1
|
Martins L, Bader M, Pesquero JB. Kinins: Locally formed peptides during inflammation with potential use in tissue regeneration. Inflamm Res 2023; 72:1957-1963. [PMID: 37750921 DOI: 10.1007/s00011-023-01799-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 07/23/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
Kinins are a set of peptides present in tissues and involved in cardiovascular regulation, inflammation, and pain. Here, we briefly comment on recent key findings on the use of kinins in regenerative medicine.
Collapse
Affiliation(s)
- Leonardo Martins
- Division of Medical Sciences, Laboratory of Transcriptional Regulation, Institute of Medical Biology of Polish Academy of Sciences (IMB-PAN), 3a Tylna St., 90-364, Łódź, Poland.
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th floor, São Paulo, 04039032, Brazil.
- Department of Biochemistry and Molecular Biology, Federal University of São Paulo, Rua Três de Maio 100, 4th floor, São Paulo, 04044-020, Brazil.
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Potsdamer Str. 58, 10785, Berlin, Germany
| | - João Bosco Pesquero
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th floor, São Paulo, 04039032, Brazil
- Department of Biophysics, Federal University of São Paulo, Rua Botucatu 862, 6th floor, São Paulo, 04023-062, Brazil
| |
Collapse
|
2
|
Song J, Du J, Tan X, Wu Z, Yuan J, Cong B. Dexmedetomidine protects the heart against ischemia reperfusion injury via regulation of the bradykinin receptors. Eur J Pharmacol 2021; 911:174493. [PMID: 34506777 DOI: 10.1016/j.ejphar.2021.174493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Dexmedetomidine (DEX) has been reported to protect the heart against ischemia reperfusion (I/R) injury. However, the exact mechanisms are still not fully understood. METHODS AND RESULTS A rat cardiac I/R injury model was induced by ligation of the left anterior descending coronary artery for 1 h and subsequent reperfusion for 2 h, and DEX was administered intravenously 30 min before ischemia. We confirmed that DEX treatment mitigated cardiac I/R injury. Interestingly, we found that DEX regulated the expression of bradykinin (BK) receptors (B1R and B2R) in rat hearts during I/R injury and enhanced the protective action of BK administered during reperfusion. Moreover, in vitro hypoxia reoxygenation (H/R) injury was induced in neonatal rat cardiomyocytes (CMs), and DEX was administered 1 h before hypoxia. The in vitro findings were consistent with the in vivo experiments. We found that an α2-adrenoceptor (α2-AR) antagonist (yohimbine) completely aborted DEX-induced B1R and B2R regulation; an adenylyl cyclase (AC) agonist (forskolin) blocked B1R downregulation, while a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) blocked B2R upregulation. The above findings indicated that DEX interacted with α2-AR in cardiomyocytes, inhibited B1R expression via suppression of AC, and stimulated B2R expression via activation of PI3K. CONCLUSIONS DEX regulates BK receptor expression and potentiates the protection of BK in cardiac I/R injury, which suggests that modulating endogenous cardioprotective factors may play an important role in DEX-induced cardioprotection.
Collapse
Affiliation(s)
- Jinchao Song
- Department of Anesthesiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, China.
| | - Jiankui Du
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Xing Tan
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Zhaotang Wu
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Jihong Yuan
- Department of Nephropathy, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Binhai Cong
- Department of Physiology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China.
| |
Collapse
|
3
|
Sörensen-Zender I, Chen R, Rong S, David S, Melk A, Haller H, Schmitt R. Binding to carboxypeptidase M mediates protective effects of fibrinopeptide Bβ 15-42. Transl Res 2019; 213:124-135. [PMID: 31401267 DOI: 10.1016/j.trsl.2019.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 02/03/2023]
Abstract
During fibrinolysis a 28-amino-acid peptide is generated besides other degradation products of fibrin. This peptide, called Bβ15-42, which is cleaved by plasmin from the end of the fibrin Bβ-chain, is protective in myocardial and renal ischemia/reperfusion injury and improves the outcome in experimental sepsis. Bβ15-42 has been shown to mediate different beneficial effects in endothelial cells through binding to vascular endothelial-cadherin. Here, we provide in vitro and in vivo evidence that Bβ15-42 has additional cell protective activity in tubular cells, which is caused by a distinct mechanism. As vascular endothelial-cadherin is not expressed by tubular cells we used ligand-receptor capture technology LRC-TriCEPS to search for tubular cell surface receptors and identified carboxypeptidase M (CBPM) as a novel binding partner of Bβ15-42. Silencing CBPM with siRNA reduced the protective potential of Bβ15-42 against tubular cell stress. Bβ15-42 inhibited the enzymatic activity of CBPM and modified the impact of CBPM on bradykinin signaling. We conclude that beneficial properties of Bβ15-42 are not restricted to endothelial cells but are also active in epithelial cells where cytoprotection depends on CBPM binding.
Collapse
Affiliation(s)
| | - Rongjun Chen
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| | - Song Rong
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| | - Sascha David
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Children's Hospital, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Medical School Hannover, Germany
| |
Collapse
|
4
|
Mesquita TRR, Miguel-dos-Santos R, Jesus ICGD, de Almeida GKM, Fernandes VA, Gomes AAL, Guatimosim S, Martins-Silva L, Ferreira AJ, Capettini LDSA, Pesquero JL, Lauton-Santos S. Ablation of B1- and B2-kinin receptors causes cardiac dysfunction through redox-nitroso unbalance. Life Sci 2019; 228:121-127. [DOI: 10.1016/j.lfs.2019.04.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 01/03/2023]
|
5
|
Deres L, Eros K, Horvath O, Bencze N, Cseko C, Farkas S, Habon T, Toth K, Halmosi R. The Effects of Bradykinin B1 Receptor Antagonism on the Myocardial and Vascular Consequences of Hypertension in SHR Rats. Front Physiol 2019; 10:624. [PMID: 31178756 PMCID: PMC6537226 DOI: 10.3389/fphys.2019.00624] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 05/02/2019] [Indexed: 01/20/2023] Open
Abstract
It is known that non-steroidal anti-inflammatory drugs increase cardiovascular (CV) morbidity and mortality. In this study, we examined whether a novel anti-inflammatory drug, bradykinin B1 receptor antagonist (FGY-1153) treatment could influence the development of hypertensive organ damages in spontaneously hypertensive rats (SHR). SHRs were treated with low (FGY-120) or high dose FGY-1153 (FGY-400) and with placebo (Control) for 26 weeks. Wistar–Kyoto rats were used as aged-matched, normotensive controls (WKY). Body weight, food consumption and blood pressure were measured regularly. Echocardiography was performed at the beginning and at the end of the study. Light and electron microscopic analysis of heart and great vessels were performed, and the extent of fibrotic areas was measured. The phosphorylation state of prosurvival Akt-1/glycogen synthase kinase (GSK)-3β pathway and the activation of signaling factors playing part in the fibrotic processes – mitogen activated protein kinases (MAPKs), and TGF-β/Smad2 – were monitored using Western-blot. Body weight and food consumption as well as the elevated blood pressure in SHRs was not influenced by FGY-1153 treatment. However, both doses of FGY-1153 treatment decreased left ventricular (LV) hypertrophy and diastolic dysfunction in hypertensive animals. Moreover systolic LV function was also preserved in FGY-120 group. Increased intima-media thickness and interstitial fibrosis were not significantly diminished in great vessels. FGY-1153 treatment inhibited the expression of TGFβ and the phosphorylation of SMAD2 in the heart. Our results suggest that the tested novel anti-inflammatory compound has no deleterious effect on CV system, moreover it exerts moderate protective effect against the development of hypertensive cardiopathy.
Collapse
Affiliation(s)
- Laszlo Deres
- Medical School, University of Pécs, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Krisztian Eros
- Medical School, University of Pécs, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Orsolya Horvath
- Medical School, University of Pécs, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Noemi Bencze
- Medical School, University of Pécs, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | | | | | - Tamas Habon
- 1st Department of Medicine, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Kalman Toth
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary.,1st Department of Medicine, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Robert Halmosi
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary.,1st Department of Medicine, Clinical Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
6
|
Levy RF, Serra AJ, Antonio EL, Dos Santos L, Bocalini DS, Pesquero JB, Bader M, Merino VF, de Oliveira HA, de Arruda Veiga EC, Silva JA, Tucci PJ. Cardiac morphofunctional characteristics of transgenic rats with overexpression of the bradykinin B1 receptor in the endothelium. Physiol Res 2017; 66:925-932. [PMID: 28937259 DOI: 10.33549/physiolres.933596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Our aim was to evaluate whether endothelial overexpressing of the bradykinin B1 receptor could be associated with altered left ventricular and myocardial performance. Echocardiography and hemodynamic were employed to assess left ventricular morphology and function in Sprague Dawley transgenic rats overexpressing the endothelial bradykinin B1 receptor (Tie2B1 rats). The myocardial inotropism was evaluated on papillary muscles contracting in vitro. In Tie2B1 animals, an enlarged left ventricular cavity and lower fractional shortening coupled with a lower rate of pressure change values indicated depressed left ventricular performance. Papillary muscle mechanics revealed that both Tie2B1 and wild-type rat groups had the same contractile capacities under basal conditions; however, in transgenic animals, there was accentuated inotropism due to post-pause potentiation. Following treatment with the Arg(9)-BK agonist, Tie2B1 papillary muscles displayed a reduction in myocardial inotropism. Endothelial B1 receptor overexpression has expanded the LV cavity and worsened its function. There was an exacerbated response of papillary muscle in vitro to a prolonged resting pause, and the use of a B1 receptor agonist impairs myocardial inotropism.
Collapse
Affiliation(s)
- R F Levy
- Universidade Federal da Paraíba, Joao Pessoa, Paraíba, Brazil, Universidade Federal de Sao Paulo, Sao Paulo, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Lin X, Bernloehr C, Hildebrandt T, Stadler FJ, Doods H, Wu D. Kinin B1 receptor blockade and ACE inhibition attenuate cardiac postinfarction remodeling and heart failure in rats. Toxicol Appl Pharmacol 2016; 305:153-160. [PMID: 27288733 DOI: 10.1016/j.taap.2016.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The aim of the present study was to evaluate the effects of the novel kinin B1 receptor antagonist BI113823 on postinfarction cardiac remodeling and heart failure, and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin 1 converting enzyme (ACE) inhibitor in rats. METHODS AND RESULTS Sprague Dawley rats were subjected to permanent occlusion of the left coronary artery. Cardiovascular function was determined at 6weeks postinfarction. Treatment with either B1 receptor antagonist (BI113823) or an ACE inhibitor (lisinopril) alone or in combination significantly reduced the heart weight-to-body weight and lung weight-to-body weight ratios, and improved postinfarction cardiac function as evidenced by greater cardiac output, the maximum rate of left ventricular pressure rise (±dP/dtmax), left ventricle ejection fraction, fractional shorting, better wall motion, and attenuation of elevated left ventricular end diastolic pressure (LVEDP). Furthermore, all three treatment groups exhibited significant reduction in cardiac interstitial fibrosis, collagen deposition, CD68 positive macrophages, neutrophils, and proinflammatory cytokine production (TNF-α and IL-1β), compared to vehicle controls. CONCLUSION The present study shows that treatment with the novel kinin B1 receptor antagonist, BI113823, reduces postinfarction cardiac remodeling and heart failure, and does not influence the cardiovascular effects of the ACE inhibitor.
Collapse
Affiliation(s)
- Xinchun Lin
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | | | | | - Florian J Stadler
- Shenzhen Engineering Laboratory for Advanced Technology of Ceramics, Shenzhen 518060, PR China.
| | - Henri Doods
- Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Dongmei Wu
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; Department of BIN Convergence Technology, Chonbuk National University, South Korea.
| |
Collapse
|
8
|
Sang H, Liu L, Wang L, Qiu Z, Li M, Yu L, Zhang H, Shi R, Yu S, Guo R, Ye R, Liu X, Zhang R. Opposite roles of bradykinin B1 and B2 receptors during cerebral ischaemia-reperfusion injury in experimental diabetic rats. Eur J Neurosci 2016; 43:53-65. [PMID: 26565562 DOI: 10.1111/ejn.13133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/29/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Hongfei Sang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ling Liu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Liumin Wang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Zhongming Qiu
- Department of Neurology; The 117th Hospital of PLA; Xihu District Hangzhou Zhejiang Province China
| | - Min Li
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Linjie Yu
- Nanjing University School of Medicine; Nanjing China
| | - Hao Zhang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruifeng Shi
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Shuhong Yu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruibing Guo
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruidong Ye
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Xinfeng Liu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Renliang Zhang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| |
Collapse
|
9
|
More AS, Kim HM, Khang G, Hildebrandt T, Bernlöhr C, Doods H, Vanhoutte PM, Wu D. Des-Arg9-bradykinin causes kinin B1 receptor mediated endothelium-independent contractions in endotoxin-treated porcine coronary arteries. Pharmacol Res 2014; 90:18-24. [DOI: 10.1016/j.phrs.2014.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/12/2014] [Accepted: 09/12/2014] [Indexed: 10/24/2022]
|
10
|
Ceravolo GS, Montezano AC, Jordão MT, Akamine EH, Costa TJ, Takano AP, Fernandes DC, Barreto-Chaves ML, Laurindo FR, Tostes RC, Fortes ZB, Chopard RP, Touyz RM, Carvalho MHC. An interaction of renin-angiotensin and kallikrein-kinin systems contributes to vascular hypertrophy in angiotensin II-induced hypertension: in vivo and in vitro studies. PLoS One 2014; 9:e111117. [PMID: 25369284 PMCID: PMC4219703 DOI: 10.1371/journal.pone.0111117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 09/24/2014] [Indexed: 01/29/2023] Open
Abstract
The kallikrein-kinin and renin-angiotensin systems interact at multiple levels. In the present study, we tested the hypothesis that the B1 kinin receptor (B1R) contributes to vascular hypertrophy in angiotensin II (ANG II)-induced hypertension, through a mechanism involving reactive oxygen species (ROS) generation and extracellular signal-regulated kinase (ERK1/2) activation. Male Wistar rats were infused with vehicle (control rats), 400 ng/Kg/min ANG II (ANG II rats) or 400 ng/Kg/min ANG II plus B1 receptor antagonist, 350 ng/Kg/min des-Arg(9)-Leu(8)-bradykinin (ANGII+DAL rats), via osmotic mini-pumps (14 days) or received ANG II plus losartan (10 mg/Kg, 14 days, gavage - ANG II+LOS rats). After 14 days, ANG II rats exhibited increased systolic arterial pressure [(mmHg) 184 ± 5.9 vs 115 ± 2.3], aortic hypertrophy; increased ROS generation [2-hydroxyethidium/dihydroethidium (EOH/DHE): 21.8 ± 2.7 vs 6.0 ± 1.8] and ERK1/2 phosphorylation (% of control: 218.3 ± 29.4 vs 100 ± 0.25]. B1R expression was increased in aortas from ANG II and ANG II+DAL rats than in aortas from the ANG II+LOS and control groups. B1R antagonism reduced aorta hypertrophy, prevented ROS generation (EOH/DHE: 9.17 ± 3.1) and ERK1/2 phosphorylation (137 ± 20.7%) in ANG II rats. Cultured aortic vascular smooth muscle cells (VSMC) stimulated with low concentrations (0.1 nM) of ANG II plus B1R agonist exhibited increased ROS generation, ERK1/2 phosphorylation, proliferating-cell nuclear antigen expression and [H3]leucine incorporation. At this concentration, neither ANG II nor the B1R agonist produced any effects when tested individually. The ANG II/B1R agonist synergism was inhibited by losartan (AT1 blocker, 10 µM), B1R antagonist (10 µM) and Tiron (superoxide anion scavenger, 10 mM). These data suggest that B1R activation contributes to ANG II-induced aortic hypertrophy. This is associated with activation of redox-regulated ERK1/2 pathway that controls aortic smooth muscle cells growth. Our findings highlight an important cross-talk between the DABK and ANG II in the vascular system and contribute to a better understanding of the mechanisms involved in vascular remodeling in hypertension.
Collapse
Affiliation(s)
- Graziela S. Ceravolo
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina, Londrina, Brazil
| | - Augusto C. Montezano
- Ottawa Health Research Institute, Kidney Research Centre, University of Ottawa, Ottawa, Canada
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Maria T. Jordão
- Department of Anatomy, Institute of Biomedical Sciences III, University of São Paulo, Sao Paulo, Brazil
| | - Eliana H. Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Tiago J. Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana P. Takano
- Department of Anatomy, Institute of Biomedical Sciences III, University of São Paulo, Sao Paulo, Brazil
| | - Denise C. Fernandes
- Vascular Biology Laboratory, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria L. Barreto-Chaves
- Department of Anatomy, Institute of Biomedical Sciences III, University of São Paulo, Sao Paulo, Brazil
| | - Francisco R. Laurindo
- Vascular Biology Laboratory, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Zuleica B. Fortes
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Renato P. Chopard
- Department of Anatomy, Institute of Biomedical Sciences III, University of São Paulo, Sao Paulo, Brazil
| | - Rhian M. Touyz
- Ottawa Health Research Institute, Kidney Research Centre, University of Ottawa, Ottawa, Canada
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Maria Helena C. Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
11
|
Marketou ME, Kontaraki J, Zacharis E, Parthenakis F, Maragkoudakis S, Gavras I, Gavras H, Vardas PE. Differential gene expression of bradykinin receptors 1 and 2 in peripheral monocytes from patients with essential hypertension. J Hum Hypertens 2014; 28:450-5. [PMID: 24401952 DOI: 10.1038/jhh.2013.133] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/07/2013] [Accepted: 11/19/2013] [Indexed: 12/26/2022]
Abstract
Bradykinin participates in various hypertensive processes, exerted via its type 1 and type 2 receptors (BKR1 and BKR2). The aim of the study was to investigate BKR1 and BK2R gene expression in peripheral monocytes in patients with essential hypertension compared with healthy individuals. Seventeen hypertensive patients (9 males, age 56 ± 7 years) and 12 healthy individuals (7 males, age 55 ± 6) participated. Mononuclear cells isolated using anti-CD14+ antibodies and mRNAs of BKR1 and BKR2 were estimated by real-time quantitative reverse transcription-PCR. Both BKR1 and BKR2 showed significantly upregulated gene expression in the group of hypertensive patients. Specifically, BKR1 gene expression was 142.1 ± 42.2 in hypertensives versus 20.2 ± 8 in controls (P = 0.024) and BKR2 was 1222.2 ± 361.6 in hypertensives versus 259.5 ± 99.1 in controls (P = 0.038). Antihypertensive treatment resulted in a decrease in BKR1 (from 142.1 ± 42.2 to 55.2 ± 17.1, P = 0.065) and in BKR2 (from 1222.2 ± 361.6 to 256.8 ± 81.8, P = 0.014) gene expression. BKR1 and BKR2 gene expression on peripheral monocytes is upregulated in essential hypertension. This may lead to functional changes in monocytes and contribute to the development of target organ damage in hypertensive patients.
Collapse
Affiliation(s)
- M E Marketou
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - J Kontaraki
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - E Zacharis
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - F Parthenakis
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - S Maragkoudakis
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| | - I Gavras
- Hypertension and Atherosclerosis Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - H Gavras
- Hypertension and Atherosclerosis Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - P E Vardas
- Cardiology Department, Heraklion University Hospital, Crete, Greece
| |
Collapse
|
12
|
Girolami JP, Blaes N, Bouby N, Alhenc-Gelas F. Genetic manipulation and genetic variation of the kallikrein-kinin system: impact on cardiovascular and renal diseases. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2014; 69:145-196. [PMID: 25130042 DOI: 10.1007/978-3-319-06683-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Genetic manipulation of the kallikrein-kinin system (KKS) in mice, with either gain or loss of function, and study of human genetic variability in KKS components which has been well documented at the phenotypic and genomic level, have allowed recognizing the physiological role of KKS in health and in disease. This role has been especially documented in the cardiovascular system and the kidney. Kinins are produced at slow rate in most organs in resting condition and/or inactivated quickly. Yet the KKS is involved in arterial function and in renal tubular function. In several pathological situations, kinin production increases, kinin receptor synthesis is upregulated, and kinins play an important role, whether beneficial or detrimental, in disease outcome. In the setting of ischemic, diabetic or hemodynamic aggression, kinin release by tissue kallikrein protects against organ damage, through B2 and/or B1 bradykinin receptor activation, depending on organ and disease. This has been well documented for the ischemic or diabetic heart, kidney and skeletal muscle, where KKS activity reduces oxidative stress, limits necrosis or fibrosis and promotes angiogenesis. On the other hand, in some pathological situations where plasma prekallikrein is inappropriately activated, excess kinin release in local or systemic circulation is detrimental, through oedema or hypotension. Putative therapeutic application of these clinical and experimental findings through current pharmacological development is discussed in the chapter.
Collapse
|
13
|
Duehrkop C, Rieben R. Ischemia/reperfusion injury: effect of simultaneous inhibition of plasma cascade systems versus specific complement inhibition. Biochem Pharmacol 2013; 88:12-22. [PMID: 24384116 DOI: 10.1016/j.bcp.2013.12.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 11/25/2013] [Accepted: 12/16/2013] [Indexed: 02/06/2023]
Abstract
Ischemia/reperfusion injury (IRI) may occur from ischemia due to thrombotic occlusion, trauma or surgical interventions, including transplantation, with subsequent reestablishment of circulation. Time-dependent molecular and structural changes result from the deprivation of blood and oxygen in the affected tissue during ischemia. Upon restoration of blood flow a multifaceted network of plasma cascades is activated, including the complement-, coagulation-, kinin-, and fibrinolytic system, which plays a major role in the reperfusion-triggered inflammatory process. The plasma cascade systems are therefore promising therapeutic targets for attenuation of IRI. Earlier studies showed beneficial effects through inhibition of the complement system using specific complement inhibitors. However, pivotal roles in IRI are also attributed to other cascades. This raises the question, whether drugs, such as C1 esterase inhibitor, which regulate more than one cascade at a time, have a higher therapeutic potential. The present review discusses different therapeutic approaches ranging from specific complement inhibition to simultaneous inhibition of plasma cascade systems for reduction of IRI, gives an overview of the plasma cascade systems in IRI as well as highlights recent findings in this field.
