1
|
Flores-Monroy J, Lezama-Martínez D, Fonseca-Coronado S, Martínez-Aguilar L. Differences in the expression of the renin angiotensin system and the kallikrein-kinin system during the course of myocardial infarction in male and female Wistar rats. J Renin Angiotensin Aldosterone Syst 2021; 21:1470320319900038. [PMID: 32458737 PMCID: PMC7268575 DOI: 10.1177/1470320319900038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: There is some evidence that components of the renin-angiotensin system and
kallikrein-kinin system are not similarly regulated in both sexes. The aim
of this work was to analyze the expression of angiotensin-converting enzyme,
angiotensin-converting enzyme 2, angiotensin 1 receptor, angiotensin 2
receptor, beta-1 receptor, and beta-2 receptor during the evolution of
myocardial infarction. Methods: Thirty-six male and 36 female Wistar rats were used. Myocardial infarction
was induced. Six groups of both sexes were formed, (n=6):
(a) sham; (b) 48 h myocardial infarction; (c) one week myocardial
infarction; (d) two weeks myocardial infarction; (e) three weeks myocardial
infarction and (f) four weeks myocardial infarction. The expression was
evaluated by real-time polymerase chain reaction on the penumbra of left
ventricle. Results: The mRNA expression of most biomarkers was lower in females than in males.
During acute infarction, an increase of all protein expression was found in
female and at two weeks while in the male only biomarker changes occurred at
three weeks. In addition, in male biomarkers mRNA expression decreased
during chronic infarction while in females it did not. Conclusions: The renin-angiotensin system and kallikrein-kinin system biomarkers
expression occurs at earlier times in the female than in the male rat. In
addition, during chronic myocardial infarction these biomarkers remained
unchanged in females while in males they decreased.
Collapse
Affiliation(s)
- Jazmín Flores-Monroy
- Laboratorio de Farmacologia del Miocardio, Universidad Nacional Autonoma de Mexico, Mexico
| | - Diego Lezama-Martínez
- Laboratorio de Farmacologia del Miocardio, Universidad Nacional Autonoma de Mexico, Mexico
| | - Salvador Fonseca-Coronado
- Laboratorio de Inmunobiología de Enfermedades Infecciosas, Universidad Nacional Autonoma de Mexico, Mexico
| | - Luisa Martínez-Aguilar
- Laboratorio de Farmacologia del Miocardio, Universidad Nacional Autonoma de Mexico, Mexico
| |
Collapse
|
2
|
Visniauskas B, Perry JC, Gomes GN, Nogueira-Pedro A, Paredes-Gamero EJ, Tufik S, Chagas JR. Intermittent hypoxia changes the interaction of the kinin-VEGF system and impairs myocardial angiogenesis in the hypertrophic heart. Physiol Rep 2021; 9:e14863. [PMID: 33991464 PMCID: PMC8123545 DOI: 10.14814/phy2.14863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Intermittent hypoxia (IH) is a feature of obstructive sleep apnea (OSA), a condition highly associated with hypertension-related cardiovascular diseases. Repeated episodes of IH contribute to imbalance of angiogenic growth factors in the hypertrophic heart, which is key in the progression of cardiovascular complications. In particular, the interaction between vascular endothelial growth factor (VEGF) and the kallikrein-kinin system (KKS) is essential for promoting angiogenesis. However, researchers have yet to investigate experimental models of IH that reproduce OSA, myocardial angiogenesis, and expression of KKS components. We examined temporal changes in cardiac angiogenesis in a mouse IH model. Adult male C57BI/6 J mice were implanted with Matrigel plugs and subjected to IH for 1-5 weeks with subsequent weekly histological evaluation of vascularization. Expression of VEGF and KKS components was also evaluated. After 3 weeks, in vivo myocardial angiogenesis and capillary density were decreased, accompanied by a late increase of VEGF and its type 2 receptor. Furthermore, IH increased left ventricular myocardium expression of the B2 bradykinin receptor, while reducing mRNA levels of B1 receptor. These results suggest that in IH, an unexpected response of the VEGF and KKS systems could explain the reduced capillary density and impaired angiogenesis in the hypoxic heart, with potential implications in hypertrophic heart malfunction.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juliana C Perry
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Guiomar N Gomes
- Departmento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jair R Chagas
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Hamid S, Rhaleb IA, Kassem KM, Rhaleb NE. Role of Kinins in Hypertension and Heart Failure. Pharmaceuticals (Basel) 2020; 13:E347. [PMID: 33126450 PMCID: PMC7692223 DOI: 10.3390/ph13110347] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
The kallikrein-kinin system (KKS) is proposed to act as a counter regulatory system against the vasopressor hormonal systems such as the renin-angiotensin system (RAS), aldosterone, and catecholamines. Evidence exists that supports the idea that the KKS is not only critical to blood pressure but may also oppose target organ damage. Kinins are generated from kininogens by tissue and plasma kallikreins. The putative role of kinins in the pathogenesis of hypertension is discussed based on human mutation cases on the KKS or rats with spontaneous mutation in the kininogen gene sequence and mouse models in which the gene expressing only one of the components of the KKS has been deleted or over-expressed. Some of the effects of kinins are mediated via activation of the B2 and/or B1 receptor and downstream signaling such as eicosanoids, nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF) and/or tissue plasminogen activator (T-PA). The role of kinins in blood pressure regulation at normal or under hypertension conditions remains debatable due to contradictory reports from various laboratories. Nevertheless, published reports are consistent on the protective and mediating roles of kinins against ischemia and cardiac preconditioning; reports also demonstrate the roles of kinins in the cardiovascular protective effects of the angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor blockers (ARBs).
Collapse
Affiliation(s)
- Suhail Hamid
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Imane A. Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Kamal M. Kassem
- Division of Cardiology, Department of Internal Medicine, University of Louisville Medical Center, Louisville, KY 40202, USA;
| | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
4
|
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
|
5
|
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. [DOI: 10.1124/jpet.113.203927] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
6
|
Inhibition of CatA: an emerging strategy for the treatment of heart failure. Future Med Chem 2013; 5:399-409. [DOI: 10.4155/fmc.13.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The lysosomal serine carboxypeptidase CatA has a very important and well-known structural function as well as a, so far, less explored catalytic function. A complete loss of the CatA protein results in the lysosomal storage disease galactosialidosis caused by intralysosomal degradation of β-galactosidase and neuraminidase 1. However, mice with a catalytically inactive CatA enzyme show no signs of this disease. This observation establishes a clear distinction between structural and catalytic functions of the CatA enzyme. Recently, several classes of orally bioavailable synthetic inhibitors of CatA have been identified. Pharmacological studies in rodents indicate a remarkable influence of CatA inhibition on cardiovascular disease progression and identify CatA as a promising novel target for the treatment of heart failure.
Collapse
|
7
|
Heart-rate changes in asphyxic preconditioning in rats depend on light-dark cycle. Open Med (Wars) 2011. [DOI: 10.2478/s11536-011-0021-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractGenerally, it is assumed that heart-rhythm disorders during hypoxia result from the interplay between the autonomic nervous system (ANS) and the direct effect of hypoxia on cardiorespiratory structures of the central nervous system and on the myocardium. Circadian variability in the ANS may substantially influence the electrical stability of the myocardium, and thus it is associated with the preconditioning protective mechanism. We designed our study using anaesthetized Wistar rats (ketamine/xylazine 100 mg/15 mg/kg, i.m., open chest experiments) to evaluate the effect of preconditioning (PC) induced by 1 to 3 cycles (1 PC–3 PC) of asphyxia (5 min. of artificial hypoventilation, VT = 0.5 ml/100 g of b.w., 20 breaths/min.) and reoxygenation (5 min. of artificial ventilation, VT = 1 ml/100 g of b.w., 50 breaths/min.) on the heart rate (HR) during followed exposure 20 minutes of hypoventilation after adaptation to a light-dark (LD) cycle of 12 hours:12 hours. Hypoxic HR increases were only minimally prevented by 1 to 2 PC pre-treatment, particularly during the dark part of the day. A statistically significant HR increase required 3 PC and was seen only in the light part of the day. We concluded that possible ANS participation in asphyxic preconditioning depends not only on the number of preconditioned cycles but also on the LD cycle, when the ANS participation in preconditioning can be effective only in the light (nonactive) period.
Collapse
|
8
|
Merino VF, Todiras M, Mori MA, Sales VMT, Fonseca RG, Saul V, Tenner K, Bader M, Pesquero JB. Predisposition to atherosclerosis and aortic aneurysms in mice deficient in kinin B1 receptor and apolipoprotein E. J Mol Med (Berl) 2009; 87:953-63. [PMID: 19618151 DOI: 10.1007/s00109-009-0501-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 05/20/2009] [Accepted: 06/29/2009] [Indexed: 01/11/2023]
Abstract
Kinin B1 receptor is involved in chronic inflammation and expressed in human atherosclerotic lesions. However, its significance for lesion development is unknown. Therefore, we investigated the effect of kinin B1 receptor deletion on the development of atherosclerosis and aortic aneurysms in apolipoprotein E-deficient (ApoE(-/-)) mice. Mice deficient both in ApoE and in kinin B1 receptor (ApoE(-/-)-B(1)(-/-)) were generated and analyzed for their susceptibility to atherosclerosis and aneurysm development under cholesterol rich-diet (western diet) and angiotensin II infusion. Kinin B1 receptor messenger RNA (mRNA) expression was significantly increased in ApoE(-/-) mice after Western-type diet. Although no difference in serum cholesterol was found between ApoE(-/-)-B(1)(-/-) and ApoE(-/-) mice under Western-type diet, aortic lesion incidence was significantly higher in ApoE(-/-)-B(1)(-/-) after this treatment. In accordance, we observed increased endothelial dysfunction in these mice. The mRNA expression of cyclic guanosine monophosphate-dependent protein kinase I, CD-11, F4/80, macrophage colony-stimulating factor, and tumor necrosis factor-alpha were increased in the aorta of double-deficient mice following Western-type diet, whereas the levels of peroxisome proliferator-activated receptor gamma protein and the activity of matrix metalloproteinase-9 activity were decreased. In addition to the increased atherosclerotic lesions, the lack of kinin B(1) receptor also increased the incidence of abdominal aortic aneurysms after angiotensin II infusion. In conclusion, our results show that kinin B(1) receptor deficiency aggravates atherosclerosis and aortic aneurysms under cholesterolemic conditions, supporting an antiatherogenic role for the kinin B(1) receptor.
