1
|
Lin L, Zicheng L, Shaohua G. Post-Acute Myocardial Infarction Heart Failure Core Genes and Relevant Signaling Pathways. J Cardiovasc Pharmacol 2023; 82:480-488. [PMID: 37678296 DOI: 10.1097/fjc.0000000000001481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
ABSTRACT There is increasing concern about heart failure after myocardial infarction and the current clinical treatment measures for ventricular remodeling. Herein, we present the results of differential gene analysis, pathway enrichment analysis, and characteristic gene screening. Our study identifies 4 core genes ( KLRC2 , SNORD105 , SNORD45B , and RNU5A-1 ) associated with post-acute myocardial infarction (AMI) heart failure. The authors discuss the significance of the identified core genes, their potential implications in immune dysfunction and heart failure, and their relevance to disease regulatory genes. The study concludes by emphasizing the importance of clinical relevance in molecular research and suggests potential therapeutic targets for post-AMI heart failure.
Collapse
Affiliation(s)
- Ling Lin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ling Zicheng
- Department of Interal Medicine, Weiting Community Health Center of Suzhou Industrial Park, Suzhou, Jiangsu, China; and
| | - Gu Shaohua
- Department of Nephrology, Kunshan Third Hospital, Suzhou, China
| |
Collapse
|
2
|
Mortensen NP, Pathmasiri W, Snyder RW, Caffaro MM, Watson SL, Patel PR, Beeravalli L, Prattipati S, Aravamudhan S, Sumner SJ, Fennell TR. Oral administration of TiO 2 nanoparticles during early life impacts cardiac and neurobehavioral performance and metabolite profile in an age- and sex-related manner. Part Fibre Toxicol 2022; 19:3. [PMID: 34986857 PMCID: PMC8728993 DOI: 10.1186/s12989-021-00444-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/23/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nanoparticles (NPs) are increasingly incorporated in everyday products. To investigate the effects of early life exposure to orally ingested TiO2 NP, male and female Sprague-Dawley rat pups received four consecutive daily doses of 10 mg/kg body weight TiO2 NP (diameter: 21 ± 5 nm) or vehicle control (water) by gavage at three different pre-weaning ages: postnatal day (PND) 2-5, PND 7-10, or PND 17-20. Cardiac assessment and basic neurobehavioral tests (locomotor activity, rotarod, and acoustic startle) were conducted on PND 20. Pups were sacrificed at PND 21. Select tissues were collected, weighed, processed for neurotransmitter and metabolomics analyses. RESULTS Heart rate was found to be significantly decreased in female pups when dosed between PND 7-10 and PND 17-20. Females dosed between PND 2-5 showed decrease acoustic startle response and when dosed between PND 7-10 showed decreased performance in the rotarod test and increased locomotor activity. Male pups dosed between PND 17-20 showed decreased locomotor activity. The concentrations of neurotransmitters and related metabolites in brain tissue and the metabolomic profile of plasma were impacted by TiO2 NP administration for all dose groups. Metabolomic pathways perturbed by TiO2 NP administration included pathways involved in amino acid and lipid metabolism. CONCLUSION Oral administration of TiO2 NP to rat pups impacted basic cardiac and neurobehavioral performance, neurotransmitters and related metabolites concentrations in brain tissue, and the biochemical profiles of plasma. The findings suggested that female pups were more likely to experience adverse outcome following early life exposure to oral TiO2 NP than male pups. Collectively the data from this exploratory study suggest oral administration of TiO2 NP cause adverse biological effects in an age- and sex-related manner, emphasizing the need to understand the short- and long-term effects of early life exposure to TiO2 NP.
Collapse
Affiliation(s)
- Ninell P Mortensen
- Discovery Sciences, RTI International, 3040 E Cornwallis Road, Research Triangle Park, NC, 27709, USA.
| | - Wimal Pathmasiri
- UNC Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, USA
| | - Rodney W Snyder
- Discovery Sciences, RTI International, 3040 E Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Maria Moreno Caffaro
- Discovery Sciences, RTI International, 3040 E Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Scott L Watson
- Discovery Sciences, RTI International, 3040 E Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Purvi R Patel
- Discovery Sciences, RTI International, 3040 E Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Lakshmi Beeravalli
- Joint School of Nanoscience and Nanoengineering, 2907 East Gate City Blvd., Greensboro, NC, 27401, USA
| | - Sharmista Prattipati
- Joint School of Nanoscience and Nanoengineering, 2907 East Gate City Blvd., Greensboro, NC, 27401, USA
| | - Shyam Aravamudhan
- Joint School of Nanoscience and Nanoengineering, 2907 East Gate City Blvd., Greensboro, NC, 27401, USA
| | - Susan J Sumner
- UNC Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, USA
| | - Timothy R Fennell
- Discovery Sciences, RTI International, 3040 E Cornwallis Road, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
3
|
Coelho SVA, Rust NM, Vellasco L, Papa MP, Pereira ASG, da Silva Palazzo MF, Juliano MA, Costa SM, Alves AMB, Cordeiro MT, Marques ETA, Scharfstein J, de Arruda LB. Contact System Activation in Plasma from Dengue Patients Might Harness Endothelial Virus Replication through the Signaling of Bradykinin Receptors. Pharmaceuticals (Basel) 2021; 14:ph14010056. [PMID: 33445640 PMCID: PMC7827195 DOI: 10.3390/ph14010056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Since exacerbated inflammation and microvascular leakage are hallmarks of dengue virus (DENV) infection, here we interrogated whether systemic activation of the contact/kallikrein-kinin system (KKS) might hamper endothelial function. In vitro assays showed that dextran sulfate, a potent contact activator, failed to generate appreciable levels of activated plasma kallikrein (PKa) in the large majority of samples from a dengue cohort (n = 70), irrespective of severity of clinical symptoms. Impaired formation of PKa in dengue-plasmas correlated with the presence of cleaved Factor XII and high molecular weight kininogen (HK), suggesting that the prothrombogenic contact system is frequently triggered during the course of infection. Using two pathogenic arboviruses, DENV or Zika virus (ZIKV), we then asked whether exogenous BK could influence the outcome of infection of human brain microvascular endothelial cells (HBMECs). Unlike the unresponsive phenotype of Zika-infected HBMECs, we found that BK, acting via B2R, vigorously stimulated DENV-2 replication by reverting nitric oxide-driven apoptosis of endothelial cells. Using the mouse model of cerebral dengue infection, we next demonstrated that B2R targeting by icatibant decreased viral load in brain tissues. In summary, our study suggests that contact/KKS activation followed by BK-induced enhancement of DENV replication in the endothelium may underlie microvascular pathology in dengue.
Collapse
Affiliation(s)
- Sharton V. A. Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Naiara M. Rust
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Lucas Vellasco
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Michelle P. Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Aline S. G. Pereira
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Matheus Ferreira da Silva Palazzo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Maria Aparecida Juliano
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| | - Simone M. Costa
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (S.M.C.); (A.M.B.A.)
| | - Ada M. B. Alves
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (S.M.C.); (A.M.B.A.)
| | - Marli T. Cordeiro
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Recife 50740-465, Brazil; (M.T.C.); (E.T.A.M.)
| | - Ernesto T. A. Marques
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Recife 50740-465, Brazil; (M.T.C.); (E.T.A.M.)
- Department of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Júlio Scharfstein
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
- Correspondence: (J.S.); (L.B.d.A.)
| | - Luciana B. de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
- Correspondence: (J.S.); (L.B.d.A.)
| |
Collapse
|
4
|
Blood-Brain Barrier Modulation to Improve Glioma Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12111085. [PMID: 33198244 PMCID: PMC7697580 DOI: 10.3390/pharmaceutics12111085] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by brain microvascular endothelial cells that are sealed by tight junctions, making it a significant obstacle for most brain therapeutics. The poor BBB penetration of newly developed therapeutics has therefore played a major role in limiting their clinical success. A particularly challenging therapeutic target is glioma, which is the most frequently occurring malignant brain tumor. Thus, to enhance therapeutic uptake in tumors, researchers have been developing strategies to modulate BBB permeability. However, most conventional BBB opening strategies are difficult to apply in the clinical setting due to their broad, non-specific modulation of the BBB, which can result in damage to normal brain tissue. In this review, we have summarized strategies that could potentially be used to selectively and efficiently modulate the tumor BBB for more effective glioma treatment.
Collapse
|
5
|
Kumar G, Dey SK, Kundu S. Functional implications of vascular endothelium in regulation of endothelial nitric oxide synthesis to control blood pressure and cardiac functions. Life Sci 2020; 259:118377. [PMID: 32898526 DOI: 10.1016/j.lfs.2020.118377] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022]
Abstract
The endothelium is the innermost vascular lining performing significant roles all over the human body while maintaining the blood pressure at physiological levels. Malfunction of endothelium is thus recognized as a biomarker linked with many vascular diseases including but not limited to atherosclerosis, hypertension and thrombosis. Alternatively, prevention of endothelial malfunctioning or regulating the functions of its associated physiological partners like endothelial nitric oxide synthase can prevent the associated vascular disorders which account for the highest death toll worldwide. While many anti-hypertensive drugs are available commercially, a comprehensive description of the key physiological roles of the endothelium and its regulation by endothelial nitric oxide synthase or vice versa is the need of the hour to understand its contribution in vascular homeostasis. This, in turn, will help in designing new therapeutics targeting endothelial nitric oxide synthase or its interacting partners present in the cellular pool. This review describes the central role of vascular endothelium in the regulation of endothelial nitric oxide synthase while outlining the emerging drug targets present in the vasculature with potential to treat vascular disorders including hypertension.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India
| | - Sanjay Kumar Dey
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India; Center for Advanced Biotechnology and Medicine, Rutgers University, NJ 08854, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India.
| |
Collapse
|
6
|
Casin KM, Kohr MJ. An emerging perspective on sex differences: Intersecting S-nitrosothiol and aldehyde signaling in the heart. Redox Biol 2020; 31:101441. [PMID: 32007450 PMCID: PMC7212482 DOI: 10.1016/j.redox.2020.101441] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of the death for both men and women. Although baseline heart physiology and the response to disease are known to differ by sex, little is known about sex differences in baseline molecular signaling, especially with regard to redox biology. In this review, we describe current research on sex differences in cardiac redox biology with a focus on the regulation of nitric oxide and aldehyde signaling. Furthermore, we argue for a new perspective on cardiovascular sex differences research, one that focuses on baseline redox biology without the elimination or disruption of sex hormones.
Collapse
Affiliation(s)
- Kevin M Casin
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
7
|
Piccirillo F, Carpenito M, Verolino G, Chello C, Nusca A, Lusini M, Spadaccio C, Nappi F, Di Sciascio G, Nenna A. Changes of the coronary arteries and cardiac microvasculature with aging: Implications for translational research and clinical practice. Mech Ageing Dev 2019; 184:111161. [PMID: 31647940 DOI: 10.1016/j.mad.2019.111161] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/28/2022]
Abstract
Aging results in functional and structural changes in the cardiovascular system, translating into a progressive increase of mechanical vessel stiffness, due to a combination of changes in micro-RNA expression patterns, autophagy, arterial calcification, smooth muscle cell migration and proliferation. The two pivotal mechanisms of aging-related endothelial dysfunction are oxidative stress and inflammation, even in the absence of clinical disease. A comprehensive understanding of the aging process is emerging as a primary concern in literature, as vascular aging has recently become a target for prevention and treatment of cardiovascular disease. Change of life-style, diet, antioxidant regimens, anti-inflammatory treatments, senolytic drugs counteract the pro-aging pathways or target senescent cells modulating their detrimental effects. Such therapies aim to reduce the ineluctable burden of age and contrast aging-associated cardiovascular dysfunction. This narrative review intends to summarize the macrovascular and microvascular changes related with aging, as a better understanding of the pathways leading to arterial aging may contribute to design new mechanism-based therapeutic approaches to attenuate the features of vascular senescence and its clinical impact on the cardiovascular system.
Collapse
Affiliation(s)
| | | | | | - Camilla Chello
- Dermatology, Università "La Sapienza" di Roma, Rome, Italy
| | | | - Mario Lusini
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | - Francesco Nappi
- Cardiac surgery, Centre Cardiologique du Nord de Saint Denis, Paris, France
| | | | - Antonio Nenna
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Rome, Italy.
| |
Collapse
|
8
|
Poderoso JJ, Helfenberger K, Poderoso C. The effect of nitric oxide on mitochondrial respiration. Nitric Oxide 2019; 88:61-72. [PMID: 30999001 DOI: 10.1016/j.niox.2019.04.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 01/04/2023]
Abstract
This article reviews the interactions between nitric oxide (NO) and mitochondrial respiration. Mitochondrial ATP synthesis is responsible for virtually all energy production in mammals, and every other process in living organisms ultimately depends on that energy production. Furthermore, both necrosis and apoptosis, that summarize the main forms of cell death, are intimately linked to mitochondrial integrity. Endogenous and exogenous •NO inhibits mitochondrial respiration by different well-studied mechanisms and several nitrogen derivatives. Instantaneously, low concentrations of •NO, specifically and reversibly inhibit cytochrome c oxidase in competition with oxygen, in several tissues and cells in culture. Higher concentrations of •NO and its derivatives (peroxynitrite, nitrogen dioxide or nitrosothiols) can cause irreversible inhibition of the respiratory chain, uncoupling, permeability transition, and/or cell death. Peroxynitrite can cause opening of the permeability transition pore and opening of this pore causes loss of cytochrome c, which in turn might contribute to peroxynitrite-induced inhibition of respiration. Therefore, the inhibition of cytochrome c oxidase by •NO may be involved in the physiological and/or pathological regulation of respiration rate, and its affinity for oxygen, which depend on reactive nitrogen species formation, pH, proton motriz force and oxygen supply to tissues.
Collapse
Affiliation(s)
- Juan José Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Hospital de Clínicas "José de San Martín", Laboratorio Del Metabolismo Del Oxígeno, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Katia Helfenberger
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th Floor, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Cecilia Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th Floor, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina.
| |
Collapse
|
9
|
de Moraes LV, Barateiro A, Sousa PM, Penha-Gonçalves C. Bradykinin Sequestration by Plasmodium berghei Infected Erythrocytes Conditions B2R Signaling and Parasite Uptake by Fetal Trophoblasts. Front Microbiol 2018; 9:3106. [PMID: 30619185 PMCID: PMC6305765 DOI: 10.3389/fmicb.2018.03106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
Plasmodium infection during pregnancy causes placental malfunction reducing fetus sustainability and leading to abortions, stillbirths, low birth weight or premature delivery. Accumulation of infected erythrocytes (IE) in the placenta is a key factor in placental malaria pathogenesis but the role played by fetal trophoblast that contact maternal blood has been neglected. Here we explore the hypothesis that interactions between Plasmodium-IE and fetal trophoblast cells impact on vasoactive alterations underlying placental dysfunction. We screened gene expression of key mediators in vasoactive pathways. We found that mRNA of bradykinin receptor 2 (B2R) and nitric oxide synthase (eNOS), as well as levels of bradykinin (BK), were decreased in late gestation placentas of pregnant Plasmodium berghei-infected mice. Co-culturing mouse trophoblasts with IE down-regulated B2R transcription and interleukin (IL)-6 secretion in a B2R-signaling dependent manner. IE showed increased levels of surface B2R and enhanced capacity to bind BK. We propose that down-regulation of B2R signaling in the course of IE–trophoblast interactions is due to BK sequestration by IE. In corroboration, levels of BK were lower in infected placentas and the positive correlation of B2R gene expression and fetal weight was disrupted by infection. This indicates that deregulation of the BK-B2R pathway is associated to placental dysfunction provoked by malaria infection. We further found that upon inhibition of B2R signaling, trophoblasts engulf IE to a lesser extent and show reduced production of IL-6. Our data suggest that BK sequestration by P. berghei represents a strategy for the parasite to ameliorate the risk of phagocytic capture by down modulating B2R activation.
Collapse
Affiliation(s)
| | - André Barateiro
- Disease Genetics, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | |
Collapse
|
10
|
Scharfstein J, Ramos PIP, Barral-Netto M. G Protein-Coupled Kinin Receptors and Immunity Against Pathogens. Adv Immunol 2017; 136:29-84. [PMID: 28950949 DOI: 10.1016/bs.ai.2017.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For decades, immunologists have considered the complement system as a paradigm of a proteolytic cascade that, acting cooperatively with the immune system, enhances host defense against infectious organisms. In recent years, advances made in thrombosis research disclosed a functional link between activated neutrophils, monocytes, and platelet-driven thrombogenesis. Forging a physical barrier, the fibrin scaffolds generated by synergism between the extrinsic and intrinsic (contact) pathways of coagulation entrap microbes within microvessels, limiting the systemic spread of infection while enhancing the clearance of pathogens by activated leukocytes. Insight from mice models of thrombosis linked fibrin formation via the intrinsic pathway to the autoactivation of factor XII (FXII) by negatively charged "contact" substances, such as platelet-derived polyphosphates and DNA from neutrophil extracellular traps. Following cleavage by FXIIa, activated plasma kallikrein (PK) initiates inflammation by liberating the nonapeptide bradykinin (BK) from an internal domain of high molecular weight kininogen (HK). Acting as a paracrine mediator, BK induces vasodilation and increases microvascular permeability via activation of endothelial B2R, a constitutively expressed subtype of kinin receptor. During infection, neutrophil-driven extravasation of plasma fuels inflammation via extravascular activation of the kallikrein-kinin system (KKS). Whether liberated by plasma-borne PK, tissue kallikrein, and/or microbial-derived proteases, the short-lived kinins activate immature dendritic cells via B2R, thus linking the infection-associated innate immunity/inflammation to the adaptive arm of immunity. As inflammation persists, a GPI-linked carboxypeptidase M removes the C-terminal arginine from the primary kinin, converting the B2R agonist into a high-affinity ligand for B1R, a GPCR subtype that is transcriptionally upregulated in injured/inflamed tissues. As reviewed here, lessons taken from studies of kinin receptor function in experimental infections have shed light on the complex proteolytic circuits that, acting at the endothelial interface, reciprocally couple immunity to the proinflammatory KKS.
Collapse
Affiliation(s)
- Julio Scharfstein
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Center of Health Sciences (CCS), Cidade Universitária, Rio de Janeiro, Brazil.
| | - Pablo I P Ramos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | | |
Collapse
|
11
|
Bubb KJ, Birgisdottir AB, Tang O, Hansen T, Figtree GA. Redox modification of caveolar proteins in the cardiovascular system- role in cellular signalling and disease. Free Radic Biol Med 2017; 109:61-74. [PMID: 28188926 DOI: 10.1016/j.freeradbiomed.2017.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/18/2017] [Accepted: 02/05/2017] [Indexed: 02/07/2023]
Abstract
Rapid and coordinated release of a variety of reactive oxygen species (ROS) such as superoxide (O2.-), hydrogen peroxide (H2O2) and peroxynitrite, in specific microdomains, play a crucial role in cell signalling in the cardiovascular system. These reactions are mediated by reversible and functional modifications of a wide variety of key proteins. Dysregulation of this oxidative signalling occurs in almost all forms of cardiovascular disease (CVD), including at the very early phases. Despite the heavily publicized failure of "antioxidants" to improve CVD progression, pharmacotherapies such as those targeting the renin-angiotensin system, or statins, exert at least part of their large clinical benefit via modulating cellular redox signalling. Over 250 proteins, including receptors, ion channels and pumps, and signalling proteins are found in the caveolae. An increasing proportion of these are being recognized as redox regulated-proteins, that reside in the immediate vicinity of the two major cellular sources of ROS, nicotinamide adenine dinucleotide phosphate oxidase (Nox) and uncoupled endothelial nitric oxide synthase (eNOS). This review focuses on what is known about redox signalling within the caveolae, as well as endogenous protective mechanisms utilized by the cell, and new approaches to targeting dysregulated redox signalling in the caveolae as a therapeutic strategy in CVD.
Collapse
Affiliation(s)
- Kristen J Bubb
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Asa Birna Birgisdottir
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; Department of Cardiothoracic and Vascular Surgery, Heart and Lung Clinic, University Hospital of North Norway, Tromsø, Norway
| | - Owen Tang
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Thomas Hansen
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| |
Collapse
|
12
|
Hein TW, Xu W, Xu X, Kuo L. Acute and Chronic Hyperglycemia Elicit JIP1/JNK-Mediated Endothelial Vasodilator Dysfunction of Retinal Arterioles. Invest Ophthalmol Vis Sci 2017; 57:4333-40. [PMID: 27556216 PMCID: PMC5015966 DOI: 10.1167/iovs.16-19990] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Hyperglycemia, a hallmark of diabetes mellitus, is associated with retinal inflammation and impairment of endothelium-dependent nitric oxide (NO)–mediated dilation of retinal arterioles. However, molecular mechanisms involved in this diminished endothelial vasodilator function remain unclear. We examined whether inflammatory stress-activated kinases, c-Jun N-terminal kinase (JNK) and p38, contribute to retinal arteriolar dysfunction during exposure to acute and chronic hyperglycemia. Methods Retinal arterioles were isolated from streptozocin-induced diabetic pigs (2 weeks; chronic hyperglycemia, 471 ± 23 mg/dL) or age-matched control pigs (euglycemia, 79 ± 5 mg/dL), and then cannulated and pressurized for vasoreactivity study. For acute hyperglycemia study, vessels from nondiabetic pigs were exposed intraluminally to high glucose (25 mM ≈ 450 mg/dL) for 2 hours, and normal glucose (5 mM ≈ 90 mg/dL) served as the control. Results Endothelium-dependent vasodilation to bradykinin was reduced in a similar manner after exposure to acute or chronic hyperglycemia. Administration of NO synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME) nearly abolished vasodilations either in control (euglycemia and normal glucose) or hyperglycemic (acute and chronic) vessels. Treatment of either acute or chronic hyperglycemic vessels with JNK inhibitor SP600125 or JNK-interacting protein-1 (JIP1) inhibitor BI-78D3, but not p38 inhibitor SB203580, preserved bradykinin-induced dilation in an L-NAME–sensitive manner. By contrast, endothelium-independent vasodilation to sodium nitroprusside was unaffected by acute or chronic hyperglycemia. Conclusions Activation of JIP1/JNK signaling in retinal arterioles during exposure to acute or chronic hyperglycemia leads to selective impairment of endothelium-dependent NO-mediated dilation. Therapeutic targeting of the vascular JNK pathway may improve retinal endothelial vasodilator function during early diabetes.