Collapse
Affiliation(s)
- Claudia Duehrkop
- Department of Clinical Research, University of Bern, Murtenstrasse 50, P.O. Box 44, CH-3010 Bern, Switzerland; Graduate School of Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Robert Rieben
- Department of Clinical Research, University of Bern, Murtenstrasse 50, P.O. Box 44, CH-3010 Bern, Switzerland.
| |
Collapse
|
14
|
Potier L, Waeckel L, Vincent MP, Chollet C, Gobeil F, Marre M, Bruneval P, Richer C, Roussel R, Alhenc-Gelas F, Bouby N. Selective kinin receptor agonists as cardioprotective agents in myocardial ischemia and diabetes. J Pharmacol Exp Ther 2013; 346:23-30. [PMID: 23591995 DOI: 10.1124/jpet.113.203927] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Cardiac ischemia is a leading cause of death, especially in diabetic patients. The diabetic ischemic heart is resistant experimentally to established cardioprotective treatments. New pharmacological approaches to cardiac protection are warranted. The kallikrein-kinin system is involved in myocardial protection in ischemia. Respective roles of B1 (B1R) and B2 (B2R) receptors remain controversial. We tested whether pharmacological activation of kinin receptors may have therapeutic effect in cardiac ischemia-reperfusion in nondiabetic (NDiab) and diabetic (Diab) mice. We assessed effect on infarct size (IS) and signaling pathways involved in myocardial protection of potent selective pharmacological agonists of B1R or B2R given at reperfusion. In NDiab mice, a B2R agonist reduced IS significantly by 47%, similarly to ramiprilat or ischemic postconditioning, via activation of phosphoinositide 3 kinase/Akt pathway leading to inhibition of glycogen synthase kinase-3β (GSK-3β). B1R agonist had no effect on IS. In contrast, in Diab mice, the B2R agonist, ramiprilat, or ischemic postconditioning failed to reduce IS but a B1R agonist significantly reduced IS by 44% via activation of phosphoinositide 3 kinase/Akt and extracellular signal-regulated kinase 1/2, both leading to GSK-3β inhibition. Differential effect of B2R or B1R agonists in NDiab and Diab mice can be linked to inactivation of B2R signaling and induction of B1R in heart of Diab mice. Thus, a pharmacological B2R agonist is cardioprotective in acute ischemia in nondiabetic animals. B1R agonist overcomes resistance of diabetic heart to cardioprotective treatments. Pharmacological activation of B1R and B2R may become a treatment for diabetic and nondiabetic patients, respectively, in acute coronary syndromes.
Collapse
MESH Headings
- Animals
- Bradykinin/administration & dosage
- Bradykinin/adverse effects
- Bradykinin/analogs & derivatives
- Bradykinin/therapeutic use
- Cardiotonic Agents/administration & dosage
- Cardiotonic Agents/adverse effects
- Cardiotonic Agents/therapeutic use
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Dose-Response Relationship, Drug
- Drug Resistance
- Gene Expression Regulation
- Glycogen Synthase Kinase 3/antagonists & inhibitors
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- MAP Kinase Signaling System/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Myocardial Ischemia/complications
- Myocardial Ischemia/drug therapy
- Myocardial Ischemia/metabolism
- Myocardial Reperfusion Injury/prevention & control
- Myocardium/metabolism
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Receptor, Bradykinin B1/agonists
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptor, Bradykinin B2/agonists
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Louis Potier
- INSERM U872, Research Center les Cordeliers, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Effects of a novel bradykinin B1 receptor antagonist and angiotensin II receptor blockade on experimental myocardial infarction in rats. PLoS One 2012; 7:e51151. [PMID: 23236443 PMCID: PMC3517424 DOI: 10.1371/journal.pone.0051151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/29/2012] [Indexed: 01/06/2023] Open
Abstract
Background The aim of the present study was to evaluate the cardiovascular effects of the novel bradykinin B1 receptor antagonist BI-113823 following myocardial infarction (MI) and to determine whether B1 receptor blockade alters the cardiovascular effects of an angiotensin II type 1 (AT1) receptor antagonist after MI in rats. Methodology/Principal Findings Sprague Dawley rats were subjected to permanent occlusion of the left descending coronary artery. Cardiovascular function was determined at 7 days post MI. Treatment with either B1 receptor antagonist (BI-113823) or AT1 receptor antagonist (irbesartan) alone or in combination improved post-MI cardiac function as evidenced by attenuation of elevated left ventricular end diastolic pressure (LVEDP); greater first derivative of left ventricular pressure (± dp/dt max), left ventricle ejection fraction, fractional shorting, and better wall motion; as we as reductions in post-MI up-regulation of matrix metalloproteinases 2 (MMP-2) and collagen III. In addition, the cardiac up-regulation of B1 receptor and AT1 receptor mRNA were markedly reduced in animals treated with BI 113823, although bradykinin B2 receptor and angiotensin 1 converting enzyme (ACE1) mRNA expression were not significantly affected by B1 receptor blockade. Conclusions/Significance The present study demonstrates that treatment with the novel B1 receptor antagonist, BI-113823 improves post-MI cardiac function and does not influence the cardiovascular effects of AT1 receptor antagonist following MI.
Collapse
|
16
|
Protective role of AT(2) and B(1) receptors in kinin B(2)-receptor-knockout mice with myocardial infarction. Clin Sci (Lond) 2012; 124:87-96. [PMID: 22849668 DOI: 10.1042/cs20120341] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AT(2)Rs [AngII (angiotensin II) type 2 receptors] contribute to the cardioprotective effects of angiotensin II receptor blockers, possibly via kinins acting on the B(1)R (B(1) receptor) and B(2)R (B(2) receptor). Recent studies have shown that a lack of B(2)R up-regulates B(1)R and AT(2)R; however, the pathophysiological relevance of such an event remains unclear. We hypothesized that up-regulation of AT(2)R and B(1)R compensates for the loss of B(2)R. Blockade of AT(2)R and/or B(1)R worsens cardiac remodelling and dysfunction following MI (myocardial infarction) in B(2)R(-/-) (B(2)-receptor-knockout mice). B(2)R(-/-) mice and WT (wild-type) controls were subjected to sham MI or MI and treated for 4 weeks with (i) vehicle, (ii) a B(1)R-ant (B(1)R antagonist; 300 μg/kg of body weight per day), (iii) an AT(2)R-ant [AT(2) receptor antagonist (PD123319); 20 mg/kg of body weight per day], or (iv) B(1)R-ant+AT(2)R-ant. B(2)R(-/-) mice had a greater MCSA (myocyte cross-sectional area) and ICF (interstitial collagen fraction) at baseline and after MI compared with WT controls. Cardiac function and increase in macrophage infiltration, TGFβ(1) (transforming growth factor β(1)) expression and ERK1/2 (extracellular-signal-regulated kinase 1/2) phosphorylation post-MI were similar in both strains. Blockade of AT(2)R or B(1)R worsened cardiac remodelling, hypertrophy and dysfunction associated with increased inflammation and ERK1/2 phosphorylation and decreased NO excretion in B(2)R(-/-) mice, which were exacerbated by dual blockade of B(1)R and AT(2)R. No such effects were seen in WT mice. Our results suggest that, in the absence of B(2)R, both B(1)R and AT(2)R play important compensatory roles in preventing deterioration of cardiac function and remodelling post-MI possibly via suppression of inflammation, TGFβ(1) and ERK1/2 signalling.
Collapse
|
17
|
Abstract
Diabetic nephropathy is the major cause of end-stage renal disease worldwide. Although the renin-angiotensin system has been implicated in the pathogenesis of diabetic nephropathy, angiotensin I-converting enzyme inhibitors have a beneficial effect on diabetic nephropathy independently of their effects on blood pressure and plasma angiotensin II levels. This suggests that the kallikrein-kinin system (KKS) is also involved in the disease. To study the role of the KKS in diabetic nephropathy, mice lacking either the bradykinin B1 receptor (B1R) or the bradykinin B2 receptor (B2R) have been commonly used. However, because absence of either receptor causes enhanced expression of the other, it is difficult to determine the precise functions of each receptor. This difficulty has recently been overcome by comparing mice lacking both receptors with mice lacking each receptor. Deletion of both B1R and B2R reduces nitric oxide (NO) production and aggravates renal diabetic phenotypes, relevant to either lack of B1R or B2R, demonstrating that both B1R and B2R exert protective effects on diabetic nephropathy presumably via NO. Here, we review previous epidemiological and experimental studies, and discuss novel insights regarding the therapeutic implications of the importance of the KKS in averting diabetic nephropathy.
Collapse
|
18
|
Pereira RL, Buscariollo BN, Corrêa-Costa M, Semedo P, Oliveira CD, Reis VO, Maquigussa E, Araújo RC, Braga TT, Soares MF, Moura IC, Malheiros DMAC, Filho APS, Keller AC, Câmara NOS. Bradykinin receptor 1 activation exacerbates experimental focal and segmental glomerulosclerosis. Kidney Int 2011; 79:1217-27. [PMID: 21412216 DOI: 10.1038/ki.2011.14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Focal and segmental glomerulosclerosis (FSGS) is one of the most important causes of end-stage renal failure. The bradykinin B1 receptor has been associated with tissue inflammation and renal fibrosis. To test for a role of the bradykinin B1 receptor in podocyte injury, we pharmacologically modulated its activity at different time points in an adriamycin-induced mouse model of FSGS. Estimated albuminuria and urinary protein to creatinine ratios correlated with podocytopathy. Adriamycin injection led to loss of body weight, proteinuria, and upregulation of B1 receptor mRNA. Early treatment with a B1 antagonist reduced albuminuria and glomerulosclerosis, and inhibited the adriamycin-induced downregulation of podocin, nephrin, and α-actinin-4 expression. Moreover, delayed treatment with antagonist also induced podocyte protection. Conversely, a B1 agonist aggravated renal dysfunction and even further suppressed the levels of podocyte-related molecules. Thus, we propose that kinin has a crucial role in the pathogenesis of FSGS operating through bradykinin B1 receptor signaling.
Collapse
Affiliation(s)
- Rafael L Pereira
- Departamento de Medicina, Laboratório de Imunologia Clínica e Experimental, Disciplina de Nefrologia, Universidade Federal de São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Blockade of the kinin receptor B1 protects from autoimmune CNS disease by reducing leukocyte trafficking. J Autoimmun 2011; 36:106-14. [PMID: 21216565 DOI: 10.1016/j.jaut.2010.11.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 11/30/2010] [Indexed: 01/05/2023]
Abstract
Disruption of the blood brain barrier (BBB) and transendothelial trafficking of immune cells into the central nervous system (CNS) are pathophysiological hallmarks of Multiple Sclerosis (MS) and its animal model, Experimental Autoimmune Encephalomyelitis (EAE). Kinins are proinflammatory peptides which are released during tissue injury including EAE. They increase vascular permeability and enhance inflammation by acting on distinct bradykinin receptors, B1R and B2R. We studied the expression of B1R and B2R and the effect of their inhibition on the disease course, BBB integrity and T cell migration following myelin oligodendrocyte glycoprotein (MOG(35-55))-induced EAE. B1R, but not B2R expression was markedly enhanced in inflammatory CNS lesions in mice and humans. Brain endothelial cells could be identified as major source of B1R protein. The severity of EAE was significantly alleviated in B1R(-/-) mice compared with wild-type (WT) controls (P<0.05). Treatment of WT mice with the B1R antagonist R715 before and after disease onset was equally effective (P<0.05) while B1R activation by R838 promoted EAE (P<0.05). B1R inhibition was accompanied by a remarkable reduction of BBB disruption and tissue inflammation. In vitro analyses revealed that B1R suppression reverses the upregulation of ICAM-I and VCAM-I at the inflamed BBB thereby limiting T cell transmigration. In contrast, blocking B2R had no significant impact on EAE. We conclude that B1R inhibition can reduce BBB damage and cell invasion during autoimmune CNS disease and may offer a novel anti-inflammatory strategy for the treatment of MS.
