51
|
Boulanger CM, Baretella O, Blaise G, Bond RA, Cai Y, Chan CKY, Chataigneau T, Chen MJ, Chen H, Cheng Y, Clement DL, Cohen RA, Collis M, Danser AHJ, de Mey J, Detremmerie CMS, Duprez D, Feletou M, Flavahan N, Gao Y, Guo Y, Hoeffner U, Houston DS, Huang IB, Huang Y, Iliano S, Junquero D, Katusic ZS, Komori K, Lee MYK, Leung SWS, Li Z, Liang SC, Liu JTC, Luscher TF, Michel F, Miller VM, Mombouli JV, Morrison K, Muldoon SM, O'Rourke S, Perrault L, Quignard JF, Rusch NJ, Sanchez-Ferrer CF, Schini-Kerth V, Shen K, Shi Y, Song E, Sun KWY, Taddei S, Tang EHC, Tuncer M, van den Ende R, Vedernikov Y, Verbeuren TJ, Webb C, Weigert A, Wong KHK, Xu C, Yang K, Ying F, Zellers T, Zhao Y, Zou Q, Shimokawa H. Tribute to Paul M. Vanhoutte, MD, PhD (1940-2019). Arterioscler Thromb Vasc Biol 2019; 39:2445-2447. [PMID: 31770032 DOI: 10.1161/atvbaha.119.313461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | | | | | | | - Yin Cai
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | | | | | | | - Hui Chen
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Yanhua Cheng
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | | | | | | | | | - Jo de Mey
- University of Southern Denmark (J.D.M.)
| | | | | | | | | | | | - Yumeng Guo
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Ute Hoeffner
- Glenmark Pharmaceuticals, Gröbenzell, Germany (U.H.)
| | | | | | - Yu Huang
- Chinese University of Hong Kong, Hong Kong, PRC (I.B.H., Y.H., F.Y.)
| | | | | | | | | | - Mary Y K Lee
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Susan W S Leung
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Zhuoming Li
- Sun Yat-sen University, Guangzhou, China (Z.L.)
| | - Sophie Chaofan Liang
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Jacky Tsz Chiu Liu
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Thomas F Luscher
- Royal Brompton & Harefield Hospitals, London, United Kingdom (T.F.L.)
| | - Frederic Michel
- University of the Witwatersrand, Johannesburg, South Africa (F.M.)
| | | | | | - Keith Morrison
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland (K.M.)
| | | | | | | | | | - Nancy J Rusch
- University of Arkansas for Medical Sciences, Little Rock (N.J.R.)
| | | | | | - Kaikai Shen
- Shanghai University of Traditional Chinese Medicine, China (K.S.)
| | - Yi Shi
- Zhongshan Hospital Fudan University, China (Y.S.)
| | - Erfei Song
- Jinan University, Guangzhou, China (E.S.)
| | - Kiwi W Y Sun
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | | | - Eva Hoi Ching Tang
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | | | | | | | | | | | - André Weigert
- Hospital S. Cruz, University of Lisbon, Portugal (A.W.)
| | - Kenneth H K Wong
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | - Cheng Xu
- Shenzhen University, China (C.X.)
| | | | - Fan Ying
- Chinese University of Hong Kong, Hong Kong, PRC (I.B.H., Y.H., F.Y.)
| | - Thomas Zellers
- University of Texas Southwestern Medical Center, Dallas (T.Z.)
| | - Yingzi Zhao
- Chinese Academy of Sciences, Shenzhen, China (Y.Z.)
| | - Qian Zou
- University of Hong Kong, PRC (Y. Cai, H.C., Y. Cheng, Y. Guo, M.Y.K.L., S.W.S.L., S.C.L., J.T.C.L., K.W.Y.S., E.H.C.T., K.H.K.W., Q.Z.)
| | | | | |
Collapse
|
52
|
Arildsen L, Andersen JV, Waagepetersen HS, Nissen JBD, Sheykhzade M. Hypermetabolism and impaired endothelium-dependent vasodilation in mesenteric arteries of type 2 diabetes mellitus db/db mice. Diab Vasc Dis Res 2019; 16:539-548. [PMID: 31364402 DOI: 10.1177/1479164119865885] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Besides being a metabolic disease, diabetes is considered a vascular disease as many of the complications relate to vascular pathologies. The aim of this study was to investigate how vascular tone and reactivity and vascular cell metabolism were affected in type 2 diabetes mellitus and whether β-hydroxybutyrate could have a positive effect as alternative energy substrate. Isolated mesenteric arteries of db/db and control mice were incubated in media containing [U-13C]glucose or [U-13C]β-hydroxybutyrate, and tissue extracts were analysed by mass spectrometry. Functional characterization was performed by wire myography to assess vasodilation and vasocontraction. Hypermetabolism of glucose and β-hydroxybutyrate was observed for mesenteric arteries of db/db mice; however, hypermetabolism was significant only with β-hydroxybutyrate as energy substrate. The functional characterization showed impaired endothelial-dependent vasodilation in mesenteric arteries of the db/db mice, whereas the contractility was unaffected. This study provides evidence that the endothelial cells are impaired, whereas the vascular smooth muscle cells are more robust and seemed less affected in the db/db mouse. Furthermore, the results indicate that hypermetabolism of energy substrates may be due to adaptive changes in the mesenteric arteries.
Collapse
Affiliation(s)
- Lene Arildsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jens Velde Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Helle Soenderby Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jakob Borre Dahl Nissen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
53
|
Circadian Variation in Vasoconstriction and Vasodilation Mediators and Baroreflex Sensitivity in Hypertensive Rats. J Circadian Rhythms 2019; 17:10. [PMID: 31673274 PMCID: PMC6798778 DOI: 10.5334/jcr.185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The purpose of this study was to evaluate the relationship between the circadian profile of the vasorelaxing substances calcitonin gene-related peptide (CGRP) and epoxyeicosatrienoic acids (EETs) and the vasconstrictive agent endothelin-1 (ET1) and the daily rhythms of cardiac hemodynamic indices (CHI) and baroreflex (BRS) in Wistar rats with 1 kidney-1 clip model of arterial hypertension (1K-1C AH). The animals were divided into 3 groups: I- sham-operated (SO), II- 4-week and III- 8-week 1K-1C AH rats. Plasma concentration of ET1, CGRP and EET’s were investigated every 4 h. In conscious freely moving 1K-1C AH rats unlike SO animals blood pressure (BP), heart period (HP) and BRS underwent significant circadian fluctuations, with more marked increase in mean values of BP in 8-week hypertensive rats in comparison to 4-week hypertensive rats (179 ± 5 vs. 162 ± 4 mm Hg, p < 0.05). These alterations correlated with more significant reduction in HP (138 ± 5 vs. 150 ± 6 ms, p < 0,05) and BRS (0.44 ± 0.04 vs. 0.58 ± 0.04 ms mm Hg–1, p < 0.05) in 8-week 1K-1C AH rats. The acrophases of BP in 8-week 1K-1C AH rats in comparison with 4-week were shifted to more late night hours (1:58 a.m. vs. 11:32 p.m.) and in both groups of animals corresponded to lowest circadian plasma levels of CGRP and EETs and to greatest level of ET1. SO rats were characterized by lower values of BP (121 ± 3 mm Hg, p < 0,05) and higher indices of HP (158 ± 2 ms, p < 0,05) and BRS (0.86 ± 0.02 ms mmHg–1, p < 0,001) in comparison with 1K-1C AH rats 4-week duration. The acrophases of BP, HP and BRS in hypertensive animals were revealed at 14.8 ± 0.5 h, 13.6 ± 0.4 h and 13.1 ± 0.2 h, which correlated with maximal circadian contents of ET1 and CGRP at 24:00 h and EETs at 12:00 h and were shifted in comparison to sham-operated group. In rats with 1K-1C AH, plasma levels of ET1, CGRP and EETs undergo circadian fluctuation with corresponding alterations in CHI and BRS which are more markedly expressed on the late stage of diseases and could be used in future for predictive, preventive, and personalized treatment of arterial hypertension.
Collapse
|
54
|
Van den Bergh G, Opdebeeck B, D'Haese PC, Verhulst A. The Vicious Cycle of Arterial Stiffness and Arterial Media Calcification. Trends Mol Med 2019; 25:1133-1146. [PMID: 31522956 DOI: 10.1016/j.molmed.2019.08.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Arterial media calcification and arterial stiffness are independent predictors of cardiovascular mortality. Both processes reinforce one another, creating a vicious cycle in which transdifferentiation of endothelial cells and vascular smooth muscle cells play a central role. Physiological functioning of vascular smooth muscle cells in the arterial medial layer greatly depends on normal endothelial cell behavior. Endothelial or intimal layer cells are the primary sensors of pathological triggers circulating in the blood during, for example, ageing or inflammation, and often can be seen as initiators of this vicious cycle. As such, the search for treatment of arterial media calcification, which until now has been mainly concentrated at the level of the vascular smooth cell, may need to be expanded to intimal layer targets.
Collapse
Affiliation(s)
- Geoffrey Van den Bergh
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium
| | - Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium.
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium
| |
Collapse
|
55
|
Krüger-Genge A, Blocki A, Franke RP, Jung F. Vascular Endothelial Cell Biology: An Update. Int J Mol Sci 2019; 20:ijms20184411. [PMID: 31500313 PMCID: PMC6769656 DOI: 10.3390/ijms20184411] [Citation(s) in RCA: 669] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
The vascular endothelium, a monolayer of endothelial cells (EC), constitutes the inner cellular lining of arteries, veins and capillaries and therefore is in direct contact with the components and cells of blood. The endothelium is not only a mere barrier between blood and tissues but also an endocrine organ. It actively controls the degree of vascular relaxation and constriction, and the extravasation of solutes, fluid, macromolecules and hormones, as well as that of platelets and blood cells. Through control of vascular tone, EC regulate the regional blood flow. They also direct inflammatory cells to foreign materials, areas in need of repair or defense against infections. In addition, EC are important in controlling blood fluidity, platelet adhesion and aggregation, leukocyte activation, adhesion, and transmigration. They also tightly keep the balance between coagulation and fibrinolysis and play a major role in the regulation of immune responses, inflammation and angiogenesis. To fulfill these different tasks, EC are heterogeneous and perform distinctly in the various organs and along the vascular tree. Important morphological, physiological and phenotypic differences between EC in the different parts of the arterial tree as well as between arteries and veins optimally support their specified functions in these vascular areas. This review updates the current knowledge about the morphology and function of endothelial cells, particularly their differences in different localizations around the body paying attention specifically to their different responses to physical, biochemical and environmental stimuli considering the different origins of the EC.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Department of Biomaterials and Healthcare, Division of Life Science and Bioprocesses, Fraunhofer Institute for Applied Polymer Research (IAP), Potsdam-Golm 14476, Germany.
- Department of Anesthesia, Pain Management and Perioperative Medicine, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 2Y9, Canada.
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine (ITERM), School of Biomedical Sciences (SBS), Chinese University of Hong Kong (CUHK), New Territories, Hong Kong, China
| | - Ralf-Peter Franke
- Central Institute for Biomedical Technology, Dep. Biomaterials, University of Ulm, Albert-Einstein-Allee 47, 89081 Ulm, Germany
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology, 01968 Senftenberg, Germany
| |
Collapse
|
56
|
Xie X, Yang C, Cui Q, Ma W, Liu J, Yao Q, Zhang Z, Xiao L, Wang N. Stachydrine Mediates Rapid Vascular Relaxation: Activation of Endothelial Nitric Oxide Synthase Involving AMP-Activated Protein Kinase and Akt Phosphorylation in Vascular Endothelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9805-9811. [PMID: 31407895 DOI: 10.1021/acs.jafc.9b03501] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Stachydrine (STA) is a constituent of citrus fruits and Leonurus heterophyllus Sweet. In the present study, we established that STA caused acute endothelium-dependent relaxation. The vascular action of STA was mediated by nitric oxide (NO) via cyclic guanosine monophosphate. Mechanistically, STA activated AMP-activated protein kinase (AMPK), protein kinase B/Akt, and endothelial NO synthase (eNOS) in vascular endothelial cells (ECs). AMPK inhibition by compound C blocked STA-induced Akt/eNOS phosphorylation, suggesting that AMPK is the upstream of Akt and eNOS. Inhibition of Akt by MK2206 blocked STA-stimulated eNOS phosphorylation without altering AMPK phosphorylation. Furthermore, we showed that STA activated AMPK via the induction of liver kinase B1 phosphorylation. These results indicated that STA can induce eNOS phosphorylation and vasorelaxation by regulating the interplay between AMPK and Akt pathways in ECs. These findings further highlighted the potential of STA as a nutritional factor in the beneficial effects of fruit intake in preventing the endothelial dysfunction-related metabolic cardiovascular diseases.