Collapse
Affiliation(s)
- Vanessa F Merino
- Department of Biophysics, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
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: 1.2] [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
|
10
|
Schriefer JA, Craig Hartman J. Section Review Cardiovascular & Renal: Bradykinin degradation: potential pathways for cardiovascular therapeutic use. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.5.11.1465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Participation of kallikrein-kinin system in different pathologies. Int Immunopharmacol 2007; 8:135-42. [PMID: 18182216 DOI: 10.1016/j.intimp.2007.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2007] [Revised: 08/01/2007] [Accepted: 08/02/2007] [Indexed: 02/07/2023]
Abstract
The general description of kinins refers to these peptides as molecules involved in vascular tone regulation and inflammation. Nevertheless, in the last years a series of evidences has shown that local hormonal systems, such as the kallikrein-kinin system, may be differently regulated and are of pivotal importance to pathophysiological control. The combined interpretations of many recent studies allow us to conclude that the kallikrein-kinin system plays broader and richer roles than those classically described until recently. In this review, we report findings concerning the participation of the kallikrein-kinin system in inflammation, cancer, and in pathologies related to cardiovascular, renal and central nervous systems.
Collapse
|
12
|
Fernandes L, Ceravolo GS, Fortes ZB, Tostes R, Santos RAS, Santos JA, Mori MAS, Pesquero JB, de Carvalho MHC. Modulation of kinin B1 receptor expression by endogenous angiotensin II in hypertensive rats. ACTA ACUST UNITED AC 2006; 136:92-7. [PMID: 16822558 DOI: 10.1016/j.regpep.2006.04.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Revised: 04/19/2006] [Accepted: 04/28/2006] [Indexed: 11/22/2022]
Abstract
We investigated the expression and localization of B1 receptor in tissues of rats submitted to a renin-dependent model of hypertension (2K-1C), and analyzed the influence of endogenous Ang II in modulating the in vivo expression of these receptors. B1 mRNA levels in the heart, kidney and thoracic aorta were quantified by real time PCR, B1 receptor protein expression was assessed by immunohistochemistry, plasma Ang II levels were analyzed by radioimmunoassay and the effects of AT1 receptor blockade were determined after losartan treatment. 2K-1C rats presented a marked increase in Ang II levels when compared to sham-operated rats. In parallel, cardiac- (but not renal and aortic) B1 mRNA levels were 15-fold higher in 2K-1C than in sham rats. In 2K-1C, B1 expression was detected in the endothelium of small cardiac arteries and in cardiomyocytes. Losartan completely reverted the increased B1 mRNA levels and significantly decreased the protein expression observed in 2K-1C rats, despite reducing, but not normalizing blood pressure. We conclude that in the 2K-1C rat, induction of cardiac B1 receptor might be tightly linked to AT1 receptor activation. These data suggest the existence of a new site of interaction between kinins and angiotensins, and might provide important contributions for a better understanding of the pathophysiology of hypertension.
Collapse
Affiliation(s)
- Liliam Fernandes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, SP 05508-900, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Couture R, Girolami JP. Putative roles of kinin receptors in the therapeutic effects of angiotensin 1-converting enzyme inhibitors in diabetes mellitus. Eur J Pharmacol 2005; 500:467-85. [PMID: 15464053 DOI: 10.1016/j.ejphar.2004.07.045] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2004] [Indexed: 02/07/2023]
Abstract
The role of endogenous kinins and their receptors in diabetes mellitus is being confirmed with the recent developments of molecular and genetic animal models. Compelling evidence suggests that the kinin B(2) receptor is organ-protective and partakes to the therapeutic effects of angiotensin 1-converting enzyme inhibitors (ACEI) and angiotensin AT(1) receptor antagonists. Benefits derive primarily from vasodilatory, antihypertensive, antiproliferative, antihypertrophic, antifibrotic, antithrombotic and antioxidant properties of kinin B(2) receptor activation. Mechanisms include the formation of nitric oxide and prostacyclin and the inhibition of NAD(P)H oxidase activity involving classical and novel signalling pathways. Kinin B(2) receptor also ameliorates insulin resistance by increasing glucose uptake and supply, and by inducing glucose transporter-4 translocation either directly or through phosphorylation of insulin receptor. The kinin B(1) receptor, which is induced by the cytokine network, growth factors and hyperglycaemia, mediates hyperalgesia, vascular hyperpermeability and leukocytes infiltration in diabetic animals. However, emerging data highlight reno- and cardio-protective effects mediated by kinin B(1) receptor under chronic ACEI therapy in diabetes mellitus. Thus, the Janus-faced of kinin receptors needs to be taken into account in future drug development. For instance, locally acting kinin B(1)/B(2) receptor agonists if used in a safe therapeutic window may represent a more rationale strategy in the prevention and management of diabetic complications. Because kinin B(2) receptor antagonists may further increase insulin resistance, the persisting dogma that restricts the development of kinin receptor analogues to antagonists (that is still relevant to abrogate pain and inflammation) needs to be revisited.
Collapse
Affiliation(s)
- Réjean Couture
- Département de Physiologie, Faculté de Médecine, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, Québec, Canada H3C 3J7.
| | | |
Collapse
|
14
|
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: 729] [Impact Index Per Article: 38.4] [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
|
15
|
McLean PG, Perretti M, Ahluwalia A. Kinin B1receptors as novel anti-inflammatory targets. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.4.2.127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Svorc P, Benacka R, Bracoková I, Kujaník S. Chronophysiological view on changes of the electrical stability of the heart and heart rate under pentobarbital sodium and ketamine/xylazine anesthesia in a hypoventilation-reoxygenation rat model. Exp Clin Cardiol 2004; 9:119-124. [PMID: 19641698 PMCID: PMC2716265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
OBJECTIVES To determine the effect of the light-dark cycle (LD) and type of anesthetic agent used on changes in the electrical stability of the heart in disorders of pulmonary ventilation. METHODS The ventricular arrhythmia threshold (VAT) and heart rate (HR) were measured in anesthetized female Wistar rats adapted to an LD regimen of 12 h:12 h (pentobarbital sodium 30 mg/kg, intraperitoneally, or ketamine/xylazine 100 mg/15 mg/kg, intramuscularly; open chest experiments) under artificial normoventilation (tidal volume 1 mL/100 g, respiratory rate 40 breaths/min) and during 20 min of hypoventilation (tidal volume 0.5 mL/100 g, respiratory rate 20 breaths/min) followed by 20 min of reoxygenation. Control and experimental groups were set according to LD cycle adaptation and type of anesthesia used: light control pentobarbital sodium group, n=67; dark control pentobarbital sodium group n=50; light hypoventilation-reoxygenation (hypo-reoxy) pentobarbital sodium group, n=26; dark hypo-reoxy pentobarbital sodium group, n=16; light control ketamine/xylazine group, n=90; dark control ketamine/xylazine group, n=57; light hypo-reoxy ketamine/xylazine group, n=13; and dark hypo-reoxy ketamine/xylayine group, n=18. RESULTS In the control groups, the LD cycle only had an effect on animals under pentobarbital sodium anesthesia (P<0.001). During hypoventilation, the VAT was significantly lower in animals under both types of anesthesia and during the light period of the LD cycle than in the control groups. During the light part of the LD cycle, the period of reoxygenation increased VAT values (P<0.001). However, during the dark period of the LD cycle the VAT values decreased. Heart rate was significantly lower in the ketamine/xylazine groups than in the pentobarbital sodium groups during the light and dark periods of the LD cycle, respectively (P<0.001). CONCLUSIONS The mechanisms mediating the effects of pentobarbital sodium anesthesia on myocardial vulnerability likely depend on the LD cycle, in contrast with the mechanisms that mediate the effects of ketamine/xylazine anesthesia, in female Wistar rats.
