51
|
Zhang W, Feng J, Cheng B, Lu Q, Chen X. Oleanolic acid protects against oxidative stress‑induced human umbilical vein endothelial cell injury by activating AKT/eNOS signaling. Mol Med Rep 2018; 18:3641-3648. [PMID: 30106101 PMCID: PMC6131357 DOI: 10.3892/mmr.2018.9354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/20/2018] [Indexed: 02/05/2023] Open
Abstract
Oxidative injury of vascular endothelial cells in the initial event of atherosclerosis (AS) in diabetes was assessed in the present study. The antioxidant effect of oleanolic acid (OA) has attracted much attention. In the present study the potential effects of OA on human umbilical vein endothelial cells (HUVECs) were investigated. Cell viability was examined using the CCK‑8 assay. The activity of oxidative stress parameters was determined using commercial kits. Flow cytometry analysis was performed to detect the level of reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and cell apoptosis. The expression levels of target genes and proteins were examined by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis. It was indicated that cell viability that was suppressed by high glucose was increased by the pretreatment of OA, and nitric oxide (NO) generation, the activities of superoxide dismutase (SOD) and catalase (CAT) were recovered by OA. By contrast, it was observed that OA decreased the MDA content. Notably, the pretreatment of OA alleviated mitochondria damage by reducing the level of ROS and maintaining MMP. In addition, apoptosis that was caused by high glucose was reduced by OA. Pro‑apoptotic genes (caspase‑3, Fas, Fasl) and anti‑apoptotic gene (Bcl‑2) expression levels were decreased and increased in the OA groups, respectively. Furthermore, the activity of AKT/endothelial nitric oxide synthase (eNOS) signaling was elevated by OA. Taken together, it was suggested that OA could protect against oxidative stress‑induced apoptosis of HUVECs, which was associated with AKT/eNOS signaling pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Geriatric Cardiovascular Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Jian Feng
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Biao Cheng
- Department of Geriatric Cardiovascular Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Qing Lu
- Department of Geriatric Cardiovascular Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xiaoping Chen
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
52
|
Sunggip C, Shimoda K, Oda S, Tanaka T, Nishiyama K, Mangmool S, Nishimura A, Numaga-Tomita T, Nishida M. TRPC5-eNOS Axis Negatively Regulates ATP-Induced Cardiomyocyte Hypertrophy. Front Pharmacol 2018; 9:523. [PMID: 29872396 PMCID: PMC5972289 DOI: 10.3389/fphar.2018.00523] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/01/2018] [Indexed: 01/19/2023] Open
Abstract
Cardiac hypertrophy, induced by neurohumoral factors, including angiotensin II and endothelin-1, is a major predisposing factor for heart failure. These ligands can induce hypertrophic growth of neonatal rat cardiomyocytes (NRCMs) mainly through Ca2+-dependent calcineurin/nuclear factor of activated T cell (NFAT) signaling pathways activated by diacylglycerol-activated transient receptor potential canonical 3 and 6 (TRPC3/6) heteromultimer channels. Although extracellular nucleotide, adenosine 5'-triphosphate (ATP), is also known as most potent Ca2+-mobilizing ligand that acts on purinergic receptors, ATP never induces cardiomyocyte hypertrophy. Here we show that ATP-induced production of nitric oxide (NO) negatively regulates hypertrophic signaling mediated by TRPC3/6 channels in NRCMs. Pharmacological inhibition of NO synthase (NOS) potentiated ATP-induced increases in NFAT activity, protein synthesis, and transcriptional activity of brain natriuretic peptide. ATP significantly increased NO production and protein kinase G (PKG) activity compared to angiotensin II and endothelin-1. We found that ATP-induced Ca2+ signaling requires inositol 1,4,5-trisphosphate (IP3) receptor activation. Interestingly, inhibition of TRPC5, but not TRPC6 attenuated ATP-induced activation of Ca2+/NFAT-dependent signaling. As inhibition of TRPC5 attenuates ATP-stimulated NOS activation, these results suggest that NO-cGMP-PKG axis activated by IP3-mediated TRPC5 channels underlies negative regulation of TRPC3/6-dependent hypertrophic signaling induced by ATP stimulation.
Collapse
Affiliation(s)
- Caroline Sunggip
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Biomedical Science and Therapeutic, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| | - Kakeru Shimoda
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Sayaka Oda
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Tomohiro Tanaka
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kazuhiro Nishiyama
- Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Akiyuki Nishimura
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Takuro Numaga-Tomita
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Creative Research Group on Cardiocirculatory Dynamism, Exploratory Research Center on Life and Living Systems, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
- Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
53
|
Suksawat M, Techasen A, Namwat N, Boonsong T, Titapun A, Ungarreevittaya P, Yongvanit P, Loilome W. Inhibition of endothelial nitric oxide synthase in cholangiocarcinoma cell lines - a new strategy for therapy. FEBS Open Bio 2018; 8:513-522. [PMID: 29632805 PMCID: PMC5881549 DOI: 10.1002/2211-5463.12388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 12/14/2017] [Accepted: 01/01/2018] [Indexed: 12/18/2022] Open
Abstract
The isoform of nitric oxide synthase (NOS) found in endothelial cells (eNOS) plays a crucial role in vasodilation. We recently reported the activation of eNOS in cholangiocarcinoma (CCA) tissues and cell lines. Moreover, we also reported that the abundance of eNOS and phosphorylated eNOS (p-eNOS), as well as its upstream regulator proteins, is significantly associated with the metastatic status of CCA patients. However, the function of eNOS in CCA progression has not been addressed. Therefore, the present study aimed to investigate the function of eNOS involved in the migration and invasion ability of CCA cell lines. The results reveal that eNOS activation significantly increases migration and invasion ability of CCA cells via the up-regulation of phosphorylated vasodilator-stimulated protein (p-VASP). A combination treatment with recombinant human vascular endothelial growth factor C and eNOS inhibitor (Nω-nitro-l-arginine methyl ester hydrochloride) resulted in the down-regulation of p-VASP, as well as a decreased migration and invasion ability of the CCA cell line. Thus, this work suggests that eNOS can serve as an attractive target to inhibit the progression of CCA.
Collapse
Affiliation(s)
- Manida Suksawat
- Department of Biochemistry Faculty of Medicine Khon Kaen University Thailand.,Cholangiocarcinoma Research Institute Khon Kaen University Thailand
| | - Anchalee Techasen
- Cholangiocarcinoma Research Institute Khon Kaen University Thailand.,Faculty of Associated Medical Science Khon Kaen University Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand
| | - Nisana Namwat
- Department of Biochemistry Faculty of Medicine Khon Kaen University Thailand.,Cholangiocarcinoma Research Institute Khon Kaen University Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand
| | - Thianrut Boonsong
- Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand.,Department of Surgery Faculty of Medicine Khon Kaen University Thailand
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute Khon Kaen University Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand.,Department of Surgery Faculty of Medicine Khon Kaen University Thailand
| | - Piti Ungarreevittaya
- Cholangiocarcinoma Research Institute Khon Kaen University Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand.,Department of Pathology Faculty of Medicine Khon Kaen University Thailand
| | - Puangrat Yongvanit
- Cholangiocarcinoma Research Institute Khon Kaen University Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand
| | - Watcharin Loilome
- Department of Biochemistry Faculty of Medicine Khon Kaen University Thailand.,Cholangiocarcinoma Research Institute Khon Kaen University Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP) Khon Kaen University Thailand
| |
Collapse
|
54
|
Madfis N, Lin Z, Kumar A, Douglas SA, Platt MO, Fan Y, McCloskey KE. Co-Emergence of Specialized Endothelial Cells from Embryonic Stem Cells. Stem Cells Dev 2018; 27:326-335. [PMID: 29320922 DOI: 10.1089/scd.2017.0205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A well-formed and robust vasculature is critical to the health of most organ systems in the body. However, the endothelial cells (ECs) forming the vasculature can exhibit a number of distinct functional subphenotypes like arterial or venous ECs, as well as angiogenic tip and stalk ECs. In this study, we investigate the in vitro differentiation of EC subphenotypes from embryonic stem cells (ESCs). Using our staged induction methods and chemically defined mediums, highly angiogenic EC subpopulations, as well as less proliferative and less migratory EC subpopulations, are derived. Furthermore, the EC subphenotypes exhibit distinct surface markers, gene expression profiles, and positional affinities during sprouting. While both subpopulations contained greater than 80% VE-cad+/CD31+ cells, the tip/stalk-like EC contained predominantly Flt4+/Dll4+/CXCR4+/Flt-1- cells, while the phalanx-like EC was composed of higher numbers of Flt-1+ cells. These studies suggest that the tip-specific EC can be derived in vitro from stem cells as a distinct and relatively stable EC subphenotype without the benefit of its morphological positioning in the sprouting vessel.
