1
|
Tropea T, Mavichak W, Evangelinos A, Brennan-Richardson C, Cottrell EC, Myers JE, Johnstone ED, Brownbill P. Fetoplacental vascular effects of maternal adrenergic antihypertensive and cardioprotective medications in pregnancy. J Hypertens 2023; 41:1675-1687. [PMID: 37694528 PMCID: PMC10552840 DOI: 10.1097/hjh.0000000000003532] [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: 03/16/2023] [Revised: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 09/12/2023]
Abstract
Maternal cardiovascular diseases, including hypertension and cardiac conditions, are associated with poor fetal outcomes. A range of adrenergic antihypertensive and cardioprotective medications are often prescribed to pregnant women to reduce major maternal complications during pregnancy. Although these treatments are not considered teratogenic, they may have detrimental effects on fetal growth and development, as they cross the fetoplacental barrier, and may contribute to placental vascular dysregulation. Medication risk assessment sheets do not include specific advice to clinicians and women regarding the safety of these therapies for use in pregnancy and the potential off-target effects of adrenergic medications on fetal growth have not been rigorously conducted. Little is known of their effects on the fetoplacental vasculature. There is also a dearth of knowledge on adrenergic receptor activation and signalling within the endothelium and vascular smooth muscle cells of the human placenta, a vital organ in the maintenance of adequate blood flow to satisfy fetal growth and development. The fetoplacental circulation, absent of sympathetic innervation, and unique in its reliance on endocrine, paracrine and autocrine influence in the regulation of vascular tone, appears vulnerable to dysregulation by adrenergic antihypertensive and cardioprotective medications compared with the adult peripheral circulation. This semi-systematic review focuses on fetoplacental vascular expression of adrenergic receptors, associated cell signalling mechanisms and predictive consequences of receptor activation/deactivation by antihypertensive and cardioprotective medications.
Collapse
Affiliation(s)
- Teresa Tropea
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Weerawaroon Mavichak
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Angelos Evangelinos
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Charlotte Brennan-Richardson
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Elizabeth C. Cottrell
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Jenny E. Myers
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Edward D. Johnstone
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Paul Brownbill
- Maternal & Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester
- St Mary's Hospital, Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
2
|
da Silva SB, Feitosa SGD, de L Alves SM, Santos RCA, Dos Anjos JV, Araújo AV. A Concise and Useful Guide to Understand How Alpha1 Adrenoceptor Antagonists Work. Mini Rev Med Chem 2022; 22:2383-2405. [PMID: 35507746 DOI: 10.2174/1389557522666220504141949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/23/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
Adrenoceptors are the receptors for the catecholamines, adrenaline and noradrenaline. They are divided in α (α1 and α2) and β (β1, β2 and β3). α1-Adrenoceptors are subdivided in α1A, α1B and α1D. Most tissues express mixtures of α1-adrenoceptors subtypes, which appear to coexist in different densities and ratios, and in most cases their responses are probably due to the activation of more than one type. The three subtypes of α1-adrenoceptors are G-protein-coupled receptors (GPCR), specifically coupled to Gq/11. Additionally, the activation of these receptors may activate other signaling pathways or different components of these pathways, which leads to a great variety of possible cellular effects. The first clinically used α1 antagonist was Prazosin, for Systemic Arterial Hypertension (SAH). It was followed by its congeners, Terazosin and Doxazosin. Nowadays, there are many classes of α-adrenergic antagonists with different selectivity profiles. In addition to SAH, the α1-adrenoceptors are used for the treatment of Benign Prostatic Hyperplasia (BPH) and urolithiasis. This antagonism may be part of the mechanism of action of tricyclic antidepressants. Moreover, the activation of these receptors may lead to adverse effects such as orthostatic hypotension, similar to what happens with the antidepressants and with some antipsychotic. Structure-activity relationships can explain, in part, how antagonists work and how selective they can be for each one of the subtypes. However, it is necessary to develop new molecules which antagonize the α1-adrenoceptors or make chemical modifications in these molecules to improve the selectivity, pharmacokinetic profile and/or reduce the adverse effects of known drugs.
Collapse
Affiliation(s)
- Sidiane B da Silva
- Laboratório de Nutrição, Atividade Física e Plasticidade Fenotípica - Centro Acadêmico de Vitória - Universidade Federal de Pernambuco. R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| | - Sidney G D Feitosa
- Departamento de Química Fundamental - Universidade Federal de Pernambuco. Av. Jornalista Aníbal Fernandes, s/n, Cidade Universitária - Recife, PE, 50740-560, Brazil
| | - Silvia M de L Alves
- Laboratório de Nutrição, Atividade Física e Plasticidade Fenotípica - Centro Acadêmico de Vitória - Universidade Federal de Pernambuco. R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| | - Ruth C A Santos
- Laboratório de Nutrição, Atividade Física e Plasticidade Fenotípica - Centro Acadêmico de Vitória - Universidade Federal de Pernambuco. R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| | - Janaína V Dos Anjos
- Departamento de Química Fundamental - Universidade Federal de Pernambuco. Av. Jornalista Aníbal Fernandes, s/n, Cidade Universitária - Recife, PE, 50740-560, Brazil
| | - Alice V Araújo
- Núcleo de Saúde Pública, Centro Acadêmico de Vitória - Universidade Federal de Pernambuco R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| |
Collapse
|
3
|
Moraga FA, Reyes RV, Ebensperger G, López V, Llanos AJ. Enhanced Vasoconstriction Mediated by α 1-Adrenergic Mechanisms in Small Femoral Arteries in Newborn Llama and Sheep Gestated at Low and High Altitudes. Front Physiol 2021; 12:697211. [PMID: 34421636 PMCID: PMC8371382 DOI: 10.3389/fphys.2021.697211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/28/2021] [Indexed: 12/01/2022] Open
Abstract
The authors previously demonstrated that newborn llama (NBLL) express high levels of α1 adrenergic receptors, which provide a potent vasoconstriction response when compared with newborn sheep (NBSH) gestated at sea level. However, data regarding the impact of chronic gestational hypobaric hypoxia on α-adrenergic vasoconstriction in the neonatal life has not been studied. We evaluated if gestation under chronic hypobaric hypoxia modifies α1-adrenergic vasoconstrictor function in NBLL and NBSH. We compared the vasoconstrictor response induced by potassium and α-adrenergic stimuli in isolated small femoral arteries of NBLL and NBSH gestated at high altitude (HA; 3,600 m) or low altitude (LA; 580 m). The maximal contraction (RMAX) and potency (EC50) to potassium, noradrenaline (NA), and phenylephrine (PHE) were larger in HA-NBLL than LA-NBLL. RMAX to potassium, NA, and PHE were lower in HA-NBSH when compared with LA-NBSH and potency results were similar. Competitive blockade with prazosin showed that RNLL LA/HA have a similar pA2. In contrast, NBSH had increased pA2 values in HA when compared with LA. Finally, small femoral arteries denudated or treated with LNAME in LA and HA lacked NO or endothelium participation in response to PHE stimulation. In contrast, NBSH displayed that denudation or blockade with LNAME support NO or endothelium participation in response to PHE activation. In conclusion, HA chronic hypoxia enhances α1 adrenergic receptor activity in small femoral arteries in NBLL to a higher degree than NBSH, implying a higher vasoconstriction function.
