Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Kidney Int ; 98(3): 543-545, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32828234

RESUMO

The role of membrane channels in juxtaglomerular cell physiology is only partially understood. Pannexin 1 is a mechanosensitive, nonjunctional channel known for its role in adenosine triphosphate release. The study by DeLalio et al. documents involvement of pannexin 1 in renin secretion by studying mice deficient in pannexin 1 in renin-secreting cells and a prorenin-secreting cell line. Pannexin 1 is believed to suppress renin secretion and thereby modify blood pressure. The commentary addresses the broader physiological implication of these observations for the regulation of renin and blood pressure.


Assuntos
Sistema Justaglomerular , Renina , Animais , Pressão Sanguínea , Células Cultivadas , Homeostase , Camundongos
2.
Am J Physiol Heart Circ Physiol ; 314(2): H350-H358, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29101168

RESUMO

Angiotensin I-converting enzyme (ACE) levels in humans are under strong genetic influence. Genetic variation in ACE has been linked to risk for and progression of cardiovascular and renal diseases. Causality has been documented in genetically modified mice, but the mechanisms underlying causality are not completely elucidated. To further document the vascular and renal consequences of a moderate genetic increase in ACE synthesis, we studied genetically modified mice carrying three copies of the ACE gene (three-copy mice) and littermate wild-type animals (two-copy mice). We investigated peripheral and renal vascular reactivity to angiotensin II and bradykinin in vivo by measuring blood pressure and renal blood flow after intravenous administration and also reactivity of isolated glomerular arterioles by following intracellular Ca2+ mobilization. Carrying three copies of the ACE gene potentiated the systemic and renal vascular responses to angiotensin II over the whole range of peptide concentration tested. Consistently, the response of isolated glomerular afferent arterioles to angiotensin II was enhanced in three-copy mice. In these mice, signaling pathways triggered by endothelial activation by bradykinin or carbachol in glomerular arterioles were also altered. Although the nitric oxide (NO) synthase (NOS)/NO pathway was not functional in arterioles of two-copy mice, in muscular efferent arterioles of three-copy mice NOS3 gene expression was induced and NO mediated the effect of bradykinin or carbachol. These data document new and unexpected vascular consequences of a genetic increase in ACE synthesis. Enhanced vasoconstrictor effect of angiotensin II may contribute to the risk for cardiovascular and renal diseases linked to genetically high ACE levels. NEW & NOTEWORTHY A moderate genetic increase in angiotensin I-converting enzyme (ACE) in mice similar to the effect of the ACE gene D allele in humans unexpectedly potentiates the systemic and renal vasoconstrictor responses to angiotensin II. It also alters the endothelial signaling pathways triggered by bradykinin or carbachol in glomerular efferent arterioles.


Assuntos
Angiotensina II/farmacologia , Pressão Arterial/efeitos dos fármacos , Arteríolas/efeitos dos fármacos , Bradicinina/farmacologia , Glomérulos Renais/irrigação sanguínea , Peptidil Dipeptidase A/biossíntese , Circulação Renal/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia , Animais , Arteríolas/enzimologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Carbacol/farmacologia , Indução Enzimática , Feminino , Genótipo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Peptidil Dipeptidase A/genética , Fenótipo
3.
Biol Chem ; 397(12): 1217-1222, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27622831

RESUMO

Genetic and pharmacological studies, clinical and experimental, focused on kallikrein-K1, kinin receptors and ACE/kininase II suggest that kinin release in the settings of ischemia or diabetes reduces organ damage, especially in the heart and kidney. Kinin bioavailability may be a limiting factor for efficacy of current kinin-potentiating drugs, like ACE inhibitors. Primary activation of kinin receptors by prototypic pharmacological agonists, peptidase-resistant, selective B1 or B2, displays therapeutic efficacy in experimental cardiac and peripheral ischemic and diabetic diseases. B1R agonism was especially efficient in diabetic animals and had no unwanted effects. Clinical development of kinin receptor agonists may be warranted.


Assuntos
Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Isquemia/metabolismo , Isquemia/terapia , Sistema Calicreína-Cinina , Animais , Diabetes Mellitus/tratamento farmacológico , Humanos , Isquemia/tratamento farmacológico , Sistema Calicreína-Cinina/efeitos dos fármacos
4.
Clin Sci (Lond) ; 130(1): 45-56, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26443866

RESUMO

Impaired skin wound healing is a major medical problem in diabetic subjects. Kinins exert a number of vascular and other actions limiting organ damage in ischaemia or diabetes, but their role in skin injury is unknown. We investigated, through pharmacological manipulation of bradykinin B1 and B2 receptors (B1R and B2R respectively), the role of kinins in wound healing in non-diabetic and diabetic mice. Using two mouse models of diabetes (streptozotocin-induced and db/db mice) and non-diabetic mice, we assessed the effect of kinin receptor activation or inhibition by subtype-selective pharmacological agonists (B1R and B2R) and antagonist (B2R) on healing of experimental skin wounds. We also studied effects of agonists and antagonist on keratinocytes and fibroblasts in vitro. Levels of Bdkrb1 (encoding B1R) and Bdkrb2 (encoding B2R) mRNAs increased 1-2-fold in healthy and wounded diabetic skin compared with in non-diabetic skin. Diabetes delayed wound healing. The B1R agonist had no effect on wound healing. In contrast, the B2R agonist impaired wound repair in both non-diabetic and diabetic mice, inducing skin disorganization and epidermis thickening. In vitro, B2R activation unbalanced fibroblast/keratinocyte proliferation and increased keratinocyte migration. These effects were abolished by co-administration of B2R antagonist. Interestingly, in the two mouse models of diabetes, the B2R antagonist administered alone normalized wound healing. This effect was associated with the induction of Ccl2 (encoding monocyte chemoattractant protein 1)/Tnf (encoding tumour necrosis factor α) mRNAs. Thus stimulation of kinin B2 receptor impairs skin wound healing in mice. B2R activation occurs in the diabetic skin and delays wound healing. B2R blockade improves skin wound healing in diabetic mice and is a potential therapeutic approach to diabetic ulcers.