Collapse
Affiliation(s)
- Xinya Xie
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | - Chunmiao Yang
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | | | - Wen Ma
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | - Jia Liu
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | - Qinyu Yao
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | - Zihui Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | - Lei Xiao
- Cardiovascular Research Center, School of Basic Medical Sciences , Xi'an Jiaotong University , Xi'an 710061 , China
| | | |
Collapse
|
57
|
Ottolini M, Hong K, Sonkusare SK. Calcium signals that determine vascular resistance. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1448. [PMID: 30884210 PMCID: PMC6688910 DOI: 10.1002/wsbm.1448] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 12/19/2022]
Abstract
Small arteries in the body control vascular resistance, and therefore, blood pressure and blood flow. Endothelial and smooth muscle cells in the arterial walls respond to various stimuli by altering the vascular resistance on a moment to moment basis. Smooth muscle cells can directly influence arterial diameter by contracting or relaxing, whereas endothelial cells that line the inner walls of the arteries modulate the contractile state of surrounding smooth muscle cells. Cytosolic calcium is a key driver of endothelial and smooth muscle cell functions. Cytosolic calcium can be increased either by calcium release from intracellular stores through IP3 or ryanodine receptors, or the influx of extracellular calcium through ion channels at the cell membrane. Depending on the cell type, spatial localization, source of a calcium signal, and the calcium-sensitive target activated, a particular calcium signal can dilate or constrict the arteries. Calcium signals in the vasculature can be classified into several types based on their source, kinetics, and spatial and temporal properties. The calcium signaling mechanisms in smooth muscle and endothelial cells have been extensively studied in the native or freshly isolated cells, therefore, this review is limited to the discussions of studies in native or freshly isolated cells. This article is categorized under: Biological Mechanisms > Cell Signaling Laboratory Methods and Technologies > Imaging Models of Systems Properties and Processes > Mechanistic Models.
Collapse
Affiliation(s)
- Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| | - Kwangseok Hong
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Physical Education, Chung-Ang University, Seoul, 06974, South Korea
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| |
Collapse
|
58
|
Taurine Regulation of Peripheral Hemodynamics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31468396 DOI: 10.1007/978-981-13-8023-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Taurine plays an important role in the modulation of cardiovascular function by acting not only within the brain but also within peripheral tissues. We found that IV injection of taurine to male rats caused hypotension and tachycardia. A single injection of taurine significantly lowered the systolic, diastolic and mean arterial pressure blood pressure in freely moving long Evans control rats. Previousely, we found that the endothelial cells express high levels of taurine transporters and GABAA receptors and showed that taurine activates GABAA receptors. Thus we suggest that the functional implication of GABAA receptors activation is the relaxation of the arterial muscularis, vasodilation and a decrease in blood pressure. Interestingly however, the effects of acute taurine injection were very different that chronic supplementation of taurine. When rats were supplemented taurine (0.05%, 4 weeks) in their drinking water, taurine has significant hypertensive properties. The increase in blood pressure was observed however only in females, males supplemented with taurine did not show an increase in systolic, diastolic or mean arterial pressure. In both genders however, taurine supplementation caused a significant tachycardia. Thus, we suggest that acute administration of taurine may be beneficial to lowering blood pressure. However, our data indicate that supplementation of taurine to females caused a significant increase in blood pressure. It remains to be seen the effect of taurine supplementation on hypertensive rats.
Collapse
|
59
|
Chen H, Vanhoutte PM, Leung SWS. Acute activation of endothelial AMPK surprisingly inhibits endothelium-dependent hyperpolarization-like relaxations in rat mesenteric arteries. Br J Pharmacol 2019; 176:2905-2921. [PMID: 31116877 DOI: 10.1111/bph.14716] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/04/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Endothelium-dependent hyperpolarizations (EDHs) contribute to the regulation of peripheral resistance. They are initiated through opening of endothelial calcium-activated potassium channels (KCa ); the potassium ions released then diffuse to the underlying smooth muscle cells, causing hyperpolarization and thus relaxation. The present study aimed to examine whether or not AMPK modulates EDH-like relaxations in rat mesenteric arteries. EXPERIMENTAL APPROACH Arterial rings were isolated for isometric tension recording. AMPK activity and protein level were measured by ELISA and western blotting respectively. KEY RESULTS The AMPK activator, AICAR, reduced ACh-induced EDH-like relaxations and increased AMPK activity in preparations with endothelium; these responses were prevented by compound C, an AMPK inhibitor. AICAR inhibited relaxations induced by SKA-31 (opener of endothelial KCa ) but did not affect potassium-induced, hyperpolarization-attributable relaxations or increase AMPK activity in preparations without endothelium. A769662, another AMPK activator, not only caused a similar inhibition of relaxations to ACh and SKA-31 in preparations with endothelium but also inhibited hyperpolarization-attributable relaxations and augmented AMPK activity in rings without endothelium. Protein levels of total AMPKα, AMPKα1, or AMPKβ1/2 were comparable between preparations with and without endothelium. CONCLUSIONS AND IMPLICATIONS Activation of endothelial AMPK, by either AICAR or A769662, acutely inhibits EDH-like relaxations of rat mesenteric arteries. Furthermore, A769662 inhibits signalling downstream of smooth muscle hyperpolarization. In view of the major blunting effect of AMPK activation on EDH-like relaxations, caution should be applied when administering therapeutic agents that activate AMPK in patients with endothelial dysfunction characterized by reduced production and/or bioavailability of NO.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Paul M Vanhoutte
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Susan W S Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
60
|
Sulodexide promotes arterial relaxation via endothelium-dependent nitric oxide-mediated pathway. Biochem Pharmacol 2019; 166:347-356. [PMID: 31014752 DOI: 10.1016/j.bcp.2019.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/19/2019] [Indexed: 12/18/2022]
Abstract
Sulodexide (SDX) is a highly purified glycosaminoglycan with antithrombotic and profibrinolytic properties and reported benefits in thrombotic and atherosclerotic vascular disorders. However, the effects of SDX on vascular function are unclear. We tested whether SDX affects vascular relaxation and examined the potential underlying mechanisms. Isolated segments of male rat abdominal aorta and mesenteric artery were suspended in a tissue bath, and the changes in arterial contraction/relaxation were measured. The α-adrenergic receptor agonist phenylephrine (Phe) (10-9-10-5 M) caused concentration-dependent aortic and mesenteric artery contraction that was reduced in tissues pretreated with SDX (1 mg/ml). In aortic and mesenteric arterial segments precontracted with submaximal concentration of Phe (3 × 10-7-6 × 10-7 M), SDX (0.001-1 mg/ml) caused concentration-dependent relaxation. To test the role of endothelium, SDX-induced relaxation was compared with that of acetylcholine (ACh), a known activator of endothelium-dependent relaxation. In Phe precontracted aorta, ACh relaxation was abolished and SDX relaxation was significantly inhibited by endothelium removal or the nitric oxide synthase (NOS) inhibitor Nω-nitro-l-arginine methyl ester (L-NAME), suggesting a role of NO. In mesenteric artery, ACh relaxation was abolished by endothelium removal, partially blocked by L-NAME, and completely blocked by a mixture of indomethacin, a cyclooxygenase inhibitor and blocker of the PGI2-cAMP pathway, and tetraethylammonium, a blocker of K+ channels and EDHF-dependent hyperpolarization pathway. In comparison, SDX relaxation of mesenteric artery was almost completely inhibited by endothelium removal or NOS inhibitor L-NAME. SDX enhanced vascular relaxation and increased nitrate/nitrite production in response to all ACh concentrations in the aorta, but only to low ACh concentrations (<10-7 M) in mesenteric artery. SDX did not affect aortic or mesenteric artery endothelium-independent relaxation to the NO donor sodium nitroprusside. Thus, SDX promotes arterial relaxation via a mechanism involving endothelium-dependent NO production; an effect that could enhance vasodilation and decrease vasoconstriction in vascular disorders.
Collapse
|
61
|
Potential Benefits of Nitrate Supplementation on Antioxidant Defense System and Blood Pressure Responses after Exercise Performance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7218936. [PMID: 31049136 PMCID: PMC6458846 DOI: 10.1155/2019/7218936] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/10/2018] [Accepted: 02/11/2019] [Indexed: 01/26/2023]
Abstract
Nitrate (NO3−) supplementation is associated with exercise performance, oxygen uptake, blood flow, and blood pressure improvement, and it can act as an antioxidant agent. This study evaluated the effects of sodium nitrate supplementation on oxidative stress markers and blood pressure responses after aerobic exercise performance in physically active males. Fourteen subjects aged 22 ± 3 years and with a BMI of 23 ± 1 kg/m2 were submitted to four exercise tests in intervals of 5 days. Nitrate supplementation (NO session) and placebo supplementation (PL session) were acute (AC) and over a period of 5 days (FD) in random order with a crossover design. Saliva was collected at basal (0′); 60 min after supplementation (60′); immediately after exercise (90′); and 15, 30, and 60 min after the test (105′, 120′, and 150′). The NO session had higher concentrations (P < 0.05) of salivary nitrite in both AC and FD treatments when compared with the PL session. There was a reduction in systolic blood pressure (SBP) only after FD in the NO session. Furthermore, uric acid and total antioxidant capacity (FRAP) salivary concentrations increased, while SOD activity and TBARS levels decreased after FD but not after AC in the NO session. The results suggest that nitrate supplemented over a period of 5 days reduced SBP and indirectly acted as an antioxidant in healthy nonsedentary young men.
Collapse
|
62
|
Important Role of Endothelial Caveolin-1 in the Protective Role of Endothelium-dependent Hyperpolarization Against Nitric Oxide-Mediated Nitrative Stress in Microcirculation in Mice. J Cardiovasc Pharmacol 2019; 71:113-126. [PMID: 29419573 DOI: 10.1097/fjc.0000000000000552] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIMS Nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH) play important roles in maintaining cardiovascular homeostasis. We have previously demonstrated that endothelial NO synthase (eNOS) plays diverse roles depending on vessel size, as a NO generating system in conduit arteries and an EDH-mediated system in resistance arteries, for which caveolin-1 (Cav-1) is involved. However, the physiological role of endothelial Cav-1 in microvessels remains to be elucidated. METHODS AND RESULTS We newly generated endothelium-specific Cav-1-knockout (eCav-1-KO) mice. eCav-1-KO mice showed loss of endothelial Cav-1/eNOS complex and had cardiac hypertrophy despite normal blood pressure. In eCav-1-KO mice, as compared to wild-type controls, the extent of eNOS phosphorylation at inhibitory Thr495 was significantly reduced in mesenteric arteries and the heart. Isometric tension and Langendorff-perfused heart experiments showed that NO-mediated responses were enhanced, whereas EDH-mediated responses were reduced in coronary microcirculation in eCav-1-KO mice. Immunohistochemistry showed increased level of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a marker of nitrative stress, in the heart from eCav-1-KO mice. S-guanylation of cardiac H-Ras in eCav-1-KO mice was also significantly increased compared with wild-type controls. CONCLUSIONS These results suggest that eCav-1 is involved in the protective role of EDH against nitrative stress caused by excessive NO to maintain cardiac microvascular homeostasis.
