1
|
Yilmaz G, Alexander JS. Impaired Peripheral Vascular Function Following Ischemic Stroke in Mice: Potential Insights into Blood Pressure Variations in the Post-Stroke Patient. PATHOPHYSIOLOGY 2024; 31:488-501. [PMID: 39311310 PMCID: PMC11417821 DOI: 10.3390/pathophysiology31030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
High systolic blood pressure and increased blood pressure variability after the onset of ischemic stroke are associated with poor clinical outcomes. One of the key determinants of blood pressure is arteriolar size, determined by vascular smooth muscle tone and vasodilatory and vasoconstrictor substances that are released by the endothelium. The aim of this study is to outline alterations in vasomotor function in isolated peripheral arteries following ischemic stroke. The reactivity of thoracic aortic segments from male C57BL/6 mice to dilators and constrictors was quantified using wire myography. Acetylcholine-induced endothelium-dependent vasodilation was impaired after ischemic stroke (LogIC50 Sham = -7.499, LogIC50 Stroke = -7.350, p = 0.0132, n = 19, 31 respectively). The vasodilatory responses to SNP were identical in the isolated aortas in the sham and stroke groups. Phenylephrine-induced vasoconstriction was impaired in the aortas isolated from the stroke animals in comparison to their sham treatment counterparts (Sham LogEC50= -6.652 vs. Stroke LogEC50 = -6.475, p < 0.001). Our study demonstrates that 24 h post-ischemic stroke, peripheral vascular responses are impaired in remote arteries. The aortas from the stroke animals exhibited reduced vasoconstrictor and endothelium-dependent vasodilator responses, while the endothelium-independent vasodilatory responses were preserved. Since both the vasodilatory and vasoconstrictor responses of peripheral arteries are impaired following ischemic stroke, our findings might explain increased blood pressure variability following ischemic stroke.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Molecular Cellular and Biomedical Sciences, CUNY School of Medicine, New York, NY 10031, USA
| | - Jonathan Steven Alexander
- Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA;
| |
Collapse
|
2
|
Ye P, Deng Y, Gu Y, Liu P, Luo J, Pu J, Chen J, Huang Y, Wang N, Ji Y, Chen S. GRK2-YAP signaling is implicated in pulmonary arterial hypertension development. Chin Med J (Engl) 2024; 137:846-858. [PMID: 38242702 PMCID: PMC10997289 DOI: 10.1097/cm9.0000000000002946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by excessive proliferation of small pulmonary arterial vascular smooth muscle cells (PASMCs), endothelial dysfunction, and extracellular matrix remodeling. G protein-coupled receptor kinase 2 (GRK2) plays an important role in the maintenance of vascular tone and blood flow. However, the role of GRK2 in the pathogenesis of PAH is unknown. METHODS GRK2 levels were detected in lung tissues from healthy people and PAH patients. C57BL/6 mice, vascular smooth muscle cell-specific Grk2 -knockout mice ( Grk2ΔSM22 ), and littermate controls ( Grk2flox/flox ) were grouped into control and hypoxia mice ( n = 8). Pulmonary hypertension (PH) was induced by exposure to chronic hypoxia (10%) combined with injection of the SU5416 (cHx/SU). The expression levels of GRK2 and Yes-associated protein (YAP) in pulmonary arteries and PASMCs were detected by Western blotting and immunofluorescence staining. The mRNA expression levels of Grk2 and Yes-associated protein ( YAP ) in PASMCs were quantified with real-time polymerase chain reaction (RT-PCR). Wound-healing assay, 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay, and 5-Ethynyl-2'-deoxyuridine (EdU) staining were performed to evaluate the proliferation and migration of PASMCs. Meanwhile, the interaction among proteins was detected by immunoprecipitation assays. RESULTS The expression levels of GRK2 were upregulated in the pulmonary arteries of patients with PAH and the lungs of PH mice. Moreover, cHx/SU-induced PH was attenuated in Grk2ΔSM22 mice compared with littermate controls. The amelioration of PH in Grk2ΔSM22 mice was accompanied by reduced pulmonary vascular remodeling. In vitro study further confirmed that GRK2 knock-down significantly altered hypoxia-induced PASMCs proliferation and migration, whereas this effect was severely intensified by overexpression of GRK2 . We also identified that GRK2 promoted YAP expression and nuclear translocation in PASMCs, resulting in excessive PASMCs proliferation and migration. Furthermore, GRK2 is stabilized by inhibiting phosphorylating GRK2 on Tyr86 and subsequently activating ubiquitylation under hypoxic conditions. CONCLUSION Our findings suggest that GRK2 plays a critical role in the pathogenesis of PAH, via regulating YAP expression and nuclear translocation. Therefore, GRK2 serves as a novel therapeutic target for PAH treatment.
Collapse
Affiliation(s)
- Peng Ye
- Division of Cardiovascular Molecular Laboratory, Third Clinical College, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Yunfei Deng
- Division of Cardiovascular Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
- Division of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yue Gu
- Division of Cardiovascular Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Pengfei Liu
- Division of Cardiovascular Molecular Laboratory, Third Clinical College, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jie Luo
- Division of Cardiovascular Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jiangqin Pu
- Division of Cardiovascular Molecular Laboratory, Third Clinical College, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jingyu Chen
- Division of Pulmonary Surgery, Wuxi People’s Hospital, Nanjing Medical University, Wuxi, Jiangsu 300247, China
| | - Yu Huang
- Institute of Vascular Medicine, The Chinese University of Hong Kong, Hongkong 999077, China
| | - Nanping Wang
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Yong Ji
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 210004, China
| | - Shaoliang Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 210004, China
- Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| |
Collapse
|
3
|
Alsufyani HA, Docherty JR. Effects of RS17053 on α 1 -adrenoceptors in rat vas deferens and aorta. Fundam Clin Pharmacol 2023; 37:1170-1178. [PMID: 37392126 DOI: 10.1111/fcp.12930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/01/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND RS17053 is classed as an α1A -adrenoceptor selective antagonist. OBJECTIVES We have examined its profile of action at all subtypes of α1 -adrenoceptor. METHODS Noradrenaline (NA) evoked contractions of rat vas deferens involve α1D -adrenoceptors in phasic contractions and α1A -adrenoceptors in tonic contractions. Contractions of rat aorta to NA involve α1D - and α1B -adrenoceptors. RESULTS RS17053 (10-5 M) shifted NA potency and virtually abolished tonic contractions to NA, with little or limited effect on phasic contractions. The α1D -adrenoceptor antagonist BMY7378 (3 × 10-7 M) significantly inhibited the remaining phasic component of the contractions, and the α1A -adrenoceptor antagonist RS100329 (10-7 M) inhibited further the residual tonic contraction. Hence, RS17053 shows high selectivity for α1A -adrenoceptors over α1D -adrenoceptors in rat vas deferens. However, RS17053 (10-5 M) produced a large shift in the potency of NA in rat aorta, with a pKB of 6.82. Large shifts of NA potency in rat aorta involve α1B -adrenoceptor blockade. CONCLUSION Results in rat vas deferens demonstrate low potency of RS17053 at α1D -adrenoceptors, but results from rat aorta can only be explained as demonstrating α1B -adrenoceptor antagonism by RS17053. RS17053 may be a useful pharmacological tool when reclassified as a mainly α1A - and to a lesser extent α1B -adrenoceptor antagonist with little effect at α1D -adrenoceptors.
Collapse
Affiliation(s)
- Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - James R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
4
|
Sung DJ, Park S, Noh HJ, Golpasandi S, Eun SH, Lee H, Kim B, Wie J, Seo MS, Park SW, Bae YM. Receptor-specific contributions of caveolae, PKC, and Src tyrosine kinase to serotonergic and adrenergic regulation of Kv channels and vasoconstriction. Life Sci 2023; 328:121903. [PMID: 37394095 DOI: 10.1016/j.lfs.2023.121903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
AIMS Caveolae are invaginated, Ω-shaped membrane structures. They are now recognized as portals for signal transduction of multiple chemical and mechanical stimuli. Notably, the contribution of caveolae has been reported to be receptor-specific. However, details of how they differentially contribute to receptor signaling remain unclear. MAIN METHODS Using isometric tension measurements, patch-clamping, and western blotting, we examined the contribution of caveolae and their related signaling pathways to serotonergic (5-HT2A receptor-mediated) and adrenergic (α1-adrenoceptor-mediated) signaling in rat mesenteric arteries. KEY FINDINGS Disruption of caveolae by methyl-β-cyclodextrin effectively blocked vasoconstriction mediated by the 5-HT2A receptor (5-HT2AR), but not by the α1-adrenoceptor. Caveolar disruption selectively impaired 5-HT2AR-mediated voltage-dependent K+ channel (Kv) inhibition, but not α1-adrenoceptor-mediated Kv inhibition. In contrast, both serotonergic and α1-adrenergic effects on vasoconstriction, as well as Kv currents, were similarly blocked by the Src tyrosine kinase inhibitor PP2. However, inhibition of protein kinase C (PKC) by either GO6976 or chelerythrine selectively attenuated the effects mediated by the α1-adrenoceptor, but not by 5-HT2AR. Disruption of caveolae decreased 5-HT2AR-mediated Src phosphorylation, but not α1-adrenoceptor-mediated Src phosphorylation. Finally, the PKC inhibitor GO6976 blocked Src phosphorylation by the α1-adrenoceptor, but not by 5-HT2AR. SIGNIFICANCE 5-HT2AR-mediated Kv inhibition and vasoconstriction are dependent on caveolar integrity and Src tyrosine kinase, but not on PKC. In contrast, α1-adrenoceptor-mediated Kv inhibition and vasoconstriction are not dependent on caveolar integrity, but rather on PKC and Src tyrosine kinase. Caveolae-independent PKC is upstream of Src activation for α1-adrenoceptor-mediated Kv inhibition and vasoconstriction.
Collapse
Affiliation(s)
- Dong Jun Sung
- Department of Sport and Health Studies, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; Sports Convergence Institute, Konkuk University, Chungju 27478, Republic of Korea; Center for Metabolic Diseases, Konkuk University, Chungju 27478, Republic of Korea; Research Institute for Biomedical & Health Science, Chungju 27478, Republic of Korea
| | - Solah Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Hyun Ju Noh
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Shadi Golpasandi
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Seo Hyeon Eun
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Hyeryeong Lee
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Bokyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Jinhong Wie
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Mi Seon Seo
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, Eulji University, Seongnam 13135, Republic of Korea.
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea.
| |
Collapse
|
5
|
Broadley KJ, Mehta D. Trace amine-induced vasoconstriction of human mammary artery and saphenous vein. Vascul Pharmacol 2023; 151:107191. [PMID: 37399882 DOI: 10.1016/j.vph.2023.107191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/15/2023] [Accepted: 06/29/2023] [Indexed: 07/05/2023]
Abstract
Sympathomimetic amines, including β-phenylethylamine (PEA), constrict animal blood vessels but their mechanism of action is not now thought to be through α-adrenoceptors and release of noradrenaline but via trace amine-associated receptors (TAARs). This information is not available for human blood vessels. Functional studies were therefore performed on human arteries and veins to establish whether they constrict to PEA and whether any constrictions are adrenoceptor-mediated. Isolated internal mammary artery or saphenous vein rings were set up in Kreb's-bicarbonate solution at 37 ± 0.5 °C gassed with O2:CO2 (95:5) under class 2 containment. Isometric contractions were measured and cumulative concentration-response curves for PEA or the α-adrenoceptor agonist, phenylephrine were established. PEA showed concentration-related contractions. The maximum was significantly greater in arteries (1.53 ± 0.31 g, n = 9) than veins (0.55 ± 0.18 g, n = 10), but not when plotted as % of KCl contractions. PEA showed slowly developing contractions plateauing at 17,3 ± 3.7 min in mammary artery. The reference α-adrenoceptor agonist, phenylephrine, exhibited more rapid onset (peak 5.0 ± 1.2 min) but non-sustained contractions. In saphenous veins, PEA (62.8 ± 10.7%) and phenylephrine (61.4 ± 9.7%, n = 4) displayed the same maximum, but phenylephrine was more potent. The α1-adrenoceptor antagonist, prazosin (1 μM), blocked phenylephrine contractions of mammary arteries but not PEA contractions in either vessel. PEA causes substantial vasoconstriction of human saphenous vein and mammary artery, which explains its vasopressor actions. This response, however, was not mediated via α1-adrenoceptors, but likely due to TAARs. The classification of PEA as a sympathomimetic amine on human blood vessels is therefore no longer valid and requires revision.