Collapse
Affiliation(s)
- Fernando A Moraga
- Laboratorio de Fisiología, Hipoxia y Función Vascular, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Roberto V Reyes
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Centro Internacional de Estudios Andinos (INCAS), Universidad de Chile, Santiago, Chile
| | - Germán Ebensperger
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Centro Internacional de Estudios Andinos (INCAS), Universidad de Chile, Santiago, Chile
| | - Vasthi López
- Laboratorio de Fisiología, Hipoxia y Función Vascular, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Aníbal J Llanos
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Centro Internacional de Estudios Andinos (INCAS), Universidad de Chile, Santiago, Chile
| |
Collapse
|
4
|
Acute-on-chronic liver disease enhances phenylephrine-induced endothelial nitric oxide release in rat mesenteric resistance arteries through enhanced PKA, PI3K/AKT and cGMP signalling pathways. Sci Rep 2019; 9:6993. [PMID: 31061522 PMCID: PMC6502824 DOI: 10.1038/s41598-019-43513-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 04/15/2019] [Indexed: 12/15/2022] Open
Abstract
Acute-on-chronic liver disease is a clinical syndrome characterized by decompensated liver fibrosis, portal hypertension and splanchnic hyperdynamic circulation. We aimed to determine whether the alpha-1 agonist phenylephrine (Phe) facilitates endothelial nitric oxide (NO) release by mesenteric resistance arteries (MRA) in rats subjected to an experimental microsurgical obstructive liver cholestasis model (LC). Sham-operated (SO) and LC rats were maintained for eight postoperative weeks. Phe-induced vasoconstriction (in the presence/absence of the NO synthase –NOS- inhibitor L-NAME) and vasodilator response to NO donor DEA-NO were analysed. Phe-induced NO release was determined in the presence/absence of either H89 (protein kinase –PK- A inhibitor) or LY 294002 (PI3K inhibitor). PKA and PKG activities, alpha-1 adrenoceptor, endothelial NOS (eNOS), PI3K, AKT and soluble guanylate cyclase (sGC) subunit expressions, as well as eNOS and AKT phosphorylation, were determined. The results show that LC blunted Phe-induced vasoconstriction, and enhanced DEA-NO-induced vasodilation. L-NAME increased the Phe-induced contraction largely in LC animals. The Phe-induced NO release was greater in MRA from LC animals. Both H89 and LY 294002 reduced NO release in LC. Alpha-1 adrenoceptor, eNOS, PI3K and AKT expressions were unchanged, but sGC subunit expression, eNOS and AKT phosphorylation and the activities of PKA and PKG were higher in MRA from LC animals. In summary, these mechanisms may help maintaining splanchnic vasodilation and hypotension observed in decompensated LC.
Collapse
|
5
|
Ishida H, Saito SY, Hishinuma E, Kitayama T, Ishikawa T. Differential contribution of calcium channels to α 1-adrenoceptor-mediated contraction is responsible for diverse responses to cooling between rat tail and iliac arteries. Eur J Pharmacol 2018; 826:9-16. [PMID: 29458039 DOI: 10.1016/j.ejphar.2018.02.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/30/2018] [Accepted: 02/16/2018] [Indexed: 01/29/2023]
Abstract
Our previous studies have shown that α1-adrenoceptors, in addition to α2-adrenoceptors, are involved in enhanced contraction of cutaneous blood vessels during cooling. The present study aimed to elucidate the mechanism underlying it. In tail and iliac arteries isolated from rats, isometric contraction was measured using a myograph and the phosphorylation level of myosin phosphatase target subunit 1 (MYPT1) was quantified by western blotting. The phenylephrine-induced contraction was enhanced by cooling to 24 °C in tail arteries, but was suppressed in iliac arteries. Endothelium denudation or treatment with iberiotoxin enhanced the phenylephrine-induced contraction in tail arteries at 37 °C; however, neither affected the contraction at 24 °C. The phenylephrine-induced contraction at 37 °C was largely suppressed by nifedipine in iliac arteries, but only slightly in tail arteries. The Rho kinase inhibitor H-1152 largely suppressed the phenylephrine-induced contraction at 24 °C, but only slightly at 37 °C, in both arteries. The phosphorylation level of MYPT1 at Thr855 in tail arteries was increased by the cooling. Taken together, these results suggest the following mechanism in regard to cooling-induced enhancement of α1-adrenoceptor-mediated contraction in tail arteries: Cooling enhances the contraction of tail arteries via α1-adrenoceptor stimulation by reducing endothelium-dependent, large-conductance Ca2+-activated K+ channel-mediated relaxation and by inducing Rho kinase-mediated Ca2+ sensitization, although the latter occurs even in iliac arteries. A smaller contribution of voltage-dependent Ca2+ channels, which are largely suppressed by cooling, to α1-adrenoceptor-mediated contraction in tail arteries seems to be more crucially involved in the appearance of the enhanced contractile response to cooling.