Collapse
|
63
|
Boudaka A, Al-Suleimani M, Al-Lawati I, Baomar H, Al-Siyabi S, Zadjali F. Downregulation of endothelial transient receptor potential vanilloid type 4 channel underlines impaired endothelial nitric oxide-mediated relaxation in the mesenteric arteries of hypertensive rats. Physiol Res 2019; 68:219-231. [PMID: 30628831 DOI: 10.33549/physiolres.933952] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The endothelium contributes to the maintenance of vasodilator tone by releasing endothelium-derived relaxing factors, including nitric oxide (NO). In hypertension, endothelial nitric oxide synthase (eNOS) produces less NO and could be one of the contributing factors to the increased peripheral vascular resistance. Agonist-induced Ca(2+) entry is essential for the activation of eNOS. The transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca(2+)-permeant cation channel, is expressed in the endothelial cells and involved in the regulation of vascular tone. The present study aimed to investigate the role of TRPV4 channel in endothelium-dependent NO-mediated relaxation of the resistance artery in hypertensive rats. Using a wire myograph, relaxation response to the TRPV4 activator, 4alpha-phorbol-12,13-didecanoate (4alphaPDD) was assessed in mesenteric arteries obtained from Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHRs). Compared to WKY, SHR demonstrated a significantly attenuated 4alphaPDD-induced endothelium-dependent NO-mediated relaxation. Immunohistochemical analysis revealed positive staining for TRPV4 in the endothelium of mesenteric artery sections in both WKY and SHR. Furthermore, TRPV4 mRNA and protein expressions in SHR were significantly lower than their expression levels in WKY rats. We conclude that 4alphaPDD-induced endothelium-dependent NO-mediated vasorelaxation is reduced in SHR and downregulation of TRPV4 could be one of the contributing mechanisms.
Collapse
Affiliation(s)
- A Boudaka
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoud, Sultanate of Oman.
| | | | | | | | | | | |
Collapse
|
64
|
Gilad D, Atiya S, Mozes-Autmazgin Z, Ben-Shushan RS, Ben-David R, Amram E, Tamir S, Chuyun D, Szuchman-Sapir A. Paraoxonase 1 in endothelial cells impairs vasodilation induced by arachidonic acid lactone metabolite. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:386-393. [PMID: 30572120 DOI: 10.1016/j.bbalip.2018.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Paraoxonase 1 (PON1) is a high density lipoprotein (HDL)-associated lactonase, which is known for its antiatherogenic properties. Previous studies in PON1 knockout (PON1KO) mice revealed that PON1KO mice have low blood pressure, which is inversely correlated with the renal levels of the cytochrome P450 -derived arachidonic acid metabolite 5,6-epoxyeicosatrienoic acid (5,6-EET). Our previous studies revealed that 5,6-EET is unstable, transforming to the δ-lactone isomer 5,6-δ-DHTL, an endothelium-derived hyperpolarizing factor (EDHF) that mediates vasodilation, and it is a potential substrate for PON1. AIM To elucidate the role of PON1 in the modulation of vascular resistance via the regulation of the lactone-containing metabolite 5,6-δ-DHTL. RESULTS In mouse resistance arteries, PON1 was found to be present and active in the endothelial layer. Vascular reactivity experiments revealed that 5,6-δ-DHTL dose-dependently dilates PON1KO mouse mesenteric arteries significantly more than wild type (w.t.) resistance arteries. Pre-incubation with HDL or rePON1 reduced 5,6-δ-DHTL-dependent vasodilation. FACS analyses and confocal microscopy experiments revealed that fluorescence-tagged rePON1 penetrates into human endothelial cells' (ECs') in both dose- and time- dependent manner, accumulate in the perinuclear compartment, and retains its lactonase activity in the cells. The presence of rePON1, but not the presence of PON1 loss-of-lactonase-activity mutant, reduced the Ca2+ influx in the ECs mediated by 5,6-δ-DHTL. CONCLUSION PON1 lactonase activity in the endothelium affects vascular dilation by regulating Ca2+ influx mediated by the lactone-containing EDHF 5,6-δ-DHTL.
Collapse
Affiliation(s)
- Dan Gilad
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Shahar Atiya
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Ziv Mozes-Autmazgin
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel
| | - Rotem Shelly Ben-Shushan
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel
| | - Raz Ben-David
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Eytan Amram
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | - Snait Tamir
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | | | - Andrea Szuchman-Sapir
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
65
|
Santisteban MM, Iadecola C. Hypertension, dietary salt and cognitive impairment. J Cereb Blood Flow Metab 2018; 38:2112-2128. [PMID: 30295560 PMCID: PMC6282225 DOI: 10.1177/0271678x18803374] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Abstract
Dementia is growing at an alarming rate worldwide. Although Alzheimer disease is the leading cause, over 50% of individuals diagnosed with Alzheimer disease have vascular lesions at autopsy. There has been an increasing appreciation of the pathogenic role of vascular risk factors in cognitive impairment caused by neurodegeneration. Midlife hypertension is a leading risk factor for late-life dementia. Hypertension alters cerebrovascular structure, impairs the major factors regulating the cerebral microcirculation, and promotes Alzheimer pathology. Experimental studies have identified brain perivascular macrophages as the major free radical source mediating neurovascular dysfunction of hypertension. Recent evidence indicates that high dietary salt may also induce cognitive impairment. Contrary to previous belief, the effect is not necessarily associated with hypertension and is mediated by a deficit in endothelial nitric oxide. Collectively, the evidence suggests a remarkable cellular diversity of the impact of vascular risk factors on the cerebral vasculature and cognition. Whereas long-term longitudinal epidemiological studies are needed to resolve the temporal relationships between vascular risk factors and cognitive dysfunction, single-cell molecular studies of the vasculature in animal models will provide a fuller mechanistic understanding. This knowledge is critical for developing new preventive, diagnostic, and therapeutic approaches for these devastating diseases of the mind.
Collapse
Affiliation(s)
- Monica M Santisteban
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
66
|
Dash JR, Mishra SK, Parida S, Singh TU, Choudhury S, Muniyappa K. TRPV4 activation in rat carotid artery in DOCA hypertension involves eNOS and endothelium-derived contractile factor (EDCF). Clin Exp Hypertens 2018; 41:564-570. [PMID: 30325243 DOI: 10.1080/10641963.2018.1523915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Aim: Role of TRPV4 channel in regulation of endothelial function in the carotid artery in deoxycorticosterone acetate (DOCA) model of hypertension in rat was studied. Methods: 8-10 weeks old albino Wistar rats divided into three groups namely Control, UNX and hypertensive animals. Vascular smooth muscle response was studied in isolated carotid artery of rat with acetylcholine, sodium nitroprusside, GSK1016790A (GSK) in presence and absence of L-NAME and indomethacin. Results: At the end of the 6th week, the mean systolic blood pressure was increased in DOCA-treated hypertensive rats (166 ± 8 mm Hg) compared to Control and UNX (125 ± 5 mm Hg). ACh (10-9 to 10-5 M) produced almost 100% relaxation in Control (Emax = 97.48 ± 1.06 %) and UNX animals (Emax = 93.16 ± 2.33 %) which was attenuated in DOCA-treated hypertensive animals (Emax = 70.85 ± 1.65 %). No significant changes seen in SNP (10-12 to 10-5 M) induced relaxation. GSK1016790A (10-12 to 10-7 M)-mediated relaxation was significantly attenuated in DOCA-treated hypertensive animals (Emax = 25.58 ± 13.60%) compared to the control (Emax = 80.59 ± 6.86%) and UNX (Emax = 87.32 ± 2.01%) animals. L-NAME (10-4 M) potently blocked GSK-induced relaxation, and a contractile response to GSK was observed in presence of L-NAME in all the three groups of animals which was sensitive to indomethacin (10-5 M). Conclusion: TRPV4 may regulate the vascular tone of rat carotid artery through an attenuated NO pathway and stimulation of the release of contractile prostanoids in the DOCA hypertensive rats.
Collapse
Affiliation(s)
- J R Dash
- a Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Bareilly , Uttar Pradesh , India
| | - S K Mishra
- a Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Bareilly , Uttar Pradesh , India
| | - S Parida
- a Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Bareilly , Uttar Pradesh , India
| | - T U Singh
- a Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Bareilly , Uttar Pradesh , India
| | - S Choudhury
- a Division of Pharmacology and Toxicology, Indian Veterinary Research Institute, Bareilly , Uttar Pradesh , India
| | - K Muniyappa
- b Department of Biochemistry, Indian 5 Institute of Science , Bangalore , India
| |
Collapse
|
67
|
Zhang B, Paffett ML, Naik JS, Jernigan NL, Walker BR, Resta TC. Cholesterol Regulation of Pulmonary Endothelial Calcium Homeostasis. CURRENT TOPICS IN MEMBRANES 2018; 82:53-91. [PMID: 30360783 DOI: 10.1016/bs.ctm.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cholesterol is a key structural component and regulator of lipid raft signaling platforms critical for cell function. Such regulation may involve changes in the biophysical properties of lipid microdomains or direct protein-sterol interactions that alter the function of ion channels, receptors, enzymes, and membrane structural proteins. Recent studies have implicated abnormal membrane cholesterol levels in mediating endothelial dysfunction that is characteristic of pulmonary hypertensive disorders, including that resulting from long-term exposure to hypoxia. Endothelial dysfunction in this setting is characterized by impaired pulmonary endothelial calcium entry and an associated imbalance that favors production vasoconstrictor and mitogenic factors that contribute to pulmonary hypertension. Here we review current knowledge of cholesterol regulation of pulmonary endothelial Ca2+ homeostasis, focusing on the role of membrane cholesterol in mediating agonist-induced Ca2+ entry and its components in the normal and hypertensive pulmonary circulation.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Michael L Paffett
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States.
| |
Collapse
|
68
|
Santos SE, Ribeiro FPRA, Menezes PMN, Duarte-Filho LAM, Quintans JSS, Quintans-Junior LJ, Silva FS, Ribeiro LAA. New insights on relaxant effects of (-)-borneol monoterpene in rat aortic rings. Fundam Clin Pharmacol 2018; 33:148-158. [PMID: 30240490 DOI: 10.1111/fcp.12417] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/31/2018] [Accepted: 09/18/2018] [Indexed: 11/28/2022]
Abstract
The monoterpene alcohol (-)-borneol has many biological effects such as sedative, anti-inflammatory, analgesic, anti-nociceptive, antithrombotic and vasorelaxant effects. Our objective in this study was to investigate the mechanism of action of (-)-borneol and determine its vasorelaxant effect. (-)-Borneol was tested on isolated aortic rings contracted with PE (10-6 m). This study was performed in the absence or in the presence of endothelium, L-NAME (100 μm), indomethacin (10 μm), TEA (1 and 10 mm), 4-AP (1 mm) or glibenclamide (1 mm) to assess the participation of EDRF, nitric oxide, prostanoids and potassium channels on the relaxing effect of (-)-borneol. In this work, (-)-borneol induced a relaxant effect in aortic rings, with and without endothelium, in a concentration-dependent manner. The pharmacological characterization obtained using L-NAME, indomethacin, TEA, 4-AP and glibenclamide demonstrates that the effect of (-)-borneol was modified in the presence of L-NAME, indomethacin and glibenclamide showing that these signal transduction pathways are involved in the relaxing effect of the monoterpene. (-)-Borneol has a vasorelaxant effect that depends on the presence of vascular endothelium, with the participation of nitric oxide and prostanoids. Also, (-)-borneol displayed a direct action on the vascular smooth muscle, greatly dependent on KATP channels.