Collapse
Affiliation(s)
| | - Roman Benacka
- Pathophysiology, Medical Faculty, Sararik University, Kosice, Slovak Republic
| | | | - Stefan Kujaník
- Pathophysiology, Medical Faculty, Sararik University, Kosice, Slovak Republic
| |
Collapse
|
17
|
Lagneux C, Adam A, Lamontagne D. A study of the mediators involved in the protection induced by exogenous kinins in the isolated rat heart. Int Immunopharmacol 2003; 3:1511-8. [PMID: 12946448 DOI: 10.1016/s1567-5769(03)00177-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The aim of this study was to determine whether endothelium-derived mediators and the endocannabinoid system were involved in the cardioprotective effects induced by exogenous kinins, namely bradykinin and its active metabolites, des-Arg(9)-bradykinin. Isolated rat hearts were submitted to a 20-min stabilisation period, followed by 90 min of low-flow ischemia (flow rate, 0.6 ml min(-1)) before a 60-min reperfusion period. Perfusion of bradykinin (BK, 30 nM) or des-Arg(9)-bradykinin (DBK, 30 nM) was initiated 1 min before the ischemia and maintained during the entire ischemic period. Perfusion with BK reduced infarct size, when measured at the end of the 60-min reperfusion. This effect was blocked by the B2-receptor antagonist, HOE140 (30 nM). Likewise, DBK reduced infarct size, effect that was blocked by the B1-receptor antagonist (30 nM Lys(0)-Leu(8)-DBK). The cardioprotective effect of both BK and DBK was abolished by the cannabinoid CB1-receptor antagonist (1 microM SR141716A), but not by the CB2-receptor antagonist (1 microM SR144528). Neither the NO synthase inhibitor, N-nitro-L-arginine (NNLA, 30 microM), the COX inhibitor, indomethacin (2.8 microM), nor the CYP450 inhibitor, clotrimazole (1 microM), prevented the cardioprotective effect of the kinins. However, a combined treatment with those three inhibitors abolished completely the ability of BK and DBK to reduce infarct size. In conclusion, exogenously administered BK and DBK exert a protective effect against ischemia in an isolated heart model. Endothelium-derived mediators such as nitric oxide, prostanoids, and endothelium-derived hyperpolarizing factor, as well as an SR141716A-sensitive mediator, appear to be involved in this beneficial effect.
Collapse
Affiliation(s)
- Caroline Lagneux
- Faculté de pharmacie, Université de Montréal, CP 6128, succursale Centre-ville, Montréal, QC, Canada H3C3J7
| | | | | |
Collapse
|
18
|
Sangsree S, Brovkovych V, Minshall RD, Skidgel RA. Kininase I-type carboxypeptidases enhance nitric oxide production in endothelial cells by generating bradykinin B1 receptor agonists. Am J Physiol Heart Circ Physiol 2003; 284:H1959-68. [PMID: 12623793 DOI: 10.1152/ajpheart.00036.2003] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Kininase I-type carboxypeptidases convert native kinin agonists for B(2) receptors into B(1) receptor agonists by specifically removing the COOH-terminal Arg residue. The membrane localization of carboxypeptidase M (CPM) and carboxypeptidase D (CPD) make them ideally situated to regulate kinin activity. Nitric oxide (NO) release from human lung microvascular endothelial cells (HLMVEC) was measured directly in real time with a porphyrinic microsensor. Bradykinin (1-100 nM) elicited a transient (5 min) peak of generation of NO that was blocked by the B(2) antagonist HOE 140, whereas B(1) agonist des-Arg(10)-kallidin caused a small linear increase in NO over 20 min. Treatment of HLMVEC with 5 ng/ml interleukin-1beta and 200 U/ml interferon-gamma for 16 h upregulated B(1) receptors as shown by an approximately fourfold increase in prolonged (>20 min) output of NO in response to des-Arg(10)-kallidin, which was blocked by the B(1) antagonist des-Arg(10)-Leu(9)-kallidin. B(2) receptor agonists bradykinin or kallidin also generated prolonged NO production in treated HLMVEC, which was significantly reduced by either a B(1) antagonist or carboxypeptidase inhibitor, and completely abolished with a combination of B(1) and B(2) receptor antagonists. Furthermore, CPM and CPD activities were increased about twofold in membrane fractions of HLMVEC treated with interleukin-1beta and interferon-gamma compared with control cells. Immunostaining localized CPD primarily in a perinuclear/Golgi region, whereas CPM was on the cell membrane. These data show that cellular kininase I-type carboxypeptidases can enhance kinin signaling and NO production by converting B(2) agonists to B(1) agonists, especially in inflammatory conditions.
Collapse
Affiliation(s)
- Sakonwun Sangsree
- Department of Pharmacology, University of Illinois College of Medicine, Chicago 60612, USA
| | | | | | | |
Collapse
|
19
|
Ignjatovic T, Tan F, Brovkovych V, Skidgel RA, Erdös EG. Activation of bradykinin B1 receptor by ACE inhibitors. Int Immunopharmacol 2002; 2:1787-93. [PMID: 12489793 DOI: 10.1016/s1567-5769(02)00146-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ACE or kininase II inhibitors are very important, widely used therapeutic agents for the treatment of a variety of diseases. Although they inhibit ACE, thus, angiotensin II release and bradykinin (BK) inactivation, this inhibition alone does not suffice to explain their successful application in medical practice. Enalaprilat and other ACE inhibitors at nanomolar concentrations activate the BK B1 receptor directly in the absence of ACE and the peptide ligands, des-Arg-kinins. The inhibitors activate at the Zn-binding pentameric consensus sequence HEXXH (195 -199) of B1, a motif also present in the active centers of ACE but absent from the BK B2 receptor. ACE inhibitors, when activating the B1 receptor, elevate intracellular calcium [Ca2+]i and release NO from cultured cells. Activation by ACE inhibitor was abolished by Ca-EDTA, a B1 receptor antagonist, by a synthetic undecapeptide representing the 192-202 sequence in the B1 receptor, and by site-directed mutagenesis of H195 to A. With the exception of the B1 receptor blocker, these agents and the mutation did not affect the actions of the peptide ligand des-Arg10-Lys1-BK. Ischemia and inflammatory cytokines induce B1 receptors and elevate its expression. Direct activation of the B1 receptor by ACE inhibitors can contribute to their therapeutic efficacy, for example, by releasing NO in vascular beds, or to some of their side effects.
Collapse
Affiliation(s)
- Tatjana Ignjatovic
- Department of Pharmacology (M/C 868), University of Illinois at Chicago College of Medicine, 835 S Wolcott Avenue, Chicago, IL 60612-7344, USA
| | | | | | | | | |
Collapse
|
20
|
Ignjatovic T, Tan F, Brovkovych V, Skidgel RA, Erdös EG. Novel mode of action of angiotensin I converting enzyme inhibitors: direct activation of bradykinin B1 receptor. J Biol Chem 2002; 277:16847-52. [PMID: 11880373 DOI: 10.1074/jbc.m200355200] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Angiotensin I converting enzyme (kininase II; ACE) inhibitors are important therapeutic agents widely used for treatment in cardiovascular and renal diseases. They inhibit angiotensin II release and bradykinin inactivation; these actions do not explain completely the clinical benefits. We found that enalaprilat and other ACE inhibitors in nanomolar concentrations activate human bradykinin B(1) receptors directly in the absence of ACE and the B(1) agonist des-Arg(10)-Lys(1)-bradykinin. These inhibitors activate at the Zn(2+)-binding consensus sequence HEXXH (195-199) in B(1), which is present also in ACE but not in the B(2) receptor. Activation elevates [Ca(2+)](i) and releases NO from endothelial or transfected cells expressing the B(1) receptor but is blocked by Ca-EDTA, a B(1) receptor antagonist, the synthetic undecapeptide sequence (192-202) of B(1), and the mutagenesis of His(195) to Ala(195). Except for the B(1) antagonist, these agents and manipulations did not block activation by a peptide ligand. Thus, Zn(2+) is essential for B(1) receptor activation by ACE inhibitors at the zinc-binding consensus sequence. Ischemia or cytokines induce abundant B(1) receptor expression. B(1) receptor activation by ACE inhibitors, a novel mode of action reported here first, can contribute to their therapeutic effects by releasing NO in the heart and to some side effects.
Collapse
Affiliation(s)
- Tatjana Ignjatovic
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
21
|
Lagneux C, Bader M, Pesquero JB, Demenge P, Ribuot C. Detrimental implication of B1 receptors in myocardial ischemia: evidence from pharmacological blockade and gene knockout mice. Int Immunopharmacol 2002; 2:815-22. [PMID: 12095172 DOI: 10.1016/s1567-5769(02)00022-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate the contribution of kinin B1 receptors in myocardial ischemia using both pharmacological blockade and gene knockout mice. MATERIAL AND METHODS Hearts (n = 6-8 per group) from wild type or homozygous B1 receptor gene knockout mice were isolated and perfused using the Langendorff technique. After a 30-min stabilisation period, the left coronary artery was occluded for 30 min followed by 60 min of reperfusion. In two separate groups of wild type hearts, B1 and B2 receptors were blocked with 3 nM of (des-Arg9, Leu8)-bradykinin and 10 nM of Hoe 140, respectively, (started 15 min before ischemia and stopped before the reperfusion). RESULTS Infarct size to risk zone (I/R) ratio was significantly reduced in hearts of knockout mice (11.3 +/- 2.1%) compared to those of wild type mice (25.7 +/- 1.7%). Furthermore, in wild type mice, I/R was significantly reduced in hearts perfused with the B1 receptor antagonist (12.8 +/- 2.4%) but not in hearts perfused with the B2 receptor antagonist (36.3 +/- 4.4%) compared to untreated hearts. Finally, a RT-PCR technique showed an activation of kinin B1 receptor gene transcription, in wild type hearts, subjected to the ischemia-reperfusion sequence. CONCLUSION This study demonstrates that B1 receptors are induced during myocardial ischemia where they could play a detrimental role in mice.
