to induce vascular hyperplasia using ex vivo organ-culture system. METHODS AND RESULTS In organ-cultured rat tail arteries, concomitant treatment with 100 ng/ml PDGF-BB and 300 nM ET-1 for 4 days induced medial hyperplasia with increased smooth muscle cell proliferation. Concomitant treatment with PDGF-BB (10-300 nM) and ET-1 (30 nM-1 μM) dose-dependently suppressed contractile responses to high K(+) and norepinephrine. This dyscontractility was accompanied by decreased α-actin protein expression. In all series of experiments, concomitant treatment with PDGF-BB and ET-1 exhibited stronger effects than sole treatment with PDGF-BB (100 ng/ml) or ET-1 (300 nM). Western blot analysis revealed that concomitant treatment with PDGF-BB and ET-1 synergistically phosphorylated extracellular signal-regulated kinase 1 and 2 (ERK1/2), Akt, and a downstream target of mammalian target of rapamycin (mTOR), p70 ribosomal S6 kinase in cultured artery. Consistently, a MAPK/ERK kinase (MEK) inhibitor, PD98059 (30 μM), a phosphoinositide 3-kinase (PI3K) inhibitor, LY294002, and an mTOR inhibitor, rapamycin (30 nM), partially restored PDGF-BB and ET-1-induced hyperplastic changes. CONCLUSIONS We evidenced for the first time at tissue level that PDGF-BB and ET-1 synergistically accelerate vascular smooth muscle hyperplastic changes and lose its contractility, at least partially through ERK1/2, Akt, and mTOR activation.
Collapse
|
17
|
Saha PK, Reddy VT, Konopleva M, Andreeff M, Chan L. The triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic-acid methyl ester has potent anti-diabetic effects in diet-induced diabetic mice and Lepr(db/db) mice. J Biol Chem 2010; 285:40581-92. [PMID: 20956520 DOI: 10.1074/jbc.m110.176545] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The triterpenoid 2-Cyano-3,12-dioxooleana-1,9-dien-28-oic-acid (CDDO) and its methyl ester (CDDO-Me) are undergoing clinical trials in cancer and leukemia therapy. Here we report that CDDO-Me ameliorates diabetes in high fat diet-fed type 2 diabetic mice and in Lepr(db/db) mice. CDDO-Me reduces proinflammatory cytokine expression in these animals. Oral CDDO-Me administration reduces total body fat, plasma triglyceride, and free fatty acid levels. It also improves glucose tolerance and insulin tolerance tests. Its potent glucose-lowering activity results from enhanced insulin action. Hyperinsulinemic-euglycemic clamp reveals an increased glucose infusion rate required to maintain euglycemia and showed a significant increase in muscle-specific insulin-stimulated glucose uptake (71% soleus, 58% gastrocnemius) and peripheral glucose clearance as documented by a 48% increase in glucose disposal rate. CDDO-Me activates AMP-activated protein kinase (AMPK) and via LKB1 activation in muscle and liver in vivo. Treatment of isolated hepatocytes with CDDO-Me directly stimulates AMPK activity and LKB1 phosphorylation and decreases acetyl-coA carboxylase activity; it also down-regulates lipogenic gene expression, suppresses gluconeogenesis, and increases glucose uptake. Inhibition of AMPK phosphorylation using compound C and lentiviral-mediated knockdown of AMPK completely blocks the CDDO-Me-induced effect on hepatocytes as well as C(2)C(12) cells. We conclude that the triterpenoid CDDO-Me has potent anti-diabetic action in diabetic mouse models that is mediated at least in part through AMPK activation. The in vivo anti-diabetogenic effects occur at a dose substantially lower than that used for anti-leukemia therapy. We suggest that CDDO-Me holds promise as a potential anti-diabetic agent.