Collapse
Affiliation(s)
- Kenneth J Broadley
- Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward Vll Avenue, Cardiff, Wales CF10 3NB, UK.
| | - Dheeraj Mehta
- Department of Cardiothoracic Surgery, University Hospital of Wales, Cardiff, Wales CF14 4XW, UK
| |
Collapse
|
6
|
Park DY, Heo W, Kang M, Ahn T, Kim D, Choi A, Birnbaumer L, Cho HJ, Kim JY. Role of TRPC3 in Right Ventricular Dilatation under Chronic Intermittent Hypoxia in 129/SvEv Mice. Int J Mol Sci 2023; 24:11284. [PMID: 37511045 PMCID: PMC10379021 DOI: 10.3390/ijms241411284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with obstructive sleep apnea (OSA) exhibit a high prevalence of pulmonary hypertension and right ventricular (RV) hypertrophy. However, the exact molecule responsible for the pathogenesis remains unknown. Given the resistance to RV dilation observed in transient receptor potential canonical 3(Trpc3)-/- mice during a pulmonary hypertension model induced by phenylephrine (PE), we hypothesized that TRPC3 also plays a role in chronic intermittent hypoxia (CIH) conditions, which lead to RV dilation and dysfunction. To test this, we established an OSA mouse model using 8- to 12-week-old 129/SvEv wild-type and Trpc3-/- mice in a customized breeding chamber that simulated sleep and oxygen cycles. Functional parameters of the RV were evaluated through analysis of cardiac cine magnetic resonance images, while histopathological examinations were conducted on cardiomyocytes and pulmonary vessels. Following exposure to 4 weeks of CIH, Trpc3-/- mice exhibited significant RV dysfunction, characterized by decreased ejection fraction, increased end-diastole RV wall thickness, and elevated expression of pathological cardiac markers. In addition, reactive oxygen species (ROS) signaling and the endothelin system were markedly increased solely in the hearts of CIH-exposed Trpc3-/- mice. Notably, no significant differences in pulmonary vessel thickness or the endothelin system were observed in the lungs of wild-type (WT) and Trpc3-/- mice subjected to 4 weeks of CIH. In conclusion, our findings suggest that TRPC3 serves as a regulator of RV resistance in response to pressure from the pulmonary vasculature, as evidenced by the high susceptibility to RV dilation in Trpc3-/- mice without notable changes in pulmonary vasculature under CIH conditions.
Collapse
Affiliation(s)
- Do-Yang Park
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Woon Heo
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Miran Kang
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Taeyoung Ahn
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - DoHyeon Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ayeon Choi
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires C1107AFF, Argentina
| | - Hyung-Ju Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Joo Young Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
7
|
Perez DM. α 1-Adrenergic Receptors: Insights into Potential Therapeutic Opportunities for COVID-19, Heart Failure, and Alzheimer's Disease. Int J Mol Sci 2023; 24:4188. [PMID: 36835598 PMCID: PMC9963459 DOI: 10.3390/ijms24044188] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
α1-Adrenergic receptors (ARs) are members of the G-Protein Coupled Receptor superfamily and with other related receptors (β and α2), they are involved in regulating the sympathetic nervous system through binding and activation by norepinephrine and epinephrine. Traditionally, α1-AR antagonists were first used as anti-hypertensives, as α1-AR activation increases vasoconstriction, but they are not a first-line use at present. The current usage of α1-AR antagonists increases urinary flow in benign prostatic hyperplasia. α1-AR agonists are used in septic shock, but the increased blood pressure response limits use for other conditions. However, with the advent of genetic-based animal models of the subtypes, drug design of highly selective ligands, scientists have discovered potentially newer uses for both agonists and antagonists of the α1-AR. In this review, we highlight newer treatment potential for α1A-AR agonists (heart failure, ischemia, and Alzheimer's disease) and non-selective α1-AR antagonists (COVID-19/SARS, Parkinson's disease, and posttraumatic stress disorder). While the studies reviewed here are still preclinical in cell lines and rodent disease models or have undergone initial clinical trials, potential therapeutics discussed here should not be used for non-approved conditions.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
8
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
9
|
Bojić MG, Todorović L, Santrač A, Mian MY, Sharmin D, Cook JM, Savić MM. Vasodilatory effects of a variety of positive allosteric modulators of GABA A receptors on rat thoracic aorta. Eur J Pharmacol 2021; 899:174023. [PMID: 33722589 DOI: 10.1016/j.ejphar.2021.174023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/18/2021] [Accepted: 03/10/2021] [Indexed: 01/06/2023]
Abstract
Different subtypes of GABAA (gamma-aminobutyric acid A) receptors, through their specific regional and cellular localization, are involved in the manifestation of various functions, both at the central and peripheral levels. We hypothesized that various non-neuronal GABAA receptors are expressed on blood vessels, through which positive allosteric modulators of GABAA receptors exhibit vasodilatory effects. This study involved two parts: one to determine the presence of α1-6 subunit GABAA receptor mRNAs in the rat thoracic aorta, and the other to determine the vasoactivity of the various selective and non-selective positive GABAA receptor modulators: zolpidem (α1-selective), XHe-III-074 (α4-selective), MP-III-022 (α5-selective), DK-I-56-1 (α6-selective), SH-I-048A and diazepam (non-selective). Reverse transcription-polymerase chain reaction (RT-PCR) analysis data demonstrated for the first time the expression of α1, α2, α3, α4 and α5 subunits in the rat thoracic aorta tissue. Tissue bath assays on isolated rat aortic rings revealed significant vasodilatory effects of diazepam, SH-I-048A, XHe-III-074, MP-III-022 and DK-I-56-1, all in terms of achieved relaxations (over 50% of relative tension decrease), as well as in terms of preventive effects on phenylephrine (PE) contraction. Diazepam was the most efficient ligand in the present study, while zolpidem showed the weakest vascular effects. In addition, diazepam-induced relaxations in the presence of antagonists PK11195 or bicuculline were significantly reduced (P < 0.001 and P < 0.05, respectively) at lower concentrations of diazepam (10-7 M and 3 × 10-7 M). The present work suggests that the observed vasoactivity is due to modulation of "vascular" GABAA receptors, which after further detailed research may provide a therapeutic target.
Collapse
Affiliation(s)
- Milica Gajić Bojić
- Center for Biomedical Research, Faculty of Medicine, University of Banja Luka, 16 Save Mrkalja St, 78000, Banja Luka, Republic of Srpska, Bosnia and Herzegovina
| | - Lidija Todorović
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Anja Santrač
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe St, 11000, Belgrade, Serbia
| | - Md Yeunus Mian
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI, 53201, USA
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI, 53201, USA
| | - James M Cook
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI, 53201, USA
| | - Miroslav M Savić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe St, 11000, Belgrade, Serbia.
| |
Collapse
|
10
|
Zhou W, Zhou R, Li N, Chen Y, Pei Y, Han L, Ren J. Vasorelaxation effect of oxysophoridine on isolated thoracicc aorta rings of rats. CHINESE J PHYSIOL 2021; 64:274-280. [DOI: 10.4103/cjp.cjp_60_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
11
|
Normetadrenaline and metadrenaline induce rat thoracic aorta/prostate contraction via α1D/1A-adrenoceptor stimulation. Eur J Pharmacol 2020; 877:173079. [DOI: 10.1016/j.ejphar.2020.173079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/31/2020] [Accepted: 03/24/2020] [Indexed: 11/23/2022]
|
12
|
Pulmonary Artery Denervation as an Innovative Treatment for Pulmonary Hypertension With and Without Heart Failure. Cardiol Rev 2020; 29:89-95. [PMID: 32032132 DOI: 10.1097/crd.0000000000000299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pulmonary hypertension (PH) is categorized into 5 groups based on etiology. The 2 most prevalent forms are pulmonary arterial hypertension (PAH) and PH due to left heart disease (PH-LHD). Therapeutic options do exist for PAH to decrease symptoms and improve functional capacity; however, the mortality rate remains high and clinical improvements are limited. PH-LHD is the most common cause of PH; however, no treatment exists and the use of PAH-therapies is discouraged. Pulmonary artery denervation (PADN) is an innovative catheter-based ablation technique targeting the afferent and efferent fibers of a baroreceptor reflex in the main pulmonary artery (PA) trunk and its bifurcation. This reflex is involved in the elevation of the PA pressure seen in PH. Since 2013, both animal trials and human trials have shown the efficacy of PADN in improving PAH, including improved hemodynamic parameters, increased functional capacity, decreased PA remodeling, and much more. PADN has been shown to decrease the rate of rehospitalization, PH-related complications, and death, and is an overall safe procedure. PADN has also been shown to be effective for PH-LHD. Additional therapeutic mechanisms and benefits of PADN are discussed along with new PADN techniques. PADN has shown efficacy and safety as a potential treatment option for PH.
Collapse
|
13
|
Modulation of vascular responses of guinea-pig aorta by non-endothelial nitric oxide: A minor role for the endothelium. Vascul Pharmacol 2019; 121:106580. [DOI: 10.1016/j.vph.2019.106580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/19/2019] [Accepted: 07/21/2019] [Indexed: 11/21/2022]
|
14
|
Abstract
PURPOSE OF REVIEW Sympathetic overactivity plays an important role in the progression of pulmonary arterial hypertension (PAH). The purpose of this review is to illustrate localization of pulmonary arterial sympathetic nerves, the key steps of pulmonary artery denervation (PADN) procedure, and to highlight clinical outcomes. RECENT FINDINGS Sympathetic nerves mostly occurred in the posterior region of the bifurcation and pulmonary trunk. Emerging preclinical data provided the potential of PADN for PAH. PADN, produced at bifurcation area, improved a profound reduction of pulmonary arterial pressure and ameliorated clinical outcomes with an exclusive ablation catheter. The application of PADN in the patients of PAH or combined pre-capillary and post-capillary PH (CpcPH) improved the hemodynamic parameters and increased 6MWD. Sympathetic overactivity aggravates PAH. PADN is a promising interventional treatment for PAH and CpcPH. Additional clinical trials are warranted to confirm the efficacy of PADN.
Collapse
|
15
|
Akinaga J, García‐Sáinz JA, S. Pupo A. Updates in the function and regulation of α 1 -adrenoceptors. Br J Pharmacol 2019; 176:2343-2357. [PMID: 30740663 PMCID: PMC6592863 DOI: 10.1111/bph.14617] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
α1 -Adrenoceptors are seven transmembrane domain GPCRs involved in numerous physiological functions controlled by the endogenous catecholamines, noradrenaline and adrenaline, and targeted by drugs useful in therapeutics. Three separate genes, whose products are named α1A -, α1B -, and α1D - adrenoceptors, encode these receptors. Although the existence of multiple α1 -adrenoceptors has been acknowledged for almost 25 years, the specific functions regulated by each subtype are still largely unknown. Despite the limited comprehension, the identification of a single class of subtype-selective ligands for the α1A - adrenoceptors, the so-called α-blockers for prostate dysfunction, has led to major improvement in therapeutics, demonstrating the need for continued efforts in the field. This review article surveys the tissue distribution of the three α1 -adrenoceptor subtypes in the cardiovascular system, genitourinary system, and CNS, highlighting the functions already identified as mediated by the predominant activation of specific subtypes. In addition, this review covers the recent advances in the understanding of the molecular mechanisms involved in the regulation of each of the α1 -adrenoceptor subtypes by phosphorylation and interaction with proteins involved in their desensitization and internalization. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Juliana Akinaga
- Department of PharmacologyInstituto de Biociências, UNESPBotucatuBrazil
| | - J. Adolfo García‐Sáinz
- Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - André S. Pupo
- Department of PharmacologyInstituto de Biociências, UNESPBotucatuBrazil
| |
Collapse
|
16
|
Docherty JR. The pharmacology of α 1-adrenoceptor subtypes. Eur J Pharmacol 2019; 855:305-320. [PMID: 31067439 DOI: 10.1016/j.ejphar.2019.04.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 01/30/2023]
Abstract
This review examines the functions of α1-adrenoceptor subtypes, particularly in terms of contraction of smooth muscle. There are 3 subtypes of α1-adrenoceptor, α1A- α1B- and α1D-adrenoceptors. Evidence is presented that the postulated α1L-adrenoceptor is simply the native α1A-adrenoceptor at which prazosin has low potency. In most isolated tissue studies, smooth muscle contractions to exogenous agonists are mediated particularly by α1A-, with a lesser role for α1D-adrenoceptors, but α1B-adrenoceptors are clearly involved in contractions of some tissues, for example, the spleen. However, nerve-evoked responses are the most crucial physiologically, so that these studies of exogenous agonists may overestimate the importance of α1A-adrenoceptors. The major α1-adrenoceptors involved in blood pressure control by sympathetic nerves are the α1D- and the α1A-adrenoceptors, mediating peripheral vasoconstrictor actions. As noradrenaline has high potency at α1D-adrenceptors, these receptors mediate the fastest response and seem to be targets for neurally released noradrenaline especially to low frequency stimulation, with α1A-adrenoceptors being more important at high frequencies of stimulation. This is true in rodent vas deferens and may be true in vasopressor nerves controlling peripheral resistance and tissue blood flow. The αlA-adrenoceptor may act mainly through Ca2+ entry through L-type channels, whereas the α1D-adrenoceptor may act mainly through T-type channels and exhaustable Ca2+ stores. α1-Adrenoceptors may also act through non-G-protein linked second messenger systems. In many tissues, multiple subtypes of α-adrenoceptor are present, and this may be regarded as the norm rather than exception, although one receptor subtype is usually predominant.