Assuntos
Antagonistas de Receptor B2 da Bradicinina/farmacologia , Bradicinina/análogos & derivados , Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Receptor B2 da Bradicinina/efeitos dos fármacos , Úlcera Cutânea/tratamento farmacológico , Pele/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Animais , Bradicinina/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Complicações do Diabetes/etiologia , Complicações do Diabetes/genética , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , RNA Mensageiro/metabolismo , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Úlcera Cutânea/etiologia , Úlcera Cutânea/metabolismo , Úlcera Cutânea/patologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
6.
J Pharmacol Exp Ther ; 352(2): 218-26, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25398240

RESUMO

Limb ischemia is a major complication of thromboembolic diseases. Diabetes worsens prognosis by impairing neovascularization. Genetic or pharmacological inactivation of the kallikrein-kinin system aggravates limb ischemia in nondiabetic animals, whereas angiotensin I-converting enzyme/kininase II inhibition improves outcome. The role of kinins in limb ischemia in the setting of diabetes is not documented. We assessed whether selective activation of kinin receptors by pharmacological agonists can influence neovascularization in diabetic mice with limb ischemia and have a therapeutic effect. Selective pseudopeptide kinin B1 or B2 receptor agonists resistant to peptidase action were administered by osmotic minipumps at a nonhypotensive dosage for 14 days after unilateral femoral artery ligation in mice previously rendered diabetic by streptozotocin. Comparison was made with ligatured, nonagonist-treated nondiabetic and diabetic mice. Diabetes reduced neovascularization, assessed by microangiography and histologic capillary density analysis, by roughly 40%. B1 receptor agonist or B2 receptor agonist similarly restored neovascularization in diabetic mice. Neovascularization in agonist-treated diabetic mice was indistinguishable from nondiabetic mice. Both treatments restored blood flow in the ischemic hindfoot, measured by laser-Doppler perfusion imaging. Macrophage infiltration increased 3-fold in the ischemic gastrocnemius muscle during B1 receptor agonist or B2 receptor agonist treatment, and vascular endothelial growth factor (VEGF) level increased 2-fold. Both treatments increased, by 50-100%, circulating CD45/CD11b-positive monocytes and CD34(+)/VEGFR2(+) progenitor cells. Thus, selective pharmacological activation of B1 or B2 kinin receptor overcomes the effect of diabetes on postischemic neovascularization and restores tissue perfusion through monocyte/macrophage mobilization. Kinin receptors are potential therapeutic targets in limb ischemia in diabetes.


Assuntos
Diabetes Mellitus Experimental/complicações , Angiopatias Diabéticas/tratamento farmacológico , Membro Posterior/irrigação sanguínea , Isquemia/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Receptor B1 da Bradicinina/agonistas , Receptor B2 da Bradicinina/agonistas , Animais , Bradicinina/administração & dosagem , Bradicinina/análogos & derivados , Bradicinina/uso terapêutico , Diabetes Mellitus Experimental/metabolismo , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/metabolismo , Isquemia/etiologia , Isquemia/metabolismo , Sistema Calicreína-Cinina/efeitos dos fármacos , Fluxometria por Laser-Doppler , Masculino , Camundongos Endogâmicos C57BL , Fluxo Sanguíneo Regional/efeitos dos fármacos , Estreptozocina/farmacologia
8.
Prog Drug Res ; 69: 145-96, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25130042

RESUMO

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.


Assuntos
Doenças Cardiovasculares/genética , Variação Genética , Calicreínas/genética , Nefropatias/genética , Cininas/genética , Animais , Fármacos Cardiovasculares/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/fisiopatologia , Predisposição Genética para Doença , Humanos , Calicreínas/metabolismo , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Nefropatias/fisiopatologia , Cininas/metabolismo , Fenótipo , Fármacos Renais/uso terapêutico , Transdução de Sinais
9.
J Cardiovasc Pharmacol ; 63(3): 274-81, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24220315

RESUMO

Coronary endothelial dysfunction is involved in cardiac ischemia-reperfusion (IR) injury. Vascular endothelial growth factor (VEGF) activates endothelial cells and exerts cardioprotective effects in isolated hearts. The recently discovered viper venom protein called increasing capillary permeability protein (ICPP) exerts VEGF-like effects in endothelial cells. We examined whether VEGF or ICPP can influence IR outcome in vivo in mice. Dosages of VEGF and ICPP were determined by preliminary blood pