1
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [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: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
2
|
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
|
3
|
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: 0.3] [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
|
4
|
Horie H, Hisatome I, Kurata Y, Yamamoto Y, Notsu T, Adachi M, Li P, Kuwabara M, Sakaguchi T, Kinugasa Y, Miake J, Koba S, Tsuneto M, Shirayoshi Y, Ninomiya H, Ito S, Kitakaze M, Yamamoto K, Yoshikawa Y, Nishimura M. α1-Adrenergic receptor mediates adipose-derived stem cell sheet-induced protection against chronic heart failure after myocardial infarction in rats. Hypertens Res 2021; 45:283-291. [PMID: 34853408 DOI: 10.1038/s41440-021-00802-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/08/2021] [Accepted: 10/15/2021] [Indexed: 11/09/2022]
Abstract
Cell-based therapy using adipose-derived stem cells (ADSCs) has emerged as a novel therapeutic approach to treat heart failure after myocardial infarction (MI). The purpose of this study was to determine whether inhibition of α1-adrenergic receptors (α1-ARs) in ADSCs attenuates ADSC sheet-induced improvements in cardiac functions and inhibition of remodeling after MI. ADSCs were isolated from fat tissues of Lewis rats. In in vitro studies using cultured ADSCs, we determined the mRNA levels of vascular endothelial growth factor (VEGF)-A and α1-AR under normoxia or hypoxia and the effects of norepinephrine and an α1-blocker, doxazosin, on the mRNA levels of angiogenic factors. Hypoxia increased α1-AR and VEGF mRNA levels in ADSCs. Norepinephrine further increased VEGF mRNA expression under hypoxia; this effect was abolished by doxazosin. Tube formation of human umbilical vein endothelial cells was promoted by conditioned media of ADSCs treated with the α1 stimulant phenylephrine under hypoxia but not by those of ADSCs pretreated with phenylephrine plus doxazosin. In in vivo studies using rats with MI, transplanted ADSC sheets improved cardiac functions, facilitated neovascularization, and suppressed fibrosis after MI. These effects were abolished by doxazosin treatment. Pathway analysis from RNA sequencing data predicted significant upregulation of α1-AR mRNA expression in transplanted ADSC sheets and the involvement of α1-ARs in angiogenesis through VEGF. In conclusion, doxazosin abolished the beneficial effects of ADSC sheets on rat MI hearts as well as the enhancing effect of norepinephrine on VEGF expression in ADSCs, indicating that ADSC sheets promote angiogenesis and prevent cardiac dysfunction and remodeling after MI via their α1-ARs.
Collapse
Affiliation(s)
- Hiromu Horie
- Division of Cardiovascular Surgery, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Yasutaka Kurata
- Department of Physiology II, Kanazawa Medical University, Uchinada, Japan.
| | - Yasutaka Yamamoto
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Tomomi Notsu
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Maaya Adachi
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Peili Li
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Masanari Kuwabara
- Intensive Care Unit and Department of Cardiology, Toranomon Hospital, Tokyo, Japan
| | - Takuki Sakaguchi
- Division of Medical Education, Department of Medical Education, Tottori University Faculty of Medicine, Yonago, Japan
| | - Yoshiharu Kinugasa
- Division of Cardiovascular Medicine, Department of Molecular Medicine and Therapeutics, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Junichiro Miake
- Department of Pharmacology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Satoshi Koba
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Motokazu Tsuneto
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Yasuaki Shirayoshi
- Division of Regenerative Medicine and Therapeutics, Tottori University Graduate School of Medical Science, Yonago, Japan
| | - Haruaki Ninomiya
- Department of Biological Regulation, Tottori University Faculty of Medicine, Yonago, Japan
| | - Shin Ito
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | | | - Kazuhiro Yamamoto
- Division of Cardiovascular Medicine, Department of Molecular Medicine and Therapeutics, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yasushi Yoshikawa
- Division of Cardiovascular Surgery, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Motonobu Nishimura
- Division of Cardiovascular Surgery, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
5
|