Collapse
Affiliation(s)
- James R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
17
|
Zhang H, Yu W, Zhang J, Xie D, Gu Y, Ye P, Chen SL. Pulmonary artery denervation improves hemodynamics and cardiac function in pulmonary hypertension secondary to heart failure. Pulm Circ 2018; 9:2045894018816297. [PMID: 30421645 PMCID: PMC6614950 DOI: 10.1177/2045894018816297] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This study aimed to determine the benefits and correlated mechanisms of pulmonary artery denervation (PADN) for heart failure (HF) pulmonary hypertension (PH). PH secondary to HF is associated with poor clinical outcomes because there is no proper therapy for it. PADN showed improved outcomes for patients with HF-PH. However, the underlying mechanisms remain unknown. Supracoronary aortic banding (SAB) was used to create HF-PH models. Sprague-Dawley rats were randomly assigned to control, SAB, sham, SAB with PADN, and SAB without PADN groups. Surgical (longitudinally damaging vessel nerves) and chemical (10% phenol applied to the surface of nerves) PADN was performed for animals in the SAB with PADN group. Morphological, echocardiographic, hemodynamic, and protein expression changes were measured four weeks thereafter. Adrenergic receptor (AR) expressions of pulmonary arteries from four HF-PH patients and four patients without PH were measured. Ten HF-PH patients who underwent PADN were followed-up for six months. SAB-induced HF-PH was achieved by 50% of animals. Surgical and chemical PADN was associated with significant improvements in pulmonary artery muscularization, hemodynamics, and right ventricular functions. In pulmonary arterial specimens from HF-PH patients, β2-AR and α1A/B-AR, as well as eNOS, were downregulated and α1D-AR was upregulated compared to those from patients without PH. PADN led to a mean increase of 84 m during the 6-min walk distance for HF-PH patients at six-month follow-up. HF-PH was characterized by downregulated β2-AR, α1A-AR, and α1B-AR and by upregulated α1D-AR. PADN is associated with significant improvements in hemodynamics and pulmonary artery remodeling.
Collapse
Affiliation(s)
- Hang Zhang
- 1 Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wande Yu
- 2 Third College, Nanjing Medical University, Nanjing, China
| | - Juan Zhang
- 1 Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dujiang Xie
- 1 Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- 1 Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Ye
- 2 Third College, Nanjing Medical University, Nanjing, China
| | - Shao-Liang Chen
- 1 Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,3 College of Pharmacy and Key Laboratory in Cooperative Innovational Center of Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Broadley KJ, Broadley HD. Non-adrenergic vasoconstriction and vasodilatation of guinea-pig aorta by β-phenylethylamine and amphetamine - Role of nitric oxide determined with L-NAME and NO scavengers. Eur J Pharmacol 2017; 818:198-205. [PMID: 29074414 DOI: 10.1016/j.ejphar.2017.10.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 10/18/2022]
Abstract
Sympathomimetic and trace amines, including β-phenylethylamine (PEA) and amphetamine, increase blood pressure and constrict isolated blood vessels. By convention this is regarded as a sympathomimetic response, however, recent studies suggest trace amine-associated receptor (TAAR) involvement. There is also uncertainty whether these amines also release nitric oxide (NO) causing opposing vasodilatation. These questions were addressed in guinea-pig isolated aorta, a species not previously examined. Guinea-pig aortic rings were set up to measure contractile tension. Cumulative concentration-response curves were constructed for the reference α-adrenoceptor agonist, phenylephrine, PEA or d-amphetamine before and in the presence of vehicles, the α1-adrenoceptor antagonist, prazosin (1µM), the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (L-NAME), or NO scavengers, curcumin and astaxanthin. Prazosin inhibited phenylephrine contractions with low affinity consistent with α1L-adrenoceptors. However, PEA and amphetamine were not antagonised, indicating non-adrenergic responses probably via TAARs. L-NAME potentiated contractions to PEA both in the absence and presence of prazosin, indicating that PEA releases NO to cause underlying opposing vasodilatation, independent of α1-adrenoceptors. L-NAME also potentiated amphetamine and phenylephrine. PEA was potentiated by the NO scavenger astaxanthin but less effectively. Curcumin, an active component of turmeric, however, inhibited PEA. Trace amines therefore constrict blood vessels non-adrenergically with an underlying NO-mediated non-adrenergic vasodilatation. This has implications in the pressor actions of these amines when NO is compromised.
Collapse
Affiliation(s)
- Kenneth J Broadley
- Division of Pharmacology, Cardiff School of Pharmacy & Pharmaceutical Sciences, Cardiff University, King Edward Vll Avenue, Cathays Park, Cardiff Wales, CF10 3NB, UK.
| | - Harrison D Broadley
- Division of Pharmacology, Cardiff School of Pharmacy & Pharmaceutical Sciences, Cardiff University, King Edward Vll Avenue, Cathays Park, Cardiff Wales, CF10 3NB, UK
| |
Collapse
|
19
|
Albee LJ, Eby JM, Tripathi A, LaPorte HM, Gao X, Volkman BF, Gaponenko V, Majetschak M. α 1-Adrenergic Receptors Function Within Hetero-Oligomeric Complexes With Atypical Chemokine Receptor 3 and Chemokine (C-X-C motif) Receptor 4 in Vascular Smooth Muscle Cells. J Am Heart Assoc 2017; 6:JAHA.117.006575. [PMID: 28862946 PMCID: PMC5586474 DOI: 10.1161/jaha.117.006575] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Recently, we provided evidence that α1‐adrenergic receptors (ARs) in vascular smooth muscle are regulated by chemokine (C‐X‐C motif) receptor (CXCR) 4 and atypical chemokine receptor 3 (ACKR3). While we showed that CXCR4 controls α1‐ARs through formation of heteromeric receptor complexes in human vascular smooth muscle cells (hVSMCs), the molecular basis underlying cross‐talk between ACKR3 and α1‐ARs is unknown. Methods and Results We show that ACKR3 agonists inhibit inositol trisphosphate production in hVSMCs on stimulation with phenylephrine. In proximity ligation assays and co‐immunoprecipitation experiments, we observed that recombinant and endogenous ACKR3 form heteromeric complexes with α1A/B/D‐AR. While small interfering RNA knockdown of ACKR3 in hVSMCs reduced α1B/D‐AR:ACKR3, CXCR4:ACKR3, and α1B/D‐AR:CXCR4 complexes, small interfering RNA knockdown of CXCR4 reduced α1B/D‐AR:ACKR3 heteromers. Phenylephrine‐induced inositol trisphosphate production from hVSMCs was abolished after ACKR3 and CXCR4 small interfering RNA knockdown. Peptide analogs of transmembrane domains 2/4/7 of ACKR3 showed differential effects on heteromerization between ACKR3, α1A/B/D‐AR, and CXCR4. While the transmembrane domain 2 peptide interfered with α1B/D‐AR:ACKR3 and CXCR4:ACKR3 heteromerization, it increased heteromerization between CXCR4 and α1A/B‐AR. The transmembrane domain 2 peptide inhibited ACKR3 but did not affect α1b‐AR in β‐arrestin recruitment assays. Furthermore, the transmembrane domain 2 peptide inhibited phenylephrine‐induced inositol trisphosphate production in hVSMCs and attenuated phenylephrine‐induced constriction of mesenteric arteries. Conclusions α1‐ARs form hetero‐oligomeric complexes with the ACKR3:CXCR4 heteromer, which is required for α1B/D‐AR function, and activation of ACKR3 negatively regulates α1‐ARs. G protein–coupled receptor hetero‐oligomerization is a dynamic process, which depends on the relative abundance of available receptor partners. Endogenous α1‐ARs function within a network of hetero‐oligomeric receptor complexes.
Collapse
Affiliation(s)
- Lauren J Albee
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| | - Jonathan M Eby
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| | - Abhishek Tripathi
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| | - Heather M LaPorte
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| | - Xianlong Gao
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, IL
| | - Matthias Majetschak
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL .,Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago Stritch School of Medicine, Maywood, IL
| |
Collapse
|
20
|
Rizvić E, Janković G, Savić MM. Elucidation of the profound antagonism of contractile action of phenylephrine in rat aorta effected by an atypical sympathomimetic decongestant. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:385-395. [PMID: 28706452 PMCID: PMC5507777 DOI: 10.4196/kjpp.2017.21.4.385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 11/15/2022]
Abstract
Vasoconstrictive properties of sympathomimetic drugs are the basis of their widespread use as decongestants and possible source of adverse responses. Insufficiently substantiated practice of combining decongestants in some marketed preparations, such are those containing phenylephrine and lerimazoline, may affect the overall contractile activity, and thus their therapeutic utility. This study aimed to examine the interaction between lerimazoline and phenylephrine in isolated rat aortic rings, and also to assess the substrate of the obtained lerimazoline-induced attenuation of phenylephrine contraction. Namely, while lower concentrations of lerimazoline (10−6 M and especially 10−7 M) expectedly tended to potentiate the phenylephrine-induced contractions, lerimazoline in higher concentrations (10−4 M and above) unexpectedly and profoundly depleted the phenylephrine concentration-response curve. Suppression of NO with NO synthase (NOS) inhibitor Nw-nitro-L-arginine methyl ester (L-NAME; 10−4 M) or NO scavanger OHB12 (10−3 M), as well as non-specific inhibition of K+-channels with tetraethylammonium (TEA; 10−3 M), have reversed lerimazoline-induced relaxation of phenylephrine contractions, while cyclooxygenase inhibitor indomethacin (10−5 M) did not affect the interaction between two vasoconstrictors. At the receptor level, non-selective 5-HT receptor antagonist methiothepin reversed the attenuating effect of lerimazoline on phenylephrine contraction when applied at 3×10−7 and 10−6 M, but not at the highest concentration (10−4 M). Neither the 5-HT1D-receptor selective antagonist BRL 15572 (10−6 M) nor 5-HT7 receptor selective antagonist SB 269970 (10−6 M) affected the lerimazoline-induced attenuation of phenylephrine activity. The mechanism of lerimazoline-induced suppression of phenylephrine contractions may involve potentiation of activity of NO and K+-channels and activation of some methiothepin-sensitive receptors, possibly of the 5-HT2B subtype.
Collapse
Affiliation(s)
- Eldina Rizvić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Goran Janković
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Miroslav M Savić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| |
Collapse
|
21
|
Vasopressors induce passive pulmonary hypertension by blood redistribution from systemic to pulmonary circulation. Basic Res Cardiol 2017; 112:21. [PMID: 28258299 DOI: 10.1007/s00395-017-0611-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 02/28/2017] [Indexed: 02/05/2023]
Abstract
Vasopressors are widely used in resuscitation, ventricular failure, and sepsis, and often induce pulmonary hypertension with undefined mechanisms. We hypothesize that vasopressor-induced pulmonary hypertension is caused by increased pulmonary blood volume and tested this hypothesis in dogs under general anesthesia. In normal hearts (model 1), phenylephrine (2.5 μg/kg/min) transiently increased right but decreased left cardiac output, associated with increased pulmonary blood volume (63% ± 11.8, P = 0.007) and pressures in the left atrium, pulmonary capillary, and pulmonary artery. However, the trans-pulmonary gradient and pulmonary vascular resistance remained stable. These changes were absent after decreasing blood volume or during right cardiac dysfunction to reduce pulmonary blood volume (model 2). During double-ventricle bypass (model 3), phenylephrine (1, 2.5 and 10 μg/kg/min) only slightly induced pulmonary vasoconstriction. Vasopressin (1U and 2U) dose-dependently increased pulmonary artery pressure (52 ± 8.4 and 71 ± 10.3%), but did not cause pulmonary vasoconstriction in normally beating hearts (model 1). Pulmonary artery and left atrial pressures increased during left ventricle dysfunction (model 4), and further increased after phenylephrine injection by 31 ± 5.6 and 43 ± 7.5%, respectively. In conclusion, vasopressors increased blood volume in the lung with minimal pulmonary vasoconstriction. Thus, this pulmonary hypertension is similar to the hemodynamic pattern observed in left heart diseases and is passive, due to redistribution of blood from systemic to pulmonary circulation. Understanding the underlying mechanisms may improve clinical management of patients who are taking vasopressors, especially those with coexisting heart disease.