Collapse
Affiliation(s)
- Caroline Lagneux
- Laboratoire du Stress Cardiovasculaire et Pathologies Associées, Université Joseph Fourier, La Tronche, France.
| | | | | | | | | |
Collapse
|
22
|
Baxter GF, Ebrahim Z. Role of bradykinin in preconditioning and protection of the ischaemic myocardium. Br J Pharmacol 2002; 135:843-54. [PMID: 11861312 PMCID: PMC1573212 DOI: 10.1038/sj.bjp.0704548] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- G F Baxter
- The Hatter Institute, University College London, London.
| | | |
Collapse
|
23
|
Babai L, Papp JG, Parratt JR, Végh Á. The antiarrhythmic effects of ischaemic preconditioning in anaesthetized dogs are prevented by atropine; role of changes in baroreceptor reflex sensitivity. Br J Pharmacol 2002; 135:55-64. [PMID: 11786480 PMCID: PMC1573109 DOI: 10.1038/sj.bjp.0704445] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Dogs, anaesthetized with chloralose and urethane, were subjected to a 25 min occlusion of the left anterior descending coronary artery. This resulted in ventricular ectopic activity, a reduction in baroreflex sensitivity (BRS, measured following the intravenous administration of phenylephrine), elevations in the epicardial ST-segment and increases in the degree of inhomogeneity of electrical activation, both measured from the ischaemic region of the left ventricular wall. 2. These changes were markedly reduced when the 25 min occlusion was preceded, 20 min earlier, by a 5 min (preconditioning) occlusion of the same coronary artery (e.g. VF during ischaemia reduced from 40% in the controls to 0%; P<0.05; BRS increased from 1.22+/-0.23 pre-occlusion to 1.61+/-0.25 mmHg ms(-1) post-occlusion in preconditioned dogs; cf. 1.28+/-0.29 to 0.45+/-0.12 mmHg ms(-1) respectively in the controls, P<0.05). 3. These beneficial effects of preconditioning were prevented by the administration, 10 min prior to the 25 min coronary artery occlusion, of atropine (1 mg kg(-1) i.v. followed by a continuous infusion of 0.04 mg kg(-1) h(-1)). For example, VF during occlusion was increased from 0% in the preconditioned dogs to 40% (P<0.05) in the presence of atropine and BRS was again reduced during occlusion (from 1.75+/-0.29 to 0.30+/-0.08 mmHg ms(-1); P<0.05). 4. We conclude that preconditioning reduces arrhythmia severity during ischaemia by favourably modifying cardiac autonomic receptor mechanism through enhancing vagal influences.
Collapse
Affiliation(s)
- László Babai
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Albert Szent-Györgyi Faculty of Medicine, Dóm tér 12, P.O. Box 427, H-6701 Hungary
| | - Julius Gy Papp
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Albert Szent-Györgyi Faculty of Medicine, Dóm tér 12, P.O. Box 427, H-6701 Hungary
- Research Unit for Cardiovascular Pharmacology, Hungarian Academy of Sciences, University of Szeged, Albert Szent-Györgyi Faculty of Medicine, Dóm tér 12, P.O. Box 427, H-6701 Hungary
| | - James R Parratt
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Albert Szent-Györgyi Faculty of Medicine, Dóm tér 12, P.O. Box 427, H-6701 Hungary
- Department of Physiology and Pharmacology, Strathclyde Institute for Biomedical Sciences, 27 Taylor Street, Glasgow G4 0NR
| | - Ágnes Végh
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Albert Szent-Györgyi Faculty of Medicine, Dóm tér 12, P.O. Box 427, H-6701 Hungary
- Author for correspondence:
| |
Collapse
|
24
|
Mazenot C, Loufrani L, Henrion D, Ribuot C, Muller-Esterl W, Godin-Ribuot D. Endothelial kinin B(1)-receptors are induced by myocardial ischaemia-reperfusion in the rabbit. J Physiol 2001; 530:69-78. [PMID: 11136859 PMCID: PMC2278396 DOI: 10.1111/j.1469-7793.2001.0069m.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Kinin B1-receptors are induced by various inflammatory stimuli. Since myocardial ischaemia-reperfusion results in inflammation, we questioned whether it could induce B1-receptor-dependent responses to des-Arg9-bradykinin (DBK). Thirty-six rabbits were submitted either to a 30 min coronary occlusion followed by a 3 h reperfusion or to a sham operation. The response to DBK was then tested in vivo on mean arterial pressure (MAP) and in vitro on isolated hearts and arterial rings. DBK induced a dose-dependent decrease in MAP in the ischaemia-reperfusion group (DBK, 10 microg kg(-1), intra-arterial: -12 +/- 2 vs. -5 +/- 2 mm Hg in the sham group, P < 0.02), which was significantly antagonised by [Leu8]-des-Arg9-bradykinin (LBK), a B1-receptor antagonist. Following ischaemia-reperfusion, isolated hearts responded to DBK by a decrease in coronary perfusion pressure greater than that of the sham group. DBK dose-dependently decreased the isometric force of isolated carotid rings (DBK, 10(-5) M: -9 +/- 2 vs. -1 +/- 2% in the sham group, P < 0.02) and mesenteric arteries (DBK, 10-6 M: -38 +/- 7% vs. -3 +/- 2 % in the sham group, P < 0.001). The vascular effects of DBK seen after ischaemia-reperfusion were significantly antagonised by LBK. The presence of B1-receptors in ischaemia-reperfusion animals was confirmed by immunolocalisation and Western blot analysis. This study demonstrates that myocardial ischaemia-reperfusion induces a global induction of functional kinin B1-receptors in the endothelium.
Collapse
Affiliation(s)
- C Mazenot
- LSCPA, UFR de Pharmacie, Universite Grenoble I, France
| | | | | | | | | | | |
Collapse
|
25
|
Cyr M, Eastlund T, Blais C, Rouleau JL, Adam A. Bradykinin metabolism and hypotensive transfusion reactions. Transfusion 2001; 41:136-50. [PMID: 11161259 DOI: 10.1046/j.1537-2995.2001.41010136.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- M Cyr
- Faculty of Pharmacy, University of Montréal, Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
26
|
Kawai H, Stevens SY, Liang CS. Renin-angiotensin system inhibition on noradrenergic nerve terminal function in pacing-induced heart failure. Am J Physiol Heart Circ Physiol 2000; 279:H3012-9. [PMID: 11087259 DOI: 10.1152/ajpheart.2000.279.6.h3012] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic angiotensin-converting enzyme (ACE) inhibition has been shown to improve cardiac sympathetic nerve terminal function in heart failure. To determine whether similar effects could be produced by angiotensin II AT(1) receptor blockade, we administered the ACE inhibitor quinapril, angiotensin II AT(1) receptor blocker losartan, or both agents together, to rabbits with pacing-induced heart failure. Chronic rapid pacing produced left ventricular dilation and decline of fractional shortening, increased plasma norepinephrine (NE), and caused reductions of myocardial NE uptake activity, NE histofluorescence profile, and tyrosine hydroxylase immunostained profile. Administration of quinapril or losartan retarded the progression of left ventricular dysfunction and attenuated cardiac sympathetic nerve terminal abnormalities in heart failure. Quinapril and losartan together produced greater effects than either agent alone. The effect of renin-angiotensin system inhibition on improvement of left ventricular function and remodeling, however, was not sustained. Our results suggest that the effects of ACE inhibitors are mediated via the reduction of angiotensin II and that angiotensin II plays a pivotal role in modulating cardiac sympathetic nerve terminal function during development of heart failure. The combined effect of ACE inhibition and angiotensin II AT(1) receptor blockade on cardiac sympathetic nerve terminal dysfunction may contribute to the beneficial effects on cardiac function in heart failure.
Collapse
Affiliation(s)
- H Kawai
- Cardiology Unit, Department of Medicine, and Department of Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | |
Collapse
|
27
|
Mazenot C, Gobeil F, Ribuot C, Regoli D, Godin-Ribuot D. Delayed myocardial protection induced by endotoxin does not involve kinin B(1)-receptors. Br J Pharmacol 2000; 131:740-4. [PMID: 11030723 PMCID: PMC1572378 DOI: 10.1038/sj.bjp.0703619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Endotoxin is known to confer a delayed protection against myocardial infarction. Lipopolysaccharide (LPS) treatment also induces the de novo synthesis of kinin B(1)-receptors that are not present in normal conditions. The aim of this study was to evaluate whether LPS-induced B(1)-receptors are implicated in the reduction of infarct size brought about by LPS. Rabbits were submitted to a 30-min coronary artery occlusion and 3-h reperfusion sequence. Six groups were studied: pretreated or not (control animals) with LPS (5 microgram kg(-1) i.v.) 24 h earlier and treated 15 min before and throughout ischaemia - reperfusion with either the B(1)-antagonist R-715 (1 mg kg(-1) h(-1)), the B(1)-agonist Sar-[D-Phe(8)]-des-Arg(9)-bradykinin (15 microgram kg(-1) h(-1)) or vehicle (saline). Infarct size and area at risk were assessed by differential staining and planimetric analysis. The presence of B(1)-receptors in LPS-pretreated animals was confirmed by a decrease in mean arterial pressure in response to B(1) stimulation. LPS-pretreatment significantly reduced infarct size (6.4+/-1.7%, of area at risk vs 24.1+/-2.5% in control animals, P<0.05). This protection was not modified by B(1)-receptor antagonism (7.4+/-2.2%, NS) or stimulation (5.2+/-1.2%, NS). Neither antagonist nor agonist modified infarct size in control animals. In conclusion, these data suggest that LPS-induced myocardial protection in the rabbit is not related to concomitant de novo B(1)-receptor induction.