Luo Z, Li M, Ye M, Ji P, Lou X, Huang J, Yao K, Zhao Y, Zhang H. Effect of Electrical Stimulation of Cervical Sympathetic Ganglia on Intraocular Pressure Regulation According to Different Circadian Rhythms in Rats. Invest Ophthalmol Vis Sci 2021; 61:40. [PMID: 32976562 PMCID: PMC7521184 DOI: 10.1167/iovs.61.11.40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose The purpose of this study was to investigate the relationship between circadian rhythm and intraocular pressure (IOP), and to explore whether electrical stimulation of cervical sympathetic ganglia (SCG) can regulate IOP via neurotransmitter distribution around the Schlemm's canal (SC) in rats. Methods Sprague Dawley rats were housed under normal (N-normal), constant dark (N-dark), and constant light (N-light) rhythms (n = 6 per group). Electrical stimulation (intermittent wave [20 hertz {Hz}, 2 mA, 10 minutes]) was used to stimulate the SCG. Atropine sulfate eye gel was applied three times a day. DiI was injected into the SCG and anterior chamber. The cross-sectional area and circumference of SC were evaluated using hematoxylin-eosin staining. Immunofluorescence staining was used to evaluate dopamine-β-hydroxylase (DβH) expression in SC endothelial (SCE) cells. Results N-Dark increased the IOP, decreased the cross-sectional area of SC, and increased DβH levels in SCE cells. Nerve projection between SC and SCG was detected, and electrical stimulation of SCG upregulated DβH expression in SCE cells. Under normal and constant light rhythms, electrical stimulation of SCG increased DβH and decreased the cross-sectional area and circumference of SC, while simultaneously increasing IOP and decreasing IOP fluctuations. After paralyzing the ciliary muscles, electrical stimulation of SCG decreased the cross-sectional area and circumference of SC under normal and constant light rhythms. Conclusions N-Dark increased DβH in SCE cells, reduced the cross-sectional area of SC, and increased IOP. Under the normal and light rhythms, electrical stimulation of SCG increased DβH in SCE cells, reduced the cross-sectional area and circumference of SC, and in turn elevated IOP and decreased IOP fluctuations.
Collapse
Affiliation(s)
- Zhaoxia Luo
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mu Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Ye
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingting Ji
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaotong Lou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingqiu Huang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Yao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Involvement of the Catecholamine Pathway in Glioblastoma Development. Cells 2021; 10:cells10030549. [PMID: 33806345 PMCID: PMC7998903 DOI: 10.3390/cells10030549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive tumor of the central nervous system (CNS). The standard of care improves the overall survival of patients only by a few months. Explorations of new therapeutic targets related to molecular properties of the tumor are under way. Even though neurotransmitters and their receptors normally function as mediators of interneuronal communication, growing data suggest that these molecules are also involved in modulating the development and growth of GBM by acting on neuronal and glioblastoma stem cells. In our previous DNA CpG methylation studies, gene ontology analyses revealed the involvement of the monoamine pathway in sequential GBM. In this follow-up study, we quantitated the expression levels of four selected catecholamine pathway markers (alpha 1D adrenergic receptor-ADRA1D; adrenergic beta receptor kinase 1 or G protein-coupled receptor kinase 2-ADRBK1/GRK2; dopamine receptor D2-DRD2; and synaptic vesicle monoamine transporter-SLC18A2) by immunohistochemistry, and compared the histological scores with the methylation levels within the promoters + genes of these markers in 21 pairs of sequential GBM and in controls. Subsequently, we also determined the promoter and gene methylation levels of the same markers in an independent database cohort of sequential GBM pairs. These analyses revealed partial inverse correlations between the catecholamine protein expression and promoter + gene methylation levels, when the tumor and control samples were compared. However, we found no differences in the promoter + gene methylation levels of these markers in either our own or in the database primary-recurrent GBM pairs, despite the higher protein expression of all markers in the primary samples. This observation suggests that regulation of catecholamine expression is only partially related to CpG methylation within the promoter + gene regions, and additional mechanisms may also influence the expression of these markers in progressive GBM. These analyses underscore the involvement of certain catecholamine pathway markers in GBM development and suggest that these molecules mediating or modulating tumor growth merit further exploration.
Collapse
|
7
|
Alves JQ, Pernomian L, Silva CD, Gomes MS, de Oliveira AM, da Silva RS. Vascular tone and angiogenesis modulation by catecholamine coordinated to ruthenium. RSC Med Chem 2020; 11:497-510. [PMID: 33479651 DOI: 10.1039/c9md00573k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/12/2020] [Indexed: 01/11/2023] Open
Abstract
Catecholamines participate in angiogenesis, an important tumor development process. However, the way catecholamines interact with their receptors has not been completely elucidated, and doubts still remain as to whether these interactions occur between catechol and/or amine sites and particular amino acid residues on the catecholamine receptors. To evaluate how catechol and amine groups contribute to angiogenesis, we immobilized the catechol site through ruthenium ion (Ru) coordination, to obtain species with the general formula [Ru(NH3)4(catecholamine-R)]Cl. We then assessed the angiogenic activity of the complexes in a chorioallantoic membrane model (CAM) and examined vascular reactivity and calcium mobilization in rat aortas and vascular cells. [Ru(NH3)4(catecholamine-R)]Cl acted as partial agonists and/or antagonists of their respective receptors and induced calcium mobilization. [Ru(NH3)4(isoproterenol)]+ [Ru(NH3)4(noradrenaline)]+, and [Ru(NH3)4(adrenaline)]+ behaved as antiangiogenic complexes, whereas [Ru(NH3)4(dopamine)]+ proved to be a proangiogenic complex. In conclusion, catecholamines and [Ru(NH3)4(catecholamine-R)]Cl can modulate angiogenesis, and catechol group availability can modify the way these complexes impact the vascular tone, suggesting that catecholamines and their receptors interact differently after catecholamine coordination to ruthenium.
Collapse
Affiliation(s)
- Jacqueline Querino Alves
- Faculty of Philosophy , Sciences and Letters of Ribeirão Preto - University of São Paulo (USP) , Department of Chemistry , Avenida Bandeirantes, 3900 , postal code 14.040-901 , Ribeirão Preto , São Paulo , Brazil
| | - Laena Pernomian
- Faculty of Pharmaceutical Sciences of Ribeirão Preto (FCFRP) - University of São Paulo (USP) , Department of Physics and Chemistry , Avenida do Café, s/n , postal code 14.040-903 , Ribeirão Preto , São Paulo , Brazil .
| | - Cássia Dias Silva
- Faculty of Philosophy , Sciences and Letters of Ribeirão Preto - University of São Paulo (USP) , Department of Chemistry , Avenida Bandeirantes, 3900 , postal code 14.040-901 , Ribeirão Preto , São Paulo , Brazil
| | - Mayara Santos Gomes
- Faculty of Pharmaceutical Sciences of Ribeirão Preto (FCFRP) - University of São Paulo (USP) , Department of Physics and Chemistry , Avenida do Café, s/n , postal code 14.040-903 , Ribeirão Preto , São Paulo , Brazil .
| | - Ana Maria de Oliveira
- Faculty of Pharmaceutical Sciences of Ribeirão Preto (FCFRP) - University of São Paulo (USP) , Department of Physics and Chemistry , Avenida do Café, s/n , postal code 14.040-903 , Ribeirão Preto , São Paulo , Brazil .
| | - Roberto Santana da Silva
- Faculty of Philosophy , Sciences and Letters of Ribeirão Preto - University of São Paulo (USP) , Department of Chemistry , Avenida Bandeirantes, 3900 , postal code 14.040-901 , Ribeirão Preto , São Paulo , Brazil.,Faculty of Pharmaceutical Sciences of Ribeirão Preto (FCFRP) - University of São Paulo (USP) , Department of Physics and Chemistry , Avenida do Café, s/n , postal code 14.040-903 , Ribeirão Preto , São Paulo , Brazil .
| |
Collapse
|
8
|
Methodological Approach to Use Fresh and Cryopreserved Vessels as Tools to Analyze Pharmacological Modulation of the Angiogenic Growth. J Cardiovasc Pharmacol 2017; 68:230-40. [PMID: 27631438 DOI: 10.1097/fjc.0000000000000407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The sprouting of new vessels is greatly influenced by the procedure chosen. We sought to optimize the experimental conditions of the angiogenic growth of fresh and cryopreserved vessels cultured in Matrigel with the aim to use this system to analyze the pharmacological modulation of the process. Segments of second-order branches of rat mesenteric resistance arteries, thoracic aorta of rat or mouse, and cryopreserved rat aorta and human femoral arteries were cultured in Matrigel for 7-21 days in different mediums, as well as in the absence of endothelial or adventitia layer. Quantification of the angiogenic growth was performed by either direct measurement of the mean length of the neovessels or by calcein AM staining and determination of fluorescence intensity and area. Fresh and cryopreserved arterial rings incubated in Matrigel exhibited a spontaneous angiogenic response that was strongly accelerated by fetal calf serum. Addition of vascular endothelial growth factor, fibroblast growth factor, endothelial growth factor, or recombinant insulin-like growth factor failed to increase aortic sprouting, unless all were added together. Removal of adventitia, but not the endothelial layer, abrogated the angiogenic response of aortic rings. Determination of the mean neovessel length is an easy and accurate method to quantify the angiogenic growth devoid of confounding factors, such as inclusion of other cellular types surrounding the neovessels. Activity of a α1-adrenoceptor agonist (phenylephrine) and its inhibition by a selective antagonist (prazosin) were analyzed to prove the usefulness of the Matrigel system to evaluate the pharmacological modulation of the angiogenic growth.
Collapse
|
9
|
Drummond PD, Dawson LF, Wood FM, Fear MW. Up-regulation of α 1-adrenoceptors in burn and keloid scars. Burns 2017; 44:582-588. [PMID: 29089212 DOI: 10.1016/j.burns.2017.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/29/2017] [Accepted: 09/17/2017] [Indexed: 12/30/2022]
Abstract
Stimulation of α1-adrenoceptors evokes inflammatory cytokine production, boosts neurogenic inflammation and pain, and influences cellular migration and proliferation. Hence, these receptors may play a role both in normal and abnormal wound healing. To investigate this, the distribution of α1-adrenoceptors in skin biopsies of burn scars (N=17), keloid scars (N=12) and unscarred skin (N=17) was assessed using immunohistochemistry. Staining intensity was greater on vascular smooth muscle in burn scars than in unscarred tissue, consistent with heightened expression of α1-adrenoceptors. In addition, expression of α1-adrenoceptors was greater on dermal nerve fibres, blood vessels and fibroblasts in keloid scars than in either burn scars or unscarred skin. These findings suggest that increased vascular expression of α1-adrenoceptors could alter circulatory dynamics both in burn and keloid scars. In addition, the augmented expression of α1-adrenoceptors in keloid tissue may contribute to processes that produce or maintain keloid scars, and might be a source of the uncomfortable sensations often associated with these scars.
Collapse
Affiliation(s)
- Peter D Drummond
- Centre for Research on Chronic Pain and Inflammatory Diseases, Murdoch University, Perth, Western Australia, Australia.
| | - Linda F Dawson
- Centre for Research on Chronic Pain and Inflammatory Diseases, Murdoch University, Perth, Western Australia, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Australia; The Fiona Wood Foundation, Perth, Western Australia, Australia; The Burns Service of Western Australia, WA Dept of Health, WA, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Surgery, University of Western Australia, Australia; The Fiona Wood Foundation, Perth, Western Australia, Australia
| |
Collapse
|
10
|
Identification of 42 Genes Linked to Stage II Colorectal Cancer Metastatic Relapse. Int J Mol Sci 2016; 17:ijms17050598. [PMID: 27136531 PMCID: PMC4881437 DOI: 10.3390/ijms17050598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer mortality. Metastasis remains the primary cause of CRC death. Predicting the possibility of metastatic relapse in early-stage CRC is of paramount importance to target therapy for patients who really need it and spare those with low-potential of metastasis. Ninety-six stage II CRC cases were stratified using high-resolution array comparative genomic hybridization (aCGH) data based on a predictive survival algorithm and supervised clustering. All genes included within the resultant copy number aberrations were each interrogated independently at mRNA level using CRC expression datasets available from public repositories, which included 1820 colon cancers, and 167 normal colon tissues. Reduced mRNA expression driven by copy number losses and increased expression driven by copy number gains revealed 42 altered transcripts (29 reduced and 13 increased transcripts) associated with metastatic relapse, short disease-free or overall survival, and/or epithelial to mesenchymal transition (EMT). Resultant genes were classified based on gene ontology (GO), which identified four functional enrichment groups involved in growth regulation, genomic integrity, metabolism, and signal transduction pathways. The identified 42 genes may be useful for predicting metastatic relapse in stage II CRC. Further studies are necessary to validate these findings.
Collapse
|
11
|
Saraf R, Mahmood F, Amir R, Matyal R. Neuropeptide Y is an angiogenic factor in cardiovascular regeneration. Eur J Pharmacol 2016; 776:64-70. [PMID: 26875634 DOI: 10.1016/j.ejphar.2016.02.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/27/2016] [Accepted: 02/09/2016] [Indexed: 12/13/2022]
Abstract
In diabetic cardiomyopathy, there is altered angiogenic signaling and increased oxidative stress. As a result, anti-angiogenic and pro-inflammatory pathways are activated. These disrupt cellular metabolism and cause fibrosis and apoptosis, leading to pathological remodeling. The autonomic nervous system and neurotransmitters play an important role in angiogenesis. Therapies that promote angiogenesis may be able to relieve the pathology in these disease states. Neuropeptide Y (NPY) is the most abundantly produced and expressed neuropeptide in the central and peripheral nervous systems in mammals and plays an important role in promoting angiogenesis and cardiomyocyte remodeling. It produces effects through G-protein-coupled Y receptors that are widely distributed and also present on the myocardium. Some of these receptors are also involved in diseased states of the heart. NPY has been implicated as a potent growth factor, causing cell proliferation in multiple systems while the NPY3-36 fragment is selective in stimulating angiogenesis and cardiomyocyte remodeling. Current research is focusing on developing a drug delivery mechanism for NPY to prolong therapy without having significant systemic consequences. This could be a promising innovation in the treatment of diabetic cardiomyopathy and ischemic heart disease.
Collapse
Affiliation(s)
- Rabya Saraf
- Department of Surgery, Division of Cardiac Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Feroze Mahmood
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Rabia Amir
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Robina Matyal
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
12
|
Pernomian L, Gomes MS, Restini CBA, Pupo AS, de Oliveira AM. Cross-talk with β2 -adrenoceptors enhances ligand affinity properties from endothelial alpha1 D -adrenoceptors that mediates carotid relaxation. ACTA ACUST UNITED AC 2013; 65:1337-46. [PMID: 23927472 DOI: 10.1111/jphp.12105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 06/18/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVES Our main objectives were to investigate the affinity properties of endothelial and muscular α1D -adrenoceptors and to characterize the cross-talk between endothelial α1D -adrenoceptors and β2 -adrenoceptors in rat carotid. METHODS Relaxation and contraction concentration-response curves for phenylephrine (α1 -adrenergic agonist) were obtained in carotid rings in absence or presence of increasing concentrations of BMY7378 (α1D -adrenergic antagonist), combined or not with increasing concentration of ICI-118,551 (β2 -adrenergic antagonist). Schild analysis was used to estimate the affinity constant from pA2 values of BMY7378. KEY FINDINGS BMY7378 produced an unsurmountable antagonism on phenylephrine-induced relaxation but a surmountable antagonism on phenylephrine-induced contraction. BMY7378 potency was higher in inhibiting the relaxation than the contraction induced by phenylephrine because the rightward shifts induced by BMY7378 were greater in the relaxation. The apparent pA2 value for BMY7378 in phenylephrine-induced relaxation was greater than in contraction. When combined with ICI-118,551, BMY7378 yielded a surmountable antagonism on phenylephrine-induced relaxation and presented a pA2 value similar to that obtained in phenylephrine-induced contraction. CONCLUSIONS Endothelial α1D -adrenoceptors, which mediates rat carotid relaxation, present high ligand affinity because of the cross-talk with β2 -adrenoceptors, which explains the higher potency of phenylephrine in inducing relaxation than contraction and the atypical unsurmountable antagonism produced by BMY7378 on phenylephrine-induced relaxation.