Collapse
Affiliation(s)
- Travis W Hein
- Department of Surgery, Baylor Scott & White Eye Institute, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| | - Wenjuan Xu
- Department of Surgery, Baylor Scott & White Eye Institute, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| | - Xin Xu
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| | - Lih Kuo
- Department of Surgery, Baylor Scott & White Eye Institute, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States 2Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Temple, Texas, United States
| |
Collapse
|
13
|
Korkmaz Y, Bloch W, Steinritz D, Baumann MA, Addicks K, Schneider K, Raab WHM. Bradykinin Mediates Phosphorylation of eNOS in Odontoblasts. J Dent Res 2016; 85:536-41. [PMID: 16723651 DOI: 10.1177/154405910608500611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
While the activation of eNOS by Akt/PKB-dependent phosphorylation, leading to NO release, and the inhibition of enzyme activity by bradykinin (BK)-mediated phosphorylation of eNOS in endothelial cells are established, the phosphorylation of eNOS in odontoblasts is unknown. To clarify the regulation of eNOS in odontoblasts by BK, we examined the phosphorylation of eNOS, Akt/PKB, and ERK1/2 in odontoblasts of rat molars. BK (10−7 M) transiently induced the phosphorylation of eNOS at Ser1177, Akt/PKB in odontoblasts, while it induced the phosphorylation of eNOS at Thr495 throughout the entire period of BK treatment. BK receptor 2 antagonist HOE 140 (10−6 M) significantly reduced signal intensities of phosphorylated-eNOS at Ser1177, Thr495, and phosphorylated-Akt/PKB. These results suggest that BK has dual effects on the activation of eNOS in odontoblasts, the Akt/PKB-dependent up-regulation of eNOS by the transient phosphorylation at Ser1177, and the ERK1/2-independent down-regulation of eNOS by the phosphorylation at Thr495.
Collapse
Affiliation(s)
- Y Korkmaz
- Department of Operative and Preventive Dentistry and Endodontics, Heinrich-Heine-University, Moorenstr. 5, 40225 Düsseldorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
14
|
Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. Pflugers Arch 2016; 468:1125-1137. [DOI: 10.1007/s00424-016-1839-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
|
15
|
Pastore MB, Talwar S, Conley MR, Magness RR. Identification of Differential ER-Alpha Versus ER-Beta Mediated Activation of eNOS in Ovine Uterine Artery Endothelial Cells. Biol Reprod 2016; 94:139. [PMID: 27170438 PMCID: PMC4946807 DOI: 10.1095/biolreprod.115.137554] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/03/2016] [Indexed: 12/19/2022] Open
Abstract
Endothelial nitric oxide (NO) production is partly responsible for maintenance of uterine vasodilatation during physiologic states of high circulating estrogen levels, e.g., pregnancy. Although 3%–5% of estrogen receptors (ER-alpha/beta) localize to the endothelial plasmalemma, these receptors are responsible for the nongenomic vasodilator responses. Estradiol induces endothelial NO synthase (eNOS) activation to increase NO production; however, it is unknown if eNOS regulation is dependent on both ERs. We hypothesize that ER-alpha and/or ER-beta are capable of changing eNOS phosphorylation and increasing NO production in uterine artery endothelial cells (UAECs). UAECs were 1) treated with vehicle or increasing concentrations (0.1–100 nM) or timed treatments (0–30 min) of estradiol and 2) pretreated with the inhibitors ICI 182,780 (nonspecific ER), 1,3-Bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride (MPP; ER-alpha specific), or 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol (PHTPP; ER-beta specific) followed by estradiol to analyze the changes in eNOS stimulatory Ser1177eNOS and Ser635eNOS versus inhibitory Thr495eNOS via Western blot analysis. UAECs were also pretreated with MPP, PHTPP, or MPP + PHTTP followed by estradiol or treated with the agonists estradiol, 4,4′,4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol, 2,3-bis(4-hydroxyphenyl)-propionitrile, or ATP to quantify total NOx levels (NO2+NO3). Estrogen and ER-alpha activation induced an increase in Ser1177eNOS and Ser635eNOS, a decrease in Thr495eNOS, and an increase in NOx levels. In contrast, ER-beta activation only reduced Thr495eNOS without changes in Ser1177eNOS or Ser635eNOS. However, ER-beta activation increased NOx levels. Lastly, the antagonism of both receptors induced a reduction in basal and stimulated NOx levels in UAECs. These data demonstrate that 1) eNOS phosphorylation changes occur via ER-alpha- and ER-beta-dependent mechanisms and 2) ER-alpha and ER-beta can both increase NO levels independently from each other.
Collapse
Affiliation(s)
- Mayra B Pastore
- Department of Obstetrics & Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin
| | - Saira Talwar
- Department of Obstetrics & Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin
| | - Meghan R Conley
- Department of Obstetrics & Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ronald R Magness
- Department of Obstetrics & Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin Department of Animal Sciences, University of Wisconsin-Madison, Madison, Wisconsin Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin University of South Florida Perinatal Research Center, Tampa, Florida
| |
Collapse
|
16
|
Maternal exposure to di-(2-ethylhexyl) phthalate exposure deregulates blood pressure, adiposity, cholesterol metabolism and social interaction in mouse offspring. Arch Toxicol 2015; 90:1211-24. [DOI: 10.1007/s00204-015-1539-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/13/2015] [Indexed: 01/28/2023]
|
17
|
Eller-Borges R, Batista WL, da Costa PE, Tokikawa R, Curcio MF, Strumillo ST, Sartori A, Moraes MS, de Oliveira GA, Taha MO, Fonseca FV, Stern A, Monteiro HP. Ras, Rac1, and phosphatidylinositol-3-kinase (PI3K) signaling in nitric oxide induced endothelial cell migration. Nitric Oxide 2015; 47:40-51. [PMID: 25819133 DOI: 10.1016/j.niox.2015.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 10/23/2022]
Abstract
The small GTP-binding proteins Ras and Rac1 are molecular switches exchanging GDP for GTP and converting external signals in response to a variety of stimuli. Ras and Rac1 play an important role in cell proliferation, cell differentiation, and cell migration. Rac1 is directly involved in the reorganization and changes in the cytoskeleton during cell motility. Nitric oxide (NO) stimulates the Ras - ERK1/2 MAP kinases signaling pathway and is involved in the interaction between Ras and the phosphatidyl-inositol-3 Kinase (PI3K) signaling pathway and cell migration. This study utilizes bradykinin (BK), which promotes endogenous production of NO, in an investigation of the role of NO in the activation of Rac1 in rabbit aortic endothelial cells (RAEC). NO-derived from BK stimulation of RAEC and incubation of the cells with the s-nitrosothiol S-nitrosoglutathione (GSNO) activated Rac1. NO-derived from BK stimulation promoted RAEC migration over a period of 12 h. The use of RAEC permanently transfected with the dominant negative mutant of Ras (Ras(N17)) or with the non-nitrosatable mutant of Ras (Ras(C118S)); and the use of specific inhibitors of: Ras, PI3K, and Rac1 resulted in inhibition of NO-mediated Rac1 activation. BK-stimulated s-nitrosylation of Ras in RAEC mediates Rac1 activation and cell migration. Inhibition of NO-mediated Rac1 activation resulted in inhibition of endothelial cell migration. In conclusion, the NO indirect activation of Rac1 involves the direct participation of Ras and PI3K in the migration of endothelial cells stimulated with BK.
Collapse
Affiliation(s)
- Roberta Eller-Borges
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Wagner L Batista
- Department of Biological Sciences, Universidade Federal de São Paulo/Campus Diadema, SP, Brazil
| | - Paulo E da Costa
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Rita Tokikawa
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Marli F Curcio
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Scheilla T Strumillo
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Adriano Sartori
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Miriam S Moraes
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Graciele A de Oliveira
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Murched O Taha
- Department of Surgery, Escola Paulista de Medicina/Universidade Federal de São Paulo, SP, Brazil
| | - Fábio V Fonseca
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western University, Cleveland, OH, USA
| | - Arnold Stern
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA; Escuela de Medicina, Universidad Espíritu Santo, Guayaquil, Ecuador.
| | - Hugo P Monteiro
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil.
| |
Collapse
|
18
|
Quillon A, Fromy B, Debret R. Endothelium microenvironment sensing leading to nitric oxide mediated vasodilation: A review of nervous and biomechanical signals. Nitric Oxide 2015; 45:20-6. [DOI: 10.1016/j.niox.2015.01.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 10/24/2022]
|
19
|
Ye BH, Lee SJ, Choi YW, Park SY, Kim CD. Preventive effect of gomisin J from Schisandra chinensis on angiotensin II-induced hypertension via an increased nitric oxide bioavailability. Hypertens Res 2014; 38:169-77. [DOI: 10.1038/hr.2014.162] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 09/04/2014] [Accepted: 10/23/2014] [Indexed: 01/18/2023]
|
20
|
Mattila JT, Thomas AC. Nitric oxide synthase: non-canonical expression patterns. Front Immunol 2014; 5:478. [PMID: 25346730 PMCID: PMC4191211 DOI: 10.3389/fimmu.2014.00478] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022] Open
Abstract
Science can move ahead by questioning established or canonical views and, so it may be with the enzymes, nitric oxide synthases (NOS). Nitric oxide (NO) is generated by NOS isoforms that are often described by their tissue-specific expression patterns. NOS1 (nNOS) is abundant in neural tissue, NOS2 is upregulated in activated macrophages and known as inducible NOS (iNOS), and NOS3 (eNOS) is abundant in endothelium where it regulates vascular tone. These isoforms are described as constitutive or inducible, but in this perspective we question the broad application of these labels. Are there instances where "constitutive" NOS (NOS1 and NOS3) are inducibly expressed; conversely, are there instances where NOS2 is constitutively expressed? NOS1 and NOS3 inducibility may be linked to post-translational regulation, making their actual patterns activity much more difficult to detect. Constitutive NOS2 expression has been observed in several tissues, especially the human pulmonary epithelium where it may regulate airway tone. These data suggest that expression of the three NOS enzymes may include non-established patterns. Such information should be useful in designing strategies to modulate these important enzymes in different disease states.