Collapse
Affiliation(s)
- Nicole Madfis
- 1 Graduate Program in Quantitative and System Biology, University of California , Merced, Merced, California
| | - Zhiqiang Lin
- 2 School of Biological Sciences and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Ashwath Kumar
- 2 School of Biological Sciences and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Simone A Douglas
- 3 Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| | - Manu O Platt
- 3 Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| | - Yuhong Fan
- 2 School of Biological Sciences and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology , Atlanta, Georgia
| | - Kara E McCloskey
- 1 Graduate Program in Quantitative and System Biology, University of California , Merced, Merced, California.,4 Department of Materials Science and Engineering, University of California , Merced, Merced, California
| |
Collapse
|
55
|
Xu S, Liu B, Yin M, Koroleva M, Mastrangelo M, Ture S, Morrell CN, Zhang DX, Fisher EA, Jin ZG. A novel TRPV4-specific agonist inhibits monocyte adhesion and atherosclerosis. Oncotarget 2018; 7:37622-37635. [PMID: 27191895 PMCID: PMC5122337 DOI: 10.18632/oncotarget.9376] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 04/29/2016] [Indexed: 11/25/2022] Open
Abstract
TRPV4 ion channel mediates vascular mechanosensitivity and vasodilation. Here, we sought to explore whether non-mechanical activation of TRPV4 could limit vascular inflammation and atherosclerosis. We found that GSK1016790A, a potent and specific small-molecule agonist of TRPV4, induces the phosphorylation and activation of eNOS partially through the AMPK pathway. Moreover, GSK1016790A inhibited TNF-α-induced monocyte adhesion to human endothelial cells. Mice given GSK1016790A showed increased phosphorylation of eNOS and AMPK in the aorta and decreased leukocyte adhesion to TNF-α-inflamed endothelium. Importantly, oral administration of GSK1016790A reduced atherosclerotic plaque formation in ApoE deficient mice fed a Western-type diet. Together, the present study suggests that pharmacological activation of TRPV4 may serve as a potential therapeutic approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Bin Liu
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Meimei Yin
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Marina Koroleva
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michael Mastrangelo
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Sara Ture
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Craig N Morrell
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - David X Zhang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, and The Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
56
|
Wang D, Gao Q, Wang T, Zhao G, Qian F, Huang J, Wang H, Zhang X, Wang Y. Green tea infusion protects against alcoholic liver injury by attenuating inflammation and regulating the PI3K/Akt/eNOS pathway in C57BL/6 mice. Food Funct 2018; 8:3165-3177. [PMID: 28782772 DOI: 10.1039/c7fo00791d] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Alcohol intake is a major risk factor for the pathogenesis of alcoholic liver diseases. Accumulating evidence suggests that green tea protects against alcoholic liver injury; however, the underlying mechanisms remain unclear. The present study investigated the role of endothelial nitric oxide synthase (eNOS) in the protective effects of green tea against alcohol-induced liver injury and inflammation. Ethanol was intragastrically administered to male C57BL/6 mice once a day, and the mice were allowed free access to green tea infusion or water for two weeks. We assessed the plasma levels of alanine aminotransferase and aspartate aminotransferase, hepatic contents of thiobarbituric acid reactive substances, malondialdehyde and triglyceride and hepatic mRNA expression of pro-inflammatory cytokines (interleukin-1β, tumor necrosis factor-α, and interleukin-6). Our results showed that compared with water alone, green tea infusion markedly reduced liver damage, hepatic oxidative stress, hepatic lipid accumulation and inflammatory response. Green tea infusion also significantly reduced hepatic nuclear factor-κB expression and its downstream inflammatory mediators (inducible nitric oxide synthase and cyclooxygenase-2) mRNA levels in ethanol-treated mice. Additionally, green tea infusion significantly activated hepatic phosphorylated phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (Akt), which are associated with the upregulation of phosphorylated eNOS expression and the increase of plasma nitric oxide levels in ethanol-treated mice. Furthermore, the protective effects of green tea infusion were considerably inhibited by the eNOS inhibitor NG-nitro-l-arginine methyl ester in ethanol-treated mice. In conclusion, our study demonstrated that the protective effects of green tea infusion on alcohol-induced liver injury and inflammation involve the modulation of the PI3K/AKT/eNOS pathway.
Collapse
Affiliation(s)
- Dongxu Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Caicedo D, Díaz O, Devesa P, Devesa J. Growth Hormone (GH) and Cardiovascular System. Int J Mol Sci 2018; 19:ijms19010290. [PMID: 29346331 PMCID: PMC5796235 DOI: 10.3390/ijms19010290] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 01/02/2023] Open
Abstract
This review describes the positive effects of growth hormone (GH) on the cardiovascular system. We analyze why the vascular endothelium is a real internal secretion gland, whose inflammation is the first step for developing atherosclerosis, as well as the mechanisms by which GH acts on vessels improving oxidative stress imbalance and endothelial dysfunction. We also report how GH acts on coronary arterial disease and heart failure, and on peripheral arterial disease, inducing a neovascularization process that finally increases flow in ischemic tissues. We include some preliminary data from a trial in which GH or placebo is given to elderly people suffering from critical limb ischemia, showing some of the benefits of the hormone on plasma markers of inflammation, and the safety of GH administration during short periods of time, even in diabetic patients. We also analyze how Klotho is strongly related to GH, inducing, after being released from the damaged vascular endothelium, the pituitary secretion of GH, most likely to repair the injury in the ischemic tissues. We also show how GH can help during wound healing by increasing the blood flow and some neurotrophic and growth factors. In summary, we postulate that short-term GH administration could be useful to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Diego Caicedo
- Department of Angiology and Vascular Surgery, Complejo Hospitalario Universitario de Pontevedra, 36701 Pontevedra, Spain.
| | - Oscar Díaz
- Department of Cardiology, Complejo Hospitalario Universitario de Pontevedra, 36701 Pontevedra, Spain.
| | - Pablo Devesa
- Research and Development, The Medical Center Foltra, 15886 Teo, Spain.
| | - Jesús Devesa
- Scientific Direction, The Medical Center Foltra, 15886 Teo, Spain.
| |
Collapse
|
58
|
Friedrich CC, Lin Y, Krannich A, Wu Y, Vacanti JP, Neville CM. Enhancing engineered vascular networks in vitro and in vivo: The effects of IGF1 on vascular development and durability. Cell Prolif 2017; 51. [PMID: 29110360 DOI: 10.1111/cpr.12387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/21/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Creation of functional, durable vasculature remains an important goal within the field of regenerative medicine. Engineered biological vasculature has the potential to restore or improve human tissue function. We hypothesized that the pleotropic effects of insulin-like growth factor 1 (IGF1) would enhance the engineering of capillary-like vasculature. MATERIALS AND METHODS The impact of IGF1 upon vasculogenesis was examined in in vitro cultures for a period of up to 40 days and as subcutaneous implants within immunodeficient mice. Co-cultures of human umbilical vein endothelial cells and human bone marrow-derived mesenchymal stem cells in collagen-fibronectin hydrogels were supplemented with either recombinant IGF1 protein or genetically engineered cells to provide sustained IGF1. Morphometric analysis was performed on the vascular networks that formed in four concentrations of IGF1. RESULTS IGF1 supplementation significantly enhanced de novo vasculogenesis both in vitro and in vivo. Effects were long-term as they lasted the duration of the study period, and included network density, vessel length, and diameter. Bifurcation density was not affected. However, the highest concentrations of IGF1 tested were either ineffective or even deleterious. Sustained IGF1 delivery was required in vivo as the inclusion of recombinant IGF1 protein had minimal impact. CONCLUSION IGF1 supplementation can be used to produce neovasculature with significantly enhanced network density and durability. Its use is a promising methodology for engineering de novo vasculature to support regeneration of functional tissue.
Collapse
Affiliation(s)
- Claudia C Friedrich
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.,Department of Anesthesiology and Intensive Care Medicine, Campus Virchow Klinikum and Campus Charité Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yunfeng Lin
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Orthopaedics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Alexander Krannich
- Department of Biostatistics, Clinical Research Unit, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yinan Wu
- Department of Biostatistics, Clinical Research Unit, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Joseph P Vacanti
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Craig M Neville
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.,Department of Orthopaedics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
59
|
Atochin D, Litvak M, Huang S, Kim YR, Huang P. Role of eNOS in water exchange index maintenance-MRI studies. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1742-6596/886/1/012002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
60
|
Costas-Insua C, Merino-Gracia J, Aicart-Ramos C, Rodríguez-Crespo I. Subcellular Targeting of Nitric Oxide Synthases Mediated by Their N-Terminal Motifs. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 111:165-195. [PMID: 29459031 DOI: 10.1016/bs.apcsb.2017.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
From a catalytic point of view, the three mammalian nitric oxide synthases (NOSs) function in an almost identical way. The N-terminal oxygenase domain catalyzes the conversion of l-arginine to l-citrulline plus ·NO in two sequential oxidation steps. Once l-arginine binds to the active site positioned above the heme moiety, two consecutive monooxygenation reactions take place. In the first step, l-arginine is hydroxylated to make Nω-hydroxy-l-arginine in a process that requires 1 molecule of NADPH and 1 molecule of O2 per mol of l-arginine reacted. In the second step, Nω-hydroxy-l-arginine, never leaving the active site, is oxidized to ·NO plus l-citrulline and 1 molecule of O2 and 0.5 molecules of NADPH are consumed. Since nitric oxide is an important signaling molecule that participates in a number of biological processes, including neurotransmission, vasodilation, and immune response, synthesis and release of ·NO in vivo must be exquisitely regulated both in time and in space. Hence, NOSs have evolved introducing in their amino acid sequences subcellular targeting motifs, most of them located at their N-termini. Deletion studies performed on recombinant, purified NOSs have revealed that part of the N-terminus of all three NOS can be eliminated with the resulting mutant enzymes still being catalytically active. Likewise, NOS isoforms lacking part of their N-terminus when transfected in cells render mislocalized, active proteins. In this review we will comment on the current knowledge of these subcellular targeting signals present in nNOS, iNOS, and eNOS.
Collapse
|
61
|
Suksawat M, Techasen A, Namwat N, Yongvanit P, Khuntikeo N, Titapun A, Koonmee S, Loilome W. Upregulation of endothelial nitric oxide synthase (eNOS) and its upstream regulators in Opisthorchis viverrini associated cholangiocarcinoma and its clinical significance. Parasitol Int 2017; 66:486-493. [DOI: 10.1016/j.parint.2016.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 04/04/2016] [Accepted: 04/29/2016] [Indexed: 12/16/2022]
|
62
|
Kurusamy S, López-Maderuelo D, Little R, Cadagan D, Savage AM, Ihugba JC, Baggott RR, Rowther FB, Martínez-Martínez S, Arco PGD, Murcott C, Wang W, Francisco Nistal J, Oceandy D, Neyses L, Wilkinson RN, Cartwright EJ, Redondo JM, Armesilla AL. Selective inhibition of plasma membrane calcium ATPase 4 improves angiogenesis and vascular reperfusion. J Mol Cell Cardiol 2017; 109:38-47. [PMID: 28684310 DOI: 10.1016/j.yjmcc.2017.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 02/04/2023]
Abstract
AIMS Ischaemic cardiovascular disease is a major cause of morbidity and mortality worldwide. Despite promising results from pre-clinical animal models, VEGF-based strategies for therapeutic angiogenesis have yet to achieve successful reperfusion of ischaemic tissues in patients. Failure to restore efficient VEGF activity in the ischaemic organ remains a major problem in current pro-angiogenic therapeutic approaches. Plasma membrane calcium ATPase 4 (PMCA4) negatively regulates VEGF-activated angiogenesis via inhibition of the calcineurin/NFAT signalling pathway. PMCA4 activity is inhibited by the small molecule aurintricarboxylic acid (ATA). We hypothesize that inhibition of PMCA4 with ATA might enhance VEGF-induced angiogenesis. METHODS AND RESULTS We show that inhibition of PMCA4 with ATA in endothelial cells triggers a marked increase in VEGF-activated calcineurin/NFAT signalling that translates into a strong increase in endothelial cell motility and blood vessel formation. ATA enhances VEGF-induced calcineurin signalling by disrupting the interaction between PMCA4 and calcineurin at the endothelial-cell membrane. ATA concentrations at the nanomolar range, that efficiently inhibit PMCA4, had no deleterious effect on endothelial-cell viability or zebrafish embryonic development. However, high ATA concentrations at the micromolar level impaired endothelial cell viability and tubular morphogenesis, and were associated with toxicity in zebrafish embryos. In mice undergoing experimentally-induced hindlimb ischaemia, ATA treatment significantly increased the reperfusion of post-ischaemic limbs. CONCLUSIONS Our study provides evidence for the therapeutic potential of targeting PMCA4 to improve VEGF-based pro-angiogenic interventions. This goal will require the development of refined, highly selective versions of ATA, or the identification of novel PMCA4 inhibitors.
Collapse
Affiliation(s)
- Sathishkumar Kurusamy
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Dolores López-Maderuelo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain
| | - Robert Little
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - David Cadagan
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Aaron M Savage
- Department of Infection, Immunity & Cardiovascular Disease & Bateson Centre, University of Sheffield, UK
| | - Jude C Ihugba
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Rhiannon R Baggott
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Farjana B Rowther
- Brain Tumor UK Neuro-oncology Research Centre, University of Wolverhampton, Wolverhampton, UK
| | - Sara Martínez-Martínez
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain
| | - Pablo Gómez-Del Arco
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain; Department of Molecular Biology, Universidad Autonoma de Madrid (C.B.M.S.O.), Madrid, Spain
| | - Clare Murcott
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Weiguang Wang
- Oncology Laboratory, Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - J Francisco Nistal
- Cardiovascular Surgery, Hospital Universitario Marqués de Valdecilla, IDIVAL, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ludwig Neyses
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK; University of Luxembourg, Luxembourg
| | - Robert N Wilkinson
- Department of Infection, Immunity & Cardiovascular Disease & Bateson Centre, University of Sheffield, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBERCV, Spain.
| | - Angel Luis Armesilla
- Cardiovascular Molecular Pharmacology Laboratory, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK; CIBERCV, Spain.
| |
Collapse
|
63
|
Wang Y, Wang S, Lei M, Boyett M, Tsui H, Liu W, Wang X. The p21-activated kinase 1 (Pak1) signalling pathway in cardiac disease: from mechanistic study to therapeutic exploration. Br J Pharmacol 2017; 175:1362-1374. [PMID: 28574147 DOI: 10.1111/bph.13872] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 01/01/2023] Open
Abstract
p21-activated kinase 1 (Pak1) is a member of the highly conserved family of serine/threonine protein kinases regulated by Ras-related small G-proteins, Cdc42/Rac1. It has been previously demonstrated to be involved in cardiac protection. Based on recent studies, this review provides an overview of the role of Pak1 in cardiac diseases including disrupted Ca2+ homoeostasis-related cardiac arrhythmias, adrenergic stress- and pressure overload-induced hypertrophy, and ischaemia/reperfusion injury. These findings demonstrate the important role of Pak1 mediated through the phosphorylation and transcriptional modification of hypertrophy and/or arrhythmia-related genes. This review also discusses the anti-arrhythmic and anti-hypertrophic, protective function of Pak1 and the beneficial effects of fingolimod (an FDA-approved sphingolipid drug), a Pak1 activator, and its ability to prevent arrhythmias and cardiac hypertrophy. These findings also highlight the therapeutic potential of Pak1 signalling in the treatment and prevention of cardiac diseases. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Yanwen Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Shunyao Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Ming Lei
- Department of Pharmacology, The University of Oxford, Oxford, UK
| | - Mark Boyett
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hoyee Tsui
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
64
|
Xu Y, Luan Y, Liu S, Sun J, Wang K, Cai J, Jiang W, Yang P, Wei F, Qu X. Kif4 regulates the expression of VEGFR1 through the PI3K/Akt signaling pathway in RAW264.7 monocytes/macrophages. Int J Mol Med 2017; 39:1285-1290. [PMID: 28350061 DOI: 10.3892/ijmm.2017.2936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 03/20/2017] [Indexed: 11/06/2022] Open
Abstract
Kinesin superfamily protein 4 (Kif4), a microtubule-based motor protein, has been shown to participate in a number of critical cellular processes, such as cell division, the intracellular transport of membranous vesicles and signal transduction. However, whether KIF4 regulates vascular endothelial growth factor (VEGF) receptor 1 (VEGFR1) expression remains unknown. Thus, in this study, in order to examine the effects of Kif4 on the expression of VEGFR1 in RAW264.7 monocytes/macrophages, Kif4 was silenced using siRNA. RT-qPCR, western blot analysis and ELISA were used to assess the expression of Kif4 and VEGFR1 up- and downstream signaling molecules, including VEGF-A, VEGFR1, soluble form of VEGFR1 (sVEGFR1), phosphorylated (p-)Akt and Akt. The silencing Kif4 inhibited the mRNA expression of VEGF (P<0.01) and p-Akt (P<0.05); however, the level of VEGF-A was increased (P<0.05) compared with the negative control siRNA-transfected group. The silencing of Kif4 decreased the VEGFR1 mRNA (P<0.05), VEGFR1 protein and sVEGFR1 levels in the cell supernatant (P<0.01). Following the application of insulin-like growth factor-1 (100 ng/ml), the specific agonist of PI3K/Akt in the Kif4 siRNA-transfected group, the VEGFR1 mRNA levels (P<0.001), the VEGFR1 protein levels and the sVEGFR1 (P<0.01) levels significantly increased; however, the levels of VEGF in the cell supernatant were decreased (P<0.05). Taken together, these findings suggest that Kif4 regulates the expression of VEGFR1 in RAW264.7 cells and that the PI3K/Akt pathway is involved in this process.
Collapse
Affiliation(s)
- Yan Xu
- Department of Stomatology and Institute of Stomatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yijun Luan
- Department of Stomatology and Institute of Stomatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shaohua Liu
- Department of Stomatology and Institute of Stomatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jintang Sun
- Institute of Basic Medical Sciences and Key Laboratory of Cardiovascular Proteomics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ketao Wang
- Department of Stomatology and Institute of Stomatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jun Cai
- Jinan Stomalogic Hospital, Jinan, Shandong 250012, P.R. China
| | - Wen Jiang
- Department of Biomedical Science, The University of Sheffield, Sheffield S10 2TN, UK
| | - Pishan Yang
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Fengcai Wei
- Department of Stomatology and Institute of Stomatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xun Qu
- Department of Stomatology and Institute of Stomatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
65
|
Interplay of myosin phosphatase and protein phosphatase-2A in the regulation of endothelial nitric-oxide synthase phosphorylation and nitric oxide production. Sci Rep 2017; 7:44698. [PMID: 28300193 PMCID: PMC5353758 DOI: 10.1038/srep44698] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/13/2017] [Indexed: 01/07/2023] Open
Abstract
The inhibitory phosphorylation of endothelial nitric oxide (NO) synthase (eNOS) at Thr497 (eNOSpThr497) by protein kinase C or RhoA-activated kinase is a major regulatory determinant of eNOS activity. The signalling mechanisms involved in the dephosphorylation of eNOSpThr497 have not yet been clarified. This study identifies myosin phosphatase (MP) holoenzyme consisting of protein phosphatase-1 catalytic subunit (PP1c) and MP target subunit-1 (MYPT1) as an eNOSpThr497 phosphatase. In support of this finding are: (i) eNOS and MYPT1 interacts in various endothelial cells (ECs) and in in vitro binding assays (ii) MYPT1 targets and stimulates PP1c toward eNOSpThr497 substrate (iii) phosphorylation of MYPT1 at Thr696 (MYPT1pThr696) controls the activity of MP on eNOSpThr497. Phosphatase inhibition suppresses both NO production and transendothelial resistance (TER) of ECs. In contrast, epigallocatechin-3-gallate (EGCG) signals ECs via the 67 kDa laminin-receptor (67LR) resulting in protein kinase A dependent activation of protein phosphatase-2A (PP2A). PP2A dephosphorylates MYPT1pThr696 and thereby stimulates MP activity inducing dephosphorylation of eNOSpThr497 and the 20 kDa myosin II light chains. Thus an interplay of MP and PP2A is involved in the physiological regulation of EC functions implying that an EGCG dependent activation of these phosphatases leads to enhanced NO production and EC barrier improvement.
Collapse
|
66
|
Freitas ACN, Silva GC, Pacheco DF, Pimenta AMC, Lemos VS, Duarte IDG, de Lima ME. The synthetic peptide PnPP-19 induces peripheral antinociception via activation of NO/cGMP/K ATP pathway: Role of eNOS and nNOS. Nitric Oxide 2017; 64:31-38. [PMID: 28087360 DOI: 10.1016/j.niox.2017.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND and purpose: The peptide PnPP-19, derived from the spider toxin PnTx2-6 (renamed as δ-CNTX-Pn1c), potentiates erectile function by activating the nitrergic system. Since NO has been studied as an antinociceptive molecule and PnPP-19 is known to induce peripheral antinociception, we intended to evaluate whether PnPP-19 could induce peripheral antinociception through activation of this pathway. EXPERIMENTAL APPROACH Nociceptive thresholds were measured by paw pressure test. PGE2 (2 μg/paw) was administered intraplantarly together with PnPP-19 and inhibitors/blockers of NOS, guanylyl cyclase and KATP channels. The nitrite concentration was accessed by Griess test. The expression and phosphorylation of eNOS and nNOS were determined by western blot. KEY RESULTS PnPP-19 (5, 10 and 20 μg/paw) induced peripheral antinociception in rats. Administration of NOS inhibitor (L-NOarg), selective nNOS inhibitor (L-NPA), guanylyl cyclase inhibitor (ODQ) and the blocker of KATP (glibenclamide) partially inhibited the antinociceptive effect of PnPP-19 (10 μg/paw). Tissue nitrite concentration increased after PnPP-19 (10 μg/paw) administration. Expression of eNOS and nNOS remained the same in all tested groups, however the phosphorylation of nNOS Ser852 (inactivation site) increased and phosphorylation of eNOS Ser1177 (activation site) decreased after PGE2 injection. Administration of PnPP-19 reverted this PGE2-induced effect. CONCLUSIONS AND IMPLICATIONS The peripheral antinociceptive effect induced by PnPP-19 is resulting from activation of NO-cGMP-KATP pathway. Activation of eNOS and nNOS might be required for such effect. Our results suggest PnPP-19 as a new drug candidate to treat pain and reinforce the importance of nNOS and eNOS activation, as well as endogenous NO release, for induction of peripheral antinociception.