Collapse
Affiliation(s)
- Pradip K Saha
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
18
|
Liu YJ, Wang XG, Tang YB, Chen JH, Lv XF, Zhou JG, Guan YY. Simvastatin Ameliorates Rat Cerebrovascular Remodeling During Hypertension via Inhibition of Volume-Regulated Chloride Channel. Hypertension 2010; 56:445-52. [DOI: 10.1161/hypertensionaha.110.150102] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Yu-Jie Liu
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Xiao-Guang Wang
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Yong-Bo Tang
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jing-Hui Chen
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Xiao-Fei Lv
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jia-Guo Zhou
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Yong-Yuan Guan
- From the Department of Pharmacology (Y.-J.L., X.-G.W., Y.-B.T., J.-H.C., X.-F.L., J.-G.Z., Y.-Y.G.), Cardiac and Cerebral Vascular Research Center (Y.-J.L., X.-G.W., Y.-B.T., X.-F.L., J.-G.Z., Y.-Y.G.), Zhongshan School of Medcine, Sun Yat-Sen University, Guangzhou, People’s Republic of China; Department of Anaesthesia (J.-H.C.), Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
19
|
Luo DX, Cheng J, Xiong Y, Li J, Xia C, Xu C, Wang C, Zhu B, Hu Z, Liao DF. Static pressure drives proliferation of vascular smooth muscle cells via caveolin-1/ERK1/2 pathway. Biochem Biophys Res Commun 2010; 391:1693-7. [DOI: 10.1016/j.bbrc.2009.12.132] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 12/23/2009] [Indexed: 10/20/2022]
|
20
|
Aoshima D, Murata T, Hori M, Ozaki H. Time-dependent phenotypic and contractile changes of pulmonary artery in chronic hypoxia-induced pulmonary hypertension. J Pharmacol Sci 2009; 110:182-90. [PMID: 19498269 DOI: 10.1254/jphs.09059fp] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Phenotypic and contractile changes in pulmonary arterial smooth muscle cells (PASMCs) were examined in rats with pulmonary hypertension induced by hypoxia. Exposure to hypoxia induced pulmonary hypertension within 1-4 weeks. Staining with BrdU revealed that proliferative activities of PASMCs peaked at 1 week of hypoxic exposure, and then moderate proliferative activity was maintained for the next 2-4 weeks. The beta-actin/alpha-actin ratio also increased at 1-2 weeks of exposure to hypoxia. Absolute contractility of the pulmonary arterial ring continuously decreased during hypoxia, whereas the basal active tonus of the pulmonary artery increased at 1-3 weeks. Nicardipine, the ETA-receptor antagonis, CI-1034 and the rho-kinase inhibitor Y27632 partially inhibited the elevated active tonus. Endothelin-1 content in the pulmonary hypertensive lung was continuously increased during exposure to hypoxia. In conclusion, the hypoxia-induced proliferative activity of PASMCs comprised a transient phase followed by a sustained phase. The change in PASMCs from a contractile to a synthetic phenotype also correlated with proliferative activity, which subsequently decreased PASMC contractility. The continuous production of endothelin-1 upon hypoxic exposure might contribute to the increased basal tonus of the pulmonary arterial wall, which might subsequently increase pulmonic arterial pressure, resulting in accelerated pulmonary hypertension.
Collapse
Affiliation(s)
- Daigo Aoshima
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
21
|
Fujimoto M, Oka T, Murata T, Hori M, Ozaki H. Fluvastatin inhibits mast cell degranulation without changing the cytoplasmic Ca2+ level. Eur J Pharmacol 2009; 602:432-8. [DOI: 10.1016/j.ejphar.2008.11.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 11/13/2008] [Accepted: 11/20/2008] [Indexed: 10/21/2022]
|
22
|
Kiyan J, Kusch A, Tkachuk S, Krämer J, Haller H, Dietz R, Smith G, Dumler I. Rosuvastatin regulates vascular smooth muscle cell phenotypic modulation in vascular remodeling: Role for the urokinase receptor. Atherosclerosis 2007; 195:254-61. [PMID: 17275828 DOI: 10.1016/j.atherosclerosis.2006.12.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 12/11/2006] [Accepted: 12/21/2006] [Indexed: 01/12/2023]
Abstract
The urokinase (uPA)/urokinase receptor (uPAR) multifunctional system is an important mediator of migration and proliferation of vascular smooth muscle cells (VSMC). However, whether uPA/uPAR-directed mechanisms are involved in the beneficial effects of 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors on vascular remodeling remains unexplored. In this study, we have investigated the effect of the hydrophilic statin rosuvastatin on neointimal remodeling, and the role of uPAR. Using an ex vivo organ and in vitro cell culture models we demonstrate that rosuvastatin decreases injury-induced neointima formation and proliferation of medial VSMC in porcine coronary arteries, as well as migration and proliferation of human coronary VSMC. Studies on the underlying mechanisms show that rosuvastatin impairs VSMC transition from their physiological contractile to the pathophysiological synthetic phenotype. These effects are mediated, at least in part, via uPAR, as confirmed by means of rosuvastatin-directed uPAR expression and uPAR silencing in both models. Our findings provide evidence that rosuvastatin modulates VSMC phenotypic changes and subsequently their proliferation and migration, and indicate the important role for uPAR in these processes. This mechanism contributes to the beneficial non-lipid lowering effect of rosuvastatin on negative vascular remodeling.