Collapse
Affiliation(s)
- C Mazenot
- Laboratoire Stress Cardiovasculaires et Pathologies Associées, Université Grenoble I, France
| | - F Gobeil
- Département de Pharmacologie, Université de Sherbrooke, Québec, Canada
| | - C Ribuot
- Laboratoire Stress Cardiovasculaires et Pathologies Associées, Université Grenoble I, France
| | - D Regoli
- Département de Pharmacologie, Université de Sherbrooke, Québec, Canada
- Author for correspondence:
| | - D Godin-Ribuot
- Laboratoire Stress Cardiovasculaires et Pathologies Associées, Université Grenoble I, France
- Author for correspondence:
| |
Collapse
|
28
|
Tschöpe C, Heringer-Walther S, Walther T. Regulation of the kinin receptors after induction of myocardial infarction: a mini-review. Braz J Med Biol Res 2000; 33:701-8. [PMID: 10829098 DOI: 10.1590/s0100-879x2000000600011] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is well known that the responses to vasoactive kinin peptides are mediated through the activation of two receptors termed bradykinin receptor B1 (B1R) and B2 (B2R). The physiologically prominent B2R subtype has certainly been the subject of more intensive efforts in structure-function studies and physiological investigations. However, the B1R activated by a class of kinin metabolites has emerged as an important subject of investigation within the study of the kallikrein-kinin system (KKS). Its inducible character under stress and tissue injury is therefore a field of major interest. Although the KKS has been associated with cardiovascular regulation since its discovery at the beginning of the last century, less is known about the B1R and B2R regulation in cardiovascular diseases like hypertension, myocardial infarction (MI) and their complications. This mini-review will summarize our findings on B1R and B2R regulation after induction of MI using a rat model. We will develop the hypothesis that differences in the expression of these receptors may be associated with a dual pathway of the KKS in the complex mechanisms of myocardial remodeling.
Collapse
Affiliation(s)
- C Tschöpe
- Department of Cardiology and Pneumology, University Hospital Benjamin Franklin, Free University of Berlin, Germany
| | | | | |
Collapse
|
29
|
Kuoppala A, Lindstedt KA, Saarinen J, Kovanen PT, Kokkonen JO. Inactivation of bradykinin by angiotensin-converting enzyme and by carboxypeptidase N in human plasma. Am J Physiol Heart Circ Physiol 2000; 278:H1069-74. [PMID: 10749699 DOI: 10.1152/ajpheart.2000.278.4.h1069] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because bradykinin (BK) appears to have cardioprotective effects ranging from improved hemodynamics to antiproliferative effects, inhibition of BK-degrading enzymes should potentiate such actions. The purpose of this study was to find out which enzymes are responsible for the degradation of BK in human plasma. Human plasma from healthy donors (n = 10) was incubated with BK in the presence or absence of specific enzyme inhibitors. At high (micromolar) concentrations, BK was mostly (>90%) degraded by carboxypeptidase N (CPN)-like activity. In contrast, at low (nanomolar) substrate concentrations, at which the velocity of the catalytic reaction is equivalent to that under physiological conditions, BK was mostly (>90%) converted into an inactive metabolite, BK-(1-7), by angiotensin-converting enzyme (ACE). BK-(1-7) was further converted by ACE into BK-(1-5), with accumulation of this active peptide. A minor fraction (<10%) of the BK was converted into another active metabolite, BK-(1-8), by CPN-like activity. The present study shows that the most critical step in plasma kinin metabolism, i.e., inactivation of BK, is mediated by ACE. Thus inhibition of plasma ACE activity would be cardioprotective by elevating the concentration of BK in the circulation.
Collapse
Affiliation(s)
- A Kuoppala
- Wihuri Research Institute, FIN-00140 Helsinki, Helsinki University Hospital, FIN-00290 Helsinki, Finland
| | | | | | | | | |
Collapse
|
30
|
Tschöpe C, Koch M, Spillmann F, Heringer-Walther S, Mochmann HC, Stauss H, Bader M, Unger T, Schultheiss HP, Walther T. Upregulation of the cardiac bradykinin B2 receptors after myocardial infarction. IMMUNOPHARMACOLOGY 1999; 44:111-7. [PMID: 10604533 DOI: 10.1016/s0162-3109(99)00116-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
An increase in myocardial bradykinin (BK) might be a mechanism to protect the heart during acute myocardial infarction (MI). To characterize the regulation of the myocardial B2 receptor during MI, we studied the expression of this BK receptor in the right ventricle (RV), left ventricle (LV) and myocardial septum (S) 24 h after left coronary ligation. Experiments were performed in male Wistar Kyoto rats (n = 10) and compared with sham operated animals (n = 6). After total RNA extraction, the myocardial B2-receptor expression was analyzed by a RNase protection assay (n = 6), using a specific probe from the coding region of the receptor gene. After 24 h, rats with MI were normotensive and showed an impaired left ventricular function. The B2-receptor expression of the LV of these rats was significantly elevated (2.3-fold) compared to sham operated rats. Furthermore, we found a dramatic upregulation of the B2 receptor in the RV (7.8-fold) and a dramatic expression of B2 receptor mRNA in S of infarcted hearts, whereas in the S of sham operated rats no B2 receptor expression could be detected. Our data show clearly that the described increase in BK during myocardial ischemia is accompanied by an elevated B2-receptor expression in the infarcted and non-infarcted parts of cardiac ventricles.
Collapse
Affiliation(s)
- C Tschöpe
- Department of Cardiology and Pneumology, University Hospital Benjamin Franklin, Free University of Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Erşahin C, Euler DE, Simmons WH. Cardioprotective effects of the aminopeptidase P inhibitor apstatin: studies on ischemia/reperfusion injury in the isolated rat heart. J Cardiovasc Pharmacol 1999; 34:604-11. [PMID: 10511138 DOI: 10.1097/00005344-199910000-00019] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Aminopeptidase P and angiotensin-converting enzyme (ACE) are responsible for the metabolism of exogenously administered bradykinin in the coronary circulation of the rat. It has been shown that ACE inhibitors decrease cytosolic enzyme release from the ischemic rat heart and reduce reperfusion-induced ventricular arrhythmias by increasing endogenous levels of bradykinin. It was hypothesized that the aminopeptidase P inhibitor apstatin could do the same. In an isolated perfused rat heart preparation subjected to global ischemia and reperfusion, both apstatin and ramiprilat (an ACE inhibitor) significantly decreased creatine kinase (CK) and lactate dehydrogenase (LDH) release. The difference between the postischemia and preischemia levels of released CK was reduced 68% by apstatin and 68% by ramiprilat compared with control. The corresponding reductions in LDH release were 74% for apstatin and 81% for ramiprilat. A combination of the inhibitors was not significantly better than either one alone. Apstatin and ramiprilat also significantly reduced the duration of reperfusion-induced ventricular fibrillation by 69 and 61%, respectively. The antiarrhythmic effect of apstatin was reversed by HOE140, a bradykinin B2-receptor antagonist, suggesting that apstatin is acting by potentiating endogenously formed bradykinin. The results demonstrate that the aminopeptidase P inhibitor apstatin is cardioprotective in this model of cardiac ischemia/ reperfusion injury.
Collapse
Affiliation(s)
- C Erşahin
- Department of Molecular and Cellular Biochemistry, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois 60153, USA
| | | | | |
Collapse
|
32
|
Sato M, Das DK, Engelman RM. Interaction of bradykinin with angiotensin, prostacyclin, and nitric oxide in myocardial preservation. Ann N Y Acad Sci 1999; 874:286-94. [PMID: 10415540 DOI: 10.1111/j.1749-6632.1999.tb09244.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This review focuses on the importance of bradykinin in myocardial preservation during ischemic arrest. Bradykinin is released from the heart spontaneously in response to ischemic stress, which may be viewed as a survival signal of the heart against ischemia. Bradykinin appears to function as a signaling molecule by controlling the release of other intracellular modulators, such as prostacyclins and nitric oxide, which also exert beneficial effects on the ischemic myocardium.
Collapse
Affiliation(s)
- M Sato
- Department of Surgery, University of Connecticut School of Medicine, Farmington 06030, USA
| | | | | |
Collapse
|
33
|
Dendorfer A, Wolfrum S, Dominiak P. Pharmacology and cardiovascular implications of the kinin-kallikrein system. JAPANESE JOURNAL OF PHARMACOLOGY 1999; 79:403-26. [PMID: 10361880 DOI: 10.1254/jjp.79.403] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Kinins are peptide hormones that can exert a significant influence on the regulation of blood pressure and vascular tone due to their vasodilatatory, natriuretic and growth modulating activity. Their cardiovascular involvement in physiological and pathophysiological situations has been studied intensively since inhibitors for angiotensin I-converting enzyme and selective receptor antagonists have become available for pharmacologically potentiating or inhibiting kinin-mediated reactions. Molecular biological analysis and the establishment of genetically modified animal models have also allowed newer information to be acquired on this subject. In this review, the components and cardiovascularly relevant mechanisms of the kinin-kallikrein system shall be described. Organ-specific effects concerning the kidneys, the vascular system, the heart and nervous tissue shall also be illustrated. On this issue, the physiological functions and pathophysiological implications of the kinin-kallikrein system should be clearly distinguished from the many, mostly endothelium-mediated protective effects which occur during ACE inhibition due to the potentiation of kinin effects. Finally, a view shall also be cast upon newly discovered targets of action, which could be exploited for therapeutically altering the kinin-kallikrein system.