Collapse
Affiliation(s)
- A C N Freitas
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil
| | - G C Silva
- Departamento Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil
| | - D F Pacheco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil; Departamento Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil
| | - A M C Pimenta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil
| | - V S Lemos
- Departamento Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil
| | - I D G Duarte
- Departamento Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil
| | - M E de Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, M.G., Brazil.
| |
Collapse
|
67
|
Miyamoto L, Yamane M, Tomida Y, Kono M, Yamaoka T, Kawasaki A, Hatano A, Tsuda K, Xu W, Ikeda Y, Tamaki T, Tsuchiya K. Nitrite Activates 5′AMP-Activated Protein Kinase-Endothelial Nitric Oxide Synthase Pathway in Human Glomerular Endothelial Cells. Biol Pharm Bull 2017; 40:1866-1872. [DOI: 10.1248/bpb.b17-00316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Licht Miyamoto
- Laboratory of Pharmacology and Physiological Sciences, Frontier Laboratory for Pharmaceutical Sciences, Institute of Biomedical Sciences, University of Tokushima Graduate School
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Megumi Yamane
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Yosuke Tomida
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Mai Kono
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Tomomi Yamaoka
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Aya Kawasaki
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Aya Hatano
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Katsunori Tsuda
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Wenting Xu
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Toshiaki Tamaki
- Department of Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, University of Tokushima Graduate School
| |
Collapse
|
68
|
Abstract
Endothelial nitric oxide (NO) synthase (eNOS) has an indispensable role in the erectile response. In the penis, eNOS activity and endothelial NO bioavailability are regulated by multiple post-translatlonal molecular mechanisms, such as eNOS phosphorylation, eNOS interaction with regulatory proteins and contractile pathways, and actions of reactive oxygen species (ROS). These mechanisms regulate eNOS-mediated responses under physiologic circumstances and provide various mechanisms whereby endothelial NO availability may be altered in states of vasculogenlc erectile dysfunction (ED), in view of the recent advances in the field of eNOS function in the penis and its role in penile erection, the emphasis in this review is placed on the mechanisms regulating eNOS activity and its interaction with the RhoA/Rho-kinase pathway in the physiology of penile erection and the pathophysiology of ED.
Collapse
Affiliation(s)
- Biljana Musicki
- Johns Hopkins Hospital, Department of Urology, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | | |
Collapse
|
69
|
Szabo C. Hydrogen sulfide, an enhancer of vascular nitric oxide signaling: mechanisms and implications. Am J Physiol Cell Physiol 2016; 312:C3-C15. [PMID: 27784679 DOI: 10.1152/ajpcell.00282.2016] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/17/2016] [Indexed: 12/15/2022]
Abstract
Nitric oxide (NO) vascular signaling has long been considered an independent, self-sufficient pathway. However, recent data indicate that the novel gaseous mediator, hydrogen sulfide (H2S), serves as an essential enhancer of vascular NO signaling. The current article overviews the multiple levels at which this enhancement takes place. The first level of interaction relates to the formation of biologically active hybrid S/N species and the H2S-induced stimulation of NO release from its various stable "pools" (e.g., nitrite). The next interactions occur on the level of endothelial calcium mobilization and PI3K/Akt signaling, increasing the specific activity of endothelial NO synthase (eNOS). The next level of interaction occurs on eNOS itself; H2S directly interacts with the enzyme: sulfhydration of critical cysteines stabilizes it in its physiological, dimeric state, thereby optimizing eNOS-derived NO production and minimizing superoxide formation. Yet another level of interaction, further downstream, occurs at the level of soluble guanylate cyclase (sGC): H2S stabilizes sGC in its NO-responsive, physiological, reduced form. Further downstream, H2S inhibits the vascular cGMP phosphodiesterase (PDE5), thereby prolonging the biological half-life of cGMP. Finally, H2S-derived polysulfides directly activate cGMP-dependent protein kinase (PKG). Taken together, H2S emerges an essential endogenous enhancer of vascular NO signaling, contributing to vasorelaxation and angiogenesis. The functional importance of the H2S/NO cooperative interactions is highlighted by the fact that H2S loses many of its beneficial cardiovascular effects when eNOS is inactive.
Collapse
Affiliation(s)
- Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
70
|
van Beijnum JR, Pieters W, Nowak-Sliwinska P, Griffioen AW. Insulin-like growth factor axis targeting in cancer and tumour angiogenesis - the missing link. Biol Rev Camb Philos Soc 2016; 92:1755-1768. [PMID: 27779364 DOI: 10.1111/brv.12306] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/15/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
Numerous molecular players in the process of tumour angiogenesis have been shown to offer potential for therapeutic targeting. Initially denoted to be involved in malignant transformation and tumour progression, the insulin-like growth factor (IGF) signalling axis has been subject to therapeutic interference, albeit with limited clinical success. More recently, IGFs and their receptors have received attention for their contribution to tumour angiogenesis, which offers novel therapeutic opportunities. Here we review the contribution of this signalling axis to tumour angiogenesis, the mechanisms of resistance to therapy and the interplay with other pro-angiogenic pathways, to offer insight in the renewed interest in the application of IGF axis targeting agents in anti-cancer combination therapies.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Wietske Pieters
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva (UNIGE), Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Arjan W Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| |
Collapse
|
71
|
Kalinowski L, Janaszak-Jasiecka A, Siekierzycka A, Bartoszewska S, Woźniak M, Lejnowski D, Collawn JF, Bartoszewski R. Posttranscriptional and transcriptional regulation of endothelial nitric-oxide synthase during hypoxia: the role of microRNAs. Cell Mol Biol Lett 2016; 21:16. [PMID: 28536619 PMCID: PMC5415778 DOI: 10.1186/s11658-016-0017-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
Abstract
Understanding the cellular pathways that regulate endothelial nitric oxide (eNOS, NOS3) expression and consequently nitric oxide (NO) bioavailability during hypoxia is a necessary aspect in the development of novel treatments for cardiovascular disorders. eNOS expression and eNOS-dependent NO cellular signaling during hypoxia promote an equilibrium of transcriptional and posttranscriptional molecular mechanisms that belong to both proapoptotic and survival pathways. Furthermore, NO bioavailability results not only from eNOS levels, but also relies on the presence of eNOS substrate and cofactors, the phosphorylation status of eNOS, and the presence of reactive oxygen species (ROS) that can inactivate eNOS. Since both NOS3 levels and these signaling pathways can also be a subject of posttranscriptional modulation by microRNAs (miRNAs), this class of short noncoding RNAs contribute another level of regulation for NO bioavailability. As miRNA antagomirs or specific target protectors could be used in therapeutic approaches to regulate NO levels, either by changing NOS3 mRNA stability or through factors governing eNOS activity, it is critical to understand their role in governing eNOS activity during hypoxa. In contrast to a large number of miRNAs reported to the change eNOS expression during hypoxia, only a few miRNAs modulate eNOS activity. Furthermore, impaired miRNA biogenesis leads to NOS3 mRNA stabilization under hypoxia. Here we discuss the recent studies that define miRNAs’ role in maintaining endothelial NO bioavailability emphasizing those miRNAs that directly modulate NOS3 expression or eNOS activity.
Collapse
Affiliation(s)
- Leszek Kalinowski
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Anna Janaszak-Jasiecka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Woźniak
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Dawid Lejnowski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - James F Collawn
- Department of Cell Biology, Developmental, and Integrative, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| |
Collapse
|
72
|
Zatorski H, Marynowski M, Fichna J. Is insulin-like growth factor 1 (IGF-1) system an attractive target inflammatory bowel diseases? Benefits and limitation of potential therapy. Pharmacol Rep 2016; 68:809-15. [PMID: 27117379 DOI: 10.1016/j.pharep.2016.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/12/2016] [Accepted: 03/04/2016] [Indexed: 01/26/2023]
Abstract
Inflammatory bowel diseases (IBD) are chronic gastrointestinal disorders with unknown etiology, whose incidence dramatically increased over the past 50 years. Currently available strategies for IBD treatment, such as biological therapies, corticosteroids, and immunosuppressive agents are effective, but their side effects and economic costs cannot be ignored. Better understanding of IBD etiology and new therapeutics are thus needed. The aim of this paper is to briefly discuss IGF-1 dependent functions, with particular focus on IGF-1 use in IBD therapy. Data collection was based on records found in medical literature. Data analysis included records published between 1984 and 2014. The IGF-1 system is involved in major physiological functions, such as cell proliferation and metabolism, and growth promotion. Most importantly IGF-1 has anti-inflammatory properties and its use in IBD treatment can be recommended. However, potential IGF-1 therapy has some limitations, which include aggravation of fibrosis in Crohn's patients and facilitated transformation to malignancy. Taken into consideration their possible side effects, IGF-1 analogs and recombinants are nonetheless a promising target for IBD therapy for a specific group of patients. Further studies, at the clinical level are thus recommended.
Collapse
Affiliation(s)
- Hubert Zatorski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Łódź, Poland
| | - Mateusz Marynowski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Łódź, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Łódź, Poland.
| |
Collapse
|
73
|
Shao Q, Fallica J, Casin KM, Murphy E, Steenbergen C, Kohr MJ. Characterization of the sex-dependent myocardial S-nitrosothiol proteome. Am J Physiol Heart Circ Physiol 2016; 310:H505-15. [PMID: 26702143 PMCID: PMC4796614 DOI: 10.1152/ajpheart.00681.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/21/2015] [Indexed: 01/23/2023]
Abstract
Premenopausal women exhibit endogenous cardioprotective signaling mechanisms that are thought to result from the beneficial effects of estrogen, which we have shown to increase protein S-nitrosylation in the heart. S-nitrosylation is a labile protein modification that increases with a number of different forms of cardioprotection, including ischemic preconditioning. Herein, we sought to identify a potential role for protein S-nitrosylation in sex-dependent cardioprotection. We utilized a Langendorff-perfused mouse heart model of ischemia-reperfusion injury with male and female hearts, and S-nitrosylation-resin-assisted capture with liquid chromatography tandem mass spectrometry to identify S-nitrosylated proteins and modification sites. Consistent with previous studies, female hearts exhibited resilience to injury with a significant increase in functional recovery compared with male hearts. In a separate set of hearts, we identified a total of 177 S-nitrosylated proteins in female hearts at baseline compared with 109 S-nitrosylated proteins in male hearts. Unique S-nitrosylated proteins in the female group included the F1FO-ATPase and cyclophilin D. We also utilized label-free peptide analysis to quantify levels of common S-nitrosylated identifications and noted that the S-nitrosylation of sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2a was nearly 70% lower in male hearts compared with female, with no difference in expression. Furthermore, we found a significant increase in endothelial nitric oxide synthase expression, phosphorylation, and total nitric oxide production in female hearts compared with males, likely accounting for the enhanced S-nitrosylation protein levels in female hearts. In conclusion, we identified a number of novel S-nitrosylated proteins in female hearts that are likely to contribute to sex-dependent cardioprotection.