Collapse
Affiliation(s)
- Hirotake Ishida
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shin-Ya Saito
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan.
| | - Eita Hishinuma
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Tomoaki Kitayama
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| |
Collapse
|
6
|
Raj RR, Subramani S. Phenylephrine Decreases Vascular Tension in Goat Arteries in Specific Circumstances. PLoS One 2016; 11:e0158551. [PMID: 27362703 PMCID: PMC4928870 DOI: 10.1371/journal.pone.0158551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/19/2016] [Indexed: 11/25/2022] Open
Abstract
Phenylephrine (PE) causes vasoconstriction through alpha adrenergic receptors. PE-induced vasodilatation has also been reported earlier in pre-constricted vessels. Here we demonstrate in spiral strips of goat arteries that addition of PE can decrease tone even from base-line levels (i.e. not pre-constricted) and show that this process requires nitric oxide (NO) and alpha adrenergic stimulation, but is cGMP-independent. Under control conditions, PE caused vasoconstriction, but under conditions where NO levels are higher, as with L-Arginine or sodium nitroprusside, PE decreased vessel tension. L-Arginine/PE combination was not able to decrease tension when alpha adrenoceptors were blocked with Phentolamine or endothelial nitric oxide synthase (eNOS) was blocked with Nω-Nitro-L-arginine (L-NNA). Propranolol, a beta blocker, was unable to prevent the reduction in tension by the L-Arginine/PE combination. Adrenaline and noradrenaline (and not isoproterenol) also reduced vessel tension in the presence of L-Arginine. Even when NO levels were not enhanced, relieving NO from having to stimulate the enzyme soluble guanylyl cyclase (sGC) (either by using sGC blockers, namely ODQ or methylene blue, or by enhancing cGMP levels (with sildenafil) which by negative feedback probably inhibits sGC) led to PE-induced reduction of vascular tension. PMA—phorbol myristate acetate—an agonist which stimulates Protein Kinase C was able to prevent the ability of PE to reduce vascular tension in a high NO environment. Our conclusion is that PE reduces vascular tension through alpha adrenoceptors if there is excess NO availability to activate a putative pathway. Though the reduction of vessel tone by PE is dependent on NO, it is independent of cGMP. Prior treatment with PMA or PE itself can prevent further PE-induced reduction of tension in a high NO environment. The results here suggest, counter-intuitively, that alpha blockers may be of help in the treatment of septic shock where nitric oxide levels are high.
Collapse
Affiliation(s)
- Renu R. Raj
- Department of Physiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Sathya Subramani
- Department of Physiology, Christian Medical College, Vellore, Tamil Nadu, India
- * E-mail:
| |
Collapse
|
7
|
Côco H, Pernomian L, Marchi KC, Gomes MS, de Andrade CR, Ramalho LNZ, Tirapelli CR, de Oliveira AM. Consequence of hyperhomocysteinaemia on α1-adrenoceptor-mediated contraction in the rat corpus cavernosum: the role of reactive oxygen species. ACTA ACUST UNITED AC 2016; 68:63-75. [PMID: 26725912 DOI: 10.1111/jphp.12486] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/12/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Our main objective was to investigate the mechanisms underlying the effects of hyperhomocysteinaemia (HHcy) on contractile response mediated by α1-adrenoceptors in the rat corpus cavernosum. METHODS Concentration-response curves for phenylephrine (PE) were obtained in strips of corpus cavernosum, in absence or after incubation with tiron, tempol or polyethylene glycol (PEG)-catalase combined or not with tempol. We also measured the superoxide anion (O2(-)) and hydrogen peroxide (H2O2) generation, superoxide dismutase (SOD) and catalase activity and α-actin expression in rat corpus cavernosum from both groups. KEY FINDINGS HHcy increased PE-induced contraction in cavernosal strips. Tiron, PEG-catalase or tempol increased PE-induced contraction in strips from control rats, but it was not altered by tiron or PEG-catalase in HHcy rats, whereas tempol reduced this response. The combination of PEG-catalase and tempol did not alter the contractile response to PE in both groups. HHcy increased O2(-) generation and SOD activity, whereas H2O2 concentration was reduced. Finally, HHcy did not alter catalase activity or expression of α-actin. CONCLUSIONS The major new finding from this study is that HHcy induced a marked increase in PE-induced contraction in rat corpus cavernosum by a mechanism that involves increased O2(-) generation and it could play a role in the pathogenesis of erectile dysfunction associated with HHcy.
Collapse
Affiliation(s)
- Hariane Côco
- Departament of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Larissa Pernomian
- Laboratory of Vascular Injury, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Katia C Marchi
- Departament of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Mayara S Gomes
- Laboratory of Vascular Injury, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Cláudia R de Andrade
- Department of Pathology and Legal Medicine, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Leandra N Z Ramalho
- Nucleus of Applied Bioprospection and Molecular Research, Faculdades Inta, Fortaleza, CE, Brazil
| | - Carlos R Tirapelli
- Laboratory of Pharmacology, Escola de Enfermagem de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Ana M de Oliveira
- Laboratory of Vascular Injury, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| |
Collapse
|
8
|
Couto GK, Davel AP, Brum PC, Rossoni LV. Double disruption of α2A- and α2C-adrenoceptors induces endothelial dysfunction in mouse small arteries: role of nitric oxide synthase uncoupling. Exp Physiol 2014; 99:1427-38. [DOI: 10.1113/expphysiol.2014.079236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Gisele K. Couto
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo SP Brazil
| | - Ana P. Davel
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas; Campinas SP Brazil
| | - Patrícia C. Brum
- School of Physical Education and Sport; University of São Paulo; São Paulo SP Brazil
| | - Luciana V. Rossoni
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo SP Brazil
| |
Collapse
|
9
|
Lin Q, Wang F, Yang R, Zheng X, Gao H, Zhang P. Effect of chronic restraint stress on human colorectal carcinoma growth in mice. PLoS One 2013; 8:e61435. [PMID: 23585898 PMCID: PMC3621827 DOI: 10.1371/journal.pone.0061435] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 03/13/2013] [Indexed: 12/31/2022] Open
Abstract
Stress alters immunological and neuroendocrinological functions. An increasing number of studies indicate that chronic stress can accelerate tumor growth, but its role in colorectal carcinoma (CRC) progression is not well understood. The aim of this study is to investigate the effects of chronic restraint stress (CRS) on CRC cell growth in nude mice and the possible underlying mechanisms. In this study, we showed that CRS increased the levels of plasma catecholamines including epinephrine (E) and norepinephrine (NE), and stimulated the growth of CRC cell-derived tumors in vivo. Treatment with the adrenoceptor (AR) antagonists phentolamine (PHE, α-AR antagonist) and propranolol (PRO, β-AR antagonist) significantly inhibited the CRS-enhanced CRC cell growth in nude mice. In addition, the stress hormones E and NE remarkably enhanced CRC cell proliferation and viability in culture, as well as tumor growth in vivo. These effects were antagonized by the AR antagonists PHE and PRO, indicating that the stress hormone-induced CRC cell proliferation is AR dependent. We also observed that the β-AR antagonists atenolol (ATE, β1- AR antagonist) and ICI 118,551 (ICI, β2- AR antagonist) inhibited tumor cell proliferation and decreased the stress hormone-induced phosphorylation of extracellular signal-regulated kinases-1/2 (ERK1/2) in vitro and in vivo. The ERK1/2 inhibitor U0126 also blocked the function of the stress hormone, suggesting the involvement of ERK1/2 in the tumor-promoting effect of CRS. We conclude that CRS promotes CRC xenograft tumor growth in nude mice by stimulating CRC cell proliferation through the AR signaling-dependent activation of ERK1/2.