Collapse
|
22
|
Wang M, Shibamoto T, Kuda Y, Tanida M, Kurata Y. Systemic vasoconstriction modulates the responses of pulmonary vasculature and airway to vasoconstrictors in anesthetized rats. Exp Lung Res 2015; 41:324-34. [PMID: 26151368 DOI: 10.3109/01902148.2015.1032448] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE The physiological responses of the pulmonary vasculature and airway to various vasoconstrictors were studied using isolated perfused lungs and pulmonary arteries, but these responses were not systematically studied in in vivo rats. We determined these responses and modulating effects of systemic circulation in anesthetized rats. METHODS We measured directly pulmonary arterial pressure (PAP), left atrial pressure (LAP), aortic blood flow, and airway pressure (AWP) to determine pulmonary vascular resistance (PVR), following injections of angiotensin II (ANG II), endothelin-1 (ET-1), vasopressin, phenylephrine and thromboxane A2 mimetic U46619 in anesthetized SD rats. RESULTS ANG II, phenylephrine and vasopressin at high doses caused strong systemic vasoconstriction and left heart overload, resulting in a transient increase in LAP and pulmonary congestion, which consequently decreased PVR. Nonetheless, prior to LAP elevation, PVR was slightly but significantly increased by ANG II and phenylephrine. In contrast, ET-1 and U46619 substantially increased PVR in the absence of LAP elevation, while vasopressin did not increase PVR. In separate experiments, PAP and AWP increased when LAP was forcedly elevated. AWP was increased by U46619 through bronchoconstriction and by the other agents through increased LAP-induced pulmonary congestion. CONCLUSION Airway constriction is induced by U46619, and pulmonary vasoconstriction is induced strongly by U46619 and ET-1, and weakly by ANG II and phenylephrine, but not by vasopressin in anesthetized rats. ANG II, vasopressin and phenylephrine exert indirectly a transient pulmonary vasodilatory action due to pulmonary congestion evoked by strong systemic vasoconstriction, which may account for weak pulmonary pressor responses to these agents.
Collapse
Affiliation(s)
- Mofei Wang
- 1Department of Physiology II, Kanazawa Medical University, Uchinada Ishikawa , Japan
| | | | | | | | | |
Collapse
|
23
|
Westcott EB, Segal SS. Perivascular innervation: a multiplicity of roles in vasomotor control and myoendothelial signaling. Microcirculation 2013; 20:217-38. [PMID: 23289720 DOI: 10.1111/micc.12035] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 12/25/2012] [Indexed: 12/30/2022]
Abstract
The control of vascular resistance and tissue perfusion reflect coordinated changes in the diameter of feed arteries and the arteriolar networks they supply. Against a background of myogenic tone and metabolic demand, vasoactive signals originating from perivascular sympathetic and sensory nerves are integrated with endothelium-derived signals to produce vasodilation or vasoconstriction. PVNs release adrenergic, cholinergic, peptidergic, purinergic, and nitrergic neurotransmitters that lead to SMC contraction or relaxation via their actions on SMCs, ECs, or other PVNs. ECs release autacoids that can have opposing actions on SMCs. Respective cell layers are connected directly to each other through GJs at discrete sites via MEJs projecting through holes in the IEL. Whereas studies of intercellular communication in the vascular wall have centered on endothelium-derived signals that govern SMC relaxation, attention has increasingly focused on signaling from SMCs to ECs. Thus, via MEJs, neurotransmission from PVNs can evoke distinct responses from ECs subsequent to acting on SMCs. To integrate this emerging area of investigation in light of vasomotor control, the present review synthesizes current understanding of signaling events that originate within SMCs in response to perivascular neurotransmission in light of EC feedback. Although often ignored in studies of the resistance vasculature, PVNs are integral to blood flow control and can provide a physiological stimulus for myoendothelial communication. Greater understanding of these underlying signaling events and how they may be affected by aging and disease will provide new approaches for selective therapeutic interventions.
Collapse
Affiliation(s)
- Erika B Westcott
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212, USA
| | | |
Collapse
|
24
|
Flacco N, Parés J, Serna E, Segura V, Vicente D, Pérez-Aso M, Noguera MA, Ivorra MD, McGrath JC, D'Ocon P. α1D-Adrenoceptors are responsible for the high sensitivity and the slow time-course of noradrenaline-mediated contraction in conductance arteries. Pharmacol Res Perspect 2013; 1:e00001. [PMID: 25505555 PMCID: PMC4184566 DOI: 10.1002/prp2.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 05/08/2013] [Accepted: 05/17/2013] [Indexed: 12/30/2022] Open
Abstract
The objective of this study was to determine whether the different time-course characteristics of α1-adrenoceptor-mediated contraction in arteries can be related to the subtypes involved. Contractile responses to noradrenaline (NA) were compared with inositol phosphate accumulation and extracellular signal-regulated kinase (ERK)1/2 phosphorylation after α1-agonist stimuli in the same vessels in the presence or absence of α1-antagonists in rat or in α1-subtype knockout (KO) mice. Aorta, where α1D-AR is the main functional subtype, had higher sensitivity to NA (in respect of inositol phosphate [IP], pERK1/2, and contractile response) than tail artery, where the α1A-adrenoceptor subtype is predominant. Furthermore, the contraction in aorta exhibited a slower decay after agonist removal and this was consistent in all strains harboring α1D-adrenoceptors (from rat, α1B-KO, and wild-type [WT] mice) but was not observed in the absence of the α1D-adrenoceptor signal (α1D-adrenoceptor blocked rat aorta or aorta from α1D-KO). IP formation paralleled α1-adrenoceptor-mediated contraction (agonist present or postagonist) in aorta and tail artery. High sensitivity to agonist and persistence of response after agonist removal is a property of α1D-adrenoceptors. Therefore, the preponderance of this subtype in noninnervated conductance arteries such as aorta allows responsiveness to circulating catecholamines and prevents abrupt changes in vessel caliber when the stimulus fluctuates. Conversely, in innervated distributing arteries, high local concentrations of NA are required to activate α1A-adrenoceptors for a response that is rapid but short lived allowing fine adjustment of the contractile tone by perivascular sympathetic nerves.
Collapse
Affiliation(s)
- Nicla Flacco
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - Jaime Parés
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - Eva Serna
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - Vanessa Segura
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - Diana Vicente
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - Miguel Pérez-Aso
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - María Antonia Noguera
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - María Dolores Ivorra
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| | - John C McGrath
- Autonomic Physiology Unit, School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow Glasgow, U.K
| | - Pilar D'Ocon
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València Valencia, Spain
| |
Collapse
|
25
|
Alpha2C-adrenoceptors play a prominent role in sympathetic constriction of porcine pulmonary arteries. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:595-603. [DOI: 10.1007/s00210-012-0741-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 02/10/2012] [Indexed: 12/28/2022]
|
26
|
Animal models of erectile dysfunction (ED): potential utility of non-human primates as a model of atherosclerosis-induced vascular ED. Int J Impot Res 2011; 24:91-100. [PMID: 22205244 DOI: 10.1038/ijir.2011.56] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Erectile dysfunction (ED) is a prevalent medical condition affecting 18 million men and their sexual partners in the United States alone. In the majority of patients, ED is related to alterations in the flow of blood to or from the penis. Undeniably, significant progress has been made in understanding the multifactorial mechanisms that modulate erectile capacity and predispose one to ED, and this, in turn, has led to the availability of more effective treatment options. Nonetheless, all current therapies have untoward side effects, and moreover, there are still no satisfactory treatments for many patients with ED. Further enhancements in the treatment of ED would logically result from both early intervention and more detailed mechanistic insight into the characteristics of the disease process per se. This fact underscores the importance of improved understanding of the initiation, development and progression of ED. However, to do so requires longitudinal studies on animal models that more closely approximate the corresponding clinical features and time course of human disease. The goal of this report is twofold. First, to provide a brief general overview of the applicability of commonly used animal models for the study of ED. The second and primary goal is to highlight the scientific rationale for using non-human primates to evaluate the impact of atherosclerosis-induced vascular disease on the penile and systemic circulatory systems. This latter goal seems especially relevant in light of the recent literature documenting a link between ED and systemic vascular disease, a finding that has major implications in an aging US male population consuming a high fat diet.
Collapse
|
27
|
Mori A, Hanada M, Sakamoto K, Nakahara T, Ishii K. Noradrenaline contracts rat retinal arterioles via stimulation of α(1A)- and α(1D)-adrenoceptors. Eur J Pharmacol 2011; 673:65-9. [PMID: 22040923 DOI: 10.1016/j.ejphar.2011.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 09/29/2011] [Accepted: 10/08/2011] [Indexed: 10/16/2022]
Abstract
The aim of this study was to characterize the α₁-adrenoceptor subtype(s) involved in the noradrenaline-induced contraction of retinal arterioles in rats. In vivo ocular fundus images were captured with a digital camera equipped with a special objective lens. By measuring changes in diameter of retinal arterioles in the fundus images, retinal vascular response was assessed. The systemic blood pressure and heart rate in the animals were also continuously recorded. Following blockade of β₁/β₂-adrenoceptors with propranolol, noradrenaline (0.03-3 μg/kg/min, i.v.) decreased the diameter of retinal arterioles and increased the mean blood pressure in a dose-dependent manner. The highest dose (3 μg/kg/min, i.v.) of noradrenaline caused a small increase in heart rate. The α(1A)-adrenoceptor antagonist RS100329 (0.1 mg/kg, i.v.) and the α(1D)-adrenoceptor antagonist BMY 7378 (1 mg/kg, i.v.) significantly prevented noradrenaline-induced contraction of retinal arterioles and pressor responses whereas the α(1B)-adrenoceptor antagonist L-765314 (1 mg/kg, i.v.) did not. The α(1A)-adrenoceptor agonist, A 61603 (0.03-0.3 μg/kg/min, i.v.), also caused contractile responses of retinal arterioles and pressor responses. These responses were almost completely prevented by RS100329 (0.1 mg/kg, i.v.), but not by BMY 7378 (1 mg/kg, i.v.). These results suggest that the contractile effects of noradrenaline on retinal arterioles and peripheral resistance vessels are, at least in part, mediated by stimulation of α(1A)- and α(1D)-adrenoceptors. Furthermore, it is likely that the α₁-adrenoceptor subtype(s) involved in rat vascular responses are similar in both retinal and peripheral circulation.
Collapse
Affiliation(s)
- Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | | | | | | | | |
Collapse
|
28
|
Gotoh Y, Andoh T, Kuraishi Y. Noradrenergic regulation of itch transmission in the spinal cord mediated by α-adrenoceptors. Neuropharmacology 2011; 61:825-31. [DOI: 10.1016/j.neuropharm.2011.05.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Revised: 05/23/2011] [Accepted: 05/24/2011] [Indexed: 11/27/2022]
|
29
|
Andersson KE. Mechanisms of penile erection and basis for pharmacological treatment of erectile dysfunction. Pharmacol Rev 2011; 63:811-59. [PMID: 21880989 DOI: 10.1124/pr.111.004515] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Erection is basically a spinal reflex that can be initiated by recruitment of penile afferents, both autonomic and somatic, and supraspinal influences from visual, olfactory, and imaginary stimuli. Several central transmitters are involved in the erectile control. Dopamine, acetylcholine, nitric oxide (NO), and peptides, such as oxytocin and adrenocorticotropin/α-melanocyte-stimulating hormone, have a facilitatory role, whereas serotonin may be either facilitatory or inhibitory, and enkephalins are inhibitory. The balance between contractant and relaxant factors controls the degree of contraction of the smooth muscle of the corpora cavernosa (CC) and determines the functional state of the penis. Noradrenaline contracts both CC and penile vessels via stimulation of α₁-adrenoceptors. Neurogenic NO is considered the most important factor for relaxation of penile vessels and CC. The role of other mediators, released from nerves or endothelium, has not been definitely established. Erectile dysfunction (ED), defined as the "inability to achieve or maintain an erection adequate for sexual satisfaction," may have multiple causes and can be classified as psychogenic, vasculogenic or organic, neurologic, and endocrinologic. Many patients with ED respond well to the pharmacological treatments that are currently available, but there are still groups of patients in whom the response is unsatisfactory. The drugs used are able to substitute, partially or completely, the malfunctioning endogenous mechanisms that control penile erection. Most drugs have a direct action on penile tissue facilitating penile smooth muscle relaxation, including oral phosphodiesterase inhibitors and intracavernosal injections of prostaglandin E₁. Irrespective of the underlying cause, these drugs are effective in the majority of cases. Drugs with a central site of action have so far not been very successful. There is a need for therapeutic alternatives. This requires identification of new therapeutic targets and design of new approaches. Research in the field is expanding, and several promising new targets for future drugs have been identified.
Collapse
Affiliation(s)
- K-E Andersson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston Salem, NC 27157, USA.
| |
Collapse
|
30
|
Burnett AL, Goldstein I, Andersson KE, Argiolas A, Christ G, Park K, Xin ZC. Future sexual medicine physiological treatment targets. J Sex Med 2011; 7:3269-304. [PMID: 21029380 DOI: 10.1111/j.1743-6109.2010.02025.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Sexual function in men and women incorporates physiologic processes and regulation of the central and peripheral nervous systems, the vascular system, and the endocrine system. There is need for state-of-the-art information as there is an evolving research understanding of the underlying molecular biological factors and mechanisms governing sexual physiologic functions. AIM To develop an evidence-based, state-of-the-art consensus report on the current knowledge of the major cellular and molecular targets of biologic systems responsible for sexual physiologic function. METHODS State-of-the-art knowledge representing the opinions of seven experts from four countries was developed in a consensus process over a 2-year period. MAIN OUTCOME MEASURES Expert opinion was based on the grading of evidence-based medical literature, widespread internal committee discussion, public presentation, and debate. RESULTS Scientific investigation in this field is needed to increase knowledge and foster development of the future line of treatments for all forms of biological-based sexual dysfunction. This article addresses the current knowledge of the major cellular and molecular targets of biological systems responsible for sexual physiologic function. Future treatment targets include growth factor therapy, gene therapy, stem and cell-based therapies, and regenerative medicine. CONCLUSIONS Scientific discovery is critically important for developing new and increasingly effective treatments in sexual medicine. Broad physiologic directions should be vigorously explored and considered for future management of sexual disorders.
Collapse
Affiliation(s)
- Arthur L Burnett
- The James Buchanan Brady Urological Institute, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Sporkova A, Perez-Rivera A, Galligan JJ. Interaction between alpha(1)- and alpha(2)-adrenoreceptors contributes to enhanced constrictor effects of norepinephrine in mesenteric veins compared to arteries. Eur J Pharmacol 2010; 643:239-46. [PMID: 20599923 DOI: 10.1016/j.ejphar.2010.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/19/2010] [Accepted: 06/10/2010] [Indexed: 02/07/2023]
Abstract
Mesenteric veins are more sensitive than arteries to the constrictor effects of sympathetic nerve stimulation and alpha-adrenoceptor agonists. We tested the hypothesis that alpha(1)- and alpha(2)-adrenoceptors interact to enhance adrenergic reactivity of mesenteric veins. We studied neurogenic and agonist-induced constrictions of mesenteric veins and arteries in vitro. Norepinephrine concentration-response curves were left-shifted in veins compared to arteries. UK 14,304 (0.01-1 microM, alpha(2)-adrenoceptor receptor agonist) did not constrict arteries or veins but enhanced constrictions and Ca(2+) signals mediated by alpha(1)-adrenoceptor stimulation in veins. Yohimbine (alpha(2)-adrenoceptor receptor antagonist) and MK912 (alpha(2C)-adrenoceptor receptor antagonist), but not alpha(2A)- or alpha(2B)-adrenoceptor antagonists, produced rightward shifts in norepinephrine concentration-response curves in veins. Pharmacological studies revealed that alpha(1D)-adrenoceptors mediate venous constrictions. Norepinephrine responses in veins from alpha(2C)-adrenoceptor knock-out (KO) mice were not different from wild type veins. Yohimbine inhibited norepinephrine constrictions in alpha(2C)-adrenoceptor KO veins suggesting that there is upregulation of other alpha(2)-adrenoceptors in alpha(2C)-KO mice. These data indicate that alpha(1D)- and alpha(2C)-adrenoceptors interact in veins but not in arteries. This interaction enhances venous adrenergic reactivity. Mesenteric vein-specific alpha(2)-adrenoceptor linked Ca(2+) and perhaps other signaling pathways account for enhanced venous adrenergic reactivity.
Collapse
Affiliation(s)
- Alexandra Sporkova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | | |
Collapse
|
32
|
Goyal R, Mittal A, Chu N, Zhang L, Longo LD. alpha(1)-Adrenergic receptor subtype function in fetal and adult cerebral arteries. Am J Physiol Heart Circ Physiol 2010; 298:H1797-806. [PMID: 20348219 PMCID: PMC2886655 DOI: 10.1152/ajpheart.00112.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 03/24/2010] [Indexed: 11/22/2022]
Abstract
In the developing fetus, cerebral artery (CA) contractility demonstrates significant functional differences from that of the adult. This may be a consequence of differential activities of alpha(1)-adrenergic receptor (alpha(1)-AR) subtypes. Thus we tested the hypothesis that maturational differences in adrenergic-mediated CA contractility are, in part, a consequence of differential expression and/or activities of alpha(1)-AR subtypes. In CA from fetal ( approximately 140 days) and nonpregnant adult sheep, we used wire myography and imaging, with simultaneous measurement of tension and intracellular Ca(2+) concentration ([Ca(2+)](i)), radioimmunoassay, and Western immunoblots to examine phenylephrine (Phe)-induced contractile responses. The alpha(1A)-AR antagonists (5-MU and WB-4101) completely inhibited Phe-induced contraction in adult but not fetal CA; however, [Ca(2+)](i) increase was reduced significantly in both age groups. The alpha(1D)-AR antagonist (BMY-7378) blocked both Phe-induced contractions and Ca(2+) responses to a significantly greater extent in adult compared with fetal CA. In both age groups, inhibition of alpha(1A)-AR and alpha(1B)-AR, but not alpha(1D)-AR, significantly reduced inositol 1,4,5-trisphosphate responses to Phe. Western immunoblots demonstrated that the alpha(1)-AR subtype expression was only approximately 20% in fetal CA compared with the adult. Moreover, in fetal CA, the alpha(1D)-AR was expressed significantly greater than the other two subtypes. Also, in fetal but not adult CA, Phe induced a significant increase in activated ERK1/2; this increase in phosphorylated ERK was blocked by alpha(1B)-AR (CEC) and alpha(1D)-AR (BMY-7378) inhibitors, but not by alpha(1A)-AR inhibitors (5-MU or WB-4101). In conclusion, in the fetal CA, alpha(1B)-AR and alpha(1D)-AR subtypes play a key role in contractile response as well as in ERK activation. We speculate that in fetal CA alpha(1B)-AR and alpha(1D)-AR subtypes may be a critical factor associated with cerebrovascular growth and function.
Collapse
Affiliation(s)
- Ravi Goyal
- Center for Perinatal Biology, Department of Physiology and Pharmacology, Loma Linda Univ., School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | |
Collapse
|
33
|
Nojimoto FD, Mueller A, Hebeler-Barbosa F, Akinaga J, Lima V, Kiguti LRDA, Pupo AS. The tricyclic antidepressants amitriptyline, nortriptyline and imipramine are weak antagonists of human and rat alpha1B-adrenoceptors. Neuropharmacology 2010; 59:49-57. [PMID: 20363235 DOI: 10.1016/j.neuropharm.2010.03.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 03/24/2010] [Accepted: 03/26/2010] [Indexed: 12/11/2022]
Abstract
Although it is long known that the tricyclic antidepressants amitriptyline, nortriptyline and imipramine inhibit the noradrenaline transporter and alpha(1)-adrenoceptors with similar affinities, which may lead to self-cancelling actions, the selectivity of these drugs for alpha(1)-adrenoceptor subtypes is unknown. The present study investigates the selectivity of amitriptyline, nortriptyline and imipramine for human recombinant and rat native alpha(1)-adrenoceptor subtypes. The selectivity of amitriptyline, nortriptyline and imipramine was investigated in HEK-293 cells expressing each of the human alpha(1)-subtypes and in rat native receptors from the vas deferens (alpha(1A)), spleen (alpha(1B)) and aorta (alpha(1D)) through [(3)H]prazosin binding, and noradrenaline-induced intracellular Ca(2+) increases and contraction assays. Amitriptyline, nortriptyline and imipramine showed considerably higher affinities for alpha(1A)- (approximately 25- to 80-fold) and alpha(1D)-adrenoceptors (approximately 10- to 25-fold) than for alpha(1B)-adrenoceptors in both contraction and [(3)H]prazosin binding assays with rat native and human receptors, respectively. In addition, amitriptyline, nortriptyline and imipramine were substantially more potent in the inhibition of noradrenaline-induced intracellular Ca(2+) increases in HEK-293 cells expressing alpha(1A)- or a truncated version of alpha(1D)-adrenoceptors which traffics more efficiently towards the cell membrane than in cells expressing alpha(1B)-adrenoceptors. Amitriptyline, nortriptyline and imipramine are much weaker antagonists of rat and human alpha(1B)-adrenoceptors than of alpha(1A)- and alpha(1D)-adrenoceptors. The differential affinities for these receptors indicate that the alpha(1)-adrenoceptor subtype which activation is most increased by the augmented noradrenaline availability resultant from the blockade of neuronal reuptake is the alpha(1B)-adrenoceptor. This may be important for the behavioural effects of these drugs.
Collapse
Affiliation(s)
- F D Nojimoto
- Department of Pharmacology, Instituto de Biociências, UNESP, Botucatu, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
34
|
Docherty JR. Subtypes of functional alpha1-adrenoceptor. Cell Mol Life Sci 2010; 67:405-17. [PMID: 19862476 PMCID: PMC11115521 DOI: 10.1007/s00018-009-0174-4] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/11/2009] [Accepted: 10/05/2009] [Indexed: 11/29/2022]
Abstract
In this review, subtypes of functional alpha1-adrenoceptor are discussed. These are cell membrane receptors, belonging to the seven-transmembrane-spanning G-protein-linked family of receptors, which respond to the physiological agonist noradrenaline. alpha1-Adrenoceptors can be divided into alpha1A-, alpha1B- and alpha1D-adrenoceptors, all of which mediate contractile responses involving Gq/11 and inositol phosphate turnover. A fourth alpha1-adrenoceptor, the alpha1L-, represents a functional phenotype of the alpha1A-adrenoceptor. alpha1-Adrenoceptor subtype knock-out mice have refined our knowledge of the functions of alpha-adrenoceptor subtypes, particuarly as subtype-selective agonists and antagonists are not available for all subtypes. alpha1-Adrenoceptors function as stimulatory receptors involved particularly in smooth muscle contraction, especially contraction of vascular smooth muscle, both in local vasoconstriction and in the control of blood pressure and temperature, and contraction of the prostate and bladder neck. Central actions are now being elucidated.