Collapse
|
20
|
Kinin B1 receptor enhances the oxidative stress in a rat model of insulin resistance: outcome in hypertension, allodynia and metabolic complications. PLoS One 2010; 5:e12622. [PMID: 20830306 PMCID: PMC2935380 DOI: 10.1371/journal.pone.0012622] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 08/12/2010] [Indexed: 01/06/2023] Open
Abstract
Background Kinin B1 receptor (B1R) is induced by the oxidative stress in models of diabetes mellitus. This study aims at determining whether B1R activation could perpetuate the oxidative stress which leads to diabetic complications. Methods and Findings Young Sprague-Dawley rats were fed with 10% D-Glucose or tap water (controls) for 8–12 weeks. A selective B1R antagonist (SSR240612) was administered acutely (3–30 mg/kg) or daily for a period of 7 days (10 mg/kg) and the impact was measured on systolic blood pressure, allodynia, protein and/or mRNA B1R expression, aortic superoxide anion (O2•−) production and expression of superoxide dismutase (MnSOD) and catalase. SSR240612 reduced dose-dependently (3–30 mg/kg) high blood pressure in 12-week glucose-fed rats, but had no effect in controls. Eight-week glucose-fed rats exhibited insulin resistance (HOMA index), hypertension, tactile and cold allodynia and significant increases of plasma levels of glucose and insulin. This was associated with higher aortic levels of O2•−, NADPH oxidase activity, MnSOD and catalase expression. All these abnormalities including B1R overexpression (spinal cord, aorta, liver and gastrocnemius muscle) were normalized by the prolonged treatment with SSR240612. The production of O2•− in the aorta of glucose-fed rats was also measured in the presence and absence of inhibitors (10–100 µM) of NADPH oxidase (apocynin), xanthine oxidase (allopurinol) or nitric oxide synthase (L-NAME) with and without Sar[D-Phe8]des-Arg9-BK (20 µM; B1R agonist). Data show that the greater aortic O2•− production induced by the B1R agonist was blocked only by apocynin. Conclusions Activation of kinin B1R increased O2•− through the activation of NADPH oxidase in the vasculature. Prolonged blockade of B1R restored cardiovascular, sensory and metabolic abnormalities by reducing oxidative stress and B1R gene expression in this model.
Collapse
|
21
|
Wende AR, Soto J, Olsen CD, Pires KMP, Schell JC, Larrieu-Lahargue F, Litwin SE, Kakoki M, Takahashi N, Smithies O, Abel ED. Loss of bradykinin signaling does not accelerate the development of cardiac dysfunction in type 1 diabetic akita mice. Endocrinology 2010; 151:3536-42. [PMID: 20501666 PMCID: PMC2940524 DOI: 10.1210/en.2010-0256] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bradykinin signaling has been proposed to play either protective or deleterious roles in the development of cardiac dysfunction in response to various pathological stimuli. To further define the role of bradykinin signaling in the diabetic heart, we examined cardiac function in mice with genetic ablation of both bradykinin B1 and B2 receptors (B1RB2R(-/-)) in the context of the Akita model of insulin-deficient type 1 diabetes (Ins2(Akita/+)). In 5-month-old diabetic and nondiabetic, wild-type and B1RB2R(-/-) mice, in vivo cardiac contractile function was determined by left-ventricular (LV) catheterization and echocardiography. Reactive oxygen species levels were measured by 2'-7'-dichlorofluorescein diacetate fluorescence. Mitochondrial function and ATP synthesis were determined in saponin-permeabilized cardiac fibers. LV systolic pressure and the peak rate of LV pressure rise and decline were decreased with diabetes but did not deteriorate further with loss of bradykinin signaling. Wall thinning and reduced ejection fractions in Akita mouse hearts were partially attenuated by B1RB2R deficiency, although other parameters of LV function were unaffected. Loss of bradykinin signaling did not increase fibrosis in Ins2(Akita/+) diabetic mouse hearts. Mitochondrial dysfunction was not exacerbated by B1RB2R deficiency, nor was there any additional increase in tissue levels of reactive oxygen species. Thus, loss of bradykinin B2 receptor signaling does not abrogate the previously reported beneficial effect of inhibition of B1 receptor signaling. In conclusion, complete loss of bradykinin expression does not worsen cardiac function or increase myocardial fibrosis in diabetes.
Collapse
MESH Headings
- Animals
- Bradykinin/metabolism
- Bradykinin/physiology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/physiopathology
- Female
- Heart/physiopathology
- Heart Diseases/etiology
- Heart Diseases/genetics
- Heart Diseases/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitochondria, Heart/pathology
- Mitochondria, Heart/physiology
- Myocardium/pathology
- Oxidative Stress/genetics
- Receptor, Bradykinin B1/deficiency
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B2/deficiency
- Receptor, Bradykinin B2/genetics
- Signal Transduction/genetics
- Time Factors
Collapse
Affiliation(s)
- Adam R Wende
- Program in Molecular Medicine and Division of Endocrinology, Metabolism, and Diabetes, University of Utah, School of Medicine, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Raslan F, Schwarz T, Meuth SG, Austinat M, Bader M, Renné T, Roosen K, Stoll G, Sirén AL, Kleinschnitz C. Inhibition of bradykinin receptor B1 protects mice from focal brain injury by reducing blood-brain barrier leakage and inflammation. J Cereb Blood Flow Metab 2010; 30:1477-86. [PMID: 20197781 PMCID: PMC2949241 DOI: 10.1038/jcbfm.2010.28] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Kinins are proinflammatory and vasoactive peptides that are released during tissue damage and may contribute to neuronal degeneration, inflammation, and edema formation after brain injury by acting on discrete bradykinin receptors, B1R and B2R. We studied the expression of B1R and B2R and the effect of their inhibition on lesion size, blood-brain barrier (BBB) disruption, and inflammatory processes after a focal cryolesion of the right parietal cortex in mice. B1R and B2R gene transcripts were significantly induced in the lesioned hemispheres of wild-type mice (P<0.05). The volume of the cortical lesions and neuronal damage at 24 h after injury in B1R(-/-) mice were significantly smaller than in wild-type controls (2.5+/-2.6 versus 11.5+/-3.9 mm(3), P<0.001). Treatment with the B1R antagonist R-715 1 h after lesion induction likewise reduced lesion volume in wild-type mice (2.6+/-1.4 versus 12.2+/-6.1 mm(3), P<0.001). This was accompanied by a remarkable reduction of BBB disruption and tissue inflammation. In contrast, genetic deletion or pharmacological inhibition of B2R had no significant impact on lesion formation or the development of brain edema. We conclude that B1R inhibition may offer a novel therapeutic strategy after acute brain injuries.
Collapse
Affiliation(s)
- Furat Raslan
- Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lack of both bradykinin B1 and B2 receptors enhances nephropathy, neuropathy, and bone mineral loss in Akita diabetic mice. Proc Natl Acad Sci U S A 2010; 107:10190-5. [PMID: 20479236 DOI: 10.1073/pnas.1005144107] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
An insertion polymorphism of the angiotensin-I converting enzyme gene (ACE) is common in humans and the higher expressing allele is associated with an increased risk of diabetic complications. The ACE polymorphism does not significantly affect blood pressure or angiotensin II levels, suggesting that the kallikrein-kinin system partly mediates the effects of the polymorphism. We have therefore explored the influence of lack of both bradykinin receptors (B1R and B2R) on diabetic nephropathy, neuropathy, and osteopathy in male mice heterozygous for the Akita diabetogenic mutation in the insulin 2 gene (Ins2). We find that all of the detrimental phenotypes observed in Akita diabetes are enhanced by lack of both B1R and B2R, including urinary albumin excretion, glomerulosclerosis, glomerular basement membrane thickening, mitochondrial DNA deletions, reduction of nerve conduction velocities and of heat sensation, and bone mineral loss. Absence of the bradykinin receptors also enhances the diabetes-associated increases in plasma thiobarbituric acid-reactive substances, mitochondrial DNA deletions, and renal expression of fibrogenic genes, including transforming growth factor beta1, connective tissue growth factor, and endothelin-1. Thus, lack of B1R and B2R exacerbates diabetic complications. The enhanced renal injury in diabetic mice caused by lack of B1R and B2R may be mediated by a combination of increases in oxidative stress, mitochondrial DNA damage and over expression of fibrogenic genes.
Collapse
|
24
|
Cardioprotective effects of a selective B(2) receptor agonist of bradykinin post-acute myocardial infarct. Am J Hypertens 2010; 23:562-8. [PMID: 20186129 DOI: 10.1038/ajh.2010.20] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The cardioprotective benefits of bradykinin are attributable to activation of its B(2) receptor (B(2)R)-mediated actions and abolished by B(2)R antagonists. The current experiments evaluated the cardioprotective potential of a potent, long-acting B(2)R-selective agonist peptide analogue of bradykinin, the compound NG291. METHODS We compared the extent of cardiac tissue damage and remodeling and expression pattern of selected genes in mice submitted to acute myocardial infarct (MI) and treated for 1 week with either NG291 [Hyp(3),Thi(5),(N)Chg(7),Thi(8)]-bradykinin or with saline delivered via osmotic minipump. RESULTS Active treatment resulted in better ejection fraction (EF) 69 +/- 1% vs. 61 +/- 3.1% (P = 0.01), (vs. 85 +/- 1.3% in sham-operated controls), fractional shortening (FS) 38 +/- 4% vs. 32 +/- 8% (NS) (vs. 53 +/- 1.2 in sham-operated controls), and fewer markers of myocyte apoptosis (TUNEL-positive nuclei 4.9 +/- 1.1% vs. 9.7 +/- 0.03%, P = 0.03). Systolic blood pressure (SBP) at end point was normal at 110 +/- 4.2 in actively treated mice, but tended to be lower at 104 +/- 4.7 mm Hg in saline controls with decreased cardiac systolic capacity. Expression patterns of selected genes to factors related to tissue injury, inflammation, and metabolism (i.e., the B(1)R, B(2)R, endothelial nitric oxide synthase (eNOS), TNF-alpha, cardiomyopathy-associated 3 (Cmya3), and pyruvate dehydrogenase kinase isoenzyme 4 (PDK4)) showed that acute MI induced significant upregulation of these genes, and active treatment prevented or attenuated this upregulation, whereas the B(2)R agonist itself produced no difference in the myocardium of sham-operated mice. CONCLUSIONS Treatment with a selective B(2)R agonist initiated at the time of induction of acute MI in mice had a beneficial effect on cardiac function, tissue remodeling, and inflammation-related tissue gene expression, which may explain its structural and functional benefits.