Collapse
MESH Headings
- Angioplasty, Balloon, Coronary/adverse effects
- Animals
- Cells, Cultured
- Coronary Vessels/cytology
- Coronary Vessels/drug effects
- Coronary Vessels/injuries
- Down-Regulation
- Female
- Fluorobenzenes/pharmacology
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Pyrimidines/pharmacology
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/metabolism
- Receptors, Urokinase Plasminogen Activator
- Rosuvastatin Calcium
- Signal Transduction
- Sulfonamides/pharmacology
- Sus scrofa
- Tunica Intima/drug effects
- Up-Regulation
- Urokinase-Type Plasminogen Activator/drug effects
Collapse
Affiliation(s)
- Julia Kiyan
- Hannover Medical School, Carl-Neuberg Strasse 1, Hannover D-30625, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Xu YG, Zhou SH, Li YG, Zheng CH, Li XP, Liu QM, Xu DM, Chen S. The mechanism underlying vascular smooth muscle cell apoptosis induced by atorvastatin may be mainly associated with down-regulation of survivin expression. Cardiovasc Drugs Ther 2007; 21:145-53. [PMID: 17372815 DOI: 10.1007/s10557-007-6018-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Studies have shown that statins may induce vascular smooth muscle cells (VSMCs) apoptosis. But its mechanisms are incompletely understood. In this study, we investigate the effects of atorvastatin and survivin antisense oligonucleotides (ASODN) on VSMCs apoptosis and the relation between survivin and VSMCs apoptosis. MATERIALS AND METHODS Cultured VSMCs were treated with atorvastatin and vascular endothelial grow factor (VEGF). Apoptosis of VSMCs at 6-72 h after treatment with atorvastatin was detected by means of Hoechst33258 staining. Survivin and Fas factors expression were detected by means of immunohistochemistry. Survivin and Fas mRNA expression were detected by means of RT-PCR. In order to determine the relations between survivin and VSMCs apoptosis, we also performed transfection of VSMCs with survivin ASODN using GenePORTER Transfection Reagent and studied the survivin protein expression by means of western blotting. RESULTS VSMCs apoptosis after treatment with atorvastatin was increased in a dose- and time-dependent manner. The expression of survivin and survivin mRNA in VSMCs was significantly down-regulated at 24 h and disappeared at 48-72 h after treatment with atorvastatin. Fas and Fas mRNA in VSMCs could only be detected at 72 h and not at 6-48 h after treatment with atorvastatin. We did not observe any effects of VEGF on VSMCs apoptosis, on survivin and survivin mRNA expression, and on Fas and Fas mRNA expression in VSMCs after treatment with atorvastatin. At 48 hours after the start of transfection, survivin protein expression was significantly reduced after transfection with 0.5, 1.0 and 2.0 microg/ml of survivin ASODN and there was no survivin protein expression after transfection with 3.0 and 4.0 microg/ml of survivin ASODN. In contrast, in the GenePORTER only and SODN studies, survivin protein expression was observed with western blotting. Hoechst33258 staining showed that treatment of VSMCs with survivin ASODN resulted in VSMCs apoptosis. CONCLUSIONS Atorvastatin induces VSMCs apoptosis in a dose- and time-dependent manner. Transfection of survivin ASODN can directly induce VSMCs apoptosis. The mechanisms of VSMCs apoptosis induced by atorvastatin may be mainly associated with down-regulation of survivin expression in VSMCs. Up-regulation of Fas in VSMCs may play a role in later stages following atorvastatin treatment.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Atorvastatin
- Blotting, Western
- Cell Nucleus/metabolism
- Cells, Cultured
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Drug Therapy, Combination
- Fas Ligand Protein/genetics
- Fas Ligand Protein/metabolism
- Heptanoic Acids/pharmacology
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oligonucleotides, Antisense/genetics