Collapse
Affiliation(s)
- Qiang Lin
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Feifei Wang
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Rong Yang
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Xinmin Zheng
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Huibao Gao
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
- * E-mail: (PZ); (HBG)
| | - Ping Zhang
- Department of Biochemistry and Molecular Cell Biology, Institute of Medical Science, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
- * E-mail: (PZ); (HBG)
| |
Collapse
|
10
|
Su J, Laursen BE, Eskildsen-Helmond Y, Horsman MR, Simonsen U. The vascular-disrupting agent, combretastatin-A4-phosphate, enhances neurogenic vasoconstriction in rat small arteries. Eur J Pharmacol 2012; 695:104-11. [DOI: 10.1016/j.ejphar.2012.08.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 08/25/2012] [Accepted: 08/27/2012] [Indexed: 11/24/2022]
|
11
|
Pernomian L, Gomes MS, de Oliveira AM. Balloon catheter injury abolishes phenylephrine-induced relaxation in the rat contralateral carotid. Br J Pharmacol 2011; 163:770-81. [PMID: 21323906 PMCID: PMC3111679 DOI: 10.1111/j.1476-5381.2011.01275.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 12/12/2010] [Accepted: 01/08/2011] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The consequences of compensatory responses to balloon catheter injury in rat carotid artery, on phenylephrine-induced relaxation and contraction in the contralateral carotid artery were studied. EXPERIMENTAL APPROACH Relaxation and contraction concentration-response curves for phenylephrine were obtained for contralateral carotid arteries in the presence of indomethacin (COX inhibitor), SC560 (COX-1 inhibitor), SC236 (COX-2 inhibitor) or 4-hydroxytetramethyl-L-piperidine-1-oxyl (tempol; superoxide dismutase mimetic). Reactive oxygen species were measured in carotid artery endothelial cells fluorimetrically with dihydroethidium. KEY RESULTS Phenylephrine-induced relaxation was abolished in contralateral carotid arteries from operated rats (E(max) = 0.01 ± 0.004 g) in relation to control (E(max) = 0.18 ± 0.005 g). Phenylephrine-induced contractions were increased in contralateral arteries (E(max) = 0.54 ± 0.009 g) in relation to control (E(max) = 0.38 ± 0.014 g). SC236 restored phenylephrine-induced relaxation (E(max) = 0.17 ± 0.004 g) and contraction (E(max) = 0.34 ± 0.018 g) in contralateral arteries. Tempol restored phenylephrine-induced relaxation (E(max) = 0.19 ± 0.012 g) and contraction (E(max) = 0.42 ± 0.014 g) in contralateral arteries, while apocynin did not alter either relaxation (E(max) = 0.01 ± 0.004 g) or contraction (E(max) = 0.54 ± 0.009 g). Dihydroethidium fluorescence was increased in contralateral samples (18 882 ± 435 U) in relation to control (10 455 ± 303 U). SC236 reduced the fluorescence in contralateral samples (8250 ± 365 U). CONCLUSIONS AND IMPLICATIONS Balloon catheter injury abolished phenylephrine-induced relaxation and increased phenylephrine-induced contraction in contralateral carotid arteries, through O(2) (-) derived from COX-2.
Collapse
Affiliation(s)
- L Pernomian
- Department of Pharmacology, School of Medicine of Ribeirão Preto, Laboratory of Pharmacology, University of São PauloRibeirão Preto, São Paulo, Brazil
| | - MS Gomes
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, Laboratory of Pharmacology, University of São PauloRibeirão Preto, São Paulo, Brazil
| | - AM de Oliveira
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, Laboratory of Pharmacology, University of São PauloRibeirão Preto, São Paulo, Brazil
| |
Collapse
|
12
|
Subramaniam G, Achike FI, Mustafa MR. Effect of acidosis on the mechanism(s) of insulin-induced vasorelaxation in normal Wistar–Kyoto (WKY) rat aorta. ACTA ACUST UNITED AC 2009; 155:70-5. [DOI: 10.1016/j.regpep.2009.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 03/29/2009] [Accepted: 04/05/2009] [Indexed: 11/29/2022]
|
13
|
Cankar K, Finderle Ž, Štrucl M. The effect of α-adrenoceptor agonists and L-NMMA on cutaneous postocclusive reactive hyperemia. Microvasc Res 2009; 77:198-203. [DOI: 10.1016/j.mvr.2008.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 09/29/2008] [Accepted: 10/27/2008] [Indexed: 11/17/2022]
|
14
|
Potentiation by yohimbine of alpha-adrenoceptor-mediated vasoconstriction in response to clonidine in the rabbit ear vein. Eur J Pharmacol 2008; 589:201-5. [PMID: 18534573 DOI: 10.1016/j.ejphar.2008.04.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 03/31/2008] [Accepted: 04/22/2008] [Indexed: 11/20/2022]
Abstract
We investigated the pharmacological profile of the vasoconstrictive response to clonidine in the isolated rabbit ear vein, and compared the characteristics of clonidine with those of noradrenaline and moxonidine. The maximal vasoconstrictive responses to clonidine and moxonidine in the rabbit ear vein were 35.94+/-11.18% and 88.78+/-11.54% of the maximum response to noradrenaline, respectively. Prazosin 0.1 microM inhibited the vasoconstriction induced by lower concentrations of noradrenaline, and the concentration-dependent response curve for noradrenaline was significantly shifted to the right by 1 microM prazosin. Yohimbine (0.1 and 0.5 microM) only decreased the vasoconstrictive response to lower concentrations of noradrenaline, but did not affect the response to higher concentrations. Vasoconstrictive responses to lower but not higher concentrations of clonidine and moxonidine were inhibited by 0.1 microM yohimbine. In contrast, the same concentration of yohimbine significantly potentiated the maximal response to a high concentration of clonidine by 24.06%. In isolated rabbit ear vein pretreated with 0.1 microM yohimbine, prazosin competitively inhibited the concentration-response curve for clonidine with a pA(2) value of 8.05+/-0.06. We conclude that clonidine acts mainly on alpha(2)-adrenoceptors to produce vasoconstriction in the rabbit ear vein; however, in the preparation pretreated with yohimbine, the clonidine-induced vasoconstriction is mediated via alpha(1)-adrenoceptors and its maximal vasoconstriction is significantly potentiated.