Collapse
Affiliation(s)
- Qin Shao
- Department of Cardiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Jonathan Fallica
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| | - Kevin M Casin
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| | - Elizabeth Murphy
- Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Charles Steenbergen
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Mark J Kohr
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland; Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| |
Collapse
|
74
|
Ali MI, Chen X, Didion SP. Heterozygous eNOS deficiency is associated with oxidative stress and endothelial dysfunction in diet-induced obesity. Physiol Rep 2015; 3:e12630. [PMID: 26660551 PMCID: PMC4760452 DOI: 10.14814/phy2.12630] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 01/08/2023] Open
Abstract
Heterozygous endothelial nitric oxide synthase (eNOS) deficiency is associated with normal endothelium-dependent responses, however, little is known regarding the mechanisms that maintain or impair endothelial function with heterozygous eNOS deficiency. The goals of this study were to (1) determine mechanism(s) which serve to maintain normal endothelial function in the absence of a single eNOS gene; and (2) to determine whether heterozygous eNOS deficiency predisposes blood vessels to endothelial dysfunction in response to a high-fat diet (HFD). Responses of carotid arteries were examined in wild-type (eNOS(+/+)) and heterozygous eNOS-deficient (eNOS(+/-)) treated with either vehicle (saline), N(G)-nitro-L-arginine (L-NNA, 100 μmol/L), an inhibitor of nitric oxide synthase, or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 1 μmol/L), an inhibitor of soluble guanylyl cyclase (sGC), and in eNOS(+/+) and eNOS(+/-) mice fed a control (10%) or a 45% HFD (kcal from fat). Responses to acetylcholine (ACh) were similar in vehicle-treated arteries from eNOS(+/+) and eNOS(+/-) mice, and were equally inhibited by L-NNA and ODQ. Phosphorylation of eNOS Ser1176, a site associated with increased eNOS activity, was significantly greater in eNOS(+/-) mice most likely as a compensatory response for the loss of a single eNOS gene. In contrast, responses to ACh were markedly impaired in carotid arteries from eNOS(+/-), but not eNOS(+/+), mice fed a HFD. Vascular superoxide levels as well as plasma levels of the pro-inflammatory cytokine interleukin-6 (IL-6) were selectively increased in HFD-fed eNOS(+/-) mice. In reconstitution experiments, IL-6 produced concentration-dependent impairment of endothelial responses as well as greater increases in NADPH-stimulated superoxide levels in arteries from eNOS(+/-) mice fed a control diet compared to eNOS(+/+) mice. Our findings of increased Ser1176-phosphorylation reveal a mechanism by which NOS- and sGC-dependent endothelial function can be maintained with heterozygous eNOS deficiency. In addition, heterozygous eNOS deficiency predisposes blood vessels to developing endothelial dysfunction in response to a HFD. The impairment produced by a HFD in eNOS(+/-) mice appears to be mediated by IL-6-induced increases in vascular superoxide. These findings serve as an important example of eNOS haploinsufficiency, one that may contribute to the development of carotid artery disease in obese humans.
Collapse
Affiliation(s)
- M Irfan Ali
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia
| | - Xunsheng Chen
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia
| | - Sean P Didion
- Department of Pharmacology and Department of Neurology, The University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
75
|
Park JH, Cho DH, Lee JY, Lee HJ, Ha Y, Ahn JH, Jo I. B56δ subunit of protein phosphatase 2A decreases phosphorylation of endothelial nitric oxide synthase at serine 116: Mechanism underlying aphidicolin-stimulated NO production. Nitric Oxide 2015; 50:46-51. [DOI: 10.1016/j.niox.2015.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/30/2015] [Accepted: 08/02/2015] [Indexed: 10/23/2022]
|
76
|
Murakami T. Kallikrein-Kinin System: An Emerging Competitor or Collaborator for VEGF in Diabetic Macular Edema? Diabetes 2015; 64:3350-2. [PMID: 26405275 DOI: 10.2337/db15-0746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Tomoaki Murakami
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
77
|
Vascular nitric oxide: Beyond eNOS. J Pharmacol Sci 2015; 129:83-94. [PMID: 26499181 DOI: 10.1016/j.jphs.2015.09.002] [Citation(s) in RCA: 494] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/11/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
As the first discovered gaseous signaling molecule, nitric oxide (NO) affects a number of cellular processes, including those involving vascular cells. This brief review summarizes the contribution of NO to the regulation of vascular tone and its sources in the blood vessel wall. NO regulates the degree of contraction of vascular smooth muscle cells mainly by stimulating soluble guanylyl cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP), although cGMP-independent signaling [S-nitrosylation of target proteins, activation of sarco/endoplasmic reticulum calcium ATPase (SERCA) or production of cyclic inosine monophosphate (cIMP)] also can be involved. In the blood vessel wall, NO is produced mainly from l-arginine by the enzyme endothelial nitric oxide synthase (eNOS) but it can also be released non-enzymatically from S-nitrosothiols or from nitrate/nitrite. Dysfunction in the production and/or the bioavailability of NO characterizes endothelial dysfunction, which is associated with cardiovascular diseases such as hypertension and atherosclerosis.
Collapse
|
78
|
Wang R, Peng L, Zhao J, Zhang L, Guo C, Zheng W, Chen H. Gardenamide A Protects RGC-5 Cells from H₂O₂-Induced Oxidative Stress Insults by Activating PI3K/Akt/eNOS Signaling Pathway. Int J Mol Sci 2015; 16:22350-67. [PMID: 26389892 PMCID: PMC4613312 DOI: 10.3390/ijms160922350] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/24/2015] [Accepted: 08/31/2015] [Indexed: 01/04/2023] Open
Abstract
Gardenamide A (GA) protects the rat retinal ganglion (RGC-5) cells against cell apoptosis induced by H₂O₂. The protective effect of GA was completely abrogated by the specific phosphoinositide 3-kinase (PI3K) inhibitor LY294002, and the specific protein kinase B (Akt) inhibitor Akt VIII respectively, indicating that the protective mechanism of GA is mediated by the PI3K/Akt signaling pathway. The specific extracellular signal-regulated kinase (ERK1/2) inhibitor PD98059 could not block the neuroprotection of GA. GA attenuated the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) induced by H₂O₂. Western blotting showed that GA promoted the phosphorylation of ERK1/2, Akt and endothelial nitric oxide synthase (eNOS), respectively, and effectively reversed the H₂O₂-inhibited phosphorylation of these three proteins. LY294002 completely inhibited the GA-activated phosphorylation of Akt, while only partially inhibiting eNOS. This evidence implies that eNOS may be activated directly by GA. PD98059 attenuated only partially the GA-induced phosphorylation of ERK1/2 with/without the presence of H₂O₂, indicating that GA may activate ERK1/2 directly. All these results put together confirm that GA protects RGC-5 cells from H₂O₂ insults via the activation of PI3K/Akt/eNOS signaling pathway. Whether the ERK1/2 signaling pathway is involved requires further investigations.
Collapse
Affiliation(s)
- Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China.
| | - Lizhi Peng
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Jiaqiang Zhao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Laitao Zhang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Cuiping Guo
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macao, Macao, China.
| | - Heru Chen
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Guangzhou 510632, China.
| |
Collapse
|
79
|
Wu Z, Yu Y, Liu C, Xiong Y, Montani JP, Yang Z, Ming XF. Role of p38 mitogen-activated protein kinase in vascular endothelial aging: interaction with Arginase-II and S6K1 signaling pathway. Aging (Albany NY) 2015; 7:70-81. [PMID: 25635535 PMCID: PMC4350325 DOI: 10.18632/aging.100722] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
p38 mitogen-activated protein kinase (p38) regulates cellular senescence and senescence-associated secretory phenotype (SASP), i.e., secretion of cytokines and/or chemokines. Previous work showed that augmented arginase-II (Arg-II) and S6K1 interact with each other to promote endothelial senescence through uncoupling of endothelial nitric oxide synthase (eNOS). Here we demonstrate eNOS-uncoupling, augmented expression/secretion of IL-6 and IL-8, elevation of p38 activation and Arg-II levels in senescent endothelial cells. Silencing Arg-II or p38α in senescent cells recouples eNOS and inhibits IL-6 and IL-8 secretion. Overexpression of Arg-II in young endothelial cells causes eNOS-uncoupling and enhances IL-6 and IL-8 expression/secretion, which is prevented by p38 inhibition or by antioxidant. Moreover, p38 activation and expression of IL-6 and KC (the murine IL-8 homologue) are increased in the heart and/or aortas of wild type (WT) old mice, which is abolished in mice with Arg-II gene deficiency (Arg-II−/−). In addition, inhibition of p38 in the old WT mice recouples eNOS function and reduces IL-6 and KC expression in the aortas and heart. Silencing Arg-II or p38α or S6K1 inhibits each other in senescence endothelial cells. Thus, Arg-II, p38, and S6K1 form a positive circuit which regulates endothelial senescence and cardiovascular aging.
Collapse
Affiliation(s)
- Zongsong Wu
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Yi Yu
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Chang Liu
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Yuyan Xiong
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Jean-Pierre Montani
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Zhihong Yang
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Xiu-Fen Ming
- Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
80
|
Aicart-Ramos C, Rodríguez-Crespo I. Binding of PDZ domains to the carboxy terminus of inducible nitric oxide synthase boosts electron transfer and NO synthesis. FEBS Lett 2015; 589:2207-12. [PMID: 26183564 DOI: 10.1016/j.febslet.2015.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/29/2015] [Accepted: 07/02/2015] [Indexed: 10/23/2022]
Abstract
iNOS lacks any phosphorylatable residue at its C-terminus despite displaying a 25-residue extension known to block electron transfer and activity. We report that C-terminal deletions of iNOS increased the cytochrome c reduction rate. Moreover, the interaction of the iNOS C-terminus with the PDZ domains of EBP50 or CAP70 resulted not only in augmented reductase activity and greater NO synthesis but also anticipated the formation of the air-stable semiquinone generated after NADPH addition. Hence, the C-terminus of iNOS regulates the activity of the enzyme, albeit, unlike nNOS and eNOS, displacement of the autoinhibitory element occurs upon binding to proteins with PDZ domains.