Collapse
Affiliation(s)
- Joshua T. Mattila
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anita C. Thomas
- Bristol Heart Institute and Bristol CardioVascular, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| |
Collapse
|
21
|
Manchini MT, Serra AJ, Feliciano RDS, Santana ET, Antônio EL, de Tarso Camillo de Carvalho P, Montemor J, Crajoinas RO, Girardi ACC, Tucci PJF, Silva JA. Amelioration of cardiac function and activation of anti-inflammatory vasoactive peptides expression in the rat myocardium by low level laser therapy. PLoS One 2014; 9:e101270. [PMID: 24991808 PMCID: PMC4081549 DOI: 10.1371/journal.pone.0101270] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 06/05/2014] [Indexed: 11/18/2022] Open
Abstract
Low-level laser therapy (LLLT) has been used as an anti-inflammatory treatment in several disease conditions, even when inflammation is a secondary consequence, such as in myocardial infarction (MI). However, the mechanism by which LLLT is able to protect the remaining myocardium remains unclear. The present study tested the hypothesis that LLLT reduces inflammation after acute MI in female rats and ameliorates cardiac function. The potential participation of the Renin-Angiotensin System (RAS) and Kallikrein-Kinin System (KKS) vasoactive peptides was also evaluated. LLLT treatment effectively reduced MI size, attenuated the systolic dysfunction after MI, and decreased the myocardial mRNA expression of interleukin-1 beta and interleukin-6 in comparison to the non-irradiated rat tissue. In addition, LLLT treatment increased protein and mRNA levels of the Mas receptor, the mRNA expression of kinin B2 receptors and the circulating levels of plasma kallikrein compared to non-treated post-MI rats. On the other hand, the kinin B1 receptor mRNA expression decreased after LLLT. No significant changes were found in the expression of vascular endothelial growth factor (VEGF) in the myocardial remote area between laser-irradiated and non-irradiated post-MI rats. Capillaries density also remained similar between these two experimental groups. The mRNA expression of the inducible nitric oxide synthase (iNOS) was increased three days after MI, however, this effect was blunted by LLLT. Moreover, endothelial NOS mRNA content increased after LLLT. Plasma nitric oxide metabolites (NOx) concentration was increased three days after MI in non-treated rats and increased even further by LLLT treatment. Our data suggest that LLLT diminishes the acute inflammation in the myocardium, reduces infarct size and attenuates left ventricle dysfunction post-MI and increases vasoactive peptides expression and nitric oxide (NO) generation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jairo Montemor
- Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brasil
| | | | | | | | | |
Collapse
|
22
|
Arya D, Chang S, DiMuzio P, Carpenter J, Tulenko TN. Sphingosine-1-phosphate promotes the differentiation of adipose-derived stem cells into endothelial nitric oxide synthase (eNOS) expressing endothelial-like cells. J Biomed Sci 2014; 21:55. [PMID: 24898615 PMCID: PMC4064270 DOI: 10.1186/1423-0127-21-55] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 05/15/2014] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Adipose tissue provides a readily available source of autologous stem cells. Adipose-derived stem cells (ASCs) have been proposed as a source for endothelial cell substitutes for lining the luminal surface of tissue engineered bypass grafts. Endothelial nitric oxide synthase (eNOS) is a key protein in endothelial cell function. Currently, endothelial differentiation from ASCs is limited by poor eNOS expression. The goal of this study was to investigate the role of three molecules, sphingosine-1-phosphate (S1P), bradykinin, and prostaglandin-E1 (PGE1) in ASC endothelial differentiation. Endothelial differentiation markers (CD31, vWF and eNOS) were used to evaluate the level of ASCs differentiation capability. RESULTS ASCs demonstrated differentiation capability toward to adipose, osteocyte and endothelial like cell phenotypes. Bradykinin, S1P and PGE were used to promote differentiation of ASCs to an endothelial phenotype. Real-time PCR showed that all three molecules induced significantly greater expression of endothelial differentiation markers CD31, vWF and eNOS than untreated cells. Among the three molecules, S1P showed the highest up-regulation on endothelial differentiation markers. Immunostaining confirmed presence of more eNOS in cells treated with S1P than the other groups. Cell growth measurements by MTT assay, cell counting and EdU DNA incorporation suggest that S1P promotes cell growth during ASCs endothelial differentiation. The S1P1 receptor was expressed in ASC-differentiated endothelial cells and S1P induced up-regulation of PI3K. CONCLUSIONS S1P up-regulates endothelial cell markers including eNOS in ASCs differentiated to endothelial like cells. This up-regulation appears to be mediated by the up-regulation of PI3K via S1P1 receptor. ASCs treated with S1P offer promising use as endothelial cell substitutes for tissue engineered vascular grafts and vascular networks.
Collapse
Affiliation(s)
| | | | | | | | - Thomas N Tulenko
- Department of Surgery, Cooper University Hospital and Cooper Medical School of Rowan University, 3 Cooper Plaza, Camden, NJ 08103, USA.
| |
Collapse
|
23
|
García C, Nuñez-Anita RE, Thebault S, Arredondo Zamarripa D, Jeziorsky MC, Martínez de la Escalera G, Clapp C. Requirement of phosphorylatable endothelial nitric oxide synthase at Ser-1177 for vasoinhibin-mediated inhibition of endothelial cell migration and proliferation in vitro. Endocrine 2014; 45:263-70. [PMID: 23640371 DOI: 10.1007/s12020-013-9964-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
Endothelial nitric oxide synthase (eNOS)-derived nitric oxide is a major vasorelaxing factor and a mediator of vasopermeability and angiogenesis. Vasoinhibins, a family of antiangiogenic prolactin fragments that include 16 K prolactin, block most eNOS-mediated vascular effects. Vasoinhibins activate protein phosphatase 2A, causing eNOS inactivation through dephosphorylation of eNOS at serine residue 1179 in bovine endothelial cells and thereby blocking vascular permeability. In this study, we examined whether human eNOS phosphorylation at S1177 (analogous to bovine S1179) influences other actions of vasoinhibins. Bovine umbilical vein endothelial cells were stably transfected with human wild-type eNOS (WT) or with phospho-mimetic (S1177D) or non-phosphorylatable (S1177A) eNOS mutants. Vasoinhibins inhibited the increases in eNOS activity, migration, and proliferation following the overexpression of WT eNOS but did not affect these responses in cells expressing S1177D and S1177A eNOS mutants. We conclude that eNOS inhibition by dephosphorylation of S1177 is fundamental for the inhibition of endothelial cell migration and proliferation by vasoinhibins.
Collapse
Affiliation(s)
- Celina García
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, QRO, Mexico
| | | | | | | | | | | | | |
Collapse
|
24
|
Qian J, Fulton D. Post-translational regulation of endothelial nitric oxide synthase in vascular endothelium. Front Physiol 2013; 4:347. [PMID: 24379783 PMCID: PMC3861784 DOI: 10.3389/fphys.2013.00347] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/11/2013] [Indexed: 01/22/2023] Open
Abstract
Nitric oxide (NO) is a short-lived gaseous signaling molecule. In blood vessels, it is synthesized in a dynamic fashion by endothelial nitric oxide synthase (eNOS) and influences vascular function via two distinct mechanisms, the activation of soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP)-dependent signaling and the S-nitrosylation of proteins with reactive thiols (S-nitrosylation). The regulation of eNOS activity and NO bioavailability is critical to maintain blood vessel function. The activity of eNOS and ability to generate NO is regulated at the transcriptional, posttranscriptional, and posttranslational levels. Post-translational modifications acutely impact eNOS activity and dysregulation of these mechanisms compromise eNOS activity and foster the development of cardiovascular diseases (CVDs). This review will intergrate past and current literature on the post-translational modifications of eNOS in both health and disease.
Collapse
Affiliation(s)
- Jin Qian
- Pulmonary and Critical Care, School of Medicine, Stanford University/VA Palo Alto Health Care System Palo Alto, CA, USA
| | - David Fulton
- Vascular Biology Center, Georgia Regents University Augusta, GA, USA
| |
Collapse
|
25
|
Zhang X, Tan F, Brovkovych V, Zhang Y, Lowry JL, Skidgel RA. Carboxypeptidase M augments kinin B1 receptor signaling by conformational crosstalk and enhances endothelial nitric oxide output. Biol Chem 2013. [PMID: 23183746 DOI: 10.1515/hsz-2012-0290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The G protein-coupled receptors (GPCRs) are the largest class of membrane proteins that play key roles in transducing extracellular signals to intracellular proteins to generate cellular responses. The kinin GPCRs, named B1 (B1R) and B2 (B2R), are responsible for mediating the biological responses to kinin peptides released from the precursor kininogens. Bradykinin (BK) or kallidin (KD) are agonists for B2Rs, whereas their carboxypeptidase (CP)-generated metabolites, des-Arg(9)-BK or des-Arg(10)-KD, are specific agonists for B1Rs. Here, we review the evidence for a critical role of membrane-bound CPM in facilitating B1R signaling by its ability to directly activate the receptor via conformational crosstalk as well as generate its specific agonist. In endothelial cells, the CPM/B1R interaction facilitates B1R-dependent high-output nitric oxide under inflammatory conditions.
Collapse
Affiliation(s)
- Xianming Zhang
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
26
|
Lowry JL, Brovkovych V, Zhang Y, Skidgel RA. Endothelial nitric-oxide synthase activation generates an inducible nitric-oxide synthase-like output of nitric oxide in inflamed endothelium. J Biol Chem 2012; 288:4174-93. [PMID: 23255592 DOI: 10.1074/jbc.m112.436022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
High levels of NO generated in the vasculature under inflammatory conditions are usually attributed to inducible nitric-oxide synthase (iNOS), but the role of the constitutively expressed endothelial NOS (eNOS) is unclear. In normal human lung microvascular endothelial cells (HLMVEC), bradykinin (BK) activates kinin B2 receptor (B2R) signaling that results in Ca(2+)-dependent activation of eNOS and transient NO. In inflamed HLMVEC (pretreated with interleukin-1β and interferon-γ), we found enhanced binding of eNOS to calcium-calmodulin at basal Ca(2+) levels, thereby increasing its basal activity that was dependent on extracellular l-Arg. Furthermore, B2R stimulation generated prolonged high output eNOS-derived NO that is independent of increased intracellular Ca(2+) and is mediated by a novel Gα(i)-, MEK1/2-, and JNK1/2-dependent pathway. This high output NO stimulated with BK was blocked with a B2R antagonist, eNOS siRNA, or eNOS inhibitor but not iNOS inhibitor. Moreover, B2R-mediated NO production and JNK phosphorylation were inhibited with MEK1/2 and JNK inhibitors or MEK1/2 and JNK1/2 siRNA but not with ERK1/2 inhibitor. BK induced Ca(2+)-dependent eNOS phosphorylation at Ser(1177), Thr(495), and Ser(114) in cytokine-treated HLMVEC, but these modifications were not dependent on JNK1/2 activation and were not responsible for prolonged NO output. Cytokine treatment did not alter the expression of B2R, Gα(q/11), Gα(i1,2), JNK, or eNOS. B2R activation in control endothelial cells enhanced migration, but in cytokine-treated HLMVEC it reduced migration. Both responses were NO-dependent. Understanding how JNK regulates prolonged eNOS-derived NO may provide new therapeutic targets for the treatment of disorders involving vascular inflammation.