Collapse
Affiliation(s)
- A Dendorfer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical University Lübeck, Germany
| | | | | |
Collapse
|
34
|
Joyeux M, Godin-Ribuot D, Yellon DM, Demenge P, Ribuot C. Heat stress response and myocardial protection. Fundam Clin Pharmacol 1999; 13:1-10. [PMID: 10027082 DOI: 10.1111/j.1472-8206.1999.tb00314.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prior whole-body hyperthermia is able to protect the myocardium against ischaemia-reperfusion injury by reducing cellular necrosis, preserving the ventricular function and preventing the occurrence of arrhythmias. These cardioprotective effects are associated with reduction of oxidative stress, preservation of the high-energy phosphate levels and synthesis of heat stress proteins. A better understanding of this powerful protective adaptation of the myocytes would be of interest for potential clinical application, and rational design of specific agents that activate this mechanism will hopefully follow soon.
Collapse
Affiliation(s)
- M Joyeux
- Laboratoire de Pharmacologie Cardiovasculaire Expérimentale-Biomolécules, Faculté de Pharmacie, Université Joseph Fourier, La Tronche, France
| | | | | | | | | |
Collapse
|
35
|
Mazenot C, Ribuot C, Demenge P, Godin-Ribuot D. Monophosphoryl lipid A, a derivative of bacterial lipopolysaccharide, fails to induce B1-receptor-dependent responses to (des-Arg9)-bradykinin in the rabbit in vivo. IMMUNOPHARMACOLOGY 1999; 41:165-8. [PMID: 10102798 DOI: 10.1016/s0162-3109(98)00065-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate whether monophosphoryl lipid A (MLA) was able to induce a hypotensive response to (des-Arg9)-bradykinin in the rabbit in vivo, by inducing B1-receptor synthesis. MATERIALS AND METHODS Arterial pressure was measured after intra-arterial administration of B1- and B2-receptor agonists and antagonists in control rabbits and in rabbits pre-treated 24 h earlier with MLA (100 microg kg(-1) i.v.) or lipopolysaccharide (LPS) (10 microg kg(-1) i.v.). RESULTS Intra-arterial bradykinin administration induced a similar dose-dependent hypotension in all groups (BK 0.25 microg kg(-1), 36 +/- 3 mm Hg, BK 1 microg kg(-1), -39 +/- 3 mm Hg, p < 0.05 vs. control conditions) that was significantly antagonised by intra-arterial HOE 140 (2 microg kg(-1)) (-5 +/- 2 mm Hg, p < 0.05). Intra-arterial (des-Arg9)-bradykinin induced a hypotensive response in the LPS-pre-treated group (DBK 1 microg kg(-1), -6 +/- 1 mm Hg, DBK 10 microg kg(-1), -10 +/- 1 mm Hg, p < 0.05 vs. control conditions) that was totally abolished by intra-arterial (des-Arg9, Leu8)-bradykinin (10 microg kg(-1) min(-1)) (+1 +/- 2 mm Hg, p < 0.05). In the control and MLA-pre-treated groups, (des-Arg9)-bradykinin had no effect. CONCLUSION MLA pre-treatment did not induce a hypotensive response to (des-Arg9)-bradykinin. We conclude that, in contrast to LPS, MLA does not induce B1-receptor synthesis, 24 h after its administration in the rabbit. Thus, the cardioprotective effects of MLA do not appear to be related to the kinin pathway.
Collapse
Affiliation(s)
- C Mazenot
- PCEBM, UFR de Pharmacie, Université Grenoble I, La Tronche, France
| | | | | | | |
Collapse
|
36
|
Li Q, Zhang J, Loro JF, Pfaffendorf M, van Zwieten PA. Bradykinin B2-receptor-mediated positive chronotropic effect of bradykinin in isolated rat atria. J Cardiovasc Pharmacol 1998; 32:452-6. [PMID: 9733359 DOI: 10.1097/00005344-199809000-00016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The positive chronotropic effect of bradykinin was investigated in isolated spontaneously beating atria of the rat. Cumulative additions of bradykinin (0.3-100 nM) caused a concentration-dependent increase in the beating rate of the atria by maximally 35+/-4 beats/min, approximately 25% of the 1 microM isoprenaline-induced maximal responses. In contrast, the active metabolite of bradykinin and selective bradykinin B1-receptor agonist, Des-Arg9-bradykinin, did not influence the spontaneous frequency of beating. Propranolol (1 microM) combined with prazosin (1 microM) did not affect the positive chronotropic effect of bradykinin. A selective bradykinin B2-receptor antagonist, Hoe 140, concentration-dependently shifted the response curves for bradykinin to the right, whereas the bradykinin B1-receptor antagonist, Lys-[Leu8]Des-Arg9-bradykinin had no effect. The tachycardic responses to bradykinin were potentiated by ramipril, an angiotensin-converting enzyme/kininase II inhibitor, but not affected by Nomega-nitro-L-arginine methyl ester hydrochloride, a nitric oxide synthesis inhibitor. Indomethacin and meclofenamate, two cyclooxygenase inhibitors, abolished the bradykinin-induced chronotropic effect. These results indicate that exogenous bradykinin induces a positive chronotropic effect that occurs independent of adrenoceptors. The bradykinin-induced chronotropic effect is mediated by bradykinin B2 receptors, whereas B1 receptors do not play a role in mediating this effect. Prostaglandins but not nitric oxide appear to be involved in bradykinin-induced positive chronotropic effect.
Collapse
Affiliation(s)
- Q Li
- Department of Pharmacotherapy, University of Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Loro JF, Zhang J, Pfaffendorf M, van Zwieten PA. Positive chronotropic activity of bradykinin in the pithed normotensive rat. Fundam Clin Pharmacol 1998; 12:77-81. [PMID: 9523188 DOI: 10.1111/j.1472-8206.1998.tb00927.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The positive chronotropic effect of bradykinin was investigated in the pithed rat preparation. Cumulative treatment with bradykinin (0.20 nmol/kg-6.59 mumol/kg, intravenous [i.v.]) caused a dose-dependent increase in heart rate (HR) by a maximum of 80 +/- 3.3 beats min-1. In contrast, the active metabolite of bradykinin and selective bradykinin B1-receptor agonist, [des-Arg9]-bradykinin did not influence the spontaneous frequency of beating. Propranolol alone reduced the bradykinin-induced increase in HR and a combination of propranolol with prazosin abolished the chronotropic effect of bradykinin. The selective bradykinin B2 receptor antagonist. Hoe 140, dose-dependently shifted the dose-response curves of bradykinin to the right, whereas the bradykinin B1 receptor antagonist, des-Arg10-[Leu9]-kallidin proved ineffective. From our experiments it may be concluded that bradykinin induces tachycardia in the pithed rat primarily by stimulating the sympathetic ganglia leading to the release of noradrenaline, which subsequently activates cardiac beta 1-adrenoceptors. The bradykinin-induced chronotropic effect is mediated by bradykinin B2-receptors, whereas B1-receptors appear not to be involved.
Collapse
Affiliation(s)
- J F Loro
- Departamento de Farmacología, Universidad de Las Palmas, Las Palmas de Gran Canaria, Spain
| | | | | | | |
Collapse
|
38
|
Bouchard JF, Chouinard J, Lamontagne D. Role of kinins in the endothelial protective effect of ischaemic preconditioning. Br J Pharmacol 1998; 123:413-20. [PMID: 9504381 PMCID: PMC1565180 DOI: 10.1038/sj.bjp.0701619] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
1. The aim of this study was to assess whether the protective effect of ischaemic preconditioning on endothelial function in coronary arteries of the rat involves kinins. 2. Isolated hearts of the rat were exposed to a 30-min low-flow ischaemia (flow rate of 1 ml min[-1]) followed by 20-min reperfusion, after which coronaries were precontracted with 0.1 microM U-46619, and the response to the endothelium-dependent vasodilator, 5-hydroxytryptamine (5-HT, 10 microM), compared to that of the endothelium-independent vasodilator, sodium nitroprusside (SNP, 3 microM). 3. In untreated hearts, ischaemia-reperfusion diminished selectively 5-HT-induced vasodilatation, compared with time-matched sham hearts. The vasodilatation to SNP was unaffected after ischaemia-reperfusion. Preconditioning (5 min of zero-flow ischaemia followed by 10 min reperfusion) in untreated hearts preserved the vasodilatation produced by 5-HT. 4. Blockade of B1 and B2 receptors with either 3 nM [Lys[0], Leu8, des-Arg9]-bradykinin (LLDBK) or 10 nM Hoe 140 (icatibant), respectively, (started 15 min before ischaemic preconditioning or a corresponding sham period and stopped just before the 20-min reperfusion period) had no effect on the vasodilatation produced by either 5-HT or SNP in sham hearts. Pretreatment with Hoe 140 did not block the protective effect of ischaemic preconditioning on the 5-HT vasodilatation. In contrast, LLDBK halved the protective effect of ischaemic preconditioning on endothelium-dependent vasodilatation. 5. Perfusion with either bradykinin or des-Arg9-bradykinin (1 nM) 30 min before and lasting throughout the ischaemia protected the endothelium. 6. In conclusion, ischaemic preconditioning affords protection to the endothelial function in coronary resistance arteries of the rat partly by activation of B1 receptors. Although exogenous BK perfusion can protect the endothelium, B2 receptors do not play an important role in this protection in the rat isolated heart.