Collapse
Affiliation(s)
- Clara Aicart-Ramos
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Spain
| | - Ignacio Rodríguez-Crespo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Spain.
| |
Collapse
|
81
|
Kameny RJ, He Y, Morris C, Sun C, Johengen M, Gong W, Raff GW, Datar SA, Oishi PE, Fineman JR. Right ventricular nitric oxide signaling in an ovine model of congenital heart disease: a preserved fetal phenotype. Am J Physiol Heart Circ Physiol 2015; 309:H157-65. [PMID: 25934095 PMCID: PMC4491525 DOI: 10.1152/ajpheart.00103.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/10/2015] [Indexed: 11/22/2022]
Abstract
We recently reported superior right ventricle (RV) performance in response to acute afterload challenge in lambs with a model of congenital heart disease with chronic left-to-right cardiac shunts. Compared with control animals, shunt lambs demonstrated increased contractility because of an enhanced Anrep effect (the slow increase in contractility following myocyte stretch). This advantageous physiological response may reflect preservation of a fetal phenotype, since the RV of shunt lambs remains exposed to increased pressure postnatally. Nitric oxide (NO) production by NO synthase (NOS) is activated by myocyte stretch and is a necessary intermediary of the Anrep response. The purpose of this study was to test the hypothesis that NO signaling is increased in the RV of fetal lambs compared with controls and shunt lambs have persistence of this fetal pattern. An 8-mm graft was placed between the pulmonary artery and aorta in fetal lambs (shunt). NOS isoform expression, activity, and association with activating cofactors were determined in fetal tissue obtained during late-gestation and in 4-wk-old juvenile shunt and control lambs. We demonstrated increased RNA and protein expression of NOS isoforms and increased total NOS activity in the RV of both shunt and fetal lambs compared with control. We also found increased NOS activation and association with cofactors in shunt and fetal RV compared with control. These data demonstrate preserved fetal NOS phenotype and NO signaling in shunt RV, which may partially explain the mechanism underlying the adaptive response to increased afterload seen in the RV of shunt lambs.
Collapse
MESH Headings
- Animals
- Aorta/surgery
- Disease Models, Animal
- Fetus/metabolism
- Heart Defects, Congenital/complications
- Heart Defects, Congenital/metabolism
- Heart Defects, Congenital/physiopathology
- Heart Ventricles/enzymology
- Heart Ventricles/metabolism
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/physiopathology
- Myocardial Contraction/physiology
- Myocytes, Cardiac
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/metabolism
- Phenotype
- Pulmonary Artery/surgery
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sheep
- Signal Transduction
Collapse
Affiliation(s)
- Rebecca Johnson Kameny
- Department of Pediatrics, University of California, San Francisco, San Francisco, California; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - Youping He
- Department of Surgery, University of California, San Francisco, San Francisco, California; and
| | - Catherine Morris
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Christine Sun
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Michael Johengen
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Wenhui Gong
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Gary W Raff
- Department of Surgery, University of California, Davis, Davis, California
| | - Sanjeev A Datar
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Peter E Oishi
- Department of Pediatrics, University of California, San Francisco, San Francisco, California; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California;
| |
Collapse
|
82
|
Reduced HRAS G12V-Driven Tumorigenesis of Cell Lines Expressing KRAS C118S. PLoS One 2015; 10:e0123918. [PMID: 25902334 PMCID: PMC4406447 DOI: 10.1371/journal.pone.0123918] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/09/2015] [Indexed: 11/19/2022] Open
Abstract
In many different human cancers, one of the HRAS, NRAS, or KRAS genes in the RAS family of small GTPases acquires an oncogenic mutation that renders the encoded protein constitutively GTP-bound and thereby active, which is well established to promote tumorigenesis. In addition to oncogenic mutations, accumulating evidence suggests that the wild-type isoforms may also be activated and contribute to oncogenic RAS-driven tumorigenesis. In this regard, redox-dependent reactions with cysteine 118 (C118) have been found to promote activation of wild-type HRAS and NRAS. We sought to determine if this residue is also important for the activation of wild-type KRAS and promotion of tumorigenesis. Thus, we mutated C118 to serine (C118S) in wild-type KRAS to block redox-dependent reactions at this site. We now report that this mutation reduced the level of GTP-bound KRAS and impaired RAS signaling stimulated by the growth factor EGF. With regards to tumorigenesis, we also report that oncogenic HRAS-transformed human cells in which endogenous KRAS was knocked down and replaced with KRASC118S exhibited reduced xenograft tumor growth, as did oncogenic HRAS-transformed KrasC118S/C118S murine cells in which the C118S mutation was knocked into the endogenous Kras gene. Taken together, these data suggest a role for redox-dependent activation of wild-type KRAS through C118 in oncogenic HRAS-driven tumorigenesis.
Collapse
|
83
|
NADPH Oxidase Activity in Cerebral Arterioles Is a Key Mediator of Cerebral Small Vessel Disease-Implications for Prevention. Healthcare (Basel) 2015; 3:233-51. [PMID: 27417759 PMCID: PMC4939544 DOI: 10.3390/healthcare3020233] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/21/2015] [Accepted: 04/08/2015] [Indexed: 02/05/2023] Open
Abstract
Cerebral small vessel disease (SVD), a common feature of brain aging, is characterized by lacunar infarcts, microbleeds, leukoaraiosis, and a leaky blood-brain barrier. Functionally, it is associated with cognitive decline, dementia, depression, gait abnormalities, and increased risk for stroke. Cerebral arterioles in this syndrome tend to hypertrophy and lose their capacity for adaptive vasodilation. Rodent studies strongly suggest that activation of Nox2-dependent NADPH oxidase activity is a crucial driver of these structural and functional derangements of cerebral arterioles, in part owing to impairment of endothelial nitric oxide synthase (eNOS) activity. This oxidative stress may also contribute to the breakdown of the blood-brain barrier seen in SVD. Hypertension, aging, metabolic syndrome, smoking, hyperglycemia, and elevated homocysteine may promote activation of NADPH oxidase in cerebral arterioles. Inhibition of NADPH oxidase with phycocyanobilin from spirulina, as well as high-dose statin therapy, may have potential for prevention and control of SVD, and high-potassium diets merit study in this regard. Measures which support effective eNOS activity in other ways-exercise training, supplemental citrulline, certain dietary flavonoids (as in cocoa and green tea), and capsaicin, may also improve the function of cerebral arterioles. Asian epidemiology suggests that increased protein intakes may decrease risk for SVD; conceivably, arginine and/or cysteine-which boosts tissue glutathione synthesis, and can be administered as N-acetylcysteine-mediate this benefit. Ameliorating the risk factors for SVD-including hypertension, metabolic syndrome, hyperglycemia, smoking, and elevated homocysteine-also may help to prevent and control this syndrome, although few clinical trials have addressed this issue to date.
Collapse
|
84
|
Molecular modeling and simulation of the human eNOS reductase domain, an enzyme involved in the release of vascular nitric oxide. J Mol Model 2014; 20:2470. [DOI: 10.1007/s00894-014-2470-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 09/14/2014] [Indexed: 01/19/2023]
|
85
|
Wilkerson BA, Argraves KM. The role of sphingosine-1-phosphate in endothelial barrier function. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1841:1403-1412. [PMID: 25009123 PMCID: PMC4169319 DOI: 10.1016/j.bbalip.2014.06.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/19/2014] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Loss of endothelial barrier function is implicated in the etiology of metastasis, atherosclerosis, sepsis and many other diseases. Studies suggest that sphingosine-1-phosphate (S1P), particularly HDL-bound S1P (HDL-S1P) is essential for endothelial barrier homeostasis and that HDL-S1P may be protective against the loss of endothelial barrier function in disease. This review summarizes evidence providing mechanistic insights into how S1P maintains endothelial barrier function, highlighting the recent findings that implicate the major S1P carrier, HDL, in the maintenance of the persistent S1P-signaling needed to maintain endothelial barrier function. We review the mechanisms proposed for HDL maintenance of persistent S1P-signaling, the evidence supporting these mechanisms and the remaining fundamental questions.
Collapse
Affiliation(s)
- Brent A Wilkerson
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., BSB650, Charleston, SC 29425, USA
| | - Kelley M Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., BSB650, Charleston, SC 29425, USA.
| |
Collapse
|
86
|
Ford H, Suri S, Nilforoushan D, Manolson M, Gong SG. Nitric oxide in human gingival crevicular fluid after orthodontic force application. Arch Oral Biol 2014; 59:1211-6. [PMID: 25108338 DOI: 10.1016/j.archoralbio.2014.07.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 05/29/2014] [Accepted: 07/17/2014] [Indexed: 01/16/2023]
Abstract
Nitric oxide (NO) is involved in bone remodelling and has been shown to play a role in regulating the rate of orthodontic tooth movement (OTM) in rat models. In humans, however, the role of NO in OTM remains less clear. In this study, NO concentration in gingival crevicular fluid (GCF) was measured in patients undergoing orthodontic treatment. Thirteen male participants (ages 11-18 years) planned for non-extraction fixed orthodontic therapy were recruited. Samples of GCF were collected from each maxillary central incisor and first and second molar immediately before (T0), 1h after (T1), and 3-4 days after (T2) application of light orthodontic forces. The maxillary second molars were not included in the appliance and served as controls. Measureable NO levels were consistently obtained from all sampled sites. Total NO levels showed significantly higher NO levels (p<0.05) at T1 at the buccal surfaces of the central incisors when compared to the first and second molars. The results indicate a possible role for NO in OTM at the pressure sites of incisors at early time points. Further studies are required to determine whether NO levels in the periodontal ligament tissues of human teeth during OTM are affected by a force gradient and the magnitude of the applied force.