Collapse
Affiliation(s)
- Jessica L Lowry
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
27
|
Antihypertensive effect of gomisin A from Schisandra chinensis on angiotensin II-induced hypertension via preservation of nitric oxide bioavailability. Hypertens Res 2012; 35:928-34. [PMID: 22534517 PMCID: PMC3434368 DOI: 10.1038/hr.2012.50] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gomisin A (GA) is a small molecular weight lignan present in Schisandra chinensis, and has been demonstrated to have vasodilatory activity. In the present study, we investigated the effect of GA on blood pressure (BP) in angiotensin II (Ang II)-induced hypertensive mice. C57/BL6 mice infused subcutaneously with Ang II (1 and 2 μg kg−1 per min for 2 weeks) showed an increase in BP with a decrease in nitric oxide (NO) metabolites in plasma, and a negative correlation between these two parameters was demonstrated. In the thoracic aorta from Ang II-induced hypertensive mice, a decrease in vascular NO that was accompanied by a diminution of phosphorylated endothelial nitric oxide synthase (eNOS), as well as by increased reactive oxygen species (ROS) production, was demonstrated. These alterations in BP, eNOS phosphorylation and ROS production in the vasculature of Ang II-treated mice were markedly and dose-dependently reversed by simultaneous administration of GA (2 and 10 μg kg−1 per min). In addition, Ang II-induced ROS production in cultured vascular cells such as endothelial cells and vascular smooth muscle cells was markedly attenuated by GA. These results suggested that GA attenuated the increase in BP via preservation of vascular NO bioavailability not only by inhibiting ROS production but also by preventing the impairment of eNOS function in the vasculature of Ang II-induced hypertensive mice.
Collapse
|
28
|
Nurmi L, Heikkilä HM, Vapaatalo H, Kovanen PT, Lindstedt KA. Downregulation of Bradykinin Type 2 Receptor Expression in Cardiac Endothelial Cells during Senescence. J Vasc Res 2012; 49:13-23. [DOI: 10.1159/000329615] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 05/20/2011] [Indexed: 11/19/2022] Open
|
29
|
Mazzanti L, Cecati M, Vignini A, D'Eusanio S, Emanuelli M, Giannubilo SR, Saccucci F, Tranquilli AL. Placental expression of endothelial and inducible nitric oxide synthase and nitric oxide levels in patients with HELLP syndrome. Am J Obstet Gynecol 2011; 205:236.e1-7. [PMID: 21700268 DOI: 10.1016/j.ajog.2011.04.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 04/05/2011] [Accepted: 04/11/2011] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To determine placental gene expression of endothelial and inducible nitric oxide synthases and measure nitric oxide levels in patients with hemolysis, elevated liver enzyme levels, and low platelet count syndrome. STUDY DESIGN Preterm placentas were obtained from 15 patients with hemolysis, elevated liver enzyme levels, and low platelet count syndrome and 30 controls matched for age, parity, and gestational age. mRNA levels were evaluated by real-time polymerase chain reaction, whereas nitric oxide and peroxynitrite production was measured by a commercially available kit. RESULTS Placental gene expression of inducible nitric oxide and endothelial nitric oxide synthases were significantly lower in the hemolysis, elevated liver enzyme levels, and low platelet count syndrome group than in controls, whereas nitric oxide and peroxynitrite production were significantly higher in hemolysis, elevated liver enzyme levels, and low platelet count syndrome compared with controls. CONCLUSION The reduced endothelial nitric oxide and inducible nitric oxide synthases gene expression in women with hemolysis, elevated liver enzyme levels, and low platelet count syndrome may indicate extreme placental dysfunction that is unable to compensate the endothelial derangement and the related hypertension. The higher nitric oxide formation found in hemolysis, elevated liver enzyme levels, and low platelet count syndrome placentas could be explained as a counteraction to the impaired fetoplacental perfusion, typical of the syndrome.
Collapse
Affiliation(s)
- Laura Mazzanti
- Department of Biochemistry, Biology and Genetics, Università Politecnica Marche, Ancona, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Increased endothelial cell-leukocyte interaction in murine schistosomiasis: possible priming of endothelial cells by the disease. PLoS One 2011; 6:e23547. [PMID: 21853150 PMCID: PMC3154496 DOI: 10.1371/journal.pone.0023547] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 07/19/2011] [Indexed: 11/21/2022] Open
Abstract
Background and Aims Schistosomiasis is an intravascular parasitic disease associated with inflammation. Endothelial cells control leukocyte transmigration and vascular permeability being modulated by pro-inflammatory mediators. Recent data have shown that endothelial cells primed in vivo in the course of a disease keep the information in culture. Herein, we evaluated the impact of schistosomiasis on endothelial cell-regulated events in vivo and in vitro. Methodology and Principal Findings The experimental groups consisted of Schistosoma mansoni-infected and age-matched control mice. In vivo infection caused a marked influx of leukocytes and an increased protein leakage in the peritoneal cavity, characterizing an inflamed vascular and cellular profile. In vitro leukocyte-mesenteric endothelial cell adhesion was higher in cultured cells from infected mice as compared to controls, either in the basal condition or after treatment with the pro-inflammatory cytokine tumor necrosis factor (TNF). Nitric oxide (NO) donation reduced leukocyte adhesion to endothelial cells from control and infected groups; however, in the later group the effect was more pronounced, probably due to a reduced NO production. Inhibition of control endothelial NO synthase (eNOS) increased leukocyte adhesion to a level similar to the one observed in the infected group. Besides, the adhesion of control leukocytes to endothelial cells from infected animals is similar to the result of infected animals, confirming that schistosomiasis alters endothelial cells function. Furthermore, NO production as well as the expression of eNOS were reduced in cultured endothelial cells from infected animals. On the other hand, the expression of its repressor protein, namely caveolin-1, was similar in both control and infected groups. Conclusion/Significance Schistosomiasis increases vascular permeability and endothelial cell-leukocyte interaction in vivo and in vitro. These effects are partially explained by a reduced eNOS expression. In addition, our data show that the disease primes endothelial cells in vivo, which keep the acquired phenotype in culture.
Collapse
|
31
|
Sobajima M, Nozawa T, Shida T, Ohori T, Suzuki T, Matsuki A, Inoue H. Repeated sauna therapy attenuates ventricular remodeling after myocardial infarction in rats by increasing coronary vascularity of noninfarcted myocardium. Am J Physiol Heart Circ Physiol 2011; 301:H548-54. [DOI: 10.1152/ajpheart.00103.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Repeated sauna therapy (ST) increases endothelial nitric oxide synthase (eNOS) activity and improves cardiac function in heart failure as well as peripheral blood flow in ischemic limbs. The present study investigates whether ST can increase coronary vascularity and thus attenuate cardiac remodeling after myocardial infarction (MI). We induced MI by ligating the left coronary artery of Wistar rats. The rats were placed in a far-infrared dry sauna at 41°C for 15 min and then at 34°C for 20 min once daily for 4 wk. Cardiac hemodynamic, histopathological, and gene analyses were performed. Despite the similar sizes of MI between the ST and non-ST groups (51.4 ± 0.3 vs. 51.1 ± 0.2%), ST reduced left ventricular (LV) end-diastolic (9.7 ± 0.4 vs. 10.7 ± 0.5 mm, P < 0.01) and end-systolic (8.6 ± 0.5 vs. 9.6 ± 0.6 mm, P < 0.01) dimensions and attenuated MI-induced increases in LV end-diastolic pressure. Cross-sectional areas of cardiomyocytes were smaller in ST rats and associated with a significant reduction in myocardial atrial natriuretic peptide mRNA levels. Vascular density was reduced in the noninfarcted myocardium of non-ST rats, and the density of cells positive for CD31 and for α-smooth muscle actin was decreased. These decreases were attenuated in ST rats compared with non-ST rats and associated with increases in myocardial eNOS and vascular endothelial growth factor mRNA levels. In conclusion, ST attenuates cardiac remodeling after MI, at least in part, through improving coronary vascularity in the noninfarcted myocardium. Repeated ST might serve as a novel noninvasive therapy for patients with MI.
Collapse
Affiliation(s)
- Mitsuo Sobajima
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Takashi Nozawa
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Takuya Shida
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Takashi Ohori
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Takayuki Suzuki
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Akira Matsuki
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Hiroshi Inoue
- The Second Department of Internal Medicine, Graduate School of Medicine, University of Toyama, Sugitani, Toyama, Japan
| |
Collapse
|
32
|
Brovkovych V, Zhang Y, Brovkovych S, Minshall RD, Skidgel RA. A novel pathway for receptor-mediated post-translational activation of inducible nitric oxide synthase. J Cell Mol Med 2011; 15:258-69. [PMID: 20015194 PMCID: PMC2888614 DOI: 10.1111/j.1582-4934.2009.00992.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 11/17/2009] [Indexed: 11/30/2022] Open
Abstract
Inducible nitric oxide synthase (iNOS) is a major source of nitric oxide during inflammation whose activity is thought to be controlled primarily at the expression level. The B1 kinin receptor (B1R) post-translationally activates iNOS beyond its basal activity via extracellular signal regulated kinase (ERK)-mediated phosphorylation of Ser(745) . Here we identified the signalling pathway causing iNOS activation in cytokine-treated endothelial cells or HEK293 cells transfected with iNOS and B1R. To allow kinetic measurements of nitric oxide release, we used a sensitive porphyrinic microsensor (response time = 10 msec.; 1 nM detection limit). B1Rs signalled through Gαi coupling as ERK and iNOS activation were inhibited by pertussis toxin. Furthermore, transfection of constitutively active mutant Gαi Q204L but not Gαq Q209L resulted in high basal iNOS-derived nitric oxide. G-βγ subunits were also necessary as transfection with the β-adrenergic receptor kinase C-terminus inhibited the response. B1R-dependent iNOS activation was also inhibited by Src family kinase inhibitor PP2 and trans-fection with dominant negative Src. Other ERK-MAP kinase members were involved as the response was inhibited by dominant negative H-Ras, Raf kinase inhibitor, ERK activation inhibitor and MEK inhibitor PD98059. In contrast, PI3 kinase inhibitor LY94002, calcium chelator 1,2-bis-(o-Aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, tetraacetoxymethyl ester (BAPTA-AM), protein kinase C inhibitor calphostin C and protein kinase C activator PMA had no effect. Angiotensin converting enzyme inhibitor enalaprilat also directly activated B1Rs to generate high output nitric oxide via the same pathway. These studies reveal a new mechanism for generating receptor-regulated high output nitric oxide in inflamed endothelium that may play an important role in the development of vascular inflammation.