Collapse
MESH Headings
- Animals
- Blood Pressure/physiology
- Body Temperature Regulation
- Drug Inverse Agonism
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Inositol Phosphates/metabolism
- Mice
- Mice, Knockout
- Muscle, Smooth/physiology
- Muscle, Smooth, Vascular/physiology
- Receptors, Adrenergic, alpha-1/classification
- Receptors, Adrenergic, alpha-1/metabolism
- Receptors, Adrenergic, alpha-1/physiology
- Second Messenger Systems/physiology
- Vasoconstriction/physiology
Collapse
Affiliation(s)
- James R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, 123, St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
35
|
Park KE, Sohn JT, Jeong YS, Sung HJ, Shin IW, Lee HK, Chung YK. Inhibitory effect of fentanyl on phenylephrine-induced contraction of the rat aorta. Yonsei Med J 2009; 50:414-21. [PMID: 19568605 PMCID: PMC2703766 DOI: 10.3349/ymj.2009.50.3.414] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/03/2008] [Accepted: 10/03/2008] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Fentanyl was reported to inhibit the alpha(1)-adrenoceptor agonist-induced contraction. The goal of this in vitro study was to identify the alpha(1)-adrenoceptor subtype primarily involved in the fentanyl-induced attenuation of phenylephrine-induced contraction in isolated endothelium-denuded rat aorta. MATERIALS AND METHODS Aortic rings were suspended in order to record isometric tension. Concentration-response curves for phenylephrine (10(-9) to 10(-5) M) were generated in the presence or absence of one of the following drugs: fentanyl (3 x 10(-7), 10(-6), 3 x10(-6) M), 5-methylurapidil (3 x10(-8), 10(-7), 3 x 10(-7) M), chloroethylclonidine (10(-5) M) and BMY 7378 (3 x 10(-9), 10(-8), 3 x 10(-8) M). Phenylephrine concentration-response curves were generated in the presence or absence of fentanyl in rings pretreated with either 3 x10(-9) M prazosin, 10(-9) M 5-methylurapidil or 3 x 10(-9) M BMY 7378. RESULTS Fentanyl (10(-6), 3 x 10(-6) M) attenuated phenylephrine-induced contraction in the rat aorta. 5-Methylurapidil and BMY 7378 produced a parallel rightward shift in the phenylephrine concentration-response curve. The pA(2) values for 5-methylurapidil and BMY 7378 were estimated to be 7.71 +/- 0.15 and 8.99 +/- 0.24, respectively. Fentanyl (10(-6) M) attenuated phenylephrine-induced contraction in rings pretreated with 10(-9) M 5-methylurapidil, but did not alter the rings when pretreated with 3 x 10(-9) M BMY 7378. Pretreatment of the rings with chloroethylclonidine showed a 72.9 +/- 2.3% reduction in phenylephrine-induced maximal contraction. CONCLUSION The results suggest that fentanyl attenuates phenylephrine-induced contraction by inhibiting the pathway involved in the alpha(1D)-adrenoceptor-mediated contraction of the rat aorta.
Collapse
Affiliation(s)
- Kyeong-Eon Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University School of Medicine, 90 Chilam-dong, Jinju, Korea
| | | | | | | | | | | | | |
Collapse
|
36
|
Görnemann T, Villalón CM, Centurión D, Pertz HH. Phenylephrine contracts porcine pulmonary veins via alpha(1B)-, alpha(1D)-, and alpha(2)-adrenoceptors. Eur J Pharmacol 2009; 613:86-92. [PMID: 19376108 DOI: 10.1016/j.ejphar.2009.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 03/20/2009] [Accepted: 04/07/2009] [Indexed: 10/20/2022]
Abstract
We have recently shown that the postjunctional alpha(2)-adrenoceptor mediating contraction of porcine pulmonary veins is of the alpha(2C)-subtype. We could also demonstrate that alpha(1)-adrenoceptors might contribute to the contraction in that blood vessel. In the present study, we aimed at characterising the alpha(1)-adrenoceptor subtype(s) involved using pharmacological and molecular biological methods. In isolated rings of porcine pulmonary veins the typical alpha(1)-adrenoceptor agonist phenylephrine caused a concentration-dependent contraction that was inhibited by the alpha(1B)-adrenoceptor selective antagonists 1-[4-(4-amino-6,7-dimethoxyquinazolin-2-yl)piperazin-1-yl]-2-[2-(isopropyl)-6-methoxyphenoxy]ethan-1-one (Rec15/2615; pA(2) 8.96+/-0.13) and 4-amino-2-[4-[1-(benzyloxycarbonyl)-2(S)-[[(1,1-dimethylethyl)amino]carbonyl]-piperazinyl]-6,7-dimethoxyquinazoline (L-765,314; pA(2) 7.22+/-0.05), as well as the alpha(1D)-adrenoceptor selective antagonist 8-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-8-azaspiro[4.5]decane-7,9-dione (BMY7378; pA(2) 8.29+/-0.15, slope of the Schild plot 0.75+/-0.09, significantly different from unity, P<0.05), but not by the alpha(1A)-adrenoceptor selective antagonists (+/-)-1,3,5-trimethyl-6-[[3-[4-((2,3-dihydro-2-hydroxymethyl)-1,4-benzodioxin-5-yl)-1-piperazinyl]propyl]amino]-2,4(1H,3H)-pyrimidinedione (B8805-033) and N-[2-(2-cyclopropylmethoxyphenoxy)ethyl]-5-chloro-alpha,alpha-dimethyl-1H-indole-3-ethanamine (RS-17053). These findings suggest that phenylephrine activates both alpha(1B)- and alpha(1D)-adrenoceptors. The observation was confirmed by reverse-transcriptase polymerase chain reaction (RT-PCR) in porcine pulmonary veins, where mRNA signals for alpha(1B)- and alpha(1D)-adrenoceptors could be detected. However, the antagonist properties of rauwolscine and yohimbine (non-subtype selective alpha(2)-adrenoceptor antagonists) against phenylephrine showed that this agonist also activates alpha(2)-adrenoceptors in pulmonary veins. This was strengthened in experiments using tissues that were stimulated with forskolin (cell permeable activator of adenylyl cyclase). Phenylephrine mimicked the effect of the selective alpha(2)-adrenoceptor agonist UK14304 by causing an inhibition of forskolin-stimulated cAMP accumulation that was blocked by rauwolscine. It is concluded that, in addition to alpha(1B)- and alpha(1D)-adrenoceptors, phenylephrine can stimulate alpha(2)-adrenoceptors in porcine pulmonary veins.
Collapse
Affiliation(s)
- Tilo Görnemann
- Freie Universität Berlin, Institut für Pharmazie, Berlin (Dahlem), Germany
| | | | | | | |
Collapse
|
37
|
Tran Quang T, Rozec B, Audigane L, Gauthier C. Investigation of the different adrenoceptor targets of nebivolol enantiomers in rat thoracic aorta. Br J Pharmacol 2009; 156:601-8. [PMID: 19210511 DOI: 10.1111/j.1476-5381.2009.00074.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Nebivolol is a highly selective beta(1)-adrenoceptor antagonist with beta(3)-adrenoceptor agonist properties and is a racemate mixture of D- and L-enantiomers. However, the cellular mechanisms of the effects of each enantiomer are not yet clear and are a matter for debate. The aim of the present experiments was to determine the adrenoceptors involved in the vascular effects of D- and L-enantiomers of nebivolol in rat thoracic aorta. EXPERIMENTAL APPROACH Responses to nebivolol enantiomers were evaluated in rings of thoracic aorta from male Sprague-Dawley rats. KEY RESULTS D-nebivolol (0.1-10 micromol.L(-1)), but not L-nebivolol, significantly shifted to the right the concentration-response curve to phenylephrine, an alpha(1)-adrenoceptor agonist, in a concentration-dependent manner. For the following experiments, aortic rings were constricted with endothelin 1 and now both enantiomers produced an endothelium-dependent relaxation of the rings involving the nitric oxide pathway. This relaxation was not modified by 1 micromol.L(-1) CGP 20,712A (beta(1)-adrenoceptor antagonist), but significantly blunted by 7 micromol.L(-1) L-74,8337 (beta(3)-adrenoceptor antagonist). However, only the vasorelaxation induced by D-nebivolol was significantly reduced by 1 micromol.L(-1) ICI 118,551 (beta(2)-adrenoceptor antagonist). CONCLUSIONS AND IMPLICATIONS Our results suggest that the nebivolol enantiomers act on different targets. D-nebivolol induced vasorelaxation by activating beta(2)- and beta(3)-adrenoceptors and antagonizing alpha(1)-adrenoceptors. L-nebivolol induced vasorelaxation by activating only beta(3)-adrenoceptors in our model. Our results emphasize that nebivolol is a beta(1)-adrenoceptor antagonist with several important pharmacological differences from other beta(1)-adrenoceptor antagonists.
Collapse
Affiliation(s)
- T Tran Quang
- INSERM, UMR915, l'Institut du Thorax, Nantes, France
| | | | | | | |
Collapse
|
38
|
Carteolol, a non-conventional partial agonist of β1-adrenoceptors, relaxes phenylephrine-constricted rat aorta through antagonism at α1-adrenoceptors. Eur J Pharmacol 2008; 590:269-75. [DOI: 10.1016/j.ejphar.2008.05.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 04/24/2008] [Accepted: 05/19/2008] [Indexed: 11/21/2022]
|
39
|
Jackson WF, Boerman EM, Lange EJ, Lundback SS, Cohen KD. Smooth muscle alpha1D-adrenoceptors mediate phenylephrine-induced vasoconstriction and increases in endothelial cell Ca2+ in hamster cremaster arterioles. Br J Pharmacol 2008; 155:514-24. [PMID: 18604236 DOI: 10.1038/bjp.2008.276] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE alpha(1)-Adrenoceptor agonists induce Ca(2+)-transients in endothelial cells (ECs) of arterioles. However, the presence of alpha(1)-adrenoceptors on arteriolar ECs has not been excluded, and the identity of alpha(1)-adrenoceptor subtypes in arterioles only has been inferred from pharmacology. Therefore, we determined which subtypes were expressed by vascular smooth muscle cells (VSMCs) and ECs, and which subtype mediated alpha(1)-adrenoceptor-induced constriction. EXPERIMENTAL APPROACH EC Ca(2+)-transients in isolated, cannulated hamster cremasteric arterioles or freshly isolated ECs were studied using Fura 2. Arteriolar diameter was measured by video microscopy. alpha(1)-Adrenoceptor expression was assessed by western blot of whole-arteriolar homogenates and real-time RT-PCR on enzymatically isolated VSMCs and ECs. KEY RESULTS Phenylephrine-induced constriction and EC Ca(2+)-transients were abolished by the alpha(1)-adrenoceptor antagonist prazosin (30 nM) in arterioles. Phenylephrine-induced constriction was inhibited by the alpha(1D)-adrenoceptor antagonist BMY 7378 (K(B)=2.96 nM) and the alpha(1A)-adrenoceptor antagonist 5-methylurapidil (K(B)=4.08 nM), suggesting a significant role for alpha(1D)-adrenoceptors. Western blots confirmed alpha(1D)-adrenoceptor expression, but did not detect alpha(1A)-adrenoceptors. VSMCs expressed alpha(1D)- and alpha(1A)-, but not alpha(1B)-, adrenoceptor transcripts. No alpha(1)-adrenoceptor transcripts were detected in ECs. Neither phenylephrine (10 microM) nor noradrenaline (0.1-1 microM) elicited Ca(2+)-transients in freshly isolated ECs, whereas the endothelium-dependent vasodilators methacholine (1 microM) and substance P (100 nM) consistently increased Ca(2+). CONCLUSIONS AND IMPLICATIONS We reject the hypothesis that hamster cremasteric arteriolar ECs express alpha(1)-adrenoceptors and conclude that alpha(1)-adrenoceptor agonists predominantly act on VSMC alpha(1D)-adrenoceptors to cause vasoconstriction and a subsequent rise in EC Ca(2+).
Collapse
Affiliation(s)
- W F Jackson
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| | | | | | | | | |
Collapse
|
40
|
Nourian Z, Mow T, Muftic D, Burek S, Pedersen ML, Matz J, Mulvany MJ. Orthostatic hypotensive effect of antipsychotic drugs in Wistar rats by in vivo and in vitro studies of alpha1-adrenoceptor function. Psychopharmacology (Berl) 2008; 199:15-27. [PMID: 18542932 DOI: 10.1007/s00213-007-1064-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 12/20/2007] [Indexed: 02/02/2023]
Abstract
RATIONALE Many antipsychotics cause orthostatic hypotension possibly due to antagonist action on resistance vessel alpha1A-adrenoceptors (alpha1A-AR). OBJECTIVE We have tested this possibility by determining in Wistar rats how the orthostatic hypotensive effect of several antipsychotic drugs compares with their affinity for adrenoceptors in mesenteric small arteries (MSA with mainly alpha1A-AR) and aorta (mainly alpha1D-AR). MATERIALS AND METHODS Using a tilt setup, orthostatic hypotension was measured in anaesthetized rats for prazosin and the antipsychotics haloperidol, sertindole, risperidone, clozapine, ziprasidone, domperidone, olanzapine, and aripiprazole. For in vitro studies, segments of MSA and aorta were mounted on a wire myograph for isometric tension recording. Cumulative concentration-response curves were constructed to phenylephrine (PE) in the absence and presence of the drugs. Apparent affinity (pA2) was calculated by Schild analysis. RESULTS Prazosin antagonized tilt-induced and PE responses in both studies (threshold 4 ng/ml, pA2 9.52 MSA, 10.1 aorta). The rank order of the potency of the antipsychotics in the tilt experiments correlated (r2 = 0.69, P = 0.01) with the pA2-values in MSA: Risperidone and sertindole had the highest potency in the tilt test (threshold 159 and 97 ng/ml) and the highest apparent affinity in MSA (pA2 8.92 and 8.78), in contrast with aripiprazole and domperidone, which had the lowest in each case (threshold 4.1 and 3.0 microg/ml, pA2 7.17 and 6.99). In aorta, the pA2 values did not correlate with the in vivo potencies; in particular, sertindole had no functional affinity in aorta. CONCLUSION We conclude that the orthostatic hypotensive effect in rats of the antipsychotic drugs investigated is mediated through alpha1A-ARs.