Collapse
Affiliation(s)
- Stefânia E Santos
- Post-Graduate Program in Semi-Arid Natural Resources (PGRNSA), University of San Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, S/N, Centro, Petrolina, PE, 56304-917, Brazil
| | - Fernanda P R A Ribeiro
- Post-Graduate Program in Semi-Arid Natural Resources (PGRNSA), University of San Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, S/N, Centro, Petrolina, PE, 56304-917, Brazil
| | - Pedro M N Menezes
- Post-Graduate Program in Semi-Arid Natural Resources (PGRNSA), University of San Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, S/N, Centro, Petrolina, PE, 56304-917, Brazil
| | - Luiz A M Duarte-Filho
- Laboratory of Experimental Pharmacology (LAFEX), University of San Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, S/N, Centro, Petrolina, PE, 56304-917, Brazil
| | - Jullyana S S Quintans
- Department of Physiology (DFS), Federal University of Sergipe, Av. Marechal Rondon, S/N, São Cristóvão, SE, 49100-000, Brazil
| | - Lucindo J Quintans-Junior
- Department of Physiology (DFS), Federal University of Sergipe, Av. Marechal Rondon, S/N, São Cristóvão, SE, 49100-000, Brazil
| | - Fabricio S Silva
- College of Pharmaceutical Sciences (CFARM), University of San Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, S/N, Centro, Petrolina, PE, 56304-917, Brazil
| | - Luciano A A Ribeiro
- College of Pharmaceutical Sciences (CFARM), University of San Francisco Valley (UNIVASF), Av. José de Sá Maniçoba, S/N, Centro, Petrolina, PE, 56304-917, Brazil
| |
Collapse
|
69
|
Nafisa A, Gray SG, Cao Y, Wang T, Xu S, Wattoo FH, Barras M, Cohen N, Kamato D, Little PJ. Endothelial function and dysfunction: Impact of metformin. Pharmacol Ther 2018; 192:150-162. [PMID: 30056057 DOI: 10.1016/j.pharmthera.2018.07.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular and metabolic diseases remain the leading cause of morbidity and mortality worldwide. Endothelial dysfunction is a key player in the initiation and progression of cardiovascular and metabolic diseases. Current evidence suggests that the anti-diabetic drug metformin improves insulin resistance and protects against endothelial dysfunction in the vasculature. Hereby, we provide a timely review on the protective effects and molecular mechanisms of metformin in preventing endothelial dysfunction and cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Asma Nafisa
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Susan G Gray
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Yingnan Cao
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| | - Tinghuai Wang
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Feroza H Wattoo
- Department of Biochemistry, PMAS Arid Agriculture University, Shamasabad, Muree Road, Rawalpindi 4600, Pakistan..
| | - Michael Barras
- Dept. of Pharmacy, Princess Alexandra Hospital, 199 Ipswich Rd, Woolloongabba, QLD 4102, Australia.
| | - Neale Cohen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia.
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| |
Collapse
|
70
|
Cheng Y, Vanhoutte PM, Leung SWS. Apolipoprotein E favours the blunting by high-fat diet of prostacyclin receptor activation in the mouse aorta. Br J Pharmacol 2018; 175:3453-3469. [PMID: 29859010 DOI: 10.1111/bph.14386] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE NO-mediated, endothelium-dependent relaxations of isolated arteries are blunted by ageing and high-fat diets, as well as by apolipoprotein E deletion. The present study was designed to test the hypothesis that apolipoprotein E deletion impairs endothelium-dependent responses to prostacyclin (IP) receptor activation. EXPERIMENTAL APPROACH Five-week-old ApoE+/+ and ApoE-/- mice were fed normal chow or high-fat diet for 29 weeks. The aortae were isolated for the measurements of isometric tension in Halpern-Mulvany myographs. Levels of proteins were assessed by Western blotting and immunofluorescence, and cyclic nucleotide levels by elisa. KEY RESULTS The IP receptor agonist, iloprost, induced endothelium-, NO-synthase- and IP-dependent relaxations in aortae of young ApoE+/+ mice. High-fat diet favoured activation of thromboxane receptors by iloprost, causing contraction. Apolipoprotein E was present in aortae of ApoE+/+ mice, especially in endothelium. Its presence was augmented by high-fat diet. Its deletion potentiated iloprost-induced relaxations in aortae of young mice and prevented the blunting of this response by high-fat diet. Levels of cAMP were higher, but those of cGMP were lower in the aorta of ApoE-/- than in ApoE+/+ mice of the same age. The levels of IP receptor protein were not different between ApoE+/+ and ApoE-/- mice. CONCLUSIONS AND IMPLICATIONS Iloprost induced an endothelium-dependent relaxation in the aorta of young healthy mice which involved both the cGMP and cAMP pathways. This response was blunted by prolonged exposure to a high-fat diet. Apolipoprotein E deletion potentiated relaxations to IP receptor activation, independently of age and diet.
Collapse
Affiliation(s)
- Yanhua Cheng
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, SAR, China
| | - Paul M Vanhoutte
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, SAR, China
| | - Susan W S Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
71
|
He D, Pan Q, Chen Z, Sun C, Zhang P, Mao A, Zhu Y, Li H, Lu C, Xie M, Zhou Y, Shen D, Tang C, Yang Z, Jin J, Yao X, Nilius B, Ma X. Treatment of hypertension by increasing impaired endothelial TRPV4-KCa2.3 interaction. EMBO Mol Med 2018; 9:1491-1503. [PMID: 28899928 PMCID: PMC5666316 DOI: 10.15252/emmm.201707725] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The currently available antihypertensive agents have undesirable adverse effects due to systemically altering target activity including receptors, channels, and enzymes. These effects, such as loss of potassium ions induced by diuretics, bronchospasm by beta‐blockers, constipation by Ca2+ channel blockers, and dry cough by ACEI, lead to non‐compliance with therapies (Moser, 1990). Here, based on new hypertension mechanisms, we explored a new antihypertensive approach. We report that transient receptor potential vanilloid 4 (TRPV4) interacts with Ca2+‐activated potassium channel 3 (KCa2.3) in endothelial cells (ECs) from small resistance arteries of normotensive humans, while ECs from hypertensive patients show a reduced interaction between TRPV4 and KCa2.3. Murine hypertension models, induced by high‐salt diet, N(G)‐nitro‐l‐arginine intake, or angiotensin II delivery, showed decreased TRPV4‐KCa2.3 interaction in ECs. Perturbation of the TRPV4‐KCa2.3 interaction in mouse ECs by overexpressing full‐length KCa2.3 or defective KCa2.3 had hypotensive or hypertensive effects, respectively. Next, we developed a small‐molecule drug, JNc‐440, which showed affinity for both TRPV4 and KCa2.3. JNc‐440 significantly strengthened the TRPV4‐KCa2.3 interaction in ECs, enhanced vasodilation, and exerted antihypertensive effects in mice. Importantly, JNc‐440 specifically targeted the impaired TRPV4‐KCa2.3 interaction in ECs but did not systemically activate TRPV4 and KCa2.3. Together, our data highlight the importance of impaired endothelial TRPV4‐KCa2.3 coupling in the progression of hypertension and suggest a novel approach for antihypertensive drug development.
Collapse
Affiliation(s)
- Dongxu He
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Qiongxi Pan
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Zhen Chen
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chunyuan Sun
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Peng Zhang
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Aiqin Mao
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Yaodan Zhu
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Hongjuan Li
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chunxiao Lu
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Mingxu Xie
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Yin Zhou
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Daoming Shen
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Zhenyu Yang
- Heart Centre, Wuxi People's Hospital, Wuxi, China
| | - Jian Jin
- School of Medicine, Jiangnan University, Wuxi, China.,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Bernd Nilius
- Department Cell Mol Medicine Laboratory Ion Channel Research Campus Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Xin Ma
- School of Medicine, Jiangnan University, Wuxi, China .,National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
72
|
Nokkari A, Abou-El-Hassan H, Mechref Y, Mondello S, Kindy MS, Jaffa AA, Kobeissy F. Implication of the Kallikrein-Kinin system in neurological disorders: Quest for potential biomarkers and mechanisms. Prog Neurobiol 2018; 165-167:26-50. [PMID: 29355711 PMCID: PMC6026079 DOI: 10.1016/j.pneurobio.2018.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.
Collapse
Affiliation(s)
- Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mark S Kindy
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA; James A. Haley VA Medical Center, Tampa, FL, USA
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Department of Medicine, Medical University of South, Charleston, SC, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Center for Neuroproteomics & Biomarkers Research, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
73
|
Vassallo DV, Almenara CCP, Broseghini-Filho GB, Teixeira AC, da Silva DCF, Angeli JK, Padilha AS. Preliminary Studies of Acute Cadmium Administration Effects on the Calcium-Activated Potassium (SKCa and BKCa) Channels and Na +/K +-ATPase Activity in Isolated Aortic Rings of Rats. Biol Trace Elem Res 2018; 183:325-334. [PMID: 28905315 DOI: 10.1007/s12011-017-1150-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/31/2017] [Indexed: 12/19/2022]
Abstract
Cadmium is an environmental pollutant closely linked with cardiovascular diseases that seems to involve endothelium dysfunction and reduced nitric oxide (NO) bioavailability. Knowing that NO causes dilatation through the activation of potassium channels and Na+/K+-ATPase, we aimed to determine whether acute cadmium administration (10 μM) alters the participation of K+ channels, voltage-activated calcium channel, and Na+/K+-ATPase activity in vascular function of isolated aortic rings of rats. Cadmium did not modify the acetylcholine-induced relaxation. After L-NAME addition, the relaxation induced by acetylcholine was abolished in presence or absence of cadmium, suggesting that acutely, this metal did not change NO release. However, tetraethylammonium (a nonselective K+ channels blocker) reduced acetylcholine-induced relaxation but this effect was lower in the preparations with cadmium, suggesting a decrease of K+ channels function in acetylcholine response after cadmium incubation. Apamin (a selective blocker of small Ca2+-activated K+ channels-SKCa), iberiotoxin (a selective blocker of large-conductance Ca2+-activated K+ channels-BKCa), and verapamil (a blocker of calcium channel) reduced the endothelium-dependent relaxation only in the absence of cadmium. Finally, cadmium decreases Na+/K+-ATPase activity. Our results provide evidence that the cadmium acute incubation unaffected the calcium-activated potassium channels (SKCa and BKCa) and voltage-calcium channels on the acetylcholine vasodilatation. In addition, acute cadmium incubation seems to reduce the Na+/K+-ATPase activity.
Collapse
Affiliation(s)
- Dalton V Vassallo
- Federal University of Espírito Santo, Graduate Program in Physiological Sciences, Vitória, ES, Brazil
| | - Camila C P Almenara
- Federal University of Espírito Santo, Graduate Program in Physiological Sciences, Vitória, ES, Brazil
| | | | - Ariane Calazans Teixeira
- Federal University of Espírito Santo, Graduate Program in Physiological Sciences, Vitória, ES, Brazil
| | - David Chaves F da Silva
- Federal University of Espírito Santo, Graduate Program in Physiological Sciences, Vitória, ES, Brazil
| | - Jhuli K Angeli
- Federal University of Espírito Santo, Graduate Program in Physiological Sciences, Vitória, ES, Brazil
| | - Alessandra S Padilha
- Federal University of Espírito Santo, Graduate Program in Physiological Sciences, Vitória, ES, Brazil.