Collapse
|
15
|
Seya Y, Fukuda T, Isobe K, Kawakami Y, Takekoshi K. Effect of norepinephrine on RhoA, MAP kinase, proliferation and VEGF expression in human umbilical vein endothelial cells. Eur J Pharmacol 2006; 553:54-60. [PMID: 17070516 DOI: 10.1016/j.ejphar.2006.09.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 09/01/2006] [Accepted: 09/18/2006] [Indexed: 01/27/2023]
Abstract
Norepinephrine is a well known major vasoconstricting factor. Recent reports suggest that norepinephrine, in addition to acting as a vasoconstricting factor, may also play several additional roles in endothelial cells. These include: 1] induction of NO release. It has been demonstrated that a small GTP-binding protein, Rho, and its downstream effecter, Rho kinase (ROCK), negatively regulate endothelial nitric oxide synthase (eNOS) production. However, it is not known whether ROCK is directly involved in norepinephrine-induced NO release. 2] Norepinephrine is reported to induce a mitogenic effect, but whether MAPKs are involved in this process is unknown. 3] Recently, we demonstrated an increase in vascular endothelial growth factor (VEGF) mRNA/protein expression in human pheochromocytoma tissue in comparison to normal adrenomedullary tissue. Thus, it is reasonable to speculate that norepinephrine may stimulate the level of VEGF mRNA. The aim of the present study was to clarify the role of norepinephrine and related endothelial adrenoceptor systems in various pathophysiological conditions, such as hypertension and in particular pheochromocytoma, using human umbilical vein endothelial cells (HUVEC). Norepinephrine-induced RhoA attenuation, through cAMP/protein kinase A (PKA) activation coupled with beta-adrenoceptors, may lead to eNOS activation in acute conditions. Norepinephrine stimulates the production of VEGF mRNA through cAMP/PKA activation coupled with beta-adrenoceptors. Norepinephrine stimulates a mitogenic effect through ERK activation coupled with the alpha(1)-adrenoceptor. In conclusion, norepinephrine stimulates eNOS activity via RhoA attenuation, VEGF mRNA synthesis and mitogenic activity in endothelial cells. We propose that an excess of norepinephrine can lead to endothelial dysfunction due to these aforementioned processes.
Collapse
Affiliation(s)
- Yumi Seya
- Department of Clinical Pathology, University of Tsukuba (Institute of Clinical Medicine), Molecular Laboratory Medicine, Graduate School of Comprehensive Human Sciences, 1-1-1 Tennoudai, Tsukuba, 305-8575, Japan
| | | | | | | | | |
Collapse
|
16
|
Mendez E, Calzada C, Ocharan E, Sierra A, Castillo C, Ramirez I, Meaney E, Meaney A, Asbun J, Miliar A, Herrera J, Ceballos G. Differential expression of α1-adrenergic receptor subtypes in coronary microvascular endothelial cells in culture. Eur J Pharmacol 2006; 546:127-33. [PMID: 16904663 DOI: 10.1016/j.ejphar.2006.06.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 06/27/2006] [Accepted: 06/28/2006] [Indexed: 11/17/2022]
Abstract
It has been postulated that in blood vessels under alpha(1)-related stimulation, the endothelial intracellular calcium concentration ([Ca(2+)](i)) increases, which is necessary to induce nitric oxide synthesis, is the result of an increase in vascular smooth muscle, which subsequently, flows into the endothelial cells through gap junctions and it is not the result of a direct adrenergic stimulation of endothelial receptors. Others, however, postulate that endothelial alpha(1D)-adrenoceptors, have a direct effect on nitric oxide synthesis. In order to clarify this phenomena, in this work we analyzed the presence of alpha(1)receptor subtypes and their functional association with nitric oxide synthesis in rat coronary microvascular endothelial cells in culture, with pharmacological, immunological and reverse transcriptase polymerase chain reaction approaches. Our results show the presence and functional coupling with nitric oxide synthesis of alpha(1A) and alpha(1D)-adrenoceptor subtypes. alpha(1B)-adrenoceptor subtype is not coupled with nitric oxide production.