Collapse
Affiliation(s)
- Z Nourian
- Department of Pharmacology, University of Aarhus, University Park 1240, 8000, Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
41
|
Rozec B, Quang TT, Noireaud J, Gauthier C. Mixed beta3-adrenoceptor agonist and alpha1-adrenoceptor antagonist properties of nebivolol in rat thoracic aorta. Br J Pharmacol 2006; 147:699-706. [PMID: 16474420 PMCID: PMC1751500 DOI: 10.1038/sj.bjp.0706648] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Nebivolol, a selective beta-adrenoceptor (beta1-AR) antagonist, induces vasodilatation by an endothelium- and NO-cGMP-dependent pathway. However, the mechanisms involved in the vascular effect of nebivolol have not been established. Thus, we evaluated the role of alpha1 and beta3-ARs in nebivolol-induced vasodilatation. The responses to nebivolol were investigated in vitro in thoracic aortic rings isolated from male Sprague-Dawley rats. Nebivolol (0.1-10 microM) significantly shifted the concentration-response curve to phenylephrine, an alpha1-AR agonist, to the right in a concentration-dependent manner (pA2 = 6.5). Conversely, the concentration-response curve to endothelin 1 (ET1) was unaffected by nebivolol. In ET1-precontracted rings, nebivolol induced a concentration-dependent relaxation, which was unaffected by nadolol (a beta1/beta2-AR antagonist) but was significantly reduced by L-748,337 (a beta3-AR antagonist), endothelium removal or pretreatment with L-NMMA (an NOS inhibitor). Similar results were obtained with a beta3-AR agonist, SR 58611A. It was concluded that, in rat aorta, nebivolol-induced relaxation results from both inhibition of alpha1-ARs and activation of beta3-ARs. In addition, we confirmed that the endothelium and the NO pathway are involved in the vascular effect of nebivolol. The identification of these vascular targets of nebivolol indicate that it has therapeutic potential for the treatment of pathological conditions associated with an elevation of sympathetic tone, such as heart failure and hypertension.
Collapse
Affiliation(s)
- Bertrand Rozec
- L'institut du thorax, Inserm U533, Faculté de Médecine, 1 rue Gaston Veil, BP 53508, Nantes F-44035, France
- Département d'anesthésie et de réanimation chirurgicale, CHRU G et R Laënnec, Nantes, France
| | - Thuy Tran Quang
- L'institut du thorax, Inserm U533, Faculté de Médecine, 1 rue Gaston Veil, BP 53508, Nantes F-44035, France
| | - Jacques Noireaud
- L'institut du thorax, Inserm U533, Faculté de Médecine, 1 rue Gaston Veil, BP 53508, Nantes F-44035, France
| | - Chantal Gauthier
- L'institut du thorax, Inserm U533, Faculté de Médecine, 1 rue Gaston Veil, BP 53508, Nantes F-44035, France
- Faculté des Sciences et Techniques, Université de Nantes, Nantes, France
- Author for correspondence:
| |
Collapse
|
42
|
Cao YX, Xu CB, Luo GG, Edvinsson L. Up-Regulation of alpha1A-Adrenoceptors in Rat Mesenteric Artery Involves Intracellular Signal Pathways. Basic Clin Pharmacol Toxicol 2006; 98:61-7. [PMID: 16433893 DOI: 10.1111/j.1742-7843.2006.pto_240.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aim of the present study was to investigate if there is an altered expression of alpha-adrenoceptors during organ culture of rat mesenteric artery segments by using a sensitive pharmacological method and molecular biological techniques. Noradrenalin (NA) induced contraction via alpha1-adrenoceptors. The contraction and alpha1A-adrenoceptor mRNA levels were elevated during organ culture. Transcriptional inhibitor actinomycin D, translational inhibitor cycloheximide, protein kinase C inhibitors (staurosporine and RO31-8220) and mitogen-activated protein kinase (MAPK) pathway inhibitors (SB386023, U0126 and SB239063) prevented the increase in NA-induced contractions. The amount of alpha1A-adrenoceptor mRNA was significantly lower in the artery segments cultured for 1 day in the presence of specific MAPK extracellular signal-regulated protein kinase1/2 pathway inhibitor SB386023 than that of the cultured controls. SB386023 did not affect alpha2-adrenoceptor mRNA level. Our results suggest that the up-regulation of alpha1A-adrenoceptors involves transcription and intracellular signal transduction via the protein kinase C and the ERK 1/2 pathways.
Collapse
Affiliation(s)
- Yong-Xiao Cao
- Division of Experimental Vascular Research, Institution of Medicine, Lund University, Sweden
| | | | | | | |
Collapse
|
43
|
Ruggieri MR, Braverman AS, Pontari MA. Combined use of alpha-adrenergic and muscarinic antagonists for the treatment of voiding dysfunction. J Urol 2005; 174:1743-8. [PMID: 16217275 PMCID: PMC3277797 DOI: 10.1097/01.ju.0000176460.62847.23] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE We provide an overview of the medical literature supporting the combined use of muscarinic and alpha-adrenergic antagonist therapy for the treatment of voiding dysfunction. MATERIALS AND METHODS The MEDLINE database (1966 to 2004) of the United States National Library of Medicine was searched for pertinent studies. RESULTS Although the mechanism of action of alpha-adrenergic antagonist therapy for voiding dysfunction has traditionally been assumed to be relaxation of the periurethral, prostatic and bladder neck smooth muscle, substantial evidence supports action at extraprostatic sites involved in micturition, including the bladder dome smooth muscle, peripheral ganglia, spinal cord and brain. Likewise the mechanism of action of anticholinergic therapy has been traditionally assumed to be inhibition of the M3 muscarinic receptor subtypes that mediate normal bladder contractions. However, M2 receptor mediates hypertrophied bladder contractions and there is evidence for an M2 component to the suprasacral control of voiding. CONCLUSIONS Based on the physiology of alpha-adrenergic and muscarinic receptors the inhibition of each one would be expected to be more beneficial than that of either alone because they would work on 2 components of detrusor function. Patients who would likely benefit from this combination therapy are men with lower urinary tract symptoms, women with urgency/frequency syndrome (overactive bladder), patients with uninhibited bladder contractions due to neurogenic bladder, and patients with pelvic pain and voiding symptoms, ie interstitial cystitis and chronic prostatitis/chronic pelvic pain syndrome.
Collapse
Affiliation(s)
- Michael R Ruggieri
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140-5104, USA.
| | | | | |
Collapse
|
44
|
Nozik-Grayck E, Whalen EJ, Stamler JS, McMahon TJ, Chitano P, Piantadosi CA. S-nitrosoglutathione inhibits alpha1-adrenergic receptor-mediated vasoconstriction and ligand binding in pulmonary artery. Am J Physiol Lung Cell Mol Physiol 2005; 290:L136-43. [PMID: 16126786 DOI: 10.1152/ajplung.00230.2005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endogenous nitric oxide donor compounds (S-nitrosothiols) contribute to low vascular tone by both cGMP-dependent and -independent pathways. We have reported that S-nitrosoglutathione (GSNO) inhibits 5-hydroxytryptamine (5-HT)-mediated pulmonary vasoconstriction via a cGMP-independent mechanism likely involving S-nitrosylation of its G protein-coupled receptor (GPCR) system. Because catecholamines, like 5-HT, constrict lung vessels via a GPCR coupled to G(q), we hypothesized that S-nitrosothiols modify the alpha1-adrenergic GPCR system to inhibit pulmonary vasoconstriction by receptor agonists, e.g., phenylephrine (PE). Rat pulmonary artery rings were pretreated for 30 min with and without an S-nitrosothiol, either GSNO or S-nitrosocysteine (CSNO), and constricted with sequential concentrations of PE (10(-8)-10(-6) M). Effective cGMP-dependence was tested in rings pretreated with soluble guanylate cyclase inhibitors {either 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or LY-83583} or G kinase inhibitor (KT-5823), and a thiol reductant [dithiothreitol (DTT)] was used to test reversibility of S-nitrosylation. Both S-nitrosothiols attenuated the PE dose response. The GSNO effect was not prevented by LY-83583, ODQ, or KT-5823, indicating cGMP independence. GSNO inhibition was reversed by DTT, consistent with S-nitrosylation or other GSNO-mediated cysteine modifications. In CSNO-treated lung protein, the alpha1-adrenergic receptor was shown to undergo S-nitrosylation in vitro using a biotin switch assay. Studies of alpha1-adrenergic receptor subtype expression and receptor density by saturation binding with 125I-HEAT showed that GSNO decreased alpha1-adrenergic receptor density but did not alter affinity for antagonist or agonist. These data demonstrate a novel cGMP-independent mechanism of reversible alpha1-adrenergic receptor inhibition by S-nitrosothiols.
Collapse
Affiliation(s)
- Eva Nozik-Grayck
- Department of Pediatrics, University of Colorado Health Science Center, Denver, CO 80262, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Martí D, Miquel R, Ziani K, Gisbert R, Ivorra MD, Anselmi E, Moreno L, Villagrasa V, Barettino D, D'Ocon P. Correlation between mRNA levels and functional role of alpha1-adrenoceptor subtypes in arteries: evidence of alpha1L as a functional isoform of the alpha1A-adrenoceptor. Am J Physiol Heart Circ Physiol 2005; 289:H1923-32. [PMID: 15951348 DOI: 10.1152/ajpheart.00288.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mRNA levels for the three alpha1-adrenoceptor subtypes, alpha1A, alpha1B, and alpha1D, were quantified by real-time RT-PCR in arteries from Wistar rats. The alpha1D-adrenoceptor was prominent in both aorta (79.0%) and mesenteric artery (68.7%), alpha1A predominated in tail (61.7%) and small mesenteric artery (73.3%), and both alpha1A- and alpha1D-subtypes were expressed at similar levels in iliac artery. The mRNA levels of the alpha1B-subtype were a minority in all vessels (1.7-11.1%). Concentration-response curves of contraction in response to phenylephrine or relaxation in response to alpha1-adrenoceptor antagonists on maximal sustained contraction induced by phenylephrine were constructed from control vessels and vessels pretreated with 100 micromol/l chloroethylclonidine (CEC) for 30 min. The significant decrease in the phenylephrine potency observed after CEC treatment together with the inhibitory potency displayed by 8-{2-[4-(2-methoxyphenyl)-1-piperazinyl]-8-azaspiro (4,5) decane-7-dionedihydrochloride} (BMY-7378, an alpha1D-adrenoceptor antagonist) confirm the relevant role of alpha1D-adrenoceptors in aorta and iliac and proximal mesenteric arteries. The potency of 5-methylurapidil (an alpha1A-adrenoceptor antagonist) and the changes in the potency of both BMY-7378 and 5-methylurapidil after CEC treatment provided evidence of a mixed population of alpha1A- and alpha1D-adrenoceptors in iliac and distal mesenteric arteries. The low potency of prazosin (pIC50 < 9) as well as the high 5-methylurapidil potency in tail and small mesenteric arteries suggest the main role of alpha1A/alpha1L-adrenoceptors with minor participation of the alpha1D-subtype. The mRNA levels and CEC treatment corroborated this pattern and confirmed that the alpha1L-adrenoceptor could be a functional isoform of the alpha1A-subtype.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Adrenergic alpha-Antagonists/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Arteries/metabolism
- DNA Primers
- Dose-Response Relationship, Drug
- Iliac Artery/drug effects
- In Vitro Techniques
- Muscle Contraction/drug effects
- Muscle Relaxation/drug effects
- Muscle, Smooth, Vascular/drug effects
- Phenylephrine/pharmacology
- Protein Isoforms
- RNA, Messenger/biosynthesis
- Rats
- Rats, Wistar
- Receptors, Adrenergic, alpha-1/biosynthesis
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/physiology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Daniel Martí
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mueed I, Bains P, Zhang L, Macleod KM. Differential participation of protein kinase C and Rho kinase in α1-adrenoceptor mediated contraction in rat arteries. Can J Physiol Pharmacol 2004; 82:895-902. [PMID: 15573150 DOI: 10.1139/y04-086] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The major functional α1-adrenoceptor in the rat aorta is of the α1Dsubtype and that in the caudal artery is of the α1Asubtype. In the present study, the participation of protein kinase C (PKC) and Rho kinase (RhoK) in contractile responses to stimulation of the α1-adrenoceptors in these two arteries was investigated. Both the PKC inhibitor Ro-318220 and the RhoK inhibitor Y-27632 significantly blocked contractile responses of the aorta to phenylephrine (PE) and the selective α1A-adrenoceptor agonist A61603. When used in combination, the inhibitors had an additive blocking effect. In the caudal artery, Y-27632 but not Ro-318220 inhibited contractile responses to PE and A61603, and, in combination, the antagonism produced was no greater than that by Y-27632 alone. Contractile responses to direct activation of PKC with phorbol 12,13-dibutyrate were much smaller and levels of CPI-17 (PKC-activated protein phosphatase inhibitor of 17 kDa) were much lower in the caudal artery than the aorta. The results suggest that both PKC and RhoK contribute independently to contractile responses to stimulation of α1D-adrenoceptors in the aorta. However, RhoK, but not PKC, participates in contractile responses to stimulation of α1A-adrenoceptors in the caudal artery. This difference may largely be due to differences between the two arteries in the extent to which PKC participates in contraction.Key words: vascular smooth muscle, α1-adrenoceptors, protein kinase C, rho kinase, phenylephrine.