- Programa de Pós-Graduação em Ciências Fisiológicas, CCS/UFES, Av. Marechal Campos, 1468. Maruípe, Vitoria, ES, 29040-091, Brazil.
| |
Collapse
|
74
|
Bautista-Niño PK, van der Stel M, Batenburg WW, de Vries R, Roks AJ, Danser AJ. Endothelium-derived hyperpolarizing factor and protein kinase G Iα activation: H 2 O 2 versus S -nitrosothiols. Eur J Pharmacol 2018; 827:112-116. [DOI: 10.1016/j.ejphar.2018.03.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 01/18/2023]
|
75
|
El Assar M, Fernández A, Sánchez-Ferrer A, Angulo J, Rodríguez-Mañas L. Multivessel analysis of progressive vascular aging in the rat: Asynchronous vulnerability among vascular territories. Mech Ageing Dev 2018; 173:39-49. [PMID: 29605639 DOI: 10.1016/j.mad.2018.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 02/26/2018] [Accepted: 03/26/2018] [Indexed: 12/31/2022]
Abstract
Aging induces vascular dysfunction, representing the major risk factor for cardiovascular disease. Our aim was to ascertain specific vulnerability of vascular territories to aging by evaluating the progressive impact of aging on vascular function in four different vascular beds: aorta, mesenteric artery (MA), coronary artery (CA), and penile corpus cavernosum (CC) from 3, 6, 9, 12, 20 or 24 months-old male rats. Contractile/relaxant responses were evaluated in organ chambers (A/CC) and wire myographs (MA/CA). Relationships of systemic biomarkers with endothelial function impairment were also determined. Although all vessels manifested aging-related impairment in endothelial vasodilation, CA was the most impacted by aging considering the onset (at 6 months) and magnitude of endothelial dysfunction (reduction by 1.5 log units in the concentration required for 50% of maximal relaxation for acetylcholine). H2O2-induced vasodilations were progressively reduced by aging in aorta, CC and CA while NO-donor-induced vasodilation was impaired by aging only in CA. Serum asymmetric dimethylarginine significantly correlated to endothelial decline in aorta, MA, and CC, while HOMA-IR was significantly associated with endothelial dysfunction in CA and MA. CA are especially vulnerable to aging-related vascular dysfunction. Correlations of vascular dysfunction with systemic biomarkers differ among vessels, further suggesting heterogeneity in aging-induced vascular impact.
Collapse
Affiliation(s)
- Mariam El Assar
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain
| | - Argentina Fernández
- Servicio de Histología, Unidad de Investigación Cardiovascular (IRYCIS/UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alberto Sánchez-Ferrer
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain
| | - Javier Angulo
- Servicio de Histología, Unidad de Investigación Cardiovascular (IRYCIS/UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Leocadio Rodríguez-Mañas
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain; Servicio de Geriatría, Hospital Universitario de Getafe, Getafe, Spain.
| |
Collapse
|
76
|
Harder DR, Rarick KR, Gebremedhin D, Cohen SS. Regulation of Cerebral Blood Flow: Response to Cytochrome P450 Lipid Metabolites. Compr Physiol 2018; 8:801-821. [PMID: 29687906 DOI: 10.1002/cphy.c170025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There have been numerous reviews related to the cerebral circulation. Most of these reviews are similar in many ways. In the present review, we thought it important to provide an overview of function with specific attention to details of cerebral arterial control related to brain homeostasis, maintenance of neuronal energy demands, and a unique perspective related to the role of astrocytes. A coming review in this series will discuss cerebral vascular development and unique properties of the neonatal circulation and developing brain, thus, many aspects of development are missing here. Similarly, a review of the response of the brain and cerebral circulation to heat stress has recently appeared in this series (8). By trying to make this review unique, some obvious topics were not discussed in lieu of others, which are from recent and provocative research such as endothelium-derived hyperpolarizing factor, circadian regulation of proteins effecting cerebral blood flow, and unique properties of the neurovascular unit. © 2018 American Physiological Society. Compr Physiol 8:801-821, 2018.
Collapse
Affiliation(s)
- David R Harder
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin, USA
| | - Kevin R Rarick
- Department of Pediatrics, Division of Critical Care, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Debebe Gebremedhin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Susan S Cohen
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
77
|
Abstract
PURPOSE OF REVIEW Although the roles of oxidant stress and redox perturbations in hypertension have been the subject of several reviews, role of thioredoxin (Trx), a major cellular redox protein in age-related hypertension remains inadequately reviewed. The purpose of this review is to bring readers up-to-date with current understanding of the role of thioredoxin in age-related hypertension. RECENT FINDINGS Age-related hypertension is a major underlying cause of several cardiovascular disorders, and therefore, intensive management of blood pressure is indicated in most patients with cardiovascular complications. Recent studies have shown that age-related hypertension was reversed and remained lowered for a prolonged period in mice with higher levels of human Trx (Trx-Tg). Additionally, injection of human recombinant Trx (rhTrx) decreased hypertension in aged wild-type mice that lasted for several days. Both Trx-Tg and aged wild-type mice injected with rhTrx were normotensive, showed increased NO production, decreased arterial stiffness, and increased vascular relaxation. These studies suggest that rhTrx could potentially be a therapeutic molecule to reverse age-related hypertension in humans. The reversal of age-related hypertension by restoring proteins that have undergone age-related modification is conceptually novel in the treatment of hypertension. Trx reverses age-related hypertension via maintaining vascular redox homeostasis, regenerating critical vasoregulatory proteins oxidized due to advancing age, and restoring native function of proteins that have undergone age-related modifications with loss-of function. Recent studies demonstrate that Trx is a promising molecule that may ameliorate or reverse age-related hypertension in older adults.
Collapse
Affiliation(s)
- Kumuda C Das
- Department of Translational and Vascular Biology, University of Texas Health Sciences Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA.
| | - Venkatesh Kundumani-Sridharan
- Department of Translational and Vascular Biology, University of Texas Health Sciences Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA
| | - Jaganathan Subramani
- Department of Translational and Vascular Biology, University of Texas Health Sciences Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA
| |
Collapse
|
78
|
Cheng Z, Shen X, Jiang X, Shan H, Cimini M, Fang P, Ji Y, Park JY, Drosatos K, Yang X, Kevil CG, Kishore R, Wang H. Hyperhomocysteinemia potentiates diabetes-impaired EDHF-induced vascular relaxation: Role of insufficient hydrogen sulfide. Redox Biol 2018. [PMID: 29524844 PMCID: PMC5854893 DOI: 10.1016/j.redox.2018.02.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Insufficient hydrogen sulfide (H2S) has been implicated in Type 2 diabetic mellitus (T2DM) and hyperhomocysteinemia (HHcy)-related cardiovascular complications. We investigated the role of H2S in T2DM and HHcy-induced endothelial dysfunction in small mesenteric artery (SMA) of db/db mice fed a high methionine (HM) diet. HM diet (8 weeks) induced HHcy in both T2DM db/db mice and non-diabetic db/+ mice (total plasma Hcy: 48.4 and 31.3 µM, respectively), and aggravated the impaired endothelium-derived hyperpolarization factor (EDHF)-induced endothelium-dependent relaxation to acetylcholine (ACh), determined by the presence of eNOS inhibitor N(ω)-nitro-L-arginine methyl ester (L-NAME) and prostacyclin (PGI2) inhibitor indomethacin (INDO), in SMA from db/db mice but not that from db/+ mice. A non-selective Ca2+-active potassium channel (KCa) opener NS309 rescued T2DM/HHcy-impaired EDHF-mediated vascular relaxation to ACh. EDHF-induced relaxation to ACh was inhibited by a non-selective KCa blocker TEA and intermediate-conductance KCa blocker (IKCa) Tram-34, but not by small-conductance KCa (SKCa) blocker Apamin. HHcy potentiated the reduction of free sulfide, H2S and cystathionine γ-lyase protein, which converts L-cysteine to H2S, in SMA of db/db mice. Importantly, a stable H2S donor DATS diminished the enhanced O2- production in SMAs and lung endothelial cells of T2DM/HHcy mice. Antioxidant PEG-SOD and DATS improved T2DM/HHcy impaired relaxation to ACh. Moreover, HHcy increased hyperglycemia-induced IKCa tyrosine nitration in human micro-vascular endothelial cells. EDHF-induced vascular relaxation to L-cysteine was not altered, whereas such relaxation to NaHS was potentiated by HHcy in SMA of db/db mice which was abolished by ATP-sensitive potassium channel blocker Glycolamide but not by KCa blockers. Conclusions Intermediate HHcy potentiated H2S reduction via CSE-downregulation in microvasculature of T2DM mice. H2S is justified as an EDHF. Insufficient H2S impaired EDHF-induced vascular relaxation via oxidative stress and IKCa inactivation in T2DM/HHcy mice. H2S therapy may be beneficial for prevention and treatment of micro-vascular complications in patients with T2DM and HHcy.
Collapse
Affiliation(s)
- Zhongjian Cheng
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA.
| | - Xinggui Shen
- Center for Cardiovascular Diseases and Sciences, Department of Pathology, Molecular and Cellular Physiology and Cell Biology and Anatomy Louisiana State University Health Sciences Center-Shreveport, New Orleans, LA 7110371103, USA
| | - Xiaohua Jiang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Huimin Shan
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Maria Cimini
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Pu Fang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Joon Young Park
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Konstantinos Drosatos
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Christopher G Kevil
- Center for Cardiovascular Diseases and Sciences, Department of Pathology, Molecular and Cellular Physiology and Cell Biology and Anatomy Louisiana State University Health Sciences Center-Shreveport, New Orleans, LA 7110371103, USA
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
79
|
Christou H, Hudalla H, Michael Z, Filatava EJ, Li J, Zhu M, Possomato-Vieira JS, Dias-Junior C, Kourembanas S, Khalil RA. Impaired Pulmonary Arterial Vasoconstriction and Nitric Oxide-Mediated Relaxation Underlie Severe Pulmonary Hypertension in the Sugen-Hypoxia Rat Model. J Pharmacol Exp Ther 2018; 364:258-274. [PMID: 29212831 PMCID: PMC5774216 DOI: 10.1124/jpet.117.244798] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Pulmonary vasoreactivity could determine the responsiveness to vasodilators and, in turn, the prognosis of pulmonary hypertension (PH). We hypothesized that pulmonary vasoreactivity is impaired, and we examined the underlying mechanisms in the Sugen-hypoxia rat model of severe PH. Male Sprague-Dawley rats were injected with Sugen (20 mg/kg s.c.) and exposed to hypoxia (9% O2) for 3 weeks, followed by 4 weeks in normoxia (Su/Hx), or treated with Sugen alone (Su) or hypoxia alone (Hx) or neither (Nx). After hemodynamic measurements, the heart was assessed for right ventricular hypertrophy (Fulton's index); the pulmonary artery, aorta, and mesenteric arteries were isolated for vascular function studies; and contractile markers were measured in pulmonary arteries using quantitative polymerase chain reaction (PCR). Other rats were used for morphometric analysis of pulmonary vascular remodeling. Right ventricular systolic pressure and Fulton's index were higher in Su/Hx versus Su, Hx, and Nx rats. Pulmonary vascular remodeling was more prominent in Su/Hx versus Nx rats. In pulmonary artery rings, contraction to high KCl (96 mM) was less in Su/Hx versus Nx and Su, and phenylephrine-induced contraction was reduced in Su/Hx versus Nx, Hx, and Su. Acetylcholine (ACh)-induced relaxation was less in Su/Hx versus Nx and Hx, suggesting reduced endothelium-dependent vasodilation. ACh relaxation was inhibited by nitric oxide synthase (NOS) and guanylate cyclase blockade in all groups, suggesting a role of the NO-cGMP pathway. Nitrate/nitrite production in response to ACh was less in Su/Hx versus Nx, supporting reduced endothelial NO production. Sodium nitroprusside (10-8 M) caused less relaxation in Su/Hx versus Nx, Hx, and Su, suggesting a decreased responsiveness of vascular smooth muscle (VSM) to vasodilators. Neither contraction nor relaxation differed in the aorta or mesenteric arteries of all groups. PCR analysis showed decreased expression of contractile markers in pulmonary artery of Su/Hx versus Nx. The reduced responsiveness to vasoconstrictors and NO-mediated vasodilation in the pulmonary, but not systemic, vessels may be an underlying mechanism of severe PH in Su/Hx rats and appears to involve attenuation of the NO relaxation pathway and a switch of pulmonary VSM cells to a synthetic less reactive phenotype.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Hannes Hudalla
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Zoe Michael
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Evgenia J Filatava
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Jun Li
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Minglin Zhu
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Jose S Possomato-Vieira
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Carlos Dias-Junior
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Stella Kourembanas
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Raouf A Khalil
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| |
Collapse
|
80
|
|
81
|
Hauck M, Noronha Martins C, Borges Moraes M, Aikawa P, da Silva Paulitsch F, Méa Plentz RD, Teixeira da Costa S, Vargas da Silva AM, Signori LU. Comparison of the effects of 1MHz and 3MHz therapeutic ultrasound on endothelium-dependent vasodilation of humans: a randomised clinical trial. Physiotherapy 2017; 105:120-125. [PMID: 29373113 DOI: 10.1016/j.physio.2017.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/17/2017] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To compare the effects of different waveforms of 1MHz and 3MHz therapeutic ultrasound on endothelial function in healthy subjects. DESIGN Randomised placebo-controlled, crossover study with concealed allocation and assessor blinding. SETTING Imaging Centre of the University Hospital. PARTICIPANTS Thirty volunteers aged between 18 and 35 years were divided into two homogeneous groups (1MHz and 3MHz). INTERVENTIONS Continuous (CUT; 0.4W/cm2SATA), pulsed (PUT; 20% duty cycle, 0.08W/cm2SATA) and placebo waveforms (equipment off) of ultrasound (1MHz and 3MHz) were randomized and applied over the brachial artery for 5minutes. MAIN OUTCOME MEASURES Endothelial function was evaluated using the flow-mediated dilation (FMD) technique. RESULTS Both 1MHz [CUT: mean difference 4%, 95% confidence interval (CI) 2 to 6%, P<0.001; PUT: mean difference 4%, 95% CI 2 to 6%, P<0.001] and 3MHz (CUT: mean difference 4%, 95% CI 2 to 6%, P<0.001; PUT: mean difference 4%, 95% CI 2 to 6%, P<0.001) of therapeutic ultrasound increased %FMD by approximately 4% compared with the placebo waveforms. The endothelium-dependent vasodilator responses were the same for both types of waves and frequencies. No differences in baseline diameter, hyperaemic flow, and nitroglycerin-mediated diameter and vasodilation were observed between groups. CONCLUSION Both CUT and PUT ultrasound waveforms improved endothelial function. The 1MHz and 3MHz frequencies of therapeutic ultrasound led to similar improvement in endothelial function in healthy volunteers. Clinical trial registration number RBR-4z5z3t.