Collapse
Affiliation(s)
- Enrique Mendez
- Laboratorio Multidisciplinario, Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, D. F. 11340, México
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
de Andrade CR, Fukada SY, Olivon VC, de Godoy MAF, Haddad R, Eberlin MN, Cunha FQ, de Souza HP, Laurindo FRM, de Oliveira AM. Alpha1D-adrenoceptor-induced relaxation on rat carotid artery is impaired during the endothelial dysfunction evoked in the early stages of hyperhomocysteinemia. Eur J Pharmacol 2006; 543:83-91. [PMID: 16828078 DOI: 10.1016/j.ejphar.2006.06.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Revised: 05/27/2006] [Accepted: 06/02/2006] [Indexed: 11/20/2022]
Abstract
Hyperhomocysteinemia is a known risk factor for cardiovascular diseases, but the underlying mechanisms of this pathology are complex. We aimed to evaluate the effect of hyperhomocysteinemia in vasorelaxations induced by alpha(1D)-adrenoceptor agonists. Vascular reactivity of rat carotid artery to the alpha-adrenoceptor agonist, phenylephrine, was enhanced in hyperhomocysteinemia. Mechanical removal of endothelium did not modify the carotid responsiveness to phenylephrine, compared to control. Phenylephrine induces endothelium-dependent relaxation, in the presence of 5-methyl urapidil (alpha(1A)-adrenoceptor antagonist). We hypothesised that endothelial-relaxant alpha(1)-adrenoceptors are impaired by hyperhomocysteinemia. Incubation with prazosin (selective alpha(1)-adrenoceptor antagonist) or BMY7378 (8-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-8-azaspiro[4,5]decane-7, 9-dione dihydrochloride) (selective alpha(1D)-adrenoceptor antagonist), similarly inhibited phenylephrine-induced relaxations in both control and hyperhomocysteinemic carotids. Immunohistochemistry showed enhanced immunoreactivity for eNOS and iNOS in hyperhomocysteinemic rats. In carotid arteries from hyperhomocysteinemic rats there was a decrease in superoxide dismutase activity and enhanced superoxide anion production. We conclude that alpha(1D)-adrenoceptors mediate endothelium-dependent relaxation triggered by phenylephrine in rat carotid artery and affect the final tone. Furthermore, the enhanced phenylephrine-induced contraction in carotid artery due to hyperhomocysteinemia is endothelium-dependent and involves a loss of the inhibitory effect of relaxant alpha(1D)-adrenoceptors by reducing NO biodisponibility.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Adrenergic alpha-Antagonists/pharmacology
- Animals
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/physiopathology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Enzyme Inhibitors/pharmacology
- Hyperhomocysteinemia/chemically induced
- Hyperhomocysteinemia/metabolism
- Hyperhomocysteinemia/physiopathology
- Male
- NG-Nitroarginine Methyl Ester/pharmacology
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/biosynthesis
- Phenylephrine/pharmacology
- Piperazines/pharmacology
- Prazosin/pharmacology
- Rats
- Rats, Wistar
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/metabolism
- Superoxide Dismutase/metabolism
- Superoxides/metabolism
- Vasoconstriction/drug effects
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Claudia Roberta de Andrade
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
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
|
19
|
Li QJ, Janssen LJ. Membrane currents in canine bronchial artery and their regulation by excitatory agonists. Am J Physiol Lung Cell Mol Physiol 2002; 282:L1358-65. [PMID: 12003793 DOI: 10.1152/ajplung.00421.2001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The bronchial vasculature plays an important role in airway physiology and pathophysiology. We investigated the ion currents in canine bronchial smooth muscle cells using patch-clamp techniques. Sustained outward K(+) current evoked by step depolarizations was significantly inhibited by tetraethylamonium (1 and 10 mM) or by charybdotoxin (10(-6) M) but was not significantly affected by 4-aminopyridine (1 or 5 mM), suggesting that it was primarily a Ca(2+)-activated K(+) current. Consistent with this, the K(+) current was markedly increased by raising external Ca(2+) to 4 mM but was decreased by nifedipine (10(-6) M) or by removing external Ca(2+). When K(+) currents were blocked (by Cs(+) in the pipette), step depolarizations evoked transient inward currents with characteristics of L-type Ca(2+) current as follows: 1) activation that was voltage dependent (threshold and maximal at -50 and -10 mV, respectively); 2) inactivation that was time dependent and voltage dependent (voltage causing 50% maximal inactivation of -26 +/- 22 mV); and 3) blockade by nifedipine (10(-6) M). The thromboxane mimetic U-46619 (10(-6) M) caused a marked augmentation of outward K(+) current (as did 10 mM caffeine) lasting only 10-20 s; this was followed by significant suppression of the K(+) current lasting several minutes. Phenylephrine (10(-4) M) also suppressed the K(+) current to a similar degree but did not cause the initial transient augmentation. None of these three agonists elicited inward current of any kind. We conclude that bronchial arterial smooth muscle expresses Ca(2+)-dependent K(+) channels and voltage-dependent Ca(2+) channels and that its excitation does not involve activation of Cl(-) channels.
Collapse
MESH Headings
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
- Animals
- Bronchial Arteries/cytology
- Bronchial Arteries/drug effects
- Bronchial Arteries/physiology
- Caffeine/pharmacology
- Calcium/metabolism
- Calcium Channel Blockers/pharmacology
- Calcium Channels, L-Type/metabolism
- Cell Separation
- Dogs
- In Vitro Techniques
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Patch-Clamp Techniques
- Potassium/metabolism
- Potassium Channel Blockers/pharmacology
- Potassium Channels, Calcium-Activated/drug effects
- Potassium Channels, Calcium-Activated/metabolism
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Q J Li
- Asthma Research Group, Father Sean O'Sullivan Research Center, St. Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada L8N 4A6
| | | |
Collapse
|
20
|
Seasholtz TM, Cai G, Wang HY, Friedman E. Selected contribution: effects of ischemia-reperfusion on vascular contractility and alpha(1)-adrenergic-receptor signaling in the rat tail artery. J Appl Physiol (1985) 2001; 91:1004-10. [PMID: 11457820 DOI: 10.1152/jappl.2001.91.2.1004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To determine the effects of ischemia-reperfusion (I/R) on alpha(1)-adrenergic-receptor (alpha(1)-AR) functions, alpha(1)-AR-mediated contraction, inositol phosphate (IP) accumulation, and alpha(1)-AR-G protein coupling were examined in the tail arteries of anesthetized rats after 60 min of ischemia and 60 min of reperfusion. The contractile response to norepinephrine (NE) was significantly increased after I/R, whereas the contractile response to KCl remained unchanged. This was accompanied by a 69% increase in NE-stimulated IP accumulation. Furthermore, receptor-stimulated coupling of alpha(1a)-AR to G alpha(q/11) proteins was increased, whereas the coupling of alpha(1b)-AR or alpha(1d)-AR to their G proteins was not altered by I/R. These changes in vascular alpha(1)-AR function occurred without concurrent alteration in expression levels of membrane alpha(1)-AR subtypes or in the associated G proteins. These data demonstrate that I/R increases alpha(1a)-AR-G(q/11) protein coupling and alpha(1)-AR-stimulated IP accumulation in the tail artery. The alterations in alpha(1)-AR signaling are associated with and may underlie the enhanced contractile response of the tail artery to adrenergic stimulation after I/R.