- Pyrroles/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Reverse Transcriptase Polymerase Chain Reaction
- Survivin
- Time Factors
- Transfection
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Yi-Guan Xu
- Cardiovascular Department, First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Vecchione C, Gentile MT, Aretini A, Marino G, Poulet R, Maffei A, Passarelli F, Landolfi A, Vasta A, Lembo G. A novel mechanism of action for statins against diabetes-induced oxidative stress. Diabetologia 2007; 50:874-80. [PMID: 17279352 DOI: 10.1007/s00125-007-0597-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS Atorvastatin exerts beneficial vascular effects in diabetes, but the underlying mechanisms are yet to be elucidated. The aim of the present study was to determine whether Rac-1 is involved in the effect of atorvastatin on oxidative stress and vascular dysfunction. MATERIALS AND METHODS Using human aortic endothelial cells (HAECs) we evaluated the effect of high glucose levels on peroxide production by dihydrodichlorofluorescein and on Rac-1 activity using immunocytochemistry to detect Rac-1 translocation to the membrane. We evaluated vascular function, peroxide production by dihydroethidium and NADPH oxidase activity in vessels from atorvastatin-treated mice. Rac-1 activity was also assessed, both by immunoprecipitation of the Rac-p21-activated kinase complex and by analysis of Rac-1 translocation to the membrane. These experiments were also conducted in vessels infected with an adenoviral vector carrying a constitutively active mutant of Rac-1. RESULTS In HAECs exposed to high glucose levels, atorvastatin prevented oxidative stress, and this protection was associated with impaired Rac-1 activation. This effect was also observed in a murine model of diabetes mellitus. More importantly, the addition of geranylgeranyl pyrophosphate (GGPP) blocked the effects of atorvastatin in both glucose-exposed HAECs and diabetic vessels. Atorvastatin failed to afford protection against vascular abnormalities in the presence of a constitutively active mutant of Rac-1. CONCLUSIONS/INTERPRETATION The results of this study demonstrate that the vascular antioxidant effect of atorvastatin in diabetes is mediated through inhibition of Rac-1 via a reduction in GGPP. Thus, selective Rac-1 inhibition should be considered in the design of novel pharmacological strategies to reduce the impact of diabetes mellitus on vascular function.
Collapse
Affiliation(s)
- C Vecchione
- Department of Angio-cardio-neurology, IRCCS Neuromed, Località Camerelle, 86077, Pozzilli (IS), Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tristano AG, Fuller K. Immunomodulatory effects of statins and autoimmune rheumatic diseases: novel intracellular mechanism involved. Int Immunopharmacol 2006; 6:1833-1846. [PMID: 17052674 DOI: 10.1016/j.intimp.2006.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 07/25/2006] [Accepted: 08/03/2006] [Indexed: 01/26/2023]
Abstract
Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, known as statins, are the most commonly prescribed agents for the treatment of hypercholesterolemia. However, the effects of statins may extend beyond their influences on serum cholesterol levels resulting in cholesterol-independent or pleiotropic effects. Clinical, animal and in vitro studies suggest that statins have additional clinical uses because of their anti-inflammatory and immunomodulatory effects, in part due to their capacity to interfere with the mevalonate pathway and inhibit prenylation of Rho family GTPases. This review focuses on the molecular mechanisms of the anti-inflammatory and immunomodulatory effects of statins. In base to all these information, we suggest that statins could have similar inhibitory effects on MAPKs pathways in cells from RA patients, including osteoclasts and fibroblasts.