Collapse
Affiliation(s)
- J F Bouchard
- Faculty of Pharmacy, Université de Montréal, Québec, Canada
| | | | | |
Collapse
|
39
|
Seyedi N, Win T, Lander HM, Levi R. Bradykinin B2-receptor activation augments norepinephrine exocytosis from cardiac sympathetic nerve endings. Mediation by autocrine/paracrine mechanisms. Circ Res 1997; 81:774-84. [PMID: 9351450 DOI: 10.1161/01.res.81.5.774] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We determined whether local bradykinin production modulates cardiac adrenergic activity. Depolarization of guinea pig heart sympathetic nerve endings (synaptosomes) with 1 to 100 mmol/L K+ caused the release of endogenous norepinephrine (10% to 50% above basal level). This release was exocytotic, because it depended on extracellular Ca2+, was inhibited by the N-type Ca(2+)-channel blocker omega-conotoxin and the protein kinase C inhibitor Ro31-8220, and was potentiated by the neuronal uptake-1 inhibitor desipramine. Typical of adrenergic terminals, norepinephrine exocytosis was enhanced by activation of prejunctional angiotensin AT1-receptors and attenuated by adrenergic alpha 2-receptors, adenosine A1-receptors, and histamine H3-receptors. Exogenous bradykinin enhanced norepinephrine exocytosis by 7% to 35% (EC50, 17 nmol/L), without inhibiting uptake 1. B2-receptor, but not B1-receptor, blockade antagonized this effect. The kininase II/angiotensin-converting enzyme inhibitor enalaprilat and the addition of kininogen or kallikrein enhanced norepinephrine exocytosis by approximately equal to 6% to 40% (EC50, 20 nmol/L) and approximately equal to 25% to 60%, respectively. This potentiation was prevented by serine protease inhibitors and was antagonized by B2-receptor blockade. Therefore, norepinephrine exocytosis is augmented when bradykinin synthesis is increased or when its breakdown is inhibited. This is the first report of a local kallikrein-kinin system in adrenergic nerve endings capable of generating enough bradykinin to activate B2-receptors in an autocrine/paracrine fashion and thus enhance norepinephrine exocytosis. This amplification process may operate in disease states, such as myocardial ischemia, associated with severalfold increases in local kinin concentrations.
Collapse
Affiliation(s)
- N Seyedi
- Department of Pharmacology, Cornell University Medical College, New York, NY 10021, USA
| | | | | | | |
Collapse
|
40
|
Rajani V, Hussain Y, Bolla BS, de Guzman FQ, Montiague RR, Igić R, Rabito SF. Attenuation of epinephrine-induced dysrhythmias by bradykinin: role of nitric oxide and prostaglandins. Am J Cardiol 1997; 80:153A-157A. [PMID: 9293970 DOI: 10.1016/s0002-9149(97)00472-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cardiac dysrhythmias are common during anesthesia and surgery. An important precipitating factor of clinically relevant arrhythmias is the introoperative use of epinephrine. Bradykinin acts as an endogenous cardioprotective substance because it suppresses ventricular dysrhythmias induced by ischemia. In this study, we investigated whether bradykinin has a protective effect, preventing the development of dysrhythmias after epinephrine infusion in rats. Because kinins are potent stimulators of the release of nitric oxide and prostaglandins from the endothelium, we investigated whether the protective effect of bradykinin is mediated by these 2 autacoids. Male Sprague-Dawley rats anesthetized with sodium pentobarbital had catheters placed into a carotid artery and both jugular veins. Arterial blood pressure and lead II of the electrocardiogram (ECG) were continuously monitored and recorded. After a steady state was achieved, 1 mg/kg enalapril, an inhibitor of angiotensin I-converting enzyme/kininase II, was given intravenously to all groups except the one treated with losartan. Bradykinin was infused at the initial rate of 0.5 microg/kg per min. Cardiac arrhythmia was induced with 7.5 microg/kg epinephrine intravenously. Dysrhythmia was assessed by counting the number of premature ventricular contractions (PVCs), runs of ventricular tachycardia (V Tach), and missing beats during the first minute after epinephrine. In untreated, control rats, epinephrine caused 10.8 +/- 2.7 PVCs, 0.8 +/- 0.2 runs of V tach, and 11.6 +/- 7.4 missing beats/min. In rats pretreated with bradykinin, the same dose of epinephrine elicited 1.2 +/- 0.5 PVCs, no runs of V tach, and 0.4 +/- 0.4 missing beats/min. This beneficial effect of bradykinin was partially reversed by N-nitro-L-arginine methyl ester (L-NAME) or indomethacin, and completely by L-NAME plus indomethacin or icatibant, but it was not affected by des-Arg9[Leu8]-bradykinin. We conclude that bradykinin, acting on the B2 receptor, attenuates epinephrine-induced dysrhythmia via a mechanism that involves the release of NO and prostaglandins. Although the mechanism is not clear, NO and prostaglandins may prevent epinephrine-induced dysrhythmia and protect the myocardium via a direct action on cardiac neurons.
Collapse
Affiliation(s)
- V Rajani
- Department of Anesthesiology and Pain Management, Cook County Hospital, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Lagneux C, Ribuot C. In vivo evidence for B1-receptor synthesis induction by heat stress in the rat. Br J Pharmacol 1997; 121:1045-6. [PMID: 9249236 PMCID: PMC1564810 DOI: 10.1038/sj.bjp.0701305] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A hypotensive effect of intravenously injected [des-Arg9]-bradykinin was found in Wistar rats following acute heat stress. This effect was similar to that of intravenously injected bradykinin and was observed 6, 18 and 24 h following an increase in rectal temperature at 42 degrees C for 20 min (H6, H18 and H24 groups, respectively). In contrast, [des-Arg9-bradykinin had no effect on blood pressure in control or sham conditions, early on (3 h) or later on (72 h) after heat stress (Ctl, H3 and H72 groups, respectively), while the response to bradykinin was maintained. The hypotension induced by [des-Arg9]-bradykinin in groups H6, H18 and H24 was comparable to that induced in rats pretreated with a small amount of endotoxin 24 h earlier (LPS group). The hypotensive response in group H24 was totally blunted by [des-Arg10]-Hoe 140 (a potent B1 receptor antagonist) infused at a rate of 10 microg min(-1). These results suggest that heat stress induces the synthesis of vascular B1 receptors in the rat.
Collapse
Affiliation(s)
- C Lagneux
- Laboratoire de Pharmacologie Cardiovasculaire Expérimentale Biomolécules, Université Joseph Fourier, La Tronche, France
| | | |
Collapse
|
42
|
Kaszala K, Végh A, Papp JG, Parratt JR. Modification by bradykinin B2 receptor blockade of protection by pacing against ischaemia-induced arrhythmias. Eur J Pharmacol 1997; 328:51-60. [PMID: 9203568 DOI: 10.1016/s0014-2999(97)83027-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In dogs, rapid cardiac pacing, by way of a pacing electrode in the right ventricle, protects against ventricular arrhythmias when a coronary artery is occluded immediately after cessation of the pacing period. This represents a form of ischaemic preconditioning. The role of bradykinin in mediating the protective effects of rapid cardiac pacing in this model was investigated using a selective antagonist of bradykinin at B2 receptors (icatibant; HOE 140). In the presence of icatibant cardiac pacing (220 beats min(-1)) resulted in more severe ischaemia (as assessed by ST-segment elevation from the pacing electrode at the end of the stimulus) and to a higher incidence of ventricular arrhythmias during the pacing protocol. When the coronary artery was occluded under such conditions the antiarrhythmic protection afforded by cardiac pacing was not seen although other indices of reduced ischaemia severity (epicardial ST-segment mapping; changes in the degree of inhomogeneity of electrical activation within the ischaemic area) were not affected by icatibant treatment. These results suggest that bradykinin is an important trigger mediator involved in the protective effects of cardiac pacing. Whether this is due to the generation of endothelium-derived protective substances (such as nitric oxide and prostacyclin) or whether it results from a direct effect on B2 receptors in cardiac myocytes is unclear.
Collapse
Affiliation(s)
- K Kaszala
- Department of Pharmacology, Albert Szent-Györgyi Medical University, Szeged, Hungary
| | | | | | | |
Collapse
|
43
|
Foucart S, Grondin L, Couture R, Nadeau R. Paradoxical action of desipramine on the modulatory effect of bradykinin on noradrenaline release in a model of metabolic anoxia in rat isolated atria. Can J Physiol Pharmacol 1997. [DOI: 10.1139/y97-048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
44
|
Foucart S, Grondin L, Couture R, Nadeau R. Modulation of noradrenaline release by B1 and B2 kinin receptors during metabolic anoxia in the rat isolated atria. Can J Physiol Pharmacol 1997. [DOI: 10.1139/y97-047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
45
|
Eich-Rathfelder S, Whalley ET, Fautz M, Hohenbleicher F, Fritz H, Siebeck M. Tachyphylaxis of the B1 kinin receptor in porcine endotoxin shock. IMMUNOPHARMACOLOGY 1997; 36:173-7. [PMID: 9228543 DOI: 10.1016/s0162-3109(97)00018-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Previous experiments in anesthetized pigs have demonstrated that blockade of the bradykinin B2 receptor in experimental endotoxin shock attenuates LPS-induced organ failure, lung dysfunction and mortality. Additional B1 receptor blockade in this situation seems to counteract the beneficial effects of B2 blockade. This suggests that the upregulation of B1 receptors during porcine LPS shock may be a useful mechanism of host defense. Furthermore, infusion of a B1 agonist during septic shock may be of therapeutic benefit. In order to prepare an experiment with B1 stimulation in LPS shock, we conducted a study in anesthetized pigs, in which the B1 receptor has been upregulated by infusion of bacterial lipopolysaccharide (LPS), by evaluating the effect of constant intravenous infusions of the B1 agonist des-Arg10-kallidin on the hypotensive response to bolus doses of this agonist. Following infusions of lipopolysaccharide from S. abortus equi, anesthetised pigs received repeated intra-arterial bolus injections of des-Arg10-kallidin before and during continuous infusions of this agonist in doses of 3, 10, 30 and 100 ng/kg/min. We found that all doses greater than 3 ng/kg/min produced attenuation of the hypotensive response produced by bolus administration of the B1 agonist des-Arg10-kallidin. We conclude that tachyphylaxis is an important feature to be considered in experiments with continuous administration of a B1 agonist in LPS shock.