Collapse
Affiliation(s)
- Irem Mueed
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | | | | | | |
Collapse
|
47
|
Bexis S, Vandeputte C, McCormick PA, Docherty JR. Deletion of inducible nitric oxide synthase decreases mesenteric vascular responsiveness in portal hypertensive mice. Eur J Pharmacol 2004; 499:325-33. [PMID: 15381055 DOI: 10.1016/j.ejphar.2004.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Revised: 07/29/2004] [Accepted: 08/03/2004] [Indexed: 02/09/2023]
Abstract
The effects of pre-hepatic portal hypertension were examined on the responsiveness of aorta and mesenteric artery from wild-type, inducible nitric oxide synthase knockout (iNOS-KO) and endothelial nitric oxide synthase knockout (eNOS-KO) mice. Mice were sham-operated or made portal hypertensive by creating a calibrated portal vein stenosis. Acetylcholine produced marked relaxations in phenylephrine (10 microM) contracted aorta and mesenteric artery from wild-type and iNOS-KO, both sham and portal hypertensive, but relaxations were abolished in vessels from eNOS-KO mice. There were no significant differences between sham and portal hypertensive animals within groups in the effects of acetylcholine. The potency of KCl was significantly increased in aorta and mesenteric artery from eNOS-KO mice. The maximum contraction to the alpha(1)-adrenoceptor agonist phenylephrine was significantly increased in aorta from eNOS-KO, as compared with wild-type mice. There were no significant differences between sham and portal hypertensive animals within each group in contractions of aorta to KCl or phenylephrine. However, in mesenteric artery, although portal hypertension did not change responsiveness in wild-type or eNOS-KO as compared to sham animals, the potency of phenylephrine was significantly reduced in portal hypertensive iNOS-KO mice as compared to shams. Hence, portal hypertension as compared to sham operation did not affect responses to vasoconstrictors in mouse aorta, but in mouse mesenteric artery portal hypertension affected vascular responses in iNOS-KO mice, suggesting that iNOS is involved in the mesenteric vascular response to portal vein ligation.
Collapse
Affiliation(s)
- Sotiria Bexis
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | | | | | |
Collapse
|
48
|
Taki N, Tanaka T, Zhang L, Suzuki F, Israilova M, Taniguchi T, Hiraizumi-Hiraoka Y, Shinozuka K, Kunitomo M, Muramatsu I. Alpha-1D adrenoceptors are involved in reserpine-induced supersensitivity of rat tail artery. Br J Pharmacol 2004; 142:647-56. [PMID: 15159276 PMCID: PMC1575040 DOI: 10.1038/sj.bjp.0705817] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. We examined reserpine-induced chemical denervation supersensitivity with special reference to alpha-1 adrenoceptor (AR) subtypes. 2. Chronic treatment with reserpine for 2 weeks depleted noradrenaline in the tail artery and spleen of rats. Noradrenaline in the thoracic aorta was negligible before and after reserpine treatment. 3. The treatment with reserpine produced supersensitivity in the contractile responses of the rat tail artery to phenylephrine, 5-HT and KCl, resulting in leftward shift of concentration-response curves (11.6-, 2.5- and 1.1-fold at EC(50) value, respectively). These results suggest a predominant sensitization of the alpha-1 AR-mediated response by reserpine treatment. 4. BMY 7378 at a concentration (30 nm) specific for blocking the alpha-1D AR subtype, but not KMD-3213 at a concentration (10 nm) selective for blocking the alpha-1A AR subtype, inhibited the supersensitivity of the phenylephrine-induced response in the reserpine-treated artery. On the other hand, the response to phenylephrine in reserpine-untreated artery was selectively inhibited by the same concentration of KMD-3213, but not by BMY 7378. Prazosin, a subtype-nonselective antagonist, blocked the responses to phenylephrine with the same potency, regardless of reserpine treatment. 5. In the thoracic aorta and spleen, no supersensitivity was produced in the responses to phenylephrine by reserpine treatment. 6. In a tissue segment-binding study using [(3)H]-prazosin, the total density and affinity of alpha-1 ARs in the rat tail artery were not changed by treatment with reserpine. However, alpha-1D AR with high affinity for BMY 7378 was significantly detected in reserpine-treated tail artery, in contrast to untreated artery. Decreases in alpha-1A AR with high affinity for KMD-3213 and alpha-1B AR with low affinities for KMD-3213 and BMY 7378 were also estimated in reserpine-treated tail artery. 7. Alpha-1D AR mRNA in rat tail artery increased to three-folds by reserpine treatment, whereas the levels of alpha-1A and 1B mRNAs were not significantly changed. 8. The present results suggest that chronic treatment with reserpine affects the expression of alpha-1 AR subtypes of rat tail artery and that the induction of alpha-1D ARs with high affinity for catecholamines is in part associated with reserpine-induced supersensitivity.
Collapse
Affiliation(s)
- Naoyuki Taki
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Takashi Tanaka
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Li Zhang
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Fumiko Suzuki
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Malika Israilova
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Takanobu Taniguchi
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Yasuko Hiraizumi-Hiraoka
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | - Kazumasa Shinozuka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo 663-8710, Japan
| | - Masaru Kunitomo
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo 663-8710, Japan
| | - Ikunobu Muramatsu
- Department of Pharmacology, School of Medicine, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
- Author for correspondence:
| |
Collapse
|
49
|
Jähnichen S, Eltze M, Pertz HH. Evidence that α1B-adrenoceptors are involved in noradrenaline-induced contractions of rat tail artery. Eur J Pharmacol 2004; 488:157-67. [PMID: 15044047 DOI: 10.1016/j.ejphar.2004.02.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2003] [Revised: 02/05/2004] [Accepted: 02/10/2004] [Indexed: 10/26/2022]
Abstract
The present study characterizes the alpha(1)-adrenoceptor subtypes mediating contractions to noradrenaline in isolated ring preparations of rat tail artery. Concentration-response (E/[A]) curves to noradrenaline were apparently monophasic (pEC(50) 6.47) but became biphasic in the presence of the selective alpha(1A)-adrenoceptor antagonist (+/-)-1,3,5-trimethyl-6-[[3-[4-((2,3-dihydro-2-hydroxymethyl)-1,4-benzodioxin-5-yl)-1-piperazinyl]propyl]amino]-2,4(1H,3H)-pyrimidinedione (B8805-033). Whereas the first phase of contraction to noradrenaline remained nearly unaffected in the presence of B8805-033 (0.03-3 microM), the second phase was concentration-dependently shifted to the right (pK(B) 8.06). In the presence of B8805-033 (3 microM), noradrenaline-induced contractions (pEC(50) 6.55) were antagonized in a competitive manner by prazosin (pK(B) 9.24), tamsulosin (pK(B) 8.55), 2-(2,6-dimethoxyphenoxyethyl)aminomethyl-1,4-benzodioxane (WB 4101; pK(B) 7.81), spiperone (pK(B) 7.69), 4-amino-2-[4-[1-(benzyloxycarbonyl)-2(S)-[[(1,1-dimethylethyl)amino]carbonyl]-piperazinyl]-6,7-dimethoxyquinazoline (L-765,314; pK(B) 7.31), 5-methylurapidil (pK(B) 6.55), 8-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-8-azaspiro[4.5]decane-7,9-dione (BMY 7378; pK(B) 6.43), and 8-[2-(1,4-benzodioxan-2-ylmethylamino)ethyl]-8-azaspiro[4.5]decane-7,9-dione (MDL 73005EF; pK(B) 5.71), and were also antagonized by 100 microM chloroethylclonidine. N-[2-(2-cyclopropylmethoxyphenoxy)ethyl]-5-chloro-alpha,alpha-dimethyl-1H-indole-3-ethanamine (RS-17053) behaved as a noncompetitive antagonist (apparent pA(2) 6.55). Antagonist affinities obtained under these experimental conditions correlated highly with affinities at native and cloned alpha(1B)-adrenoceptors. Pretreatment of arterial rings with B8805-033 (3 microM) followed by receptor inactivation with chloroethylclonidine (100 microM) yielded monophasic E/[A] curves to noradrenaline (pEC(50) 6.14). Noradrenaline-induced contractions were competitively antagonized by tamsulosin (pK(B) 10.32), 5-methylurapidil (pK(B) 8.66), RS-17053 (pK(B) 8.44), B8805-033 (pK(B) 7.87), BMY 7378 (pK(B) 6.54), and L-765,314 (pK(B) 6.41). Antagonist affinities obtained under these experimental conditions correlated highly with affinities at native and cloned alpha(1A)-adrenoceptors. It is concluded that the contraction to noradrenaline in rat tail artery is mediated by both alpha(1B)- and alpha(1A)-adrenoceptors, each component of contraction being separable by use of selective alpha(1A)-adrenoceptor blockade and alpha(1B)-adrenoceptor alkylation, respectively.
Collapse
Affiliation(s)
- Sven Jähnichen
- Institute of Pharmacy, Free University of Berlin, Königin-Luise-Str. 2+4, Berlin 14195, Germany
| | | | | |
Collapse
|
50
|
Hague C, Uberti MA, Chen Z, Hall RA, Minneman KP. Cell surface expression of alpha1D-adrenergic receptors is controlled by heterodimerization with alpha1B-adrenergic receptors. J Biol Chem 2004; 279:15541-9. [PMID: 14736874 DOI: 10.1074/jbc.m314014200] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
alpha(1)-Adrenergic receptors (ARs) belong to the large Class I G protein-coupled receptor superfamily and comprise three subtypes (alpha(1A), alpha(1B), and alpha(1D)). Previous work with heterologously expressed C-terminal green fluorescent protein (GFP)-tagged alpha(1)-ARs showed that alpha(1A)- and alpha(1B)-ARs localize to the plasma membrane, whereas alpha(1D)-ARs accumulate intracellularly. We recently showed that alpha(1D)- and alpha(1B)-ARs form heterodimers, whereas alpha(1D)- and alpha(1A)-ARs do not. Here, we examined the role of heterodimerization in regulating alpha(1D)-AR localization using both confocal imaging of GFP- or CFP-tagged alpha(1)-ARs and a luminometer-based surface expression assay in HEK293 cells. Co-expression with alpha(1B)-ARs caused alpha(1D)-ARs to quantitatively translocate to the cell surface, but co-expression with alpha(1A)-ARs did not. Truncation of the alpha(1B)-AR extracellular N terminus or intracellular C terminus had no effect on surface expression of alpha(1D)-ARs, suggesting primary involvement of the hydrophobic core. Co-transfection with an uncoupled mutant alpha(1B)-AR (Delta12alpha(1B)) increased both alpha(1D)-AR surface expression and coupling to norepinephrine-stimulated Ca(2+) mobilization. Finally, GFP-tagged alpha(1D)-ARs were not detected on the cell surface when expressed in rat aortic smooth muscle cells that express no endogenous ARs, but were almost exclusively localized on the surface when expressed in DDT(1)MF-2 cells, which express endogenous alpha(1B)-ARs. These studies demonstrate that alpha(1B)/alpha(1D)-AR heterodimerization controls surface expression and functional coupling of alpha(1D)-ARs, the N- and C-terminal domains are not involved in this interaction, and that alpha(1B)-AR G protein coupling is not required. These observations may be relevant to many other Class I G protein-coupled receptors, where the functional consequences of heterodimerization are still poorly understood.
Collapse
Affiliation(s)
- Chris Hague
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|