Collapse
Affiliation(s)
- H Ford
- Department of Orthodontics, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - S Suri
- Department of Orthodontics, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - D Nilforoushan
- Department of Orthodontics, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - M Manolson
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - S G Gong
- Department of Orthodontics, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada; Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
87
|
Oliver JL, Kavoussi PK, Smith RP, Woodson RI, Corbett ST, Costabile RA, Palmer LA, Lysiak JJ. The Role of Regulatory Proteins and S‐nitrosylation of Endothelial Nitric Oxide Synthase in the Human Clitoris: Implications for Female Sexual Function. J Sex Med 2014; 11:1927-35. [DOI: 10.1111/jsm.12576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
88
|
Aicart-Ramos C, Sánchez-Ruiloba L, Gómez-Parrizas M, Zaragoza C, Iglesias T, Rodríguez-Crespo I. Protein kinase D activity controls endothelial nitric oxide synthesis. J Cell Sci 2014; 127:3360-72. [PMID: 24928905 DOI: 10.1242/jcs.148601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) regulates key functions of the endothelium, such as angiogenesis or vessel repair in processes involving endothelial nitric oxide synthase (eNOS) activation. One of the effector kinases that become activated in endothelial cells upon VEGF treatment is protein kinase D (PKD). Here, we show that PKD phosphorylates eNOS, leading to its activation and a concomitant increase in NO synthesis. Using mass spectrometry, we show that the purified active kinase specifically phosphorylates recombinant eNOS on Ser1179. Treatment of endothelial cells with VEGF or phorbol 12,13-dibutyrate (PDBu) activates PKD and increases eNOS Ser1179 phosphorylation. In addition, pharmacological inhibition of PKD and gene silencing of both PKD1 and PKD2 abrogate VEGF signaling, resulting in a clear diminished migration of endothelial cells in a wound healing assay. Finally, inhibition of PKD in mice results in an almost complete disappearance of the VEGF-induced vasodilatation, as monitored through determination of the diameter of the carotid artery. Hence, our data indicate that PKD is a new regulatory kinase of eNOS in endothelial cells whose activity orchestrates mammalian vascular tone.
Collapse
Affiliation(s)
- Clara Aicart-Ramos
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Lucía Sánchez-Ruiloba
- Instituto de Investigaciones Biomédicas "Alberto Sols". CSIC-UAM, C/Arturo Duperier, Madrid 28029, Spain CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Carlos Zaragoza
- Cardiovascular Research Unit University Francisco de Vitoria/Hospital Ramón y Cajal, Ctra Colmenar Viejo Km 9,100, Madrid 28034, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols". CSIC-UAM, C/Arturo Duperier, Madrid 28029, Spain CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ignacio Rodríguez-Crespo
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid, Madrid 28040, Spain
| |
Collapse
|
89
|
Higashi Y, Quevedo HC, Tiwari S, Sukhanov S, Shai SY, Anwar A, Delafontaine P. Interaction between insulin-like growth factor-1 and atherosclerosis and vascular aging. FRONTIERS OF HORMONE RESEARCH 2014; 43:107-24. [PMID: 24943302 DOI: 10.1159/000360571] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The process of vascular aging encompasses alterations in the function of endothelial (ECs) and vascular smooth muscle cells (VSMCs) via oxidation, inflammation, cell senescence and epigenetic modifications, increasing the probability of atherosclerosis. Aged vessels exhibit decreased endothelial antithrombogenic properties, increased reactive oxygen species generation, inflammatory signaling and migration of VSMCs to the subintimal space, impaired angiogenesis and increased elastin degradation. The key initiating step in atherogenesis is subendothelial accumulation of apolipoprotein B-containing low-density lipoproteins resulting in activation of ECs and recruitment of monocytes. Activated ECs secrete 'chemokines' that interact with cognate chemokine receptors on monocytes and promote directional migration. Recruitment of immune cells establishes a proinflammatory status, further causing elevated oxidative stress, which in turn triggers a series of events including apoptotic or necrotic death of vascular and nonvascular cells. Increased oxidative stress is also considered to be a key factor in mechanisms of aging-associated changes in tissue integrity and function. Experimental evidence indicates that insulin-like growth factor-1 exerts antioxidant, anti-inflammatory and pro-survival effects on the vasculature, reducing atherosclerotic plaque burden and promoting features of atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Yusuke Higashi
- Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, La., USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Mayra PR, Rosalina VL, López G, Iruretagoyena J, Magness R. [Regulation of uterine blood flow. II. Functions of estrogen and estrogen receptor α/β in genomic and non-genomic actions of the uterine endothelium]. ACTA ACUST UNITED AC 2014; 79:218-228. [PMID: 26113751 DOI: 10.4067/s0717-75262014000300011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pregnancy is marked by changes and cardiovascular adaptations that are important for the maintenance and growth of the placenta and fetus. During this period, the uterine vascular adaptations manifest changes that can be classified as short or long term and they related to adaptations for vasodilation, angiogenic or remodeling. Estrogen and the classical estrogen receptors (ERs), ER-α and ER-β, have been shown to be partially responsible for facilitating this dramatic increase in uterine blood flow needed during pregnancy. This literature review discusses the basis for structural diversity and functional selectivity of ERs by estrogen, the role of ERs on the genomic and non-genomic effects in endothelial cells of uterine arteries (UAEC). These themes integrate scientific knowledge about the molecular regulation of UAEC to maintain the physiological increase in uteroplacental perfusion observed during normal pregnancy.
Collapse
Affiliation(s)
- Pastore R Mayra
- Programa de Endocrinología y Fisiología Reproductiva, Universidad de Wisconsin-Madison, Wisconsin, USA ; Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Villalón L Rosalina
- Programa de Endocrinología y Fisiología Reproductiva, Universidad de Wisconsin-Madison, Wisconsin, USA ; Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Gladys López
- Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento Perinatal, Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Jesús Iruretagoyena
- Departamento Perinatal, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento de Obstetricia y Ginecología de la División de Medicina Materno-Fetal. Universidad de Wisconsin-Madison, Wisconsin, USA
| | - Ronald Magness
- Programa de Endocrinología y Fisiología Reproductiva, Universidad de Wisconsin-Madison, Wisconsin, USA ; Laboratorio de Investigación Perinatal, Departamento de Obstetricia y Ginecología y División de Ciencias de la Reproducción, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento Perinatal, Universidad de Wisconsin-Madison, Wisconsin, USA ; Departamento de Ciencias de Animales, Universidad de Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
91
|
Apicidin-Resistant HA22T Hepatocellular Carcinoma Cells strongly activated the Wnt/β-Catenin Signaling Pathway and MMP-2 Expression via the IGF-IR/PI3K/Akt Signaling Pathway Enhancing Cell Metastatic Effect. Biosci Biotechnol Biochem 2014; 77:2397-404. [DOI: 10.1271/bbb.130503] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
92
|
Zhu M, Chen J, Yin H, Jiang H, Wen M, Miao C. Propofol protects human umbilical vein endothelial cells from cisplatin-induced injury. Vascul Pharmacol 2014; 61:72-9. [DOI: 10.1016/j.vph.2014.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/24/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023]
|
93
|
Sánchez-Ruiloba L, Aicart-Ramos C, García-Guerra L, Pose-Utrilla J, Rodríguez-Crespo I, Iglesias T. Protein kinase D interacts with neuronal nitric oxide synthase and phosphorylates the activatory residue serine 1412. PLoS One 2014; 9:e95191. [PMID: 24740233 PMCID: PMC3989272 DOI: 10.1371/journal.pone.0095191] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/24/2014] [Indexed: 12/20/2022] Open
Abstract
Neuronal Nitric Oxide Synthase (nNOS) is the biosynthetic enzyme responsible for nitric oxide (·NO) production in muscles and in the nervous system. This constitutive enzyme, unlike its endothelial and inducible counterparts, presents an N-terminal PDZ domain known to display a preference for PDZ-binding motifs bearing acidic residues at -2 position. In a previous work, we discovered that the C-terminal end of two members of protein kinase D family (PKD1 and PKD2) constitutes a PDZ-ligand. PKD1 has been shown to regulate multiple cellular processes and, when activated, becomes autophosphorylated at Ser916, a residue located at -2 position of its PDZ-binding motif. Since nNOS and PKD are spatially enriched in postsynaptic densities and dendrites, the main objective of our study was to determine whether PKD1 activation could result in a direct interaction with nNOS through their respective PDZ-ligand and PDZ domain, and to analyze the functional consequences of this interaction. Herein we demonstrate that PKD1 associates with nNOS in neurons and in transfected cells, and that kinase activation enhances PKD1-nNOS co-immunoprecipitation and subcellular colocalization. However, transfection of mammalian cells with PKD1 mutants and yeast two hybrid assays showed that the association of these two enzymes does not depend on PKD1 PDZ-ligand but its pleckstrin homology domain. Furthermore, this domain was able to pull-down nNOS from brain extracts and bind to purified nNOS, indicating that it mediates a direct PKD1-nNOS interaction. In addition, using mass spectrometry we demonstrate that PKD1 specifically phosphorylates nNOS in the activatory residue Ser1412, and that this phosphorylation increases nNOS activity and ·NO production in living cells. In conclusion, these novel findings reveal a crucial role of PKD1 in the regulation of nNOS activation and synthesis of ·NO, a mediator involved in physiological neuronal signaling or neurotoxicity under pathological conditions such as ischemic stroke or neurodegeneration.
Collapse
Affiliation(s)
- Lucía Sánchez-Ruiloba
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Aicart-Ramos
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Lucía García-Guerra
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Rodríguez-Crespo
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense de Madrid (UCM), Madrid, Spain
- * E-mail: (IRC); (TI)
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail: (IRC); (TI)
| |
Collapse
|
94
|
García C, Nuñez-Anita RE, Thebault S, Arredondo Zamarripa D, Jeziorsky MC, Martínez de la Escalera G, Clapp C. Requirement of phosphorylatable endothelial nitric oxide synthase at Ser-1177 for vasoinhibin-mediated inhibition of endothelial cell migration and proliferation in vitro. Endocrine 2014; 45:263-70. [PMID: 23640371 DOI: 10.1007/s12020-013-9964-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
Endothelial nitric oxide synthase (eNOS)-derived nitric oxide is a major vasorelaxing factor and a mediator of vasopermeability and angiogenesis. Vasoinhibins, a family of antiangiogenic prolactin fragments that include 16 K prolactin, block most eNOS-mediated vascular effects. Vasoinhibins activate protein phosphatase 2A, causing eNOS inactivation through dephosphorylation of eNOS at serine residue 1179 in bovine endothelial cells and thereby blocking vascular permeability. In this study, we examined whether human eNOS phosphorylation at S1177 (analogous to bovine S1179) influences other actions of vasoinhibins. Bovine umbilical vein endothelial cells were stably transfected with human wild-type eNOS (WT) or with phospho-mimetic (S1177D) or non-phosphorylatable (S1177A) eNOS mutants. Vasoinhibins inhibited the increases in eNOS activity, migration, and proliferation following the overexpression of WT eNOS but did not affect these responses in cells expressing S1177D and S1177A eNOS mutants. We conclude that eNOS inhibition by dephosphorylation of S1177 is fundamental for the inhibition of endothelial cell migration and proliferation by vasoinhibins.