Collapse
Affiliation(s)
- Larissa Pernomian
- Faculty of Pharmaceutical Sciences from Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
13
|
van Doorn WG, Çelikel FG, Pak C, Harkema H. Delay of iris flower senescence by cytokinins and jasmonates. PHYSIOLOGIA PLANTARUM 2013; 148:105-20. [PMID: 22974423 DOI: 10.1111/j.1399-3054.2012.01690.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/10/2012] [Indexed: 05/18/2023]
Abstract
It is not known whether tepal senescence in Iris flowers is regulated by hormones. We applied hormones and hormone inhibitors to cut flowers and isolated tepals of Iris × hollandica cv. Blue Magic. Treatments with ethylene or ethylene antagonists indicated lack of ethylene involvement. Auxins or auxin inhibitors also did not change the time to senescence. Abscisic acid (ABA) hastened senescence, but an inhibitor of ABA synthesis (norflurazon) had no effect. Gibberellic acid (GA3 ) slightly delayed senescence in some experiments, but in other experiments it was without effect, and gibberellin inhibitors [ancymidol or 4-hydroxy-5-isopropyl-2-methylphenyltrimethyl ammonium chloride-1-piperidine carboxylate (AMO-1618)] were ineffective as well. Salicylic acid (SA) also had no effect. Ethylene, auxins, GA3 and SA affected flower opening, therefore did reach the flower cells. Jasmonates delayed senescence by about 2.0 days. Similarly, cytokinins delayed senescence by about 1.5-2.0 days. Antagonists of the phosphatidylinositol signal transduction pathway (lithium), calcium channels (niguldipine and verapamil), calmodulin action [fluphenazine, trifluoroperazine, phenoxybenzamide and N-(6-aminohexyl)-5-chloro-1-naphtalenesulfonamide hydrochloride (W-7)] or protein kinase activity [1-(5-isoquinolinesulfonyl)-2-methylpiperazine hydrochloride (H-7), N-[2-(methylamino)ethyl]-5-isoquinolinesulfonamide hydrochloride (H-8) and N-(2-aminoethyl)-5-isoquinolinesulfonamide dihydrochloride (H-9)] had no effect on senescence, indicating no role of a few common signal transduction pathways relating to hormone effects on senescence. The results indicate that tepal senescence in Iris cv. Blue Magic is not regulated by endogenous ethylene, auxin, gibberellins or SA. A role of ABA can at present not be excluded. The data suggest the hypothesis that cytokinins and jasmonates are among the natural regulators.
Collapse
Affiliation(s)
- Wouter G van Doorn
- Agrotechnology and Food Sciences Group (AFSG), Wageningen University Research Centre, Wageningen, The Netherlands.
| | | | | | | |
Collapse
|
14
|
Sarkar C, Chakroborty D, Basu S. Neurotransmitters as regulators of tumor angiogenesis and immunity: the role of catecholamines. J Neuroimmune Pharmacol 2012; 8:7-14. [PMID: 22886869 DOI: 10.1007/s11481-012-9395-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022]
Abstract
The growing tumor employs various strategies to establish its growth, progression and spread in the host. Angiogenesis or formation of new blood vessels from existing ones and escape from immune surveillance are the two critical steps that ensure proper establishment and growth of the newly formed tumor. Thus understanding the novel pathways associated with tumor angiogenesis and immunity may lead to the development of newer therapeutic strategies using the regulators of these pathways to improve patient outcomes. These two pivotal steps in the process of tumorigenesis are governed by plethora of endogenous factors. The neuroendocrine molecules, which include the catecholamine neurotransmitters, dopamine, norepinephrine and epinephrine are of growing interest considering their varied and diverse regulatory roles both in the process of tumor angiogenesis and tumor immunity. This review focuses on the emerging roles of catecholamines in modulating tumor angiogenesis and immunity, and also discusses the probable molecular mechanisms of their actions. Understanding of this new group of endogenous regulators of tumor growth may lead to the development of newer therapeutic approaches for the treatment of cancer.