Collapse
Affiliation(s)
- M Hauck
- Institute of Biological Sciences, Federal University of Rio Grande, RS, Brazil
| | - C Noronha Martins
- Institute of Biological Sciences, Federal University of Rio Grande, RS, Brazil
| | - M Borges Moraes
- Institute of Biological Sciences, Federal University of Rio Grande, RS, Brazil
| | - P Aikawa
- Institute of Biological Sciences, Federal University of Rio Grande, RS, Brazil
| | | | - R Della Méa Plentz
- Graduate Programme in Healthy Sciences, Federal University of Health Sciences of Porto Alegre, RS, Brazil
| | - S Teixeira da Costa
- Graduate Programme in Physiotherapy and Rehabilitation, Federal University of Santa Maria, RS, Brazil
| | - A M Vargas da Silva
- Graduate Programme in Physiotherapy and Rehabilitation, Federal University of Santa Maria, RS, Brazil
| | - L U Signori
- Institute of Biological Sciences, Federal University of Rio Grande, RS, Brazil; Graduate Programme in Physiotherapy and Rehabilitation, Federal University of Santa Maria, RS, Brazil.
| |
Collapse
|
82
|
Experimental preeclampsia in rats affects vascular gene expression patterns. Sci Rep 2017; 7:14807. [PMID: 29093568 PMCID: PMC5665945 DOI: 10.1038/s41598-017-14926-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/06/2017] [Indexed: 01/03/2023] Open
Abstract
Normal pregnancy requires adaptations of the maternal vasculature. During preeclampsia these adaptations are not well established, which may be related to maternal hypertension and proteinuria. The effects of preeclampsia on the maternal vasculature are not yet fully understood. We aimed to evaluate gene expression in aortas of pregnant rats with experimental preeclampsia using a genome wide microarray. Aortas were isolated from pregnant Wistar outbred rats with low-dose LPS-induced preeclampsia (ExpPE), healthy pregnant (Pr), non-pregnant and low-dose LPS-infused non-pregnant rats. Gene expression was measured by microarray and validated by real-time quantitative PCR. Gene Set Enrichment Analysis was performed to compare the groups. Functional analysis of the aorta was done by isotonic contraction measurements while stimulating aortic rings with potassium chloride. 526 genes were differentially expressed, and positive enrichment of “potassium channels”, “striated muscle contraction”, and “neuronal system” gene sets were found in ExpPE vs. Pr. The potassium chloride-induced contractile response of ExpPE aortic rings was significantly decreased compared to this response in Pr animals. Our data suggest that potassium channels, neuronal system and (striated) muscle contraction in the aorta may play a role in the pathophysiology of experimental preeclampsia. Whether these changes are also present in preeclamptic women needs further investigation.
Collapse
|
83
|
Lapi D, Mastantuono T, Di Maro M, Varanini M, Colantuoni A. Low-Frequency Components in Rat Pial Arteriolar Rhythmic Diameter Changes. J Vasc Res 2017; 54:344-358. [PMID: 29065409 DOI: 10.1159/000478984] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 06/21/2017] [Indexed: 01/04/2023] Open
Abstract
This study aimed to analyze the frequency components present in spontaneous rhythmic diameter changes in rat pial arterioles. Pial microcirculation was visualized by fluorescence microscopy. Rhythmic luminal variations were evaluated via computer-assisted methods. Spectral analysis was carried out on 30-min recordings under baseline conditions and after administration of acetylcholine (Ach), papaverine (Pap), Nω-nitro-L-arginine (L-NNA) prior to Ach, indomethacin (INDO), INDO prior to Ach, charybdotoxin and apamin, and charybdotoxin and apamin prior to Ach. Under baseline conditions all arteriolar orders showed 3 frequency components in the ranges of 0.0095-0.02, 0.02-0.06, and 0.06-0.2 Hz, another 2 in the ranges of 0.2-2.0 and 2.5-4.5 Hz, and another ultra-low-frequency component in the range of 0.001-0.0095 Hz. Ach caused a significant increase in the spectral density of the frequency components in the range of 0.001-0.2 Hz. Pap was able to slightly increase spectral density in the ranges of 0.001-0.0095 and 0.0095-0.02 Hz. L-NNA mainly attenuated arteriolar responses to Ach. INDO prior to Ach did not affect the endothelial response to Ach. Charybdotoxin and apamin, suggested as endothelium-derived hyperpolarizing factor inhibitors, reduced spectral density in the range of 0.001-0.0095 Hz before and after Ach administration. In conclusion, regulation of the blood flow distribution is due to several mechanisms, one of which is affected by charibdotoxin and apamin, modulating the vascular tone.
Collapse
Affiliation(s)
- Dominga Lapi
- Department of Clinical Medicine and Surgery, Federico II University Medical School, Naples, Italy
| | | | | | | | | |
Collapse
|
84
|
Zaborska KE, Wareing M, Austin C. Comparisons between perivascular adipose tissue and the endothelium in their modulation of vascular tone. Br J Pharmacol 2017; 174:3388-3397. [PMID: 27747871 PMCID: PMC5610163 DOI: 10.1111/bph.13648] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/16/2016] [Accepted: 09/28/2016] [Indexed: 01/06/2023] Open
Abstract
The endothelium is an established modulator of vascular tone; however, the recent discovery of the anti-contractile nature of perivascular adipose tissue (PVAT) suggests that the fat, which surrounds many blood vessels, can also modulate vascular tone. Both the endothelium and PVAT secrete vasoactive substances, which regulate vascular function. Many of these factors are common to both the endothelium and PVAT; therefore, this review will highlight the potential shared mechanisms in the modulation of vascular tone. Endothelial dysfunction is a hallmark of many vascular diseases, including hypertension and obesity. Moreover, PVAT dysfunction is now being reported in several cardio-metabolic disorders. Thus, this review will also discuss the mechanistic insights into endothelial and PVAT dysfunction in order to evaluate whether PVAT modulation of vascular contractility is similar to that of the endothelium in health and disease. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- K E Zaborska
- Institute of Cardiovascular SciencesUniversity of ManchesterUK
| | - M Wareing
- Maternal and Fetal Health Research Centre, Institute of Human DevelopmentUniversity of ManchesterUK
| | - C Austin
- Faculty of Health and Social CareEdge Hill UniversityUK
| |
Collapse
|
85
|
Roles of Nitric Oxide and Prostaglandins in the Sustained Antihypertensive Effects of Acanthospermum hispidum DC. on Ovariectomized Rats with Renovascular Hypertension. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:2492483. [PMID: 29234376 PMCID: PMC5632478 DOI: 10.1155/2017/2492483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/10/2017] [Accepted: 08/16/2017] [Indexed: 01/30/2023]
Abstract
Although Acanthospermum hispidum is used in Brazilian folk medicine as an antihypertensive, no study evaluated its effects on a renovascular hypertension and ovariectomy model. So, this study investigated the mechanisms involved in the antihypertensive effects of an ethanol-soluble fraction obtained from A. hispidum (ESAH) using two-kidney-one-clip hypertension in ovariectomized rats (2K1C plus OVT). ESAH was orally administered at doses of 30, 100, and 300 mg/kg, daily, for 28 days, after 5 weeks of surgery. Enalapril (15 mg/kg) and hydrochlorothiazide (25 mg/kg) were used as standard drugs. Diuretic activity was evaluated on days 1, 7, 14, 21, and 28. Systolic, diastolic, and mean blood pressure and heart rate were recorded. Serum creatinine, urea, thiobarbituric acid reactive substances, nitrosamine, nitrite, aldosterone, vasopressin levels, and ACE activity were measured. The vascular reactivity and the role of nitric oxide (NO) and prostaglandins (PG) in the vasodilator response of ESAH on the mesenteric vascular bed (MVB) were also investigated. ESAH treatment induced an important saluretic and antihypertensive response, therefore recovering vascular reactivity in 2K1C plus OVT-rats. This effect was associated with a reduction of oxidative and nitrosative stress with a possible increase in the NO bioavailability. Additionally, a NO and PG-dependent vasodilator effect was observed on the MEV.
Collapse
|
86
|
Wang X, Khalil RA. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:241-330. [PMID: 29310800 DOI: 10.1016/bs.apha.2017.08.002] [Citation(s) in RCA: 388] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that degrade various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation through removal of the propeptide domain from their latent zymogen form. MMPs are often secreted in an inactive proMMP form, which is cleaved to the active form by various proteinases including other MMPs. MMPs degrade various protein substrates in ECM including collagen and elastin. MMPs could also influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in vascular tissue remodeling during various biological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension, preeclampsia, atherosclerosis, aneurysm formation, as well as excessive venous dilation and lower extremity venous disease. MMPs are often regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs may serve as biomarkers and potential therapeutic targets for certain vascular disorders.