Collapse
Affiliation(s)
- Antonio G Tristano
- Pharmaceutical and Administrative Sciences Department, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | | |
Collapse
|
26
|
Abstract
BACKGROUND Changes in the proteoglycan metabolism of the intima of arteries belong to the initial lesions of atherosclerosis (AS). The accumulation of proteoglycans, alterations of pericellular glycoproteins and modulations of collagen turnover also play a fundamental role in the progression of AS. They influence lipid retention, cell behavior and calcinosis. The decisive role played by the matrix metalloproteinases (MMPs) and their inhibiting factors (tissue inhibitors of metalloproteinases [TIMPs]) in these processes is not yet fully understood and therefore the subject of this overview. The causes of the abrupt change of a long-term existing stabile AS to a vulnerable plaque as well as the participation of age-related vascular wall remodeling in the progression of AS also remain open questions. DISCUSSION Apart from the well-known risk factors for AS, less well-known influences like the disturbances of gene expression in vascular smooth muscle cells affect an MMP/TIMP imbalance. The various consequences of this imbalance range from intima cell proliferation as an early change in AS as well as accelerated progression to the destabilization of fibrous plaques by increased collagenolysis as well as the formation of aneurysms. Infectious or toxic influences may trigger these mechanisms; an involvement of age-related vessel wall changes should also be considered. The prognostic significance of circulating MMP concentrations for the existence of instabile plaques are of great interest, as is the plaque stabilizing effect of statins by suppression of MMPs. CONCLUSIONS MMPs navigate the behavior of vascular wall cells in different AS stages, in adaptive remodeling, in normal aging and in non-atherosclerotic vessel disease. The clinical relevance of a disturbance in the MMP/TIMP balance is demonstrated firstly by the initiation of AS due to migration and proliferation of intima cells and secondly in the collagenolysis, necrotic transformation and apoptosis of existing fibrous lesions resulting in instabile rupture proned plaques. Investigations into the genetic typing of MMPs and the results of experimental gene deficiency models have significantly contributed to the clarification of these facts.
Collapse
|
27
|
McGillicuddy FC, O'Toole D, Hickey JA, Gallagher WM, Dawson KA, Keenan AK. TGF-beta1-induced thrombospondin-1 expression through the p38 MAPK pathway is abolished by fluvastatin in human coronary artery smooth muscle cells. Vascul Pharmacol 2006; 44:469-75. [PMID: 16624629 DOI: 10.1016/j.vph.2006.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 02/13/2006] [Accepted: 03/09/2006] [Indexed: 10/24/2022]
Abstract
Thrombospondin-1 (TSP-1) and transforming growth factor-beta1 (TGF-beta1) are both implicated in the pathogenesis of in-stent restenosis. This study evaluated the hypothesis that the HMG-CoA reductase inhibitor fluvastatin inhibits TGF-beta1 induced TSP-1 expression via inhibition of p38 mitogen activated protein kinase (MAPK) phosphorylation in human coronary artery smooth muscle cells (HCASMC) and may therefore have anti-restenosis potential. Fluvastatin significantly reduced TSP-1 mRNA and protein expression in HCASMC in a concentration-dependent manner with a significant reduction in expression observed after treatment with 0.25 microM fluvastatin. TGF-beta1 (5 ng/ml) induced phosphorylation of p38 MAPK and induced TSP-1 mRNA and protein expression in HCASMC. Fluvastatin abolished TGF-beta1-induced phosphorylation of p38 MAPK and TGF-beta1-induced TSP-1 expression. Blockade of the p38 MAPK pathway with the upstream inhibitor SB-203580 also abolished TGF-beta1-induced TSP-1 expression. We conclude that fluvastatin decreases expression of TSP-1 and abolishes the ability of TGF-beta1 to induce TSP-1 expression in HCASMC; this may be achieved by preventing signalling through the p38 MAPK pathway. Targeted delivery of fluvastatin may therefore be a useful therapeutic objective for prevention of the intimal hyperplasia associated with in-stent restenosis.
Collapse
MESH Headings
- Adult
- Cells, Cultured
- Coronary Restenosis/prevention & control
- Coronary Vessels/drug effects
- Coronary Vessels/enzymology
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Fatty Acids, Monounsaturated/pharmacology
- Fatty Acids, Monounsaturated/therapeutic use
- Fluvastatin
- Gene Expression Regulation
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use
- Imidazoles/pharmacology
- Indoles/pharmacology
- Indoles/therapeutic use
- MAP Kinase Signaling System
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Phosphorylation
- Pyridines/pharmacology
- RNA, Messenger/metabolism
- Thrombospondin 1/genetics
- Thrombospondin 1/metabolism
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta1
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Fiona C McGillicuddy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|