Collapse
Affiliation(s)
- T M Seasholtz
- Department of Pharmacology and Physiology, MCP Hahnemann School of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | | | | | |
Collapse
|
21
|
Tuttle JL, Falcone JC. Nitric oxide release during alpha1-adrenoceptor-mediated constriction of arterioles. Am J Physiol Heart Circ Physiol 2001; 281:H873-81. [PMID: 11454593 DOI: 10.1152/ajpheart.2001.281.2.h873] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined endothelial modulation of norepinephrine (NE)-mediated constriction in isolated, cannulated, first-order arterioles from skeletal muscle of rats. Acute arteriolar constrictor responses to NE (10(-9) to 10(-7) M) were significantly (P < 0.05) enhanced after either endothelial denudation or inhibition of nitric oxide synthase with NG-monomethyl-L-arginine (10(-4) M, 30 min). In contrast, arteriolar constrictions to NE were not different after treatment with either the cyclooxygenase inhibitor diclofenac (10(-6) M, 30 min) or the K+-channel blocker tetrabutylammonium (5 x 10(-5) M, 30 min). We also measured arteriolar responses to the vasoconstrictor PGF2alpha; responses were not altered by any of the experimental treatments, which indicates that this phenomenon is not ubiquitous to all vasoconstricting agents. Mechanistically, we examined vascular smooth muscle (VSM) and endothelial cell calcium. Both NE and PGF2alpha significantly increased VSM cell calcium measurements; however, endothelial cell calcium was significantly increased with NE or phenylephrine (an alpha1-adrenergic agonist) but not with PGF2alpha or UK-14304 (an alpha2-adrenergic agonist). Together these findings suggest that in rat cremaster first-order arterioles, NE stimulates an increase in VSM calcium via adrenergic receptors with subsequent increase in endothelial cell calcium, possibly via stimulation of alpha1-adrenergic receptors on the arteriolar endothelium. The burst in endothelial cell calcium may then lead to the production of nitric oxide, which diffuses to the VSM, attenuates constriction, and maintains at least some minimal level of blood flow.
Collapse
Affiliation(s)
- J L Tuttle
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky 40292, USA
| | | |
Collapse
|
22
|
Janssen LJ, Lu-Chao H, Netherton S. Responsiveness of canine bronchial vasculature to excitatory stimuli and to cooling. Am J Physiol Lung Cell Mol Physiol 2001; 280:L930-7. [PMID: 11290517 DOI: 10.1152/ajplung.2001.280.5.l930] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Changes in bronchial vascular tone, in part due to cooling during ventilation, may contribute to altered control of airflow during airway inflammation, asthma, and exercise-induced bronchoconstriction. We investigated the responses of canine bronchial vasculature to excitatory stimuli and cooling. Electrical stimulation evoked contractions in only some (8 of 88) tissues; these were phentolamine sensitive and augmented by N(omega)-nitro-L-arginine. However, sustained contractions were evoked in all tissues by phenylephrine [concentration evoking a half-maximal response (EC(50)) approximately 2 microM] or the thromboxane A(2) mimetic U-46619 (EC(50) approximately 5 nM) and less so by beta,gamma-methylene-ATP or histamine. Cooling to room temperature markedly suppressed ( approximately 75%) adrenergic responses but had no significant effect against U-46619 responses. Adrenergic responses, but not those to U-46619, were accompanied by an increase in intracellular Ca(2+) concentration. Chelerythrine (protein kinase C antagonist) markedly antagonized adrenergic responses (mean maxima reduced 39% in artery and 86% in vein) but had no significant effect against U-46619, whereas genistein (a nonspecific tyrosine kinase inhibitor) essentially abolished responses to both agonists. We conclude that cooling of the airway wall dramatically interferes with adrenergic control of bronchial perfusion but has little effect on thromboxane-mediated vasoconstriction.
Collapse
Affiliation(s)
- L J Janssen
- Asthma Research Group, Firestone Institute for Respiratory Health, St. Joseph's Hospital and Department of Medicine, McMaster University, Hamilton, Ontario, Canada L8N 3Z5.
| | | | | |
Collapse
|
23
|
McMillon RK, Townsley MI. Alpha-adrenergic vasoreactivity of canine intrapulmonary bronchial arteries in pacing-induced heart failure. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:H1392-402. [PMID: 10516174 DOI: 10.1152/ajpheart.1999.277.4.h1392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We hypothesized that pacing-induced congestive heart failure alters alpha-adrenergic constriction in intrapulmonary bronchial arteries. Cumulative dose responses to norepinephrine (NE), phenylephrine (PE), acetylcholine (ACh) and sodium nitroprusside (SNP) were determined in pressurized vessel segments. ED(50) values for NE and PE were higher for control (-5.34 +/- 0.09 and -4.27 +/- 0.08 M, respectively) vs. paced (-5.73 +/- 0.10 and -5.06 +/- 0.28 M, respectively) groups. Prazosin increased the ED(50) values for NE and PE in both control and paced groups. Yohimbine decreased NE ED(50) in the control group only. Endothelium removal or nitric oxide synthase (NOS) inhibition decreased control but not paced NE ED(50). Maximum vasodilation and sensitivity (i.e., -ED(50) values) were decreased for ACh but were similar for SNP in paced vs. control groups. Secondary segments were more reactive than paired primary segments in both groups, although pacing effects on ED(50) were unrelated to branching order. In conclusion, adrenergic constriction of canine intrapulmonary bronchial arteries is predominantly mediated via alpha(1)-adrenoreceptors and is enhanced after pacing. Endothelium-derived relaxing factor(s) normally opposes alpha-adrenergic vasoconstriction but not after pacing in this vasculature.