Collapse
Affiliation(s)
- Xi Wang
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
87
|
Lu T, Wang XL, Chai Q, Sun X, Sieck GC, Katusic ZS, Lee HC. Role of the endothelial caveolae microdomain in shear stress-mediated coronary vasorelaxation. J Biol Chem 2017; 292:19013-19023. [PMID: 28924052 DOI: 10.1074/jbc.m117.786152] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
In this study, we determined the role of caveolae and the ionic mechanisms that mediate shear stress-mediated vasodilation (SSD). We found that both TRPV4 and SK channels are targeted to caveolae in freshly isolated bovine coronary endothelial cells (BCECs) and that TRPV4 and KCa2.3 (SK3) channels are co-immunoprecipitated by anti-caveolin-1 antibodies. Acute exposure of BCECs seeded in a capillary tube to 10 dynes/cm2 of shear stress (SS) resulted in activation of TRPV4 and SK currents. However, after incubation with HC067047 (TRPV4 inhibitor), SK currents could no longer be activated by SS, suggesting SK channel activation by SS was mediated through TRPV4. SK currents in BCECs were also activated by isoproterenol or by GSK1016790A (TRPV4 activator). In addition, preincubation of isolated coronary arterioles with apamin (SK inhibitor) resulted in a significant diminution of SSD whereas preincubation with HC067047 produced vasoconstriction by SS. Exposure of BCECs to SS (15 dynes/cm2 16 h) enhanced the production of nitric oxide and prostacyclin (PGI2) and facilitated the translocation of TRPV4 to the caveolae. Inhibition of TRPV4 abolished the SS-mediated intracellular Ca2+ ([Ca2+] i ) increase in BCECs. These results indicate a dynamic interaction in the vascular endothelium among caveolae TRPV4 and SK3 channels. This caveolae-TRPV4-SK3 channel complex underlies the molecular and ionic mechanisms that modulate SSD in the coronary circulation.
Collapse
Affiliation(s)
- Tong Lu
- From the Department of Cardiovascular Medicine
| | | | - Qiang Chai
- From the Department of Cardiovascular Medicine.,the Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Science, Jinan 250062, China
| | | | - Garry C Sieck
- Department of Physiology and Biomedical Engineering, and
| | - Zvonimir S Katusic
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota 55905 and
| | - Hon-Chi Lee
- From the Department of Cardiovascular Medicine,
| |
Collapse
|
88
|
Park BG, Shin WS, Oh S, Park GM, Kim NI, Lee S. A novel antihypertension agent, sargachromenol D from marine brown algae, Sargassum siliquastrum, exerts dual action as an L-type Ca 2+ channel blocker and endothelin A/B 2 receptor antagonist. Bioorg Med Chem 2017; 25:4649-4655. [PMID: 28720331 DOI: 10.1016/j.bmc.2017.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
We isolated the novel vasoactive marine natural products, (5E,10E)-14-hydroxy-2,6,10-trimethylpentadeca-5,10-dien-4-one (4) and sargachromenol D (5), from Sargassum siliquastrum collected from the coast of the East Sea in South Korea by using activity-guided HPLC purification. The compounds effectively dilated depolarization (50mMK+)-induced basilar artery contraction with EC50 values of 3.52±0.42 and 1.62±0.63μM, respectively, but only sargachromenol D (5) showed a vasodilatory effect on endothelin-1 (ET-1)-induced basilar artery contraction (EC50=9.8±0.6μM). These results indicated that sargachromenol D (5) could act as a dual antagonist of l-type Ca2+ channel and endothelin A/B2 receptors. Moreover, sargachromenol D (5) lowered blood pressure in spontaneous hypertensive rats (SHRs) 2h after oral treatment at a dose of 80mg/kg dose and the effect was maintained for 24h. Based on our ex vivo and in vivo experiments, we propose that sargachromenol D (5) is a strong candidate for the treatment of hypertension that is not controlled by conventional drugs, in particular, severe-, type II diabetes-, salt-sensitive, and metabolic disease-induced hypertension.
Collapse
Affiliation(s)
- Byong-Gon Park
- Department of Physiology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea; Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| | - Woon-Seob Shin
- Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea; Department of Microbiology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| | - Sangtae Oh
- Department of Basic Science, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| | - Gab-Man Park
- Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea; Department of Environmental Medical Biology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| | - Nam Ik Kim
- Department of Physical Education, Catholic Kwandong University College of Education, Gangneung 25601, Republic of Korea
| | - Seokjoon Lee
- Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea; Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea.
| |
Collapse
|
89
|
Feijóo-Bandín S, Aragón-Herrera A, Rodríguez-Penas D, Portolés M, Roselló-Lletí E, Rivera M, González-Juanatey JR, Lago F. Relaxin-2 in Cardiometabolic Diseases: Mechanisms of Action and Future Perspectives. Front Physiol 2017; 8:599. [PMID: 28868039 PMCID: PMC5563388 DOI: 10.3389/fphys.2017.00599] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/03/2017] [Indexed: 12/13/2022] Open
Abstract
Despite the great effort of the medical community during the last decades, cardiovascular diseases remain the leading cause of death worldwide, increasing their prevalence every year mainly due to our new way of life. In the last years, the study of new hormones implicated in the regulation of energy metabolism and inflammation has raised a great interest among the scientific community regarding their implications in the development of cardiometabolic diseases. In this review, we will summarize the main actions of relaxin, a pleiotropic hormone that was previously suggested to improve acute heart failure and that participates in both metabolism and inflammation regulation at cardiovascular level, and will discuss its potential as future therapeutic target to prevent/reduce cardiovascular diseases.
Collapse
Affiliation(s)
- Sandra Feijóo-Bandín
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and University Clinical HospitalSantiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades CardiovascularesMadrid, Spain
| | - Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and University Clinical HospitalSantiago de Compostela, Spain
| | - Diego Rodríguez-Penas
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and University Clinical HospitalSantiago de Compostela, Spain
| | - Manuel Portolés
- Centro de Investigación Biomédica en Red de Enfermedades CardiovascularesMadrid, Spain
- Cardiocirculatory Unit, Health Research Institute of La Fe University HospitalValencia, Spain
| | - Esther Roselló-Lletí
- Centro de Investigación Biomédica en Red de Enfermedades CardiovascularesMadrid, Spain
- Cardiocirculatory Unit, Health Research Institute of La Fe University HospitalValencia, Spain
| | - Miguel Rivera
- Centro de Investigación Biomédica en Red de Enfermedades CardiovascularesMadrid, Spain
- Cardiocirculatory Unit, Health Research Institute of La Fe University HospitalValencia, Spain
| | - José R. González-Juanatey
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and University Clinical HospitalSantiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades CardiovascularesMadrid, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research and University Clinical HospitalSantiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades CardiovascularesMadrid, Spain
| |
Collapse
|
90
|
Tirloni CAS, Lívero FADR, Palozi RAC, Silveira RCA, Vasconcelos PCDP, Souza RIC, Dos Santos AC, de Almeida VP, Manfron Budel J, de Souza LM, Gasparotto Junior A. Ethnopharmacological investigations of the cardio-renal properties of a native species from the region of Pantanal, state of Mato Grosso do Sul, Brazil. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:125-134. [PMID: 28549859 DOI: 10.1016/j.jep.2017.05.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acanthospermum hispidum DC. is an important medicinal herb that belongs to family Asteraceae, popularly used as a diuretic and hypotensive in the region of Pantanal, state of Mato Grosso do Sul, Brazil. Despite the relevance of this species throughout the country, there are no detailed studies about its possible ethnobotanical indication. AIM To carry out a detailed ethnopharmacological investigation of the cardio-renal properties of A. hispidum. MATERIALS AND METHODS First, a detailed morpho-anatomical study with the purpose of characterizing and providing quality control parameters for the species was carried out. Then, purified aqueous extract (ESAH) was obtained and a detailed phytochemical investigation about its main secondary metabolites was performed. In addition, a thorough acute toxicological study was conducted to evaluate the actual toxic effects of this preparation. Finally, the possible diuretic and hypotensive effects of ESAH on male Wistar rats (30, 100, 300mg/kg; intraduodenally) were evaluated, and using pharmacological antagonists or inhibitors, the involvement of prostaglandin/cAMP and nitric oxide/cGMP pathway and potassium channels in ESAH-induced hypotension was investigated. RESULTS The analyses performed by liquid chromatography-mass spectrometry showed that the main secondary metabolites present in ESAH were phenolic compounds, such as caffeoylquinic acids (chlorogenic acid), dicaffeoylquinic acids and glycosylated flavonoids (quercetin glucoside and galactoside). ESAH did not induce any acute toxic effects and did not affect the urinary volume or renal excretion of electrolytes in Wistar rats. On the other hand, intraduodenal administration of ESAH induces a significant acute hypotensive effect. Previous treatment with N(G)-nitro-L-arginine methyl ester, methylene blue, or tetraethylammonium fully avoided the hypotensive effect of ESAH. All other parameters were not affected by treatment with ESAH. CONCLUSION Data obtained in this study allow us to suggest that ESAH obtained from A. hispidum presents an important acute hypotensive effect, which appears to be dependent on the nitric oxide/cGMP pathway. This study presents new evidences about the therapeutic potential of this species when acute hypotensive response is required.
Collapse
Affiliation(s)
- Cleide Adriane Signor Tirloni
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Francislaine Aparecida Dos Reis Lívero
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Rhanany Alan Calloi Palozi
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Renan César Alves Silveira
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Paulo César de Paula Vasconcelos
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Roosevelt Isaias Carvalho Souza
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Ariany Carvalho Dos Santos
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil
| | - Valter Paes de Almeida
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Jane Manfron Budel
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Lauro Mera de Souza
- Institute of Research Pelé Pequeno Príncipe, Pequeno Príncipe Faculty, Curitiba, PR, Brazil.
| | - Arquimedes Gasparotto Junior
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Rodovia Dourados-Itahum, km 12, P.O. Box 533, 79.804-970 Dourados, MS, Brazil.
| |
Collapse
|
91
|
Loader J, Meziat C, Watts R, Lorenzen C, Sigaudo-Roussel D, Stewart S, Reboul C, Meyer G, Walther G. Effects of Sugar-Sweetened Beverage Consumption on Microvascular and Macrovascular Function in a Healthy Population. Arterioscler Thromb Vasc Biol 2017; 37:1250-1260. [DOI: 10.1161/atvbaha.116.308010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
Objective—
To assess vascular function during acute hyperglycemia induced by commercial sugar-sweetened beverage (SSB) consumption and its effect on underlying mechanisms of the nitric oxide pathway.
Approach and Results—
In a randomized, single-blind, crossover trial, 12 healthy male participants consumed 600 mL (20 oz.) of water or a commercial SSB across 2 visits. Endothelial and vascular smooth muscle functions were assessed in the microcirculation using laser speckle contrast imaging coupled with iontophoresis and in the macrocirculation using brachial artery ultrasound with flow- and nitrate-mediated dilation. Compared with water, SSB consumption impaired microvascular and macrovascular endothelial function as indicated by a decrease in the vascular response to acetylcholine iontophoresis (208.3±24.3 versus 144.2±15.7%,
P
<0.01) and reduced flow-mediated dilation (0.019±0.002 versus 0.014±0.002%/s,
P
<0.01), respectively. Systemic vascular smooth muscle remained preserved. Similar decreases in endothelial function were observed during acute hyperglycemia in an in vivo rat model. However, function was fully restored by treatment with the antioxidants,
N
-acetylcysteine and apocynin. In addition, ex vivo experiments revealed that although the production of reactive oxygen species was increased during acute hyperglycemia, the bioavailability of nitric oxide in the endothelium was decreased, despite no change in the activation state of endothelial nitric oxide synthase.
Conclusions—
To our knowledge, this is the first study to assess the vascular effects of acute hyperglycemia induced by commercial SSB consumption alone. These findings suggest that SSB-mediated endothelial dysfunction is partly due to increased oxidative stress that decreases nitric oxide bioavailability.
Clinical Trial Registration—
URL:
https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=366442&isReview=true
. Australian New Zealand Clinical Trials Registry Number: ACTRN12614000614695.