Collapse
Affiliation(s)
- Viktor Brovkovych
- Department of Pharmacology, University of Illinois College of MedicineChicago, IL, USA
| | - Yongkang Zhang
- Department of Pharmacology, University of Illinois College of MedicineChicago, IL, USA
| | - Svitlana Brovkovych
- Department of Pharmacology, University of Illinois College of MedicineChicago, IL, USA
| | - Richard D Minshall
- Department of Pharmacology, University of Illinois College of MedicineChicago, IL, USA
- Center for Lung and Vascular Biology, University of Illinois College of MedicineChicago, IL, USA
| | - Randal A Skidgel
- Department of Pharmacology, University of Illinois College of MedicineChicago, IL, USA
- Center for Lung and Vascular Biology, University of Illinois College of MedicineChicago, IL, USA
| |
Collapse
|
33
|
Wang P, Koehle MS, Rupert JL. No association between alleles of the bradykinin receptor-B2 gene and acute mountain sickness. Exp Biol Med (Maywood) 2010; 235:737-40. [DOI: 10.1258/ebm.2010.009325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The pathophysiological mechanism(s) of the development of acute mountain sickness (AMS) is still unclear. Although the chance of developing AMS and the severity of the condition are influenced by ascent rate and altitude attained, previous history is a reliable predictor of subsequent affliction, and some individuals and families are clearly predisposed, suggesting a genetic component to susceptibility. As the vasodilator bradykinin may be involved in acclimatization to altitude, we hypothesized that variants in genes encoding components of this pathway might play a role in AMS susceptibility. We tested this by looking for associations between two functional polymorphisms (the in/del polymorphism +9/−9 [rs5810761] and the single-nucleotide polymorphism C − 58T [rs1799722]) of BDKRB2 (the gene encoding the bradykinin receptor B2) and susceptibility to AMS in an altitude-exposed Nepalese population. Lowland attendees ( n = 233) at a religious festival at 4380 m in the Nepalese Himalaya were recruited and assessed for AMS by clinical evaluation and Lake Louise score (LLS). Those with a clinical diagnosis of AMS and an LLS ≥3 were designated AMS+ ( n = 100) and those without a diagnosis of AMS and with an LLS <3 were categorized as AMS− ( n = 117). DNA was prepared from buccal cells, genotyped for the two polymorphisms and allele frequencies compared between the two cohorts. No association was found between alleles at either polymorphism and susceptibility to AMS ( P > 0.50), although C − 58T heterozygotes were significantly more common ( P < 0.001, χ2 = 49.6) in the subjects than would be predicted if the population was in Hardy–Weinberg equilibrium. The results of our association study do not support the hypothesis that variants in BDKRB2 influence altitude tolerance in a lowland Nepalese population; however, the deviation from Hardy–Weinberg equilibrium observed for the C − 58T polymorphism could be explained by self-selection for altitude tolerance in the festival attendees.
Collapse
Affiliation(s)
- Pei Wang
- School of Human Kinetics, University of British Columbia
| | - Michael S Koehle
- School of Human Kinetics, University of British Columbia
- Department of Family Practice, Allan McGavin Sport Medicine Centre, University of British Columbia, Vancouver, BC, Canada
| | - Jim L Rupert
- School of Human Kinetics, University of British Columbia
| |
Collapse
|
34
|
Ramirez-Sanchez I, Maya L, Ceballos G, Villarreal F. (-)-epicatechin activation of endothelial cell endothelial nitric oxide synthase, nitric oxide, and related signaling pathways. Hypertension 2010; 55:1398-405. [PMID: 20404222 DOI: 10.1161/hypertensionaha.109.147892] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent reports indicate that (-)-epicatechin can exert cardioprotective actions, which may involve endothelial nitric oxide synthase (eNOS)-mediated nitric oxide production in endothelial cells. However, the mechanism by which (-)-epicatechin activates eNOS remains unclear. In this study, we proposed to identify the intracellular pathways involved in (-)-epicatechin-induced effects on eNOS, using human coronary artery endothelial cells in culture. Treatment of cells with (-)-epicatechin led to time- and dose-dependent effects that peaked at 10 minutes at 1 mumol/L. (-)-Epicatechin treatment activates eNOS via serine 633 and serine 1177 phosphorylation and threonine 495 dephosphorylation. Using specific inhibitors, we have established the participation of the phosphatidylinositol 3-kinase pathway in eNOS activation. (-)-Epicatechin induces eNOS uncoupling from caveolin-1 and its association with calmodulin-1, suggesting the involvement of intracellular calcium. These results allowed us to propose that (-)-epicatechin effects may be dependent on actions exerted at the cell membrane level. To test this hypothesis, cells were treated with the phospholipase C inhibitor U73122, which blocked (-)-epicatechin-induced eNOS activation. We also demonstrated inositol phosphate accumulation in (-)-epicatechin-treated cells. The inhibitory effects of the preincubation of cells with the calmodulin-dependent kinase II (CaMKII) inhibitor KN-93 indicate that (-)-epicatechin-induced eNOS activation is at least partially mediated via the Ca(2+)/CaMKII pathway. The (-)-epicatechin stereoisomer catechin was only partially able to stimulate nitric oxide production in cells. Together, these results strongly suggest the presence of a cell surface acceptor-effector for the cacao flavanol (-)-epicatechin, which may mediate its cardiovascular effects.
Collapse
|
35
|
Côté J, Savard M, Bovenzi V, Dubuc C, Tremblay L, Tsanaclis AM, Fortin D, Lepage M, Gobeil F. Selective tumor blood-brain barrier opening with the kinin B2 receptor agonist [Phe(8)psi(CH(2)NH)Arg(9)]-BK in a F98 glioma rat model: an MRI study. Neuropeptides 2010; 44:177-85. [PMID: 20080302 DOI: 10.1016/j.npep.2009.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 12/10/2009] [Accepted: 12/14/2009] [Indexed: 11/17/2022]
Abstract
Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain barrier (BBB). One approach for transporting drugs across the BBB involves the activation of bradykinin-B2 receptors (BK-B2R). Our objective was to pharmacologically characterize the BBB permeability induced by the synthetic biostable BK-B2R analogue [Phe(8)psi(CH(2)NH)Arg(9)]-BK (R523) in F98 glioma-implanted Fischer rats. On day 10 post-inoculation, we detected the presence of B2R in the tumor cells and the peritumoral microvasculature (RT-PCR and immunohistochemistry). We assessed BBB permeability before and after the intracarotid (i.c.) infusion of R523 (0.1ml/min for 5min; 2.5, 10, and 50nmol/kg/min) using non-invasive dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with the different sized-contrast agents Gd-DTPA (0.5kDa) and Gadomer (17kDa) (0.25mmol/kg via the caudal vein). T(1)-weighted images were analyzed for the presence or absence of contrast enhancement within and surrounding the tumor area and mathematically processed to yield a contrast agent distribution volume (CADV), which was used as an indicator of vascular permeability. Our results showed that the agonist R523 increased, in a dose-dependent manner, the CADV indexes of Gd-DTPA and Gadomer, with a maximum 2-fold increase in brain uptake of both CA. The increase in CADV induced by R523 (10nmol/kg/min) was prevented by the B2R antagonist HOE140 (20nmol/kg/min, i.c.) and the nitric oxide synthase inhibitor L-NA (5mg/kg, i.v.) but not by the B1R antagonist R892 (20nmol/kg/min, i.c.) or the cyclooxygenase inhibitor Meclofenamate (5mg/kg, i.v.). The BBB permeabilizing effect of R523 (10nmol/kg/min) lasted for <1h and was accompanied by a dose-related fall in arterial blood pressure. We concluded that R523 allows the extravasation of hydrophilic macromolecular agents (17kDa) into tumor tissues by inducing selective tumor BBB permeability via B2R- and NO-dependent mechanisms.
Collapse
Affiliation(s)
- Jérôme Côté
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kuhr F, Lowry J, Zhang Y, Brovkovych V, Skidgel RA. Differential regulation of inducible and endothelial nitric oxide synthase by kinin B1 and B2 receptors. Neuropeptides 2010; 44:145-54. [PMID: 20045558 PMCID: PMC2830320 DOI: 10.1016/j.npep.2009.12.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 12/01/2009] [Accepted: 12/04/2009] [Indexed: 02/07/2023]
Abstract
Kinins are vasoactive peptides that play important roles in cardiovascular homeostasis, pain and inflammation. After release from their precursor kininogens, kinins or their C-terminal des-Arg metabolites activate two distinct G protein-coupled receptors (GPCR), called B2 (B2R) or B1 (B1R). The B2R is expressed constitutively with a wide tissue distribution. In contrast, the B1R is not expressed under normal conditions but is upregulated by tissue insult or inflammatory mediators. The B2R is considered to mediate many of the acute effects of kinins while the B1R is more responsible for chronic responses in inflammation. Both receptors can couple to Galphai and Galphaq families of G proteins to release mediators such as nitric oxide (NO), arachidonic acid, prostaglandins, leukotrienes and endothelium-derived hyperpolarizing factor and can induce the release of other inflammatory agents. The focus of this review is on the different transduction events that take place upon B2R and B1R activation in human endothelial cells that leads to generation of NO via activation of different NOS isoforms. Importantly, B2R-mediated eNOS activation leads to a transient ( approximately 5min) output of NO in control endothelial cells whereas in cytokine-treated endothelial cells, B1R activation leads to very high and prolonged ( approximately 90min) NO production that is mediated by a novel signal transduction pathway leading to post-translational activation of iNOS.
Collapse
Affiliation(s)
- F Kuhr
- Department of Pharmacology, University of Illinois at Chicago, College of Medicine, 835 South Wolcott, (M/C 868), Chicago, IL 60612, United States
| | | | | | | | | |
Collapse
|
37
|
Clapp C, Thebault S, Jeziorski MC, Martínez De La Escalera G. Peptide hormone regulation of angiogenesis. Physiol Rev 2009; 89:1177-215. [PMID: 19789380 DOI: 10.1152/physrev.00024.2009] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is now apparent that regulation of blood vessel growth contributes to the classical actions of hormones on development, growth, and reproduction. Endothelial cells are ideally positioned to respond to hormones, which act in concert with locally produced chemical mediators to regulate their growth, motility, function, and survival. Hormones affect angiogenesis either directly through actions on endothelial cells or indirectly by regulating proangiogenic factors like vascular endothelial growth factor. Importantly, the local microenvironment of endothelial cells can determine the outcome of hormone action on angiogenesis. Members of the growth hormone/prolactin/placental lactogen, the renin-angiotensin, and the kallikrein-kinin systems that exert stimulatory effects on angiogenesis can acquire antiangiogenic properties after undergoing proteolytic cleavage. In view of the opposing effects of hormonal fragments and precursor molecules, the regulation of the proteases responsible for specific protein cleavage represents an efficient mechanism for balancing angiogenesis. This review presents an overview of the actions on angiogenesis of the above-mentioned peptide hormonal families and addresses how specific proteolysis alters the final outcome of these actions in the context of health and disease.