Collapse
Affiliation(s)
- R K McMillon
- Department of Physiology, University of South Alabama, Mobile, Alabama 36688, USA
| | | |
Collapse
|
24
|
Nishina H, Ozaki T, Hanson MA, Poston L. Mechanisms of noradrenaline-induced vasorelaxation in isolated femoral arteries of the neonatal rat. Br J Pharmacol 1999; 127:809-12. [PMID: 10433486 PMCID: PMC1566105 DOI: 10.1038/sj.bjp.0702641] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/1999] [Revised: 03/31/1999] [Accepted: 04/06/1999] [Indexed: 11/09/2022] Open
Abstract
Isolated arteries from the femoral circulation of Wistar rats mounted on a small vessel myograph demonstrated age related tension development to noradrenaline (NA, 1 x 10(-8) - 5 x 10(-5) M) day 20 greater than day 10 (P<0.005); day 100 greater than day 20 (P<0.001) and depolarizing potassium (125 mM) buffer day 20 greater than day 10 (P<0.001). NA evoked dilatation in femoral arteries from neonatal rats (10 days) when added to unstimulated vessels or to those preconstricted with the thromboxane mimetic, U46619. Relaxation to NA was inhibited by L-NAME (0.1 mM) (P<0.001), endothelial removal (P<0.001) and the alpha2-adrenoceptor antagonist, yohimbine (0.1 microM) (P<0.001). Alpha1- or beta-adrenoceptor antagonism was without effect. Relaxation was evoked in femoral arteries of the 10-day-old rats by the alpha2-adrenoceptor agonist UK14304 (1 x 10(-8) - 5 x 10(-5) M). This relaxation was also abolished by L-NAME (0.1 mM) (P<0.001) or endothelial removal (P<0.001). Alpha2-adrenoceptor-mediated vasorelaxation was the predominant response to NA stimulation in femoral arteries of the neonatal rat. These responses were endothelium-dependent and were NO-mediated.
Collapse
Affiliation(s)
- H Nishina
- Department of Obstetrics and Gynaecology, University College London
| | | | | | | |
Collapse
|
25
|
Segarra G, Medina P, Revert F, Masiá S, Vila JM, Such L, Aldasoro M. Modulation of adrenergic contraction of dog pulmonary arteries by nitric oxide and prostacyclin. GENERAL PHARMACOLOGY 1999; 32:583-9. [PMID: 10382861 DOI: 10.1016/s0306-3623(98)00285-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The aim of this work was to investigate the influence of endothelium-derived nitric oxide and prostaglandins on the contractile responses of isolated dog pulmonary arteries to electrical field stimulation and noradrenaline. Electrical field stimulation (1-8 Hz, 20 v, 0.25 ms duration, for 30 s) produced frequency-dependent contractions that were abolished by tetrodotoxin, guanethidine and, prazosin (all at 10(-6) M). Noradrenaline induced concentration-dependent contractions with an EC50, of 1.85 x 10(-6) M. The increases in tension induced by electrical stimulation and noradrenaline were of greater magnitude in arteries denuded of endothelium. In segments with endothelium, N(G)-nitro-L-arginine methyl ester (10(-4) M) or indomethacin (10(-5) M) had no effects on the basal tone, but significantly enhanced the neurogenic and noradrenaline-induced contractions. The potentiation by N(G)-nitro-L-arginine methyl ester of electrical stimulation-induced contractile responses was partially reversed by L-arginine (10(-4) M). In the presence of N(G)-nitro-L-arginine methyl ester together with indomethacin the electrical stimulation-induced contractile responses were higher than those obtained when only N(G)-nitro-L-arginine methyl ester or indomethacin was used. N(G)-nitro-L-arginine methyl ester and indomethacin did not influence neurogenic-induced contractile responses of endothelium-denuded arteries. The results suggest that endothelial cells of isolated dog pulmonary arteries depress the contractile response to electrical field stimulation of intramural nerves and that endothelium-derived dilator prostaglandins and nitric oxide may interact to inhibit contractile effects of adrenergic stimulation.
Collapse
Affiliation(s)
- G Segarra
- Department of Physiology, University of Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
26
|
Zschauer AO, Sielczak MW, Wanner A. Altered contractile sensitivity of isolated bronchial artery to phenylephrine in ovalbumin-sensitized rabbits. J Appl Physiol (1985) 1999; 86:1721-7. [PMID: 10233140 DOI: 10.1152/jappl.1999.86.5.1721] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We tested the hypothesis that atopy and/or allergic lung inflammation enhances alpha1-adrenoceptor-mediated contractions of the bronchial artery. Bronchial arterial resistance vessels were isolated from rabbits that had undergone either systemic ovalbumin (OVA) sensitization followed by saline aerosol challenge (OVA/saline rabbits), or OVA sensitization followed by OVA aerosol challenge (OVA/OVA rabbits), or no sensitization followed by saline aerosol challenge (control rabbits). In OVA/OVA rabbits, bronchoalveolar lavage and lung histology revealed lymphocytic and eosinophilic inflammation. Arterial rings were contracted with phenylephrine (PE). In endothelium-intact arteries isolated from OVA/saline and OVA/OVA rabbits, PE responsiveness was enhanced compared with that of arteries isolated from controls. The nitric oxide synthase (NOS) inhibitor NG-nitro-L-arginine methyl ester increased the contractile response to PE in all three experimental groups to a similar degree, suggesting that depressed NOS activity was not involved in the enhanced PE responsiveness in OVA/saline and OVA/OVA rabbits. After endothelium removal, arteries from OVA/saline and control rabbits showed similar PE responsiveness, indicating that the enhancement of PE responsiveness was endothelium dependent, possibly due to an endothelial constricting factor. In OVA/OVA rabbits, endothelium-denuded arteries showed decreased PE responsiveness compared with the other two groups; this difference was abolished by NG-nitro-L-arginine methyl ester. We conclude that systemic sensitization with OVA per se enhances PE-induced contractions of isolated bronchial arteries in rabbits by an endothelium-dependent mechanism and that allergic lung inflammation attenuates this effect by increased nonendothelial NOS activity.
Collapse
Affiliation(s)
- A O Zschauer
- Division of Pulmonary and Critical Care Medicine, Mount Sinai Medical Center, University of Miami School of Medicine, Miami Beach, Florida 33140, USA.
| | | | | |
Collapse
|