Collapse
Affiliation(s)
- Celina García
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, QRO, Mexico
| | | | | | | | | | | | | |
Collapse
|
95
|
Kypreos KE, Zafirovic S, Petropoulou PI, Bjelogrlic P, Resanovic I, Traish A, Isenovic ER. Regulation of endothelial nitric oxide synthase and high-density lipoprotein quality by estradiol in cardiovascular pathology. J Cardiovasc Pharmacol Ther 2014; 19:256-68. [PMID: 24414281 DOI: 10.1177/1074248413513499] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Estrogens have been recognized, in the last 3 decades, as important hormones in direct and indirect modulation of vascular health. In addition to their direct benefit on cardiovascular health, the presence of esterified estrogen in the lipid core of high-density lipoprotein (HDL) particles indirectly contributes to atheroprotection by significantly improving HDL quality and functionality. Estrogens modulate their physiological activity via genomic and nongenomic mechanisms. Genomic mechanisms are thought to be mediated directly by interaction of the hormone receptor complex with the hormone response elements that regulate gene expression. Nongenomic mechanisms are thought to occur via interaction of the estrogen with membrane-bound receptors, which rapidly activate intracellular signaling without binding of the hormone receptor complex to its hormone response elements. Estradiol in particular mediates early and late endothelial nitric oxide synthase (eNOS) activation via interaction with estrogen receptors through both nongenomic and genomic mechanisms. In the vascular system, the primary endogenous source of nitric oxide (NO) generation is eNOS. Nitric oxide primarily influences blood vessel relaxation, the heart rate, and myocyte contractility. The abnormalities in expression and/or functions of eNOS lead to the development of cardiovascular diseases, both in animals and in humans. Although considerable research efforts have been dedicated to understanding the mechanisms of action of estradiol in regulating cardiac eNOS, more research is needed to fully understand the details of such mechanisms. This review focuses on recent findings from animal and human studies on the regulation of eNOS and HDL quality by estradiol in cardiovascular pathology.
Collapse
Affiliation(s)
- Kyriakos E Kypreos
- 1Department of Medicine, University of Patras Medical School, Pharmacology Laboratory, Panepistimioupolis, Rio, Greece
| | | | | | | | | | | | | |
Collapse
|
96
|
Georgakis GV, Younes A. From Rapa Nui to rapamycin: targeting PI3K/Akt/mTOR for cancer therapy. Expert Rev Anticancer Ther 2014; 6:131-40. [PMID: 16375650 DOI: 10.1586/14737140.6.1.131] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
One of the most prominent pathways explored in the area of targeted therapy is the PI3K/Akt/mTOR pathway, which plays a central role in cell survival and proliferation. Deregulation of this pathway has been implicated in the promotion of cancer cell growth and survival. Inhibition of several steps of this pathway has been shown to confer favorable antitumor activity in a variety of cancer types. This article provides a brief analysis of the PI3K/Akt/mTOR pathway, its importance in tumor pathogenesis and the current status of preclinical and clinical studies targeting signaling components of this pathway.
Collapse
Affiliation(s)
- Georgios V Georgakis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
97
|
Qian J, Fulton D. Post-translational regulation of endothelial nitric oxide synthase in vascular endothelium. Front Physiol 2013; 4:347. [PMID: 24379783 PMCID: PMC3861784 DOI: 10.3389/fphys.2013.00347] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/11/2013] [Indexed: 01/22/2023] Open
Abstract
Nitric oxide (NO) is a short-lived gaseous signaling molecule. In blood vessels, it is synthesized in a dynamic fashion by endothelial nitric oxide synthase (eNOS) and influences vascular function via two distinct mechanisms, the activation of soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP)-dependent signaling and the S-nitrosylation of proteins with reactive thiols (S-nitrosylation). The regulation of eNOS activity and NO bioavailability is critical to maintain blood vessel function. The activity of eNOS and ability to generate NO is regulated at the transcriptional, posttranscriptional, and posttranslational levels. Post-translational modifications acutely impact eNOS activity and dysregulation of these mechanisms compromise eNOS activity and foster the development of cardiovascular diseases (CVDs). This review will intergrate past and current literature on the post-translational modifications of eNOS in both health and disease.
Collapse
Affiliation(s)
- Jin Qian
- Pulmonary and Critical Care, School of Medicine, Stanford University/VA Palo Alto Health Care System Palo Alto, CA, USA
| | - David Fulton
- Vascular Biology Center, Georgia Regents University Augusta, GA, USA
| |
Collapse
|
98
|
Lopez-Quintero SV, Cancel LM, Pierides A, Antonetti D, Spray DC, Tarbell JM. High glucose attenuates shear-induced changes in endothelial hydraulic conductivity by degrading the glycocalyx. PLoS One 2013; 8:e78954. [PMID: 24260138 PMCID: PMC3832508 DOI: 10.1371/journal.pone.0078954] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 09/17/2013] [Indexed: 11/19/2022] Open
Abstract
Diabetes mellitus is a risk factor for cardiovascular disease; however, the mechanisms through which diabetes impairs homeostasis of the vasculature have not been completely elucidated. The endothelium interacts with circulating blood through the surface glycocalyx layer, which serves as a mechanosensor/transducer of fluid shear forces leading to biomolecular responses. Atherosclerosis localizes typically in regions of low or disturbed shear stress, but in diabetics, the distribution is more diffuse, suggesting that there is a fundamental difference in the way cells sense shear forces. In the present study, we examined the effect of hyperglycemia on mechanotranduction in bovine aortic endothelial cells (BAEC). After six days in high glucose media, we observed a decrease in heparan sulfate content coincident with a significant attenuation of the shear-induced hydraulic conductivity response, lower activation of eNOS after exposure to shear, and reduced cell alignment with shear stress. These studies are consistent with a diabetes-induced change to the glycocalyx altering endothelial response to shear stress that could affect the distribution of atherosclerotic plaques.
Collapse
Affiliation(s)
- Sandra V. Lopez-Quintero
- Department of Biomedical Engineering, The City College of The City University of New York, New York, New York, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, United States of America
| | - Limary M. Cancel
- Department of Biomedical Engineering, The City College of The City University of New York, New York, New York, United States of America
| | - Alexis Pierides
- Department of Biomedical Engineering, The City College of The City University of New York, New York, New York, United States of America
| | - David Antonetti
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - David C. Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, United States of America
| | - John M. Tarbell
- Department of Biomedical Engineering, The City College of The City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
99
|
Kumazoe M, Kim Y, Bae J, Takai M, Murata M, Suemasu Y, Sugihara K, Yamashita S, Tsukamoto S, Huang Y, Nakahara K, Yamada K, Tachibana H. Phosphodiesterase 5 inhibitor acts as a potent agent sensitizing acute myeloid leukemia cells to 67-kDa laminin receptor-dependent apoptosis. FEBS Lett 2013; 587:3052-7. [PMID: 23916810 DOI: 10.1016/j.febslet.2013.07.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/04/2013] [Accepted: 07/18/2013] [Indexed: 01/19/2023]
Abstract
(-)-Epigallocatechin-3-O-gallate (EGCG), a polyphenol in green tea, induces apoptosis in acute myeloid leukemia (AML) cells without affecting normal cells. In this study, we observed that cGMP acts as a cell death mediator of the EGCG-induced anti-AML effect through acid sphingomyelinase activation. EGCG activated the Akt/eNOS axis, a well-known mechanism in vascular cGMP upregulation. We also observed that a major cGMP negative regulator, phosphodiesterase 5, was overexpressed in AML cells, and PDE5 inhibitor, an anti-erectile dysfunction drug, synergistically enhanced the anti-AML effect of EGCG. This combination regimen killed AML cells via overexpressed 67-kDa laminin receptors.
Collapse
MESH Headings
- Apoptosis/drug effects
- Catechin/analogs & derivatives
- Catechin/pharmacology
- Cyclic GMP/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 5/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Drug Synergism
- Enzyme Activation/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- HL-60 Cells
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Oncogene Protein v-akt/genetics
- Oncogene Protein v-akt/metabolism
- Phosphodiesterase 5 Inhibitors/pharmacology
- Primary Cell Culture
- Receptors, Laminin/agonists
- Receptors, Laminin/genetics
- Receptors, Laminin/metabolism
- Signal Transduction
- Sphingomyelin Phosphodiesterase/genetics
- Sphingomyelin Phosphodiesterase/metabolism
Collapse
Affiliation(s)
- Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Ning WH, Zhao K. Propionyl-L-carnitine induces eNOS activation and nitric oxide synthesis in endothelial cells via PI3 and Akt kinases. Vascul Pharmacol 2013; 59:76-82. [PMID: 23850990 DOI: 10.1016/j.vph.2013.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/09/2013] [Accepted: 07/04/2013] [Indexed: 11/20/2022]
Abstract
Propionyl-l-carnitine (PLC) is a natural short-chain derivative of l-carnitine (LC), a natural amino acid that plays an important role in fatty acid metabolism. Recent studies suggest that PLC has vascular protective effects. Because of the importance of endothelial nitric oxide synthase (eNOS) and its product, antiatherogenic molecule nitric oxide (NO), in vascular endothelial function, we sought to elucidate that if PLC would stimulate eNOS and its upstream activators Akt and phosphatidylinositol 3-kinase (PI3 Kinase) in cultured human aortic endothelial cells (HAEC). PLC caused eNOS phosphorylation at Ser-1177, and dominant negative Akt and a novel Akt-selective inhibitor MK-2206 inhibited both PLC-mediated phosphorylation and activation of the enzyme. PI3 kinase inhibition also blocked the phosphorylation and activation of eNOS by PLC. Studies with specific drug inhibitors PD173955 and PP2 showed that the non-receptor tyrosine kinase, src, is an upstream stimulator of the PI3 kinase-Akt pathway in this pathway. In addition, PLC significantly decreased intracellular ATP/ADP ratio and activate AMPK, subsequently leading to Src activation. Finally, we demonstrated that the effects of PLC to augment eNOS activity were associated with a net increase in NO release from endothelial cells. NO production following incubation with PLC was abolished in endothelial cells coincubated with L-NAME, PD173955, LY294002, MK-2206 and compound C. In conclusion, PLC, via AMPK/Src-mediated signaling that leads to activation of PI3 kinase and Akt, stimulates eNOS, leading to increased production of NO.
Collapse
Affiliation(s)
- Wen-hu Ning
- Department of Emergency, the Forth Clinical Hospital, Harbin Medical University, Heilongjiang Harbin 150001, China.
| | | |
Collapse
|