Collapse
Affiliation(s)
- Chandrani Sarkar
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
15
|
Musilli C, Karam JP, Paccosi S, Muscari C, Mugelli A, Montero-Menei CN, Parenti A. Pharmacologically active microcarriers for endothelial progenitor cell support and survival. Eur J Pharm Biopharm 2012; 81:609-16. [PMID: 22561954 DOI: 10.1016/j.ejpb.2012.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 04/13/2012] [Accepted: 04/18/2012] [Indexed: 12/29/2022]
Abstract
The regenerative potential of endothelial progenitor cell (EPC)-based therapies is limited due to poor cell viability and minimal retention following application. Neovascularization can be improved by means of scaffolds supporting EPCs. The aim of the present study was to investigate whether human early EPCs (eEPCs) could be efficiently cultured on pharmacologically active microcarriers (PAMs), made with poly(d,l-lactic-coglycolic acid) and coated with adhesion/extracellular matrix molecules. They may serve as a support for stem cells and may be used as cell carriers providing a controlled delivery of active protein such as the angiogenic factor, vascular endothelial growth factor-A (VEGF-A). eEPC adhesion to fibronectin-coated PAMs (FN-PAMs) was assessed by means of microscopic evaluation and by means of Alamar blue assay. Phospho ERK(1/2) and PARP-1 expression was measured by means of Western blot to assess the survival effects of FN-PAMs releasing VEGF-A (FN-VEGF-PAMs). The Alamar blue assay or a modified Boyden chamber assay was employed to assess proliferative or migratory capacity, respectively. Our data indicate that eEPCs were able to adhere to empty FN-PAMs within a few hours. FN-VEGF-PAMs increased the ability of eEPCs to adhere to them and strongly supported endothelial-like phenotype and cell survival. Moreover, the release of VEGF-A by FN-PAMs stimulated in vitro HUVEC migration and proliferation. These data strongly support the use of PAMs for supporting eEPC growth and survival and for stimulating resident mature human endothelial cells.
Collapse
Affiliation(s)
- Claudia Musilli
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
Seasonal changes in patterns of gene expression in avian song control brain regions. PLoS One 2012; 7:e35119. [PMID: 22529977 PMCID: PMC3329558 DOI: 10.1371/journal.pone.0035119] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 03/13/2012] [Indexed: 01/23/2023] Open
Abstract
Photoperiod and hormonal cues drive dramatic seasonal changes in structure and function of the avian song control system. Little is known, however, about the patterns of gene expression associated with seasonal changes. Here we address this issue by altering the hormonal and photoperiodic conditions in seasonally-breeding Gambel's white-crowned sparrows and extracting RNA from the telencephalic song control nuclei HVC and RA across multiple time points that capture different stages of growth and regression. We chose HVC and RA because while both nuclei change in volume across seasons, the cellular mechanisms underlying these changes differ. We thus hypothesized that different genes would be expressed between HVC and RA. We tested this by using the extracted RNA to perform a cDNA microarray hybridization developed by the SoNG initiative. We then validated these results using qRT-PCR. We found that 363 genes varied by more than 1.5 fold (>log2 0.585) in expression in HVC and/or RA. Supporting our hypothesis, only 59 of these 363 genes were found to vary in both nuclei, while 132 gene expression changes were HVC specific and 172 were RA specific. We then assigned many of these genes to functional categories relevant to the different mechanisms underlying seasonal change in HVC and RA, including neurogenesis, apoptosis, cell growth, dendrite arborization and axonal growth, angiogenesis, endocrinology, growth factors, and electrophysiology. This revealed categorical differences in the kinds of genes regulated in HVC and RA. These results show that different molecular programs underlie seasonal changes in HVC and RA, and that gene expression is time specific across different reproductive conditions. Our results provide insights into the complex molecular pathways that underlie adult neural plasticity.