Collapse
Affiliation(s)
- Jordan Loader
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Cindy Meziat
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Rani Watts
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Christian Lorenzen
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Dominique Sigaudo-Roussel
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Simon Stewart
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Cyril Reboul
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Gregory Meyer
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| | - Guillaume Walther
- From the Avignon University, LAPEC EA4278, F-84000, Avignon, France (J.L., C.M., C.R., G.M., G.W.); Centre of Research Excellence to Reduce Inequality in Heart Disease, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne (J.L.); School of Exercise Science, Australian Catholic University, Melbourne (J.L., R.W., C.L., G.W.); Laboratory of Tissue Biology and Therapeutic Engineering, UMR5305, University of Lyon 1, France (D.S.-R.); and The Mary MacKillop Institute for
| |
Collapse
|
92
|
Zaborska KE, Edwards G, Austin C, Wareing M. The Role of O-GlcNAcylation in Perivascular Adipose Tissue Dysfunction of Offspring of High-Fat Diet-Fed Rats. J Vasc Res 2017; 54:79-91. [PMID: 28376507 PMCID: PMC5569708 DOI: 10.1159/000458422] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/27/2017] [Indexed: 12/31/2022] Open
Abstract
Perivascular adipose tissue (PVAT), which reduces vascular contractility, is dysfunctional in the male offspring of rats fed a high-fat diet (HFD), partially due to a reduced NO bioavailability. O-GlcNAcylation of eNOS decreases its activity, thus we investigated the role of O-GlcNAcylation in the prenatal programming of PVAT dysfunction. Female Sprague-Dawley rats were fed either a control (10% fat) or an obesogenic HFD (45% fat) diet for 12 weeks prior to mating, and throughout pregnancy and lactation. Offspring were weaned onto the control diet and were killed at 12 and 24 weeks of age. Mesenteric arteries from the 12-week-old offspring of HFD dams (HFDO) contracted less to U46619; these effects were mimicked by glucosamine in control arteries. PVAT from 12- and 24-week-old controls, but not from HFDO, exerted an anticontractile effect. Glucosamine attenuated the anticontractile effect of PVAT in the vessels from controls but not from HFDO. AMP-activated protein kinase (AMPK) activation (with A769662) partially restored an anticontractile effect in glucosamine-treated controls and HFDO PVAT. Glucosamine decreased AMPK activity and expression in HFDO PVAT, although phosphorylated eNOS expression was only reduced in that from males. The loss of anticontractile effect of HFDO PVAT is likely to result from increased O-GlcNAcylation, which decreased AMPK activity and, in males, decreased NO bioavailability.
Collapse
Affiliation(s)
- Karolina E Zaborska
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
93
|
Ruamyod K, Watanapa WB, Shayakul C. Testosterone rapidly increases Ca 2+-activated K + currents causing hyperpolarization in human coronary artery endothelial cells. J Steroid Biochem Mol Biol 2017; 168:118-126. [PMID: 28223151 DOI: 10.1016/j.jsbmb.2017.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022]
Abstract
Testosterone has endothelium-dependent vasodilatory effects on the coronary artery, with some reports suggesting endothelial ion channel involvement. This study employed the whole-cell patch clamp technique to investigate the effect of testosterone on ion channels in human coronary artery endothelial cells (HCAECs) and the mechanisms involved. We found that 0.03-3μM testosterone significantly induced a rapid, concentration-dependent increase in total HCAEC current (EC50, 71.96±1.66nM; maximum increase, 59.13±8.37%; mean±SEM). The testosterone-enhanced currents consisted of small- and large-conductance Ca2+-activated K+ currents (SKCa and BKCa currents), but not Cl- and nonselective cation currents. Either a non-permeant testosterone conjugate or the non-aromatizable androgen dihydrotestosterone (DHT) could increase HCAEC currents as well. The androgen receptor antagonist flutamide prevented this testosterone, testosterone conjugate, and DHT effect, while the estrogen receptor antagonist fulvestrant did not. Incubating HCAECs with pertussis toxin or protein kinase A inhibitor H-89 largely inhibited the testosterone effect, while pre-incubation with phospholipase C inhibitor U-73122, prostacyclin inhibitor indomethacin, nitric oxide synthase inhibitor L-NAME or cytochrome P450 inhibitor MS-PPOH, did not. Finally, testosterone application induced HCAEC hyperpolarization within minutes; this effect was prevented by SKCa and BKCa current inhibitors apamin and iberiotoxin. This is the first electrophysiological demonstration of androgen-induced KCa current increase, leading to hyperpolarization, in any endothelial cell, and the first report of SKCa as a testosterone target. Our data show that testosterone rapidly increased whole-cell HCAEC SKCa and BKCa currents via a surface androgen receptor, Gi/o protein, and protein kinase A. This mechanism may explain rapid testosterone-induced coronary vasodilation seen in vivo.
Collapse
Affiliation(s)
- Katesirin Ruamyod
- Department of Physiology Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Wattana B Watanapa
- Department of Physiology Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Chairat Shayakul
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
94
|
Biochemical and Biological Attributes of Matrix Metalloproteinases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:1-73. [PMID: 28413025 DOI: 10.1016/bs.pmbts.2017.02.005] [Citation(s) in RCA: 812] [Impact Index Per Article: 101.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that are involved in the degradation of various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation of their latent zymogen form. MMPs are often secreted as inactive pro-MMP form which is cleaved to the active form by various proteinases including other MMPs. MMPs cause degradation of ECM proteins such as collagen and elastin, but could influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in tissue remodeling during various physiological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair, as well as in pathological conditions such as myocardial infarction, fibrotic disorders, osteoarthritis, and cancer. Increases in specific MMPs could play a role in arterial remodeling, aneurysm formation, venous dilation, and lower extremity venous disorders. MMPs also play a major role in leukocyte infiltration and tissue inflammation. MMPs have been detected in cancer, and elevated MMP levels have been associated with tumor progression and invasiveness. MMPs can be regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs have been proposed as biomarkers for numerous pathological conditions and are being examined as potential therapeutic targets in various cardiovascular and musculoskeletal disorders as well as cancer.
Collapse
|
95
|
Chen Y, Peng W, Raffetto JD, Khalil RA. Matrix Metalloproteinases in Remodeling of Lower Extremity Veins and Chronic Venous Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:267-299. [PMID: 28413031 DOI: 10.1016/bs.pmbts.2017.02.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The veins of the lower extremity are equipped with efficient wall, contractile vascular smooth muscle (VSM), and competent valves in order to withstand the high venous hydrostatic pressure in the lower limb and allow unidirectional movement of deoxygenated blood toward the heart. The vein wall structure and function are in part regulated by matrix metalloproteinases (MMPs). MMPs are zinc-dependent endopeptidases that are secreted as inactive pro-MMPs by different cells in the venous wall including fibroblasts, VSM, and leukocytes. Pro-MMPs are activated by other MMPs, proteinases, and other endogenous and exogenous activators. MMPs degrade various extracellular matrix (ECM) proteins including collagen and elastin, and could affect other cellular processes including endothelium-mediated dilation, VSM cell migration, and proliferation as well as modulation of Ca2+ signaling and contraction in VSM. It is thought that increased lower limb venous hydrostatic pressure increases hypoxia-inducible factors and other MMP inducers such as extracellular matrix metalloproteinase inducer, leading to increased MMP expression/activity, ECM protein degradation, vein wall relaxation, and venous dilation. Vein wall inflammation and leukocyte infiltration cause additional increases in MMPs, and further vein wall dilation and valve degradation, that could lead to chronic venous disease and varicose veins (VVs). VVs are often presented as vein wall dilation and tortuosity, incompetent venous valves, and venous reflux. Different regions of VVs show different MMP levels and ECM proteins with atrophic regions showing high MMP levels/activity and little ECM compared to hypertrophic regions with little or inactive MMPs and abundant ECM. Treatment of VVs includes compression stockings, venotonics, sclerotherapy, or surgical removal. However, these approaches do not treat the cause of VVs, and other lines of treatment may be needed. Modulation of endogenous tissue inhibitors of metalloproteinases (TIMPs), and exogenous synthetic MMP inhibitors may provide new approaches in the management of VVs.
Collapse
Affiliation(s)
- Yunfei Chen
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Wei Peng
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joseph D Raffetto
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
96
|
Dual NEP/ECE inhibition improves endothelial function in mesenteric resistance arteries of 32-week-old SHR. Hypertens Res 2017; 40:738-745. [DOI: 10.1038/hr.2017.38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/22/2016] [Accepted: 01/23/2017] [Indexed: 01/06/2023]
|
97
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 620] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
98
|
Byon CH, Heath JM, Chen Y. Redox signaling in cardiovascular pathophysiology: A focus on hydrogen peroxide and vascular smooth muscle cells. Redox Biol 2016; 9:244-253. [PMID: 27591403 PMCID: PMC5011184 DOI: 10.1016/j.redox.2016.08.015] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress represents excessive intracellular levels of reactive oxygen species (ROS), which plays a major role in the pathogenesis of cardiovascular disease. Besides having a critical impact on the development and progression of vascular pathologies including atherosclerosis and diabetic vasculopathy, oxidative stress also regulates physiological signaling processes. As a cell permeable ROS generated by cellular metabolism involved in intracellular signaling, hydrogen peroxide (H2O2) exerts tremendous impact on cardiovascular pathophysiology. Under pathological conditions, increased oxidase activities and/or impaired antioxidant systems results in uncontrolled production of ROS. In a pro-oxidant environment, vascular smooth muscle cells (VSMC) undergo phenotypic changes which can lead to the development of vascular dysfunction such as vascular inflammation and calcification. Investigations are ongoing to elucidate the mechanisms for cardiovascular disorders induced by oxidative stress. This review mainly focuses on the role of H2O2 in regulating physiological and pathological signals in VSMC.
Collapse
Affiliation(s)
| | - Jack M Heath
- Department of Pathology, Birmingham, AL 35294, USA
| | - Yabing Chen
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA.
| |
Collapse
|
99
|
Addison MP, Singh TU, Parida S, Choudhury S, Kasa JK, Sukumaran SV, Darzi SA, Kandasamy K, Singh V, Kumar D, Mishra SK. NO synthase inhibition attenuates EDHF-mediated relaxation induced by TRPV4 channel agonist GSK1016790A in the rat pulmonary artery: Role of TxA2. Pharmacol Rep 2016; 68:620-6. [DOI: 10.1016/j.pharep.2016.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/01/2016] [Accepted: 01/04/2016] [Indexed: 11/26/2022]
|
100
|
Loss of anti-contractile effect of perivascular adipose tissue in offspring of obese rats. Int J Obes (Lond) 2016; 40:1205-14. [PMID: 27102050 PMCID: PMC4973217 DOI: 10.1038/ijo.2016.62] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/26/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
Rationale: Maternal obesity pre-programmes offspring to develop obesity and associated cardiovascular disease. Perivascular adipose tissue (PVAT) exerts an anti-contractile effect on the vasculature, which is reduced in hypertension and obesity. Objective: The objective of this study was to determine whether maternal obesity pre-programmes offspring to develop PVAT dysfunction in later life. Methods: Female Sprague–Dawley rats were fed a diet containing 10% (control) or 45% fat (high fat diet, HFD) for 12 weeks prior to mating and during pregnancy and lactation. Male offspring were killed at 12 or 24 weeks of age and tension in PVAT-intact or -denuded mesenteric artery segments was measured isometrically. Concentration–response curves were constructed to U46619 and norepinephrine. Results: Only 24-week-old HFD offspring were hypertensive (P<0.0001), although the anti-contractile effect of PVAT was lost in vessels from HFD offspring of each age. Inhibition of nitric oxide (NO) synthase with 100 μMl-NMMA attenuated the anti-contractile effect of PVAT and increased contractility of PVAT-denuded arteries (P<0.05, P<0.0001). The increase in contraction was smaller in PVAT-intact than PVAT-denuded vessels from 12-week-old HFD offspring, suggesting decreased PVAT-derived NO and release of a contractile factor (P<0.07). An additional, NO-independent effect of PVAT was evident only in norepinephrine-contracted vessels. Activation of AMP-activated kinase (with 10 μM A769662) was anti-contractile in PVAT-denuded (P<0.0001) and -intact (P<0.01) vessels and was due solely to NO in controls; the AMPK effect was similar in HFD offspring vessels (P<0.001 and P<0.01, respectively) but was partially NO-independent. Conclusions: The diminished anti-contractile effects of PVAT in offspring of HFD dams are primarily due to release of a PVAT-derived contractile factor and reduced NO bioavailability.
Collapse
|