Collapse
Affiliation(s)
- Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico.
| | | | | | | |
Collapse
|
38
|
Giustiniani J, Couloubaly S, Baillet A, Pourci ML, Cantaloube I, Fourniat C, Paul JL, Poüs C. Basal endothelial nitric oxide synthase (eNOS) phosphorylation on Ser(1177) occurs in a stable microtubule- and tubulin acetylation-dependent manner. Exp Cell Res 2009; 315:3509-20. [PMID: 19632222 DOI: 10.1016/j.yexcr.2009.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 07/17/2009] [Accepted: 07/18/2009] [Indexed: 02/07/2023]
Abstract
To better understand the relationship between the subcellular compartmentalization of endothelial nitric oxide synthase (eNOS) and its function in endothelial cells, we addressed the roles of the microtubule network and of its dynamics in organizing Golgi-bound eNOS. We found that part of Golgi-bound eNOS localizes to the trans-Golgi network and/or to trans-Golgi network-derived vesicles and membrane tubules that are organized preferentially by stable microtubules. Also, while most of cellular eNOS was recovered in a detergent-resistant microtubule-enriched subcellular fraction, its recovery was impaired after total microtubule disassembly, but not after selective disassembly of dynamic microtubules or after microtubule stabilization. Basal eNOS phosphorylation on Ser(1177) further required the association of the trans-Golgi network to stable microtubules and was enhanced by microtubule stabilization. We finally show that the involvement of stable microtubules in basal eNOS phosphorylation involved alpha-tubulin acetylation. Microtubule-dependent organization of subcellular eNOS and control over its phosphorylation would thus be essential for endothelial cells to maintain their basal eNOS function.
Collapse
Affiliation(s)
- J Giustiniani
- Laboratoire de Biochimie et Biologie Cellulaire, Univ. Paris-Sud 11, JE 2493, IFR141, Faculté de Pharmacie, Châtenay-Malabry, France
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Yeh JC, Otte LA, Frangos JA. Regulation of G protein-coupled receptor activities by the platelet-endothelial cell adhesion molecule, PECAM-1. Biochemistry 2008; 47:9029-39. [PMID: 18672896 DOI: 10.1021/bi8003846] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It is becoming increasingly evident that the cell-cell junction is a major signaling center. Here we show that the Galphaq/11 subunit of heterotrimeric G proteins forms a complex with platelet-endothelial cell adhesion molecule 1 (PECAM-1), a junctional protein that has been shown to be involved in mechanosignaling in endothelial cells. To understand the role of PECAM-1 in this complex, we determined the critical regions of PECAM-1 involved in this interaction. By expressing truncated forms of PECAM-1 in human embryonic kidney (HEK293) cells, we found that the cytoplasmic domain of PECAM-1 is not required for its association with Galphaq/11. Domain swapping of PECAM-1 with intracellular cell adhesion molecule 1 (ICAM-1), a protein that does not form a complex with Galphaq/11, provides evidence that the extracellular domain of PECAM-1 is critical for this interaction. This result also suggests that PECAM-1 does not directly interact with Galphaq/11. Coexpression of bradykinin receptor B2 (BKRB2), a Galphaq/11-coupled receptor, with PECAM-1 enhances formation of the PECAM-1-Galphaq/11 complex, suggesting an interaction between PECAM-1 and BKRB2. Co-immunoprecipitation experiments indicate that these two molecules indeed form a complex when expressed in HEK293 cells. Activation of ERK1/2 by bradykinin in HUVEC is enhanced when PECAM-1 expression is inhibited by transfection of small interference RNA against PECAM-1. Taken together, our results provide evidence of interaction of PECAM-1 with BKRB2 and of its possible role in regulating G protein-coupled receptor (GPCR) and G protein functions.
Collapse
Affiliation(s)
- Jiunn-chern Yeh
- La Jolla Bioengineering Institute, 505 Coast Boulevard South, Suite 406, La Jolla, California 92037, USA
| | | | | |
Collapse
|
40
|
Martins AH, Alves JM, Trujillo CA, Schwindt TT, Barnabé GF, Motta FLT, Guimaraes AO, Casarini DE, Mello LE, Pesquero JB, Ulrich H. Kinin-B2 receptor expression and activity during differentiation of embryonic rat neurospheres. Cytometry A 2008; 73:361-8. [PMID: 18302192 DOI: 10.1002/cyto.a.20519] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neural progenitor cells were isolated from rat fetal telencephalon and proliferate as neurospheres in the presence of EGF, FGF-2, and heparin. In the absence of these growth factors, neurospheres differentiate into neurons, astrocytes, and oligodendrocytes. Using an embryonal carcinoma cell line as in vitro differentiation model, we have already demonstrated the presence of an autocrine loop system between kinin-B2 receptor activity and secretion of its ligand bradykinin (BK) as prerequisites for final neuronal differentiation (Martins et al., J Biol Chem 2005; 280: 19576-19586). The aim of this study was to verify the activity of the kallikrein-kinin system (KKS) during neural progenitor cell differentiation. Immunofluorescence studies and flow cytometry analysis revealed increases in glial fibrillary acidic protein and beta-3 tubulin expression and decrease in the number of nestin-positive cells along neurospheres differentiation, indicating the transition of neural progenitor cells to astrocytes and neurons. Kinin-B2 receptor expression and activity, secretion of BK into the medium, and presence of high-molecular weight kininogen suggest the participation of the KKS in neurosphere differentiation. Functional kinin-B2 receptors and BK secretion indicate an autocrine loop during neurosphere differentiation to neurons, astrocytes, and oligodendrocytes, reflecting events occurring during early brain development.
Collapse
Affiliation(s)
- Antonio H Martins
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Angiotensin II (Ang II) signaling is mediated by two receptor subtypes, type 1 (AT(1)) and type 2 (AT(2)). The activation of AT(1) receptors is responsible for the development of Ang II-dependent hypertension, whereas the activation of AT(2) receptor is thought to play a counter-regulatory protective role in the regulation of blood pressure that opposes the AT(1) receptor-mediated vasoconstriction. However, the precise mechanisms by which increased numbers of AT(2) receptors counterbalance the AT(1)-mediated actions of Ang II are unknown. We have demonstrated that the abdominal aortic banding in mice and rats and the 2-kidney, 1-clip Goldblatt model of hypertension in mice induces up-regulation of AT(2) receptors in the pressure-overloaded thoracic aorta. In these hypertensive animals, the AT(1)-receptor antagonists but not calcium antagonist abolish up-regulation of the aortic AT(2) receptor as well as blood pressure elevation, suggesting that the pressure-overload up-regulates the aortic AT(2) receptor by Ang II via the activation of AT(1) receptor. Ang II binding to up-regulated AT(2) receptors induces vasodilation in these aortas through bradykinin B(2)-receptor-mediated phosphorylation of endothelial nitric oxide synthase (eNOS) at Ser(633) and Ser(1177) via a protein kinase A-dependent signaling pathway, resulting in sustained production of nitric oxide. These studies provide evidence that the vascular AT(2) receptor is up-regulated in the course of hypertension through the activation of AT(1) receptor, thereby activating a vasodilatory pathway in vessels through the AT(2) receptor via the bradykinin/nitric oxide/cGMP. This issue is important because the antihypertensive effect of AT(1)-receptor blockers is, at least in part, dependent on AT(2)-receptor activation.
Collapse
Affiliation(s)
- Katsutoshi Yayama
- Laboratory of Cardiovascular Pharmacology, Department of Biopharmaceutical Sciences, Kobe Gakuin University, Minatojima, Kobe, Japan
| | | |
Collapse
|
42
|
Zhang Y, Brovkovych V, Brovkovych S, Tan F, Lee BS, Sharma T, Skidgel RA. Dynamic receptor-dependent activation of inducible nitric-oxide synthase by ERK-mediated phosphorylation of Ser745. J Biol Chem 2007; 282:32453-61. [PMID: 17804409 DOI: 10.1074/jbc.m706242200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nitric oxide (NO) is a pleiotropic regulator of vascular function, and its overproduction by inducible nitric-oxide synthase (iNOS) in inflammatory conditions plays an important role in the pathogenesis of vascular diseases. iNOS activity is thought to be regulated primarily at the level of expression to generate "high output" NO compared with constitutive NO synthases. Here we show iNOS activity is acutely up-regulated by activation of the B1-kinin receptor (B1R) in human endothelial cells or transfected HEK293 cells to generate 2.5-5-fold higher NO than that stimulated by Arg alone. Increased iNOS activity was dependent on B1R activation of the MAPK ERK. In HEK293 cells transfected with human iNOS and B1R, ERK phosphorylated iNOS on Ser745 as determined by Western analysis using phospho-Ser antibody, in vitro kinase assays with activated ERK, and MALDI-TOF mass spectrometry. Mutation of Ser745 to Ala did not affect basal iNOS activity but eliminated iNOS phosphorylation and activation in response to B1R agonist. Mutation of Ser745 to Asp resulted in a basally hyperactive iNOS whose activity was not further increased by B1R agonist. ERK and phospho-ERK (after B1R activation) were co-localized with iNOS as determined by confocal fluorescence microscopy. Furthermore, ERK co-immunoprecipitated with iNOS. The discovery that iNOS can be phosphorylated by ERK and acutely activated by receptor-mediated signaling reveals a new level of regulation for this isoform. These findings provide a novel therapeutic target to explore in the treatment of vascular inflammatory diseases.
Collapse
Affiliation(s)
- Yongkang Zhang
- Department of Pharmacology, Protein Research Laboratory, University of Illinois at Chicago College of Medicine 60612, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Li C, Ruan L, Sood SG, Papapetropoulos A, Fulton D, Venema RC. Role of eNOS phosphorylation at Ser-116 in regulation of eNOS activity in endothelial cells. Vascul Pharmacol 2007; 47:257-64. [PMID: 17822962 PMCID: PMC2128865 DOI: 10.1016/j.vph.2007.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2007] [Accepted: 07/27/2007] [Indexed: 11/20/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) catalyzes the conversion of L-arginine to L-citrulline and nitric oxide (NO), an important modulator of vascular function. eNOS is regulated post-translationally through phosphorylation/dephosphorylation at a number of specific phosphorylation sites including Ser-116 in the bovine eNOS sequence. Whether phosphorylation of eNOS at Ser-116 in endothelial cells is stimulatory or inhibitory has not previously been definitively determined. In this study we show that mimicking phosphorylation of eNOS at Ser-116 by Asp mutation reduces basal NO release from endothelial cells. Preventing phosphorylation at this site by Ala mutation increases the amount of NO release from endothelial cells in response to agonist stimulation. In addition, mimicking phosphorylation of Ser-116 increases eNOS association with caveolin-1 and reduces the vascular reactivity of intact aortic rings. eNOS phosphorylation at Ser-116, therefore, appears to contribute to negative modulation of eNOS activity and hence to regulation of vascular tone.