Collapse
MESH Headings
- Animals
- Blood Pressure/drug effects
- Bradykinin Receptor Antagonists
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Interactions
- Infusions, Intravenous
- Injections, Intra-Arterial
- Kallidin/administration & dosage
- Kallidin/analogs & derivatives
- Kallidin/pharmacology
- Kallidin/therapeutic use
- Lipopolysaccharides/toxicity
- Receptor, Bradykinin B1
- Receptors, Bradykinin/agonists
- Receptors, Bradykinin/physiology
- Shock, Septic/chemically induced
- Shock, Septic/drug therapy
- Shock, Septic/metabolism
- Swine
- Tachyphylaxis/physiology
- Up-Regulation
Collapse
Affiliation(s)
- S Eich-Rathfelder
- Department of Surgery, Klinikum Innenstadt, Ludwig-Maximilians University, Munich, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Feng J, Yamaguchi N, Foucart S, Chahine R, Lamontagne D, Nadeau R. Transient ischemia inhibits nonexocytotic release of norepinephrine following sustained ischemia in rat heart: is bradykinin involved? Can J Physiol Pharmacol 1997. [DOI: 10.1139/y97-072] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
47
|
Duncan AM, Burrell LM, Kladis A, Campbell DJ. Angiotensin and bradykinin peptides in rats with myocardial infarction. J Card Fail 1997; 3:41-52. [PMID: 9110254 DOI: 10.1016/s1071-9164(97)90007-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Angiotensin II (Ang II) stimulates cardiac hypertrophy and fibrosis, whereas bradykinin [BK-(1-9)] has cardioprotective actions and reduces infarct size following myocardial infarction. METHODS AND RESULTS We investigated whether myocardial infarction and cardiac failure are associated with changes in circulating and tissue levels of angiotensin and bradykinin peptides. Myocardial infarction was produced in rats by coronary artery ligation and confirmed by electrocardiogram. Ang II, Ang I, BK-(1-9), and its metabolite BK-(1-7) were measured 1, 2, 3, 7, and 28 days after myocardial infarction. In comparison with sham operated rats, myocardial infarction reduced blood pressure and body weight, and produced cardiac hypertrophy and cardiac failure. Myocardial infarction increased plasma renin and ACE activity, reduced plasma angiotensinogen, and increased Ang II levels in plasma, aorta, kidney, and lung. Ang II levels in whole cardiac ventricles were similar in infarct and sham operated rats, but were positively correlated with heart weight/body weight ratio in infarct rats 3, 7, and 28 days after infarction. In a separate study of cardiac regions, Ang II levels were similar in infarct and sham operated rats, except at 7 days post surgery when right ventricular Ang II levels were higher in infarct rats. In infarct rats, Ang II levels were higher in the right ventricle and in the infarct than in the non-infarcted left ventricle at 7 days, but these differences were not apparent at 28 days after infarction. BK-(1-9) levels were increased in the heart and lung on days 2 and 3 post infarction, but not in the aorta or kidney. A decrease in BK-(1-7)/BK-(1-9) ratio suggested reduced metabolism of BK-(1-9) to BK-(1-7) in infarcted hearts. CONCLUSIONS The transient activation of the circulating renin angiotensin system, and increased Ang II levels in the aorta, kidney, and lung may contribute to the systemic responses to myocardial infarction and cardiac failure. The correlations between cardiac Ang II levels and heart weight/body weight ratio noted for whole cardiac ventricles support a role for local Ang II levels in the process of myocardial remodeling post infarction. The increased cardiac BK-(1-9) levels in the acute phase of myocardial infarction were consistent with a role for this peptide in cardioprotection and limitation of infarct size.
Collapse
Affiliation(s)
- A M Duncan
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | | | | | | |
Collapse
|
48
|
Feng J, Chahine R, Yamaguchi N, Lamontagne D, Nadeau R. Brief repetitive ischemia: effect on norepinephrine release, arrhythmias, and functional recovery in isolated perfused rat heart. Can J Physiol Pharmacol 1996. [DOI: 10.1139/y96-150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Veelken R, Glabasnia A, Stetter A, Hilgers KF, Mann JF, Schmieder RE. Epicardial bradykinin B2 receptors elicit a sympathoexcitatory reflex in rats. Hypertension 1996; 28:615-21. [PMID: 8843887 DOI: 10.1161/01.hyp.28.4.615] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Bradykinin may be generated in the heart during ischemia and is involved in nociception. We tested the hypothesis that bradykinin elicits a sympathoexcitatory reflex in rats by stimulating cardiac afferent nerve fibers. Rats were implanted with femoral catheters for measurement of blood pressure and heart rate, a bipolar electrode for measurement of renal sympathetic nerve activity, and a pericardial catheter for intrapericardial injection of substances. Rats were slightly anesthetized with hexobarbital so pain reactions were prevented. Graded doses of bradykinin (2.5, 12, 25 micrograms) were injected intravenously or intrapericardially into control rats, intrapericardially after vagotomy, intrapericardially after intrapericardial pretreatment with the bradykinin B2 receptor antagonist Hoe 140, and intrapericardially after cardiac autonomic blockade (intrapericardial pretreatment with 10% procaine). For comparison, the serotonin 5-HT3 agonist phenylbiguanide, a substance known to elicit sympathoinhibitory reflexes by cardiac vagal afferents, and adenosine, putatively inducing sympathoexcitatory responses via the heart, were applied intrapericardially. Bradykinin increased blood pressure when administered intrapericardially but decreased blood pressure when injected intravenously; both intrapericardial and intravenous bradykinin increased renal sympathetic nerve activity. Intrapericardial adenosine had no effect on circulatory control. Intrapericardial pretreatment with the B2 receptor antagonist Hoe 140 completely inhibited the increases of blood pressure and renal sympathetic nerve activity in response to intrapericardial bradykinin but did not affect the responses to intrapericardial phenylbiguanide. Bilateral cervical vagotomy abolished the decreases of blood pressure, heart rate, and renal sympathetic nerve activity after intrapericardial phenylbiguanide but did not influence the responses to intrapericardial bradykinin. Cardiac autonomic blockade with intrapericardial procaine abolished all responses to bradykinin and phenylbiguanide. We conclude that cardiac bradykinin elicits a sympathoexcitatory reflex by epicardial B2 receptors in rats. The afferent portion of the reflex is most likely contained within sympathetic cardiac afferent fibers. Bradykinin may contribute to increased sympathetic nerve activity in pathophysiological situations of coronary artery disease and cardiac ischemia.
Collapse
Affiliation(s)
- R Veelken
- Department of Internal Medicine, University of Erlangen-Nürnberg, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Gouin L, Cardinal R, Adam A, Drapeau G, Nadeau R. Kinin-induced prolongation of action-potential duration in right ventricular muscle from rat: involvement of B1 and B2 receptors. J Cardiovasc Pharmacol 1996; 28:337-43. [PMID: 8856493 DOI: 10.1097/00005344-199608000-00023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Previous work has shown that, in rat ventricular muscle, bradykinin (BK) causes a dose-dependent increase in action potential duration (APD), an action that may be responsible for APD prolongation by captopril (kininase II). To determine which kinin receptor might be involved in APD prolongation, we studied the effects of B1- and B2-receptor agonists, as well as those of antagonists and mergepta (a kininase I inhibitor) added during BK superfusion. Action potentials were recorded by using the standard glass microelectrode technique in rat ventricular muscle preparations. Action-potential characteristics were compared between preparations superfused with peptide/drug-free Tyrode's solution (control group) and preparations superfused with peptide/drug-containing solution. APD was significantly longer in preparations superfused with BK (10(-8) M) than in the control group. The APD prolongation induced by BK, a known B2-receptor agonist, was significantly reduced by Hoe 140 (a B2 antagonist) and also by Lys[Leu8]des-Arg9-BK (a B1 antagonist), an action presumably related to inhibition of B1 receptor stimulation by the BK metabolite des-Arg9-BK. When mergepta was added in the presence of BK, APD prolongation by BK was significantly reduced, an effect that could have been related to reduced B1-receptor stimulation after inhibition of the endogenous generation of des-Arg9-BK by kininase I. Sar4-[d-Phe8]des-Arg9-BK, a B1-receptor agonist that is not degraded by kininase II, also prolonged APD. We conclude that both B1 and B2 receptors may be involved in APD prolongation induced in rat ventricular muscle preparations.
Collapse
Affiliation(s)
- L Gouin
- Faculté de médecine, Département de pharmacologie, Hôpital du Sacré-Coeur de Montréal, Université de Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|