Collapse
|
25
|
Jones WK, Fan GC, Liao S, Zhang JM, Wang Y, Weintraub NL, Kranias EG, Schultz JE, Lorenz J, Ren X. Peripheral nociception associated with surgical incision elicits remote nonischemic cardioprotection via neurogenic activation of protein kinase C signaling. Circulation 2009; 120:S1-9. [PMID: 19752352 DOI: 10.1161/circulationaha.108.843938] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although remote ischemic stimuli have been shown to elicit cardioprotection against ischemia/reperfusion injury, there is little known about the effects of nonischemic stimuli. We previously described a remote cardioprotective effect of nonischemic surgical trauma (abdominal incision) called remote preconditioning of trauma (RPCT). In the present study, we elucidate mechanisms underlying this phenomenon. METHODS AND RESULTS We used a murine model of myocardial infarction to evaluate ischemia/reperfusion injury, and either abdominal surgical incision, or application of topical capsaicin, to elicit cardioprotection. We show that the cardioprotective effect of RPCT is initiated by skin nociception, and requires neurogenic signaling involving spinal nerves and activation of cardiac sensory and sympathetic nerves. Our results demonstrate bradykinin-dependent activation and repression, respectively, of PKCepsilon and PKCdelta in myocardium after RPCT, and we show involvement of the K(ATP) channels in cardioprotection. Finally, we show that topical application of capsaicin, which selectively activates C sensory fibers in the skin, mimics the cardioprotective effect of RPCT against myocardial infarction. CONCLUSIONS Nontraumatic nociceptive preconditioning represents a novel therapeutic strategy for cardioprotection with great potential clinical utility.
Collapse
Affiliation(s)
- W Keith Jones
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, OH 45267-0575, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bryant J, Shariat-Madar Z. Human plasma kallikrein-kinin system: physiological and biochemical parameters. Cardiovasc Hematol Agents Med Chem 2009; 7:234-50. [PMID: 19689262 PMCID: PMC4905712 DOI: 10.2174/187152509789105444] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The plasma kallikrein-kinin system (KKS) plays a critical role in human physiology. The KKS encompasses coagulation factor XII (FXII), the complex of prekallikrein (PK) and high molecular weight kininogen (HK). The conversion of plasma prekallikrein to kallikrein by the activated FXII and in response to numerous different stimuli leads to the generation of bradykinin (BK) and activated HK (HKa, an antiangiogenic peptide). BK is a proinflammatory peptide, a pain mediator and potent vasodilator, leading to robust accumulation of fluid in the interstitium. Systemic production of BK, HKa with the interplay between BK bound-BK receptors and the soluble form of HKa are key to angiogenesis and hemodynamics. KKS has been implicated in the pathogenesis of inflammation, hypertension, endotoxemia, and coagulopathy. In all these cases increased BK levels is the hallmark. In some cases, the persistent production of BK due to the deficiency of the blood protein C1-inhibitor, which controls FXII, is detrimental to the survival of the patients with hereditary angioedema (HAE). In others, the inability of angiotensin converting enzyme (ACE) to degrade BK leads to elevated BK levels and edema in patients on ACE inhibitors. Thus, the mechanisms that interfere with BK liberation or degradation would lead to blood pressure dysfunction. In contrast, anti-kallikrein treatment could have adverse effects in hemodynamic changes induced by vasoconstrictor agents. Genetic models of kallikrein deficiency are needed to evaluate the quantitative role of kallikrein and to validate whether strategies designed to activate or inhibit kallikrein may be important for regulating whole-body BK sensitivity.
Collapse
Affiliation(s)
- J.W. Bryant
- Pfizer Global Research and Development, CVMED Exploratory, Groton, CT 06340
| | - z Shariat-Madar
- School of Pharmacy, Department of Pharmacology, University of Mississippi, University, MS 38677-1848
| |
Collapse
|
27
|
Westermann D, Walther T, Savvatis K, Escher F, Sobirey M, Riad A, Bader M, Schultheiss HP, Tschöpe C. Gene deletion of the kinin receptor B1 attenuates cardiac inflammation and fibrosis during the development of experimental diabetic cardiomyopathy. Diabetes 2009; 58:1373-81. [PMID: 19276445 PMCID: PMC2682670 DOI: 10.2337/db08-0329] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Diabetic cardiomyopathy is associated with increased mortality in patients with diabetes. The underlying pathology of this disease is still under discussion. We studied the role of the kinin B1 receptor on the development of experimental diabetic cardiomyopathy. RESEARCH DESIGN AND METHODS We utilized B1 receptor knockout mice and investigated cardiac inflammation, fibrosis, and oxidative stress after induction of streptozotocin (STZ)-induced diabetes. Furthermore, the left ventricular function was measured by pressure-volume loops after 8 weeks of diabetes. RESULTS B1 receptor knockout mice showed an attenuation of diabetic cardiomyopathy with improved systolic and diastolic function in comparison with diabetic control mice. This was associated with a decreased activation state of the mitogen-activated protein kinase p38, less oxidative stress, as well as normalized cardiac inflammation, shown by fewer invading cells and no increase in matrix metalloproteinase-9 as well as the chemokine CXCL-5. Furthermore, the profibrotic connective tissue growth factor was normalized, leading to a reduction in cardiac fibrosis despite severe hyperglycemia in mice lacking the B1 receptor. CONCLUSIONS These findings suggest that the B1 receptor is detrimental in diabetic cardiomyopathy in that it mediates inflammatory and fibrotic processes. These insights might have useful implications on future studies utilizing B1 receptor antagonists for treatment of human diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Dirk Westermann
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Thomas Walther
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Konstantinos Savvatis
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Felcicitas Escher
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Meike Sobirey
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Alexander Riad
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - Heinz-Peter Schultheiss
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Corresponding author: Carsten Tschöpe,
| |
Collapse
|
28
|
Côté J, Savard M, Bovenzi V, Bélanger S, Morin J, Neugebauer W, Larouche A, Dubuc C, Gobeil F. Novel kinin B1 receptor agonists with improved pharmacological profiles. Peptides 2009; 30:788-95. [PMID: 19150636 DOI: 10.1016/j.peptides.2008.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 12/17/2008] [Accepted: 12/18/2008] [Indexed: 10/21/2022]
Abstract
There is some evidence to suggest that inducible kinin B1 receptors (B1R) may play beneficial and protecting roles in cardiovascular-related pathologies such as hypertension, diabetes, and ischemic organ diseases. Peptide B1R agonists bearing optimized pharmacological features (high potency, selectivity and stability toward proteolysis) hold promise as valuable therapeutic agents in the treatment of these diseases. In the present study, we used solid-phase methodology to synthesize a series of novel peptide analogues based on the sequence of Sar[dPhe(8)]desArg(9)-bradykinin, a relatively stable peptide agonist with moderate affinity for the human B1R. We evaluated the pharmacological properties of these peptides using (1) in vitro competitive binding experiments on recombinant human B1R and B2R (for index of selectivity determination) in transiently transfected human embryonic kidney 293 cells (HEK-293T cells), (2) ex vivo vasomotor assays on isolated human umbilical veins expressing endogenous human B1R, and (3) in vivo blood pressure tests using anesthetized lipopolysaccharide-immunostimulated rabbits. Key chemical modifications at the N-terminus, the positions 3 and 5 on Sar[dPhe(8)]desArg(9)-bradykinin led to potent analogues. For example, peptides 18 (SarLys[Hyp(3),Cha(5), dPhe(8)]desArg(9)-bradykinin) and 20 (SarLys[Hyp(3),Igl(5), dPhe(8)]desArg(9)-bradykinin) outperformed the parental molecule in terms of affinity, functional potency and duration of action in vitro and in vivo. These selective agonists should be valuable in future animal and human studies to investigate the potential benefits of B1R activation.
Collapse
Affiliation(s)
- Jérôme Côté
- Department of Pharmacology, Université de Sherbrooke, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Austinat M, Braeuninger S, Pesquero JB, Brede M, Bader M, Stoll G, Renné T, Kleinschnitz C. Blockade of Bradykinin Receptor B1 but Not Bradykinin Receptor B2 Provides Protection From Cerebral Infarction and Brain Edema. Stroke 2009; 40:285-93. [DOI: 10.1161/strokeaha.108.526673] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Madeleine Austinat
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - Stefan Braeuninger
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - João B. Pesquero
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - Marc Brede
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - Michael Bader
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - Guido Stoll
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - Thomas Renné
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| | - Christoph Kleinschnitz
- From Department of Neurology (M.A., S.B., G.S., C.K.), Department of Anesthesiology (M.Brede), and Institute for Clinical Biochemistry and Pathobiochemistry (T.R.), University of Würzburg, Würzburg, Germany; Departamento de Biofisica (J.B.P.), Universidade Federal de São Paulo, São Paulo, Brazil; Max-Delbrück-Center for Molecular Medicine (M.Bader), Berlin-Buch, Germany
| |
Collapse
|
30
|
Dabek J, Kulach A, Smolka G, Wilczok T, Scieszka J, Gasior Z. Expression of genes encoding kinin receptors in peripheral blood mononuclear cells from patients with acute coronary syndromes. Intern Med J 2008; 38:892-6. [DOI: 10.1111/j.1445-5994.2008.01718.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
31
|
Xu J, Carretero OA, Shesely EG, Rhaleb NE, Yang JJ, Bader M, Yang XP. The kinin B1 receptor contributes to the cardioprotective effect of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers in mice. Exp Physiol 2008; 94:322-9. [PMID: 19060116 DOI: 10.1113/expphysiol.2008.045583] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent studies have shown that inhibition of angiotensin-converting enzyme (ACE) or angiotensin II receptors causes upregulation of the B(1) receptor (B(1)R). Here we tested the hypothesis that activation of the B(1)R partly contributes to the cardiac beneficial effect of ACE inhibitor (ACEi) and angiotensin II receptor blockers (ARB). B(1)R knockout mice (B(1)R(-/-)) and C57Bl/6J (wild-type control animals, WT) were subjected to myocardial infarction (MI) by ligating the left anterior descending coronary artery. Three weeks after MI, each strain of mice was treated with vehicle, ACEi (ramipril, 2.5 mg kg(-1) day(-1) in drinking water) or ARB (valsartan, 40 mg kg(-1) day(-1) in drinking water) for 5 weeks. We found that: (1) compared with WT mice, B(1)R(-/-) mice that underwent sham surgery had slightly but significantly increased left ventricular (LV) diastolic dimension, LV mass and myocyte size, whereas systolic blood pressure, cardiac function and collagen deposition did not differ between strains; (2) MI leads to LV hypertrophy, chamber dilatation and dysfunction similarly in both WT and B(1)R(-/-) mice; and (3) ACEi and ARB improved cardiac function and remodelling in both strains; however, these benefits were significantly diminished in B(1)R(-/-) mice. Our data suggest that kinins, acting via the B(1)R, participate in the cardioprotective effects of ACEi and ARB.
Collapse
Affiliation(s)
- Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202-2689, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Xi L, Das A, Zhao ZQ, Merino VF, Bader M, Kukreja RC. Loss of myocardial ischemic postconditioning in adenosine A1 and bradykinin B2 receptors gene knockout mice. Circulation 2008; 118:S32-7. [PMID: 18824766 DOI: 10.1161/circulationaha.107.752865] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ischemic postconditioning (PostC) is a recently described cardioprotective modality against reperfusion injury, through series of brief reflow interruptions applied at the very onset of reperfusion. It is proposed that PostC can activate a complex cellular signaling cascade, in which cell membrane receptors could serve as the upstream triggers of PostC. However, the exact subtypes of such receptors remain controversial or uninvestigated. To this context, the purpose of present study was to determine the definitive role of adenosine A(1) and bradykinin B(1) and B(2) receptors in PostC. METHODS AND RESULTS The hearts isolated from adult male C57BL/6J wild-type mice or the mice lacking adenosine A(1), or bradykinin B(1) or B(2) receptors subjected to zero-flow global ischemia and reperfusion in a Langendorff model. PostC, consisting of 6 cycles of 10 seconds of reperfusion and 10 seconds of ischemia, demonstrated significantly reduced myocardial infarct size (22.8+/-3.1%, mean+/-SEM) as compared with the non-PostC wild-type controls (35.1+/-2.8%, P<0.05). The infarct-limiting protection of PostC was absent in adenosine A(1) receptor knockout mice (34.9+/-2.7%) or bradykinin B(2) receptor knockout mice (33.3+/-1.7%) and was partially attenuated in bradykinin B(1) receptor-deficient mice (25.6+/-2.9%; P>0.05). On the other hand, PostC did not significantly alter postischemic cardiac contractile function and coronary flow. CONCLUSIONS With the use of three distinctive strains of gene knockout mice, the current study has provided the first conclusive evidence showing PostC-induced infarct-limiting cardioprotection could be triggered by activation of multiple types of cell membrane receptors, which include adenosine A(1) and bradykinin B(2) receptors.
Collapse
Affiliation(s)
- Lei Xi
- Division of Cardiology, Box 980204, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-042, Richmond, VA 23298-0204, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Westermann D, Lettau O, Sobirey M, Riad A, Bader M, Schultheiss HP, Tschöpe C. Doxorubicin cardiomyopathy-induced inflammation and apoptosis are attenuated by gene deletion of the kinin B1 receptor. Biol Chem 2008; 389:713-8. [PMID: 18627295 DOI: 10.1515/bc.2008.070] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Clinical use of the anthracycline doxorubicin (DOX) is limited by its cardiotoxic effects, which are attributed to the induction of apoptosis. To elucidate the possible role of the kinin B1 receptor (B1R) during the development of DOX cardiomyopathy, we studied B1R knockout mice (B1R(-/-)) by investigating cardiac inflammation and apoptosis after induction of DOX-induced cardiomyopathy. DOX control mice showed cardiac dysfunction measured by pressure-volume loops in vivo. This was associated with a reduced activation state of AKT, as well as an increased bax/bcl2 ratio in Western blots, indicating cardiac apoptosis. Furthermore, mRNA levels of the proinflammatory cytokine interleukin 6 were increased in the cardiac tissue. In DOX B1R(-/-) mice, cardiac dysfunction was improved compared to DOX control mice, which was associated with normalization of the bax/bcl-2 ratio and interleukin 6, as well as AKT activation state. These findings suggest that B1R is detrimental in DOX cardiomyopathy in that it mediates the inflammatory response and apoptosis. These insights might have useful implications for future studies utilizing B1R antagonists for treatment of human DOX cardiomyopathy.
Collapse
Affiliation(s)
- Dirk Westermann
- Charité-Universitätsmedizin Berlin, Department of Cardiology and Pneumology, Campus Benjamin Franklin, D-12200 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Compensatory function of bradykinin B1 receptor in the inhibitory effect of captopril on cardiomyocyte hypertrophy and cardiac fibroblast proliferation in neonatal rats. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200807010-00014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
35
|
Duka A, Kintsurashvili E, Duka I, Ona D, Hopkins TA, Bader M, Gavras I, Gavras H. Angiotensin-converting enzyme inhibition after experimental myocardial infarct: role of the kinin B1 and B2 receptors. Hypertension 2008; 51:1352-7. [PMID: 18347228 DOI: 10.1161/hypertensionaha.107.108506] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We sought to define the contribution of each of the 2 kinin receptors (bradykinin 1 receptor [B(1)R] and bradykinin 2 receptor [B(2)R]) to the cardioprotection of angiotensin-converting enzyme (ACE) inhibition after acute myocardial infarct. Wild-type mice and gene knockout mice missing either B(1)R or B(2)R were submitted to coronary ligation with or without concurrent ACE inhibition and had evaluation of left ventricular systolic capacity by assessment of fractional shortening (FS). Baseline FS was similar in all of the animals and remained unchanged in sham-operated ones. At 3 weeks after myocardial infarct, in the wild-type group there was a 27% reduction of FS (P<0.5) without ACE inhibition and 8% with ACE inhibition; in the B(1)R(-/-) groups the FS was reduced by 24% and was no different (at 28%) with ACE inhibition; in the B(2)R(-/-) groups, however, the FS was decreased by 39% and with ACE inhibition was decreased further by 52%. Analysis of bradykinin receptor gene expression in hearts showed that when one receptor was missing, the other became significantly upregulated; but the B(1)R remained highly overexpressed in the B(2)R(-/-) mice throughout, whereas the overexpressed B(2)R became significantly suppressed in the B(1)R(-/-) mice in a manner quantitatively and directionally similar to that of wild-type mice. We conclude that both bradykinin receptors contribute to the cardioprotective bradykinin-mediated effect of ACE inhibition, not only the B(2)R as believed previously; but, whereas with potentiated bradykinin in the absence of B(1)R, the upregulation of B(2)R is simply insufficient to provide full cardioprotection, in the absence of B(2)R, the upregulated B(1)R actually seems to inflict further tissue damage.
Collapse
Affiliation(s)
- Arvi Duka
- Hypertension and Atherosclerosis Section, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Westermann D, Schultheiss HP, Tschöpe C. New perspective on the tissue kallikrein–kinin system in myocardial infarction: Role of angiogenesis and cardiac regeneration. Int Immunopharmacol 2008; 8:148-54. [DOI: 10.1016/j.intimp.2007.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Revised: 07/19/2007] [Accepted: 07/23/2007] [Indexed: 11/17/2022]
|
37
|
Yao YY, Yin H, Shen B, Chao L, Chao J. Tissue kallikrein and kinin infusion rescues failing myocardium after myocardial infarction. J Card Fail 2007; 13:588-96. [PMID: 17826650 PMCID: PMC4519013 DOI: 10.1016/j.cardfail.2007.04.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 03/26/2007] [Accepted: 04/23/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Tissue kallikrein is a serine proteinase that generates the vasoactive kinin peptide, which produces vasodilatory, angiogenic, and antiapoptotic effects. In this study, we investigated the effect of a stable supply of kallikrein and kinin on ventricular remodeling and blood vessel growth in rats after myocardial infarction. METHODS AND RESULTS At 1 week after coronary artery ligation, tissue kallikrein or kinin was infused through a minipump for 4 weeks. At 5 weeks after myocardial infarction, kallikrein and kinin infusion significantly improved cardiac contractility and reduced diastolic dysfunction without affecting systolic blood pressure. Kallikrein and kinin infusion significantly increased capillary density in the noninfarcted region. Kallikrein and kinin infusion also reduced heart weight/body weight ratio, cardiomyocyte size, and atrial natriuretic peptide and brain natriuretic peptide expression in the noninfarcted area. Moreover, kallikrein and kinin infusion inhibited interstitial collagen deposition, collagen fraction volume, and collagen I and collagen III mRNA levels, transforming growth factor (TGF)-beta1 and plasminogen activator inhibitor-1 expression, and Smad2 phosphorylation. The effects of kallikrein and kinin on cardiac remodeling were associated with increased nitric oxide levels and reduced NADPH oxidase expression and activity, superoxide formation, and malondialdehyde levels. Furthermore, in cultured cardiac fibroblasts, kinin inhibited angiotensin II-stimulated TGF-beta1 production, and the effect was blocked by icatibant. CONCLUSION These results indicate that a subdepressor dose of kallikrein or kinin can restore impaired cardiac function in rats with postinfarction heart failure by inhibiting hypertrophy and fibrosis and promoting angiogenesis through increased nitric oxide formation and suppression of oxidative stress and TGF-beta1 expression.
Collapse
Affiliation(s)
- Yu-Yu Yao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
38
|
Lauton-Santos S, Guatimosim S, Castro CH, Oliveira FA, Almeida AP, Dias-Peixoto MF, Gomes MA, Pessoa P, Pesquero JL, Pesquero JB, Bader M, Cruz JS. Kinin B1 receptor participates in the control of cardiac function in mice. Life Sci 2007; 81:814-22. [PMID: 17714737 DOI: 10.1016/j.lfs.2007.06.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2007] [Revised: 06/22/2007] [Accepted: 06/29/2007] [Indexed: 10/23/2022]
Abstract
The kinins have an important role in control of the cardiovascular system. They have been associated with protective effects in the heart tissue. Kinins act through stimulation of two 7-transmembrane G protein-coupled receptors, denoted B(1) and B(2) receptors. However, the physiological relevance of B(1) receptor in the heart has not been clearly established. Using B(1) kinin receptor gene knock-out mice we tested the hypothesis that the B(1) receptor plays an important role in the control of baseline cardiac function. We examined the functional aspects of the intact heart and also in the isolated cardiomyocytes to study intracellular Ca(2+) cycling by using confocal microscopy and whole-cell voltage clamp techniques. We measured heart rate, diastolic and systolic tension, contraction and relaxation rates and, coronary perfusion pressure. Whole-cell voltage clamp was performed to measure L-type Ca(2+) current (I(Ca,L)). The hearts from B(1)(-/-) mice showed smaller systolic tension. The average values for WT and B(1)(-/-) mice were 2.6+/-0.04 g vs. 1.6+/-0.08 g, respectively. This result can be explained, at least in part, by the decrease in the Ca(2+) transient (3.1+/-0.06 vs. 3.4+/-0.09 for B(1)(-/-) and WT, respectively). There was an increase in I(Ca,L) at depolarized membrane potentials. Interestingly, the inactivation kinetics of I(Ca,L) was statistically different between the groups. The coronary perfusion pressure was higher in the hearts from B(1)(-/-) mice indicating an increase in coronary resistance. This result can be explained by the significant reduction of eNOS (NOS-3) expression in the aorta of B(1)(-/-) mice. Collectively, our results demonstrate that B(1) receptor exerts a fundamental role in the mammalian cardiac function.
Collapse
Affiliation(s)
- Sandra Lauton-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yin H, Chao J, Bader M, Chao L. Differential role of kinin B1 and B2 receptors in ischemia-induced apoptosis and ventricular remodeling. Peptides 2007; 28:1383-9. [PMID: 17644219 PMCID: PMC2067250 DOI: 10.1016/j.peptides.2007.05.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 05/15/2007] [Accepted: 05/16/2007] [Indexed: 11/17/2022]
Abstract
We investigated the role of kinin receptors in cardiac remodeling after ischemia/reperfusion (I/R). Bradykinin injection improved cardiac contractility, diastolic function, reduced infarct size and prevented left ventricular thinning after I/R, whereas des-Arg(9)-BK injection had no protective effects. Bradykinin, but not des-Arg(9)-BK, reduced cardiomyocyte apoptosis and increased Akt and GSK-3beta phosphorylation. Furthermore, myocardial infarct size was similar between wild type and B2 knockout mice after I/R, but significantly reduced in kinin B1 receptor knockout mice. These results indicate that the kinin B2 receptor, but not the B1 receptor, protects against I/R-induced cardiac dysfunction by inhibiting apoptosis and limiting ventricular remodeling.
Collapse
Affiliation(s)
- Hang Yin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425-2211, USA
| | | | | | | |
Collapse
|
40
|
Kakoki M, McGarrah RW, Kim HS, Smithies O. Bradykinin B1 and B2 receptors both have protective roles in renal ischemia/reperfusion injury. Proc Natl Acad Sci U S A 2007; 104:7576-81. [PMID: 17452647 PMCID: PMC1855073 DOI: 10.1073/pnas.0701617104] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To explore the role of the kallikrein-kinin system in relation to ischemia/reperfusion injury in the kidney, we generated mice lacking both the bradykinin B1 and B2 receptor genes (B1RB2R-null, Bdkrb1-/-/Bdkrb2-/-) by deleting the genomic region encoding the two receptors. In 4-month-old mice, blood pressures were not significantly different among B1RB2R-null, B2R-null (Bdkrb2-/-), and WT mice. After 30 min of bilateral renal artery occlusion and 24 h of reperfusion, mortality rates, renal histological and functional changes, 8-hydroxy-2'-deoxyguanosine levels in total DNA, mtDNA deletions, and the number of TUNEL-positive cells in the kidneys increased progressively in the following order (from lowest to highest): WT, B2R-null, and B1RB2R-null mice. Increases in mRNA levels of TGF-beta1, connective tissue growth factor, and endothelin-1 after ischemia/reperfusion injury were also exaggerated in the same order (from lowest to highest): WT, B2R-null, and B1RB2R-null. Thus, both the B1 and B2 bradykinin receptors play an important role in reducing DNA damage, apoptosis, morphological and functional kidney changes, and mortality during renal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Masao Kakoki
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
- *To whom correspondence should be addressed. E-mail: or
| | - Robert W. McGarrah
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
| | - Hyung-Suk Kim
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
| | - Oliver Smithies
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525
- *To whom correspondence should be addressed. E-mail: or
| |
Collapse
|
41
|
Wang PHM, Cenedeze MA, Pesquero JB, Pacheco-Silva A, Câmara NOS. Influence of bradykinin B1 and B2 receptors in the immune response triggered by renal ischemia-reperfusion injury. Int Immunopharmacol 2006; 6:1960-5. [PMID: 17161349 DOI: 10.1016/j.intimp.2006.07.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Accepted: 07/20/2006] [Indexed: 10/24/2022]
Abstract
Bradykinin B1 receptors are exclusively expressed in inflamed tissues. For this reason, they have been related with the outcomes of several pathologies. Ischemia-reperfusion injury is caused by the activation of inflammatory and cytoprotective genes, such as macrophage chemoattractant protein-1 and heme oxygenase-1, respectively. This study was aimed to analyze the involvement of bradykinin B1 and B2 receptors (B1R and B2R) in tissue response after renal ischemia-reperfusion injury. For that, B1R (B1-/-), B2R (B2-/-) knockout animals and its control (wild-type mice, B1B2+/+) were subjected to renal bilateral ischemia, followed by 24, 48 and 120 h of reperfusion. At these time points, blood serum samples were collected for creatinine and urea dosages. Kidneys were harvested for histology and molecular analyses by real-time PCR. At 24 and 48 h of reperfusion, B1-/- group resulted in the lowest serum creatinine and urea levels, indicating less renal damage, which was proved by renal histology. Renal protection associated with B1-/- mice was also related with higher expression of HO-1 and lower expression of MCP-1. In conclusion, the absence of B1R had a protective role against inflammatory responses developed after renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Pamella Huey Mei Wang
- Laboratório de Imunologia Clínica e Experimental. Division of Nephrology. Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
42
|
Freise H, Palmes D, Spiegel HU. Inhibition of Angiotensin-Converting Enzyme Reduces Rat Liver Reperfusion Injury Via Bradykinin-2-Receptor. J Surg Res 2006; 134:231-7. [PMID: 16513137 DOI: 10.1016/j.jss.2005.10.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/16/2005] [Accepted: 10/24/2005] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bradykinin is both a potent vasodilatator and a central inflammatory mediator. Similar to findings in myocardial reperfusion injury, bradykinin might mediate the protective effects of angiotensin-converting enzyme (ACE) inhibition after liver ischemia via increased bradykinin-2-receptor (B-2) stimulation. On the other hand, B-2-inhibition has been shown to reduce liver reperfusion injury. This study was designed to investigate the role of Bradykinin in hepatic reperfusion injury. MATERIALS AND METHODS Twenty eight rats were allocated randomly to Sham procedure (Sham), 30-min normothermic ischemia (ischemia), ischemia with Ramiprilat (ACE-I), or ischemia with Ramiprilat and B-2-inhibitor HOE 140 (ACE-I+B-2-I). Liver microcirculation and leukocyte adherence were investigated using intravital microscopy 30 min after reperfusion (n = 7 per group). In addition, serum activities of AST and ALT were measured for 7 days (n = 28). RESULTS Ischemia was associated with a loss of perfused sinusoids, sinusoidal vasoconstriction, and a reduction in microvascular blood flow. Permanent leukocyte adherence increased both in sinusoids and in postsinusoidal venoles. ACE-I restored sinusoidal perfusion, normalized vasoregulation, maintained sinusoidal blood flow, and inhibited leukocyte adhesion. ACE-I+B-2-I abolished the protective effects linked to ACE-I. Ischemia-induced liver cell injury after 5 h of reperfusion was ameliorated by ACE-I. In the ACE-I+B-2-I group, reduction in liver cell injury was reversed. CONCLUSION After hepatic ischemia, ACE-I reduced reperfusion injury in a B-2-dependent manner. These results suggest a pivotal role for bradykinin in the treatment of reperfusion injury by Ramiprilat, mediating sinusoidal dilation and blunting hepatic inflammation.
Collapse
Affiliation(s)
- Hendrik Freise
- Department of General Surgery, Surgical Research, University Hospital Muenster, Muenster, Germany.
| | | | | |
Collapse
|
43
|
Fortin JP, Gera L, Bouthillier J, Stewart JM, Adam A, Marceau F. Endogenous aminopeptidase N decreases the potency of peptide agonists and antagonists of the kinin B1 receptors in the rabbit aorta. J Pharmacol Exp Ther 2005; 314:1169-76. [PMID: 15919764 DOI: 10.1124/jpet.105.088799] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The B(1) receptor for kinins is selectively stimulated by bradykinin-related fragments lacking the C-terminal arginine, des-arginine(9)-bradykinin (des-Arg(9)-BK), and Lys-des-Arg(9)-BK. The latter peptide is the optimal agonist at the human and rabbit receptor. The B(1) receptor is inducible as a function of inflammatory conditions in the vasculature. We studied the effect of endogenously expressed peptidases on the potency of ligands of this receptor in an established bioassay, the rabbit aorta contractility. The potency measured for agonists (EC(50)) or antagonists (pA(2) scale) in this assay was compared with the affinity of each agent determined using [(3)H]Lys-des-Arg(9)-BK binding competition in cultured aortic smooth muscle cells and with the competition K(i) for the hydrolysis of the aminopeptidase chromogenic substrate L-Ala-p-nitroanilide by smooth muscle cell membranes. The contractile potency of the agonist Lys-des-Arg(9)-BK is decreased by in situ metabolism, and aminopeptidase N mediates most of the distortion (inhibited by amastatin but not efficiently by puromycin). At the other end of the spectrum, the fully protected agonist Sar-[D-Phe(8)]des-Arg(9)-BK is not significantly potentiated by peptidase inhibitors. A similar distortion of apparent potency was observed for some peptide antagonists used in the contractility assay, B-10350 (Lys-Lys-[Hyp(3), Igl(5), d-Tic(7), CpG(8)]des-Arg(9)-BK) and Lys-[Leu(8)]des-Arg(9)-BK being intensely potentiated by amastatin treatment and effective L-Ala-p-nitroanilide competitors. N-Protected peptide antagonists or a nonpeptide antagonist of the B(1) receptor were not potentiated by amastatin. The coexpression of aminopeptidase N and the kinin B(1) receptor in rabbit arterial tissue is of interest for the inactivation of the high-affinity agonist Lys-des-Arg(9)-BK and for the design of hydrosoluble antagonist drugs.
Collapse
Affiliation(s)
- Jean-Philippe Fortin
- Centre de Recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Der Sarkissian S, Huentelman MJ, Stewart J, Katovich MJ, Raizada MK. ACE2: A novel therapeutic target for cardiovascular diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2005; 91:163-98. [PMID: 16009403 DOI: 10.1016/j.pbiomolbio.2005.05.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypertension afflicts over 65 million Americans and poses an increased risk for cardiovascular morbidity such as stroke, myocardial infarction and end-stage renal disease resulting in significant mortality. Overactivity of the renin-angiotensin system (RAS) has been identified as an important determinant that is implicated in the etiology of these diseases and therefore represents a major target for therapy. In spite of the successes of drugs inhibiting various elements of the RAS, the incidence of hypertension and cardiovascular diseases remain steadily on the rise. This has lead many investigators to seek novel and innovative approaches, taking advantage of new pathways and technologies, for the control and possibly the cure of hypertension and related pathologies. The main objective of this review is to forward the concept that gene therapy and the genetic targeting of the RAS is the future avenue for the successful control and treatment of hypertension and cardiovascular diseases. We will present argument that genetic targeting of angiotensin-converting enzyme 2 (ACE2), a newly discovered member of the RAS, is ideally poised for this purpose. This will be accomplished by discussion of the following: (i) summary of our current understanding of the RAS with a focus on the systemic versus tissue counterparts as they relate to hypertension and other cardiovascular pathologies; (ii) the newly discovered ACE2 enzyme with its physiological and pathophysiological implications; (iii) summary of the current antihypertensive pharmacotherapy and its limitations; (iv) the discovery and design of ACE inhibitors; (v) the emerging concepts for ACE2 drug design; (vi) the current status of genetic targeting of the RAS; (vii) the potential of ACE2 as a therapeutic target for hypertension and cardiovascular disease treatment; and (viii) future perspectives for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Shant Der Sarkissian
- Department of Physiology and Functional Genomics, College of Medicine, and the McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
45
|
Souza DG, Teixeira MM. The balance between the production of tumor necrosis factor-alpha and interleukin-10 determines tissue injury and lethality during intestinal ischemia and reperfusion. Mem Inst Oswaldo Cruz 2005; 100 Suppl 1:59-66. [PMID: 15962100 DOI: 10.1590/s0074-02762005000900011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A major goal in the treatment of acute ischemia of a vascular territory is to restore blood flow to normal values, i.e. to "reperfuse" the ischemic vascular bed. However, reperfusion of ischemic tissues is associated with local and systemic leukocyte activation and trafficking, endothelial barrier dysfunction in postcapillary venules, enhanced production of inflammatory mediators and great lethality. This phenomenon has been referred to as "reperfusion injury" and several studies demonstrated that injury is dependent on neutrophil recruitment. Furthermore, ischemia and reperfusion injury is associated with the coordinated activation of a series of cytokines and adhesion molecules. Among the mediators of the inflammatory cascade released, TNF-alpha appears to play an essential role for the reperfusion-associated injury. On the other hand, the release of IL-10 modulates pro-inflammatory cytokine production and reperfusion-associated tissue injury. IL-1beta, PAF and bradykinin are mediators involved in ischemia and reperfusion injury by regulating the balance between TNF-alpha and IL-10 production. Strategies that enhance IL-10 and/or prevent TNF-alpha concentration may be useful as therapeutic adjuvants in the treatment of the tissue injury that follows ischemia and reperfusion.
Collapse
Affiliation(s)
- Danielle G Souza
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brasil
| | | |
Collapse
|
46
|
Leeb-Lundberg LMF, Marceau F, Müller-Esterl W, Pettibone DJ, Zuraw BL. International union of pharmacology. XLV. Classification of the kinin receptor family: from molecular mechanisms to pathophysiological consequences. Pharmacol Rev 2005; 57:27-77. [PMID: 15734727 DOI: 10.1124/pr.57.1.2] [Citation(s) in RCA: 742] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Kinins are proinflammatory peptides that mediate numerous vascular and pain responses to tissue injury. Two pharmacologically distinct kinin receptor subtypes have been identified and characterized for these peptides, which are named B1 and B2 and belong to the rhodopsin family of G protein-coupled receptors. The B2 receptor mediates the action of bradykinin (BK) and lysyl-bradykinin (Lys-BK), the first set of bioactive kinins formed in response to injury from kininogen precursors through the actions of plasma and tissue kallikreins, whereas the B(1) receptor mediates the action of des-Arg9-BK and Lys-des-Arg9-BK, the second set of bioactive kinins formed through the actions of carboxypeptidases on BK and Lys-BK, respectively. The B2 receptor is ubiquitous and constitutively expressed, whereas the B1 receptor is expressed at a very low level in healthy tissues but induced following injury by various proinflammatory cytokines such as interleukin-1beta. Both receptors act through G alpha(q) to stimulate phospholipase C beta followed by phosphoinositide hydrolysis and intracellular free Ca2+ mobilization and through G alpha(i) to inhibit adenylate cyclase and stimulate the mitogen-activated protein kinase pathways. The use of mice lacking each receptor gene and various specific peptidic and nonpeptidic antagonists have implicated both B1 and B2 receptors as potential therapeutic targets in several pathophysiological events related to inflammation such as pain, sepsis, allergic asthma, rhinitis, and edema, as well as diabetes and cancer. This review is a comprehensive presentation of our current understanding of these receptors in terms of molecular and cell biology, physiology, pharmacology, and involvement in human disease and drug development.
Collapse
Affiliation(s)
- L M Fredrik Leeb-Lundberg
- Division of Cellular and Molecular Pharmacology, Department of Experimental Medical Science, Lund University, BMC, A12, SE-22184 Lund, Sweden.
| | | | | | | | | |
Collapse
|
47
|
Xu J, Carretero OA, Sun Y, Shesely EG, Rhaleb NE, Liu YH, Liao TD, Yang JJ, Bader M, Yang XP. Role of the B1 kinin receptor in the regulation of cardiac function and remodeling after myocardial infarction. Hypertension 2005; 45:747-53. [PMID: 15699461 PMCID: PMC4593484 DOI: 10.1161/01.hyp.0000153322.04859.81] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kinins exert cardioprotective effects via 2 G-protein-coupled receptors, B1 and B2. Using B1 kinin receptor gene knockout mice (B1-/-), we tested the hypotheses that the B1 receptor plays an important role in preservation of cardiac function, whereas lack of B1 may accelerate cardiac remodeling and dysfunction after myocardial infarction, and that B2 receptors may compensate for lack of B1, whereas blockade of B2 receptors in B1-/- mice may cause further deterioration of cardiac function and remodeling. Female B1-/- mice and wild-type controls (C57BL/6J, B1+/+) underwent sham surgery or myocardial infarction and were treated with either vehicle or B2-antagonist (icatibant, 500 microg/kg per day, subcutaneous) for 8 weeks. We found that in sham myocardial infarction, B1-/- mice had a larger left ventricular diastolic chamber dimension both initially and at 4 to 8 weeks compared with B1+/+. Left ventricular mass and myocyte size were also larger in B1-/- with sham operation than in B1+/+, although cardiac function did not differ between strains. After myocardial infarction, cardiac remodeling and function were similar in both strains, although B1-/- mice tended to have lower blood pressure. Blockade of B2 receptors tended to worsen cardiac remodeling and dysfunction in B1-/- but not in B1+/+. These results may suggest that B2 receptors play an important role in compensating for lack of B1 receptors in mice with myocardial infarction. Dual blockade of both B1 and B2 eliminates this compensation, leading to further deterioration of cardiac dysfunction and remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Jiang Xu
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Mich 48202-2689, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Calixto JB, Medeiros R, Fernandes ES, Ferreira J, Cabrini DA, Campos MM. Kinin B1 receptors: key G-protein-coupled receptors and their role in inflammatory and painful processes. Br J Pharmacol 2004; 143:803-18. [PMID: 15520046 PMCID: PMC1575942 DOI: 10.1038/sj.bjp.0706012] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2004] [Revised: 08/03/2004] [Accepted: 09/10/2004] [Indexed: 01/25/2023] Open
Abstract
Kinins are a family of peptides implicated in several pathophysiological events. Most of their effects are likely mediated by the activation of two G-protein-coupled receptors: B(1) and B(2). Whereas B(2) receptors are constitutive entities, B(1) receptors behave as key inducible molecules that may be upregulated under some special circumstances. In this context, several recent reports have investigated the importance of B(1) receptor activation in certain disease models. Furthermore, research on B(1) receptors in the last years has been mainly focused in determining the mechanisms and pathways involved in the process of induction. This was essentially favoured by the advances obtained in molecular biology studies, as well as in the design of selective and stable peptide and nonpeptide kinin B(1) receptor antagonists. Likewise, development of kinin B(1) receptor knockout mice greatly helped to extend the evidence about the relevance of B(1) receptors during pathological states. In the present review, we attempted to remark the main advances achieved in the last 5 years about the participation of kinin B(1) receptors in painful and inflammatory disorders. We have also aimed to point out some groups of chronic diseases, such as diabetes, arthritis, cancer or neuropathic pain, in which the strategic development of nonpeptidic oral-available and selective B(1) receptor antagonists could have a potential relevant therapeutic interest.
Collapse
Affiliation(s)
- João B Calixto
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88049-900 Florianópolis, SC, Brazil.
| | | | | | | | | | | |
Collapse
|
49
|
Souza DG, Lomez ESL, Pinho V, Pesquero JB, Bader M, Pesquero JL, Teixeira MM. Role of bradykinin B2 and B1 receptors in the local, remote, and systemic inflammatory responses that follow intestinal ischemia and reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2004; 172:2542-8. [PMID: 14764727 DOI: 10.4049/jimmunol.172.4.2542] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The administration of bradykinin may attenuate ischemia and reperfusion (I/R) injury by acting on B(2)Rs. Blockade of B(2)R has also been shown to ameliorate lesions associated with I/R injury. In an attempt to explain these contradictory results, the objective of the present work was to investigate the role of and interaction between B(1) and B(2) receptors in a model of intestinal I/R injury in mice. The bradykinin B(2)R antagonist (HOE 140) inhibited reperfusion-induced inflammatory tissue injury and delayed lethality. After I/R, there was an increase in the expression of B(1)R mRNA that was prevented by HOE 140. In mice that were deficient in B(1)Rs (B(1)R(-/-) mice), inflammatory tissue injury was abrogated, and lethality was delayed and partially prevented. Pretreatment with HOE 140 reversed the protective anti-inflammatory and antilethality effects provided by the B(1)R(-/-) phenotype. Thus, B(2)Rs are a major driving force for B(1)R activation and consequent induction of inflammatory injury and lethality. In contrast, activation of B(2)Rs may prevent exacerbated tissue injury and lethality, an effect unmasked in B(1)R(-/-) mice and likely dependent on the vasodilatory actions of B(2)Rs. Blockade of B(1)Rs could be a more effective strategy than B(2) or B(1)/B(2) receptor blockade for the treatment of the inflammatory injuries that follow I/R.
Collapse
Affiliation(s)
- Danielle G Souza
- Departamentos de. Bioquímica e Imunologia, e Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
50
|
Souza DG, Pinho V, Pesquero JL, Lomez ES, Poole S, Juliano L, Correa A, de A Castro MS, Teixeira MM. Role of the bradykinin B2 receptor for the local and systemic inflammatory response that follows severe reperfusion injury. Br J Pharmacol 2003; 139:129-39. [PMID: 12746231 PMCID: PMC1573815 DOI: 10.1038/sj.bjp.0705200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. Bradykinin (BK) appears to play an important role in the development and maintenance of inflammation. Here, we assessed the role of the BK B(2) receptor for the injuries that occur after ischemia and reperfusion (I/R) of the territory irrigated by the superior mesenteric artery. 2. Tissue (lung and duodenum) kallikrein activity increased after ischemia with greater enhancement after reperfusion. A selective inhibitor of tissue kallikrein, Phenylacetyl-Phe-Ser-Arg-N-(2,3-dinitrophenyl)-ethylenediamine (TKI, 0.001-10 mg ml(-1)), inhibited kallikrein activity in a concentration-dependent manner in vitro. In vivo, pretreatment with TKI (30 mg kg(-1)) prevented the extravasation of plasma and the recruitment of neutrophils. 3. Similarly, the bradykinin B(2) receptor antagonists, HOE 140 (0.01-1.0 mg kg(-1)) or FR173657 (10.0 mg kg(-1)), inhibited reperfusion-induced increases in vascular permeability and the recruitment of neutrophils in the intestine and lungs. 4. In a model of more severe I/R injury, HOE 140 (1.0 mg kg(-1)) inhibited the increase in vascular permeability, neutrophil recruitment, haemorrhage and tissue pathology. Furthermore, HOE 140 significantly inhibited the elevations of TNF-alpha in tissue and serum and partially prevented lethality. This was associated with an increase in the concentrations of IL-10 in tissue and serum. 5. Thus, our results demonstrate that, following intestinal I/R injury, there is an increase in tissue kallikrein activity and activation of BK B(2) receptors. B(2) receptor activation is essential for the development of inflammatory tissue injury and lethality. These results contrast with those of others showing that BK mostly exerts a protective role during I/R injury.
Collapse
Affiliation(s)
- Danielle G Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jorge L Pesquero
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Eliane S Lomez
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Steve Poole
- Division of Endocrinology, National Institute for Biological Standards and Control, South Mimms, Potters Bar, EN6 3QG, U.K
| | - Luiz Juliano
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ary Correa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - M Salete de A Castro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Author for correspondence:
| |
Collapse
|