Collapse
Affiliation(s)
- Chunying Li
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912, U.S.A
| | - Ling Ruan
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912, U.S.A
| | - Sarika G. Sood
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912, U.S.A
| | - Andreas Papapetropoulos
- G.P. Livanos and M. Simou Laboratories, Evangelismos Hospital, Department of Critical Care and Pulmonary Services, University of Athens School of Medicine, Athens, Greece
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912, U.S.A
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912, U.S.A
| | - Richard C. Venema
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912, U.S.A
- Department of Pediatrics, Medical College of Georgia, Augusta, GA 30912, U.S.A
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912, U.S.A
| |
Collapse
|
44
|
Dimitropoulou C, Chatterjee A, McCloud L, Yetik-Anacak G, Catravas JD. Angiotensin, bradykinin and the endothelium. Handb Exp Pharmacol 2007:255-94. [PMID: 16999222 DOI: 10.1007/3-540-32967-6_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Angiotensins and kinins are endogenous peptides with diverse biological actions; as such, they represent current and future targets of therapeutic intervention. The field of angiotensin biology has changed significantly over the last 50 years. Our original understanding of the crucial role of angiotensin II in the regulation of vascular tone and electrolyte homeostasis has been expanded to include the discovery of new angiotensins, their important role in cardiovascular inflammation and the development of clinically useful synthesis inhibitors and receptor antagonists. While less applied progress has been achieved in the kinin field, there are continuous discoveries in bradykinin physiology and in the complexity of kinin interactions with other proteins. The present review focuses on mechanisms and interactions of angiotensins and kinins that deal specifically with vascular endothelium.
Collapse
Affiliation(s)
- C Dimitropoulou
- Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912-2500, USA
| | | | | | | | | |
Collapse
|
45
|
Madeddu P, Emanueli C, El-Dahr S. Mechanisms of Disease: the tissue kallikrein–kinin system in hypertension and vascular remodeling. ACTA ACUST UNITED AC 2007; 3:208-21. [PMID: 17389890 DOI: 10.1038/ncpneph0444] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Accepted: 01/16/2007] [Indexed: 11/09/2022]
Abstract
The pathogenesis of arterial hypertension often involves a rise in systemic vascular resistance (vasoconstriction and vascular remodeling) and impairment of salt excretion in the kidney (inappropriate salt retention despite elevated blood pressure). Experimental and clinical evidence implicate an imbalance between endogenous vasoconstrictor and vasodilator systems in the development and maintenance of hypertension. Kinins (bradykinin and lys-bradykinin) are endogenous vasodilators and natriuretic peptides known best for their ability to antagonize angiotensin-induced vasoconstriction and sodium retention. In humans, angiotensin-converting enzyme inhibitors, a potent class of antihypertensive agents, lower blood pressure at least partially by favoring enhanced kinin accumulation in plasma and target tissues. The beneficial actions of kinins in renal and cardiovascular disease are largely mediated by nitric oxide and prostaglandins, and extend beyond their recognized role in lowering blood pressure to include cardioprotection and nephroprotection. This article is a review of exciting, recently generated genetic, biochemical and clinical data from studies that have examined the importance of the tissue kallikrein-kinin system in protection from hypertension, vascular remodeling and renal fibrosis. Development of novel therapeutic approaches to bolster kinin activity in the vascular wall and in specific compartments in the kidney might be a highly effective strategy for the treatment of hypertension and its complications, including cardiac hypertrophy and renal failure.
Collapse
Affiliation(s)
- Paolo Madeddu
- Experimental Cardiovascular Medicine, Bristol Heart Institute, Bristol University, Bristol, UK.
| | | | | |
Collapse
|
46
|
Silva CLM, Tamura EK, Macedo SMD, Cecon E, Bueno-Alves L, Farsky SHP, Ferreira ZS, Markus RP. Melatonin inhibits nitric oxide production by microvascular endothelial cells in vivo and in vitro. Br J Pharmacol 2007; 151:195-205. [PMID: 17375079 PMCID: PMC2013957 DOI: 10.1038/sj.bjp.0707225] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE We have previously shown that melatonin inhibits bradykinin-induced NO production by endothelial cells in vitro. The purpose of this investigation was to extend this observation to an in vivo condition and to explore the mechanism of action of melatonin. EXPERIMENTAL APPROACH RT-PCR assays were performed with rat cultured endothelial cells. The putative effect of melatonin upon arteriolar tone was investigated by intravital microscopy while NO production by endothelial cells in vitro was assayed by fluorimetry, and intracellular Ca(2+) measurements were assayed by confocal microscopy. KEY RESULTS No expression of the mRNA for the melatonin synthesizing enzymes, arylalkylamine N-acetyltransferase and hydroxyindole-O-methyltransferase, or for the melatonin MT(2) receptor was detected in microvascular endothelial cells. Melatonin fully inhibited L-NAME-sensitive bradykinin-induced vasodilation and also inhibited NO production induced by histamine, carbachol and 2-methylthio ATP, but did not inhibit NO production induced by ATP or alpha, beta-methylene ATP. None of its inhibitory effects was prevented by the melatonin receptor antagonist, luzindole. In nominally Ca(2+)-free solution, melatonin reduced intracellular Ca(2+) mobilization induced by bradykinin (40%) and 2-methylthio ATP (62%) but not Ca(2+) mobilization induced by ATP. CONCLUSIONS AND IMPLICATIONS We have confirmed that melatonin inhibited NO production both in vivo and in vitro. In addition, the melatonin effect was selective for some G protein-coupled receptors and most probably reflects an inhibition of Ca(2+) mobilization from intracellular stores.
Collapse
Affiliation(s)
- C L M Silva
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo São Paulo, Brazil
| | - E K Tamura
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo São Paulo, Brazil
| | - S M D Macedo
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo São Paulo, Brazil
| | - E Cecon
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo São Paulo, Brazil
| | - L Bueno-Alves
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo São Paulo, Brazil
| | - S H P Farsky
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo São Paulo, Brazil
| | - Z S Ferreira
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo São Paulo, Brazil
| | - R P Markus
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo São Paulo, Brazil
- Author for correspondence:
| |
Collapse
|
47
|
Cayla C, Todiras M, Iliescu R, Saul VV, Gross V, Pilz B, Chai G, Merino VF, Pesquero JB, Baltatu OC, Bader M. Mice deficient for both kinin receptors are normotensive and protected from endotoxin-induced hypotension. FASEB J 2007; 21:1689-98. [PMID: 17289925 DOI: 10.1096/fj.06-7175com] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Kinins play a central role in the modulation of cardiovascular function and in the pathophysiology of inflammation. These peptides mediate their effects by binding to two specific G-protein coupled receptors named B1 and B2. To evaluate the full functional relevance of the kallikrein-kinin system, we generated mice lacking both kinin receptors (B1B2-/-). Because of the close chromosomal position of both kinin receptor genes, B1B2-/- mice could not be obtained by simple breeding of the single knockout lines. Therefore, we inactivated the B1 receptor gene by homologous recombination in embryonic stem cells derived from B2-deficient animals. The B1B2-/- mice exhibited undetectable levels of mRNAs for both receptors and a lack of response to bradykinin (B2 agonist) and des-Arg9-bradykinin (B1 agonist), as attested by contractility studies with isolated smooth muscle tissues. B1B2-/- mice are healthy and fertile, and no sign of cardiac abnormality was detected. They are normotensive but exhibit a lower heart rate than controls. Furthermore, kinin receptor deficiency affects the pathogenesis of endotoxin-induced hypotension. While blood pressure decreased markedly in wild-type mice and B2-/- and moderately in B1-/- mice after bacterial lipopolysaccharide (LPS) injection, blood pressure remained unchanged in B1B2-/- mice. These results clearly demonstrate a pivotal role of kinins and their receptors in hypotension induced by endotoxemia in mice.
Collapse
Affiliation(s)
- Cécile Cayla
- Max-Delbrück-Center for Molecular Medicine, D-13092 Berlin-Buch, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mount PF, Kemp BE, Power DA. Regulation of endothelial and myocardial NO synthesis by multi-site eNOS phosphorylation. J Mol Cell Cardiol 2007; 42:271-9. [PMID: 16839566 DOI: 10.1016/j.yjmcc.2006.05.023] [Citation(s) in RCA: 378] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Revised: 05/03/2006] [Accepted: 05/31/2006] [Indexed: 02/07/2023]
Abstract
The controlled regulation of nitric oxide (NO) synthesis in endothelial cells and cardiomyocytes by the endothelial form of nitric oxide synthase (eNOS or NOS3) is essential for cardiovascular health. In recent years, a picture of complex and precise regulation of eNOS activity involving multi-site phosphorylation of specific serine and threonine residues has emerged. Regulation of endothelial NO synthesis by multi-site eNOS phosphorylation occurs in response to a wide variety of humoral, mechanical and pharmacological stimuli. This regulation involves numerous kinases and phosphatases, as well as interactions with other aspects of eNOS regulation such as Ca(2+) flux, protein-protein interactions and regulation of subcellular localization. Phosphorylation of eNOS-Ser(1177) close to the carboxy-terminal is a critical requirement for eNOS activation. In addition, phosphorylation of eNOS-Ser(633) in the flavin mononucleotide (FMN) binding domain also increases eNOS activity and appears particularly important for the maintenance of NO synthesis after initial activation by Ca(2+) flux and Ser(1177) phosphorylation. In contrast, NO synthesis is inhibited by phosphorylation of eNOS-Thr(495), which interferes with the binding of calmodulin to the eNOS calmodulin-binding domain. Regulated phosphorylation of eNOS also occurs at eNOS-Ser(114) and eNOS-Ser(615); however, the functions of these phosphorylation sites remain controversial. This review summarizes the present knowledge of the regulation of NO synthesis by multi-site eNOS phosphorylation and its relationship to other mechanisms of eNOS regulation. This progress in understanding important mechanisms controlling endothelial NO synthesis creates new opportunities to understand and potentially treat cardiovascular diseases characterized by deficient NO synthesis.
Collapse
Affiliation(s)
- Peter F Mount
- Department of Nephrology, Austin Health, Studley Road, Heidelberg 3084, Victoria, Australia.
| | | | | |
Collapse
|
49
|
|
50
|
Carey RM, Park J. Role of Angiotensin Type 2 Receptors in Vasodilation of Resistance and Capacitance Vessels. Hypertension 2006; 48:824-5. [PMID: 17015780 DOI: 10.1161/01.hyp.0000244109.55948.bc] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|