Collapse
|
17
|
Letavernier B, Zafrani L, Nassar D, Perez J, Levi C, Bellocq A, Mesnard L, Sachon E, Haymann JP, Aractingi S, Faussat AM, Baud L, Letavernier E. Calpains Contribute to Vascular Repair in Rapidly Progressive Form of Glomerulonephritis: Potential Role of Their Externalization. Arterioscler Thromb Vasc Biol 2012; 32:335-42. [DOI: 10.1161/atvbaha.111.240242] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Béatrice Letavernier
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Lara Zafrani
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Dany Nassar
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Joëlle Perez
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Charlène Levi
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Agnès Bellocq
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Laurent Mesnard
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Emmanuelle Sachon
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Jean-Philippe Haymann
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Selim Aractingi
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Anne-Marie Faussat
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Laurent Baud
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Emmanuel Letavernier
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| |
Collapse
|
18
|
Jensen BC, Swigart PM, Montgomery MD, Simpson PC. Functional alpha-1B adrenergic receptors on human epicardial coronary artery endothelial cells. Naunyn Schmiedebergs Arch Pharmacol 2010; 382:475-82. [PMID: 20857090 PMCID: PMC2991196 DOI: 10.1007/s00210-010-0558-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 08/30/2010] [Indexed: 11/25/2022]
Abstract
Alpha-1-adrenergic receptors (α1-ARs) regulate coronary arterial blood flow by binding catecholamines, norepinephrine (NE), and epinephrine (EPI), causing vasoconstriction when the endothelium is disrupted. Among the three α1-AR subtypes (α1A, α1B, and α1D), the α1D subtype predominates in human epicardial coronary arteries and is functional in human coronary smooth muscle cells (SMCs). However, the presence or function of α1-ARs on human coronary endothelial cells (ECs) is unknown. Here we tested the hypothesis that human epicardial coronary ECs express functional α1-ARs. Cultured human epicardial coronary artery ECs were studied using quantitative real-time reverse transcription polymerase chain reaction, radioligand binding, immunoblot, and (3)H-thymidine incorporation. The α1B-subtype messenger ribonucleic acid (mRNA) was predominant in cultured human epicardial coronary ECs (90-95% of total α1-AR mRNA), and total α1-AR binding density in ECs was twice that in coronary SMCs. Functionally, NE and EPI through the α1B subtype activated extracellular signal-regulated kinase (ERK) in ECs, stimulated phosphorylation of EC endothelial nitric oxide synthase (eNOS), and increased deoxyribonucleic acid (DNA) synthesis. These results are the first to demonstrate α1-ARs on human coronary ECs and indicate that the α1B subtype is predominant. Our findings provide another potential mechanism for adverse cardiac effects of drug antagonists that nonselectively inhibit all three α1-AR subtypes.
Collapse
Affiliation(s)
- Brian C Jensen
- Cardiology Division, VA Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | |
Collapse
|
19
|
Tilan J, Kitlinska J. Sympathetic Neurotransmitters and Tumor Angiogenesis-Link between Stress and Cancer Progression. JOURNAL OF ONCOLOGY 2010; 2010:539706. [PMID: 20508839 PMCID: PMC2874925 DOI: 10.1155/2010/539706] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 03/18/2010] [Indexed: 01/11/2023]
Abstract
Recent evidence supports a longstanding hypothesis that chronic stress can influence tumor growth and progression. It has been shown that sympathetic neurotransmitters, such as catecholamines and neuropeptides, can affect both cancer cell growth and tumor vascularization. Depending on neurotransmitter and type of tumor, these effects can be both stimulatory and inhibitory. Norepinephrine (NE) and epinephrine (E) are potent stimulators of vascularization, acting both by inducing the release of angiogenic factors from tumor cells and directly on endothelial cell (EC) functions. As a result, activation of the adrenergic system increases growth of various types of tumors and has been shown to mediate stress-induced augmentation of tumor progression. Dopamine (DA), on the other hand, interferes with VEGF signaling in endothelial cells, blocks its angiogenic functions and inhibits tumor growth. Another sympathetic neurotransmitter coreleased with NE, neuropeptide Y (NPY), directly stimulates angiogenesis. However, proangiogenic actions of NPY can be altered by its direct effect on tumor cell proliferation and survival. In consequence, NPY can either stimulate or inhibit tumor growth, depending on tumor type. Hence, sympathetic neurotransmitters are powerful modulators of tumor growth and can become new targets in cancer therapy.
Collapse
Affiliation(s)
- Jason Tilan
- Department of Physiology & Biophysics, Georgetown University, Basic Science Building 231A, 3900 Reservoir Rd., NW, Washington, DC 20007, USA
| | - Joanna Kitlinska
- Department of Physiology & Biophysics, Georgetown University, Basic Science Building 231A, 3900 Reservoir Rd., NW, Washington, DC 20007, USA
| |
Collapse
|
20
|
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: 141] [Impact Index Per Article: 9.4] [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
|
21
|
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: 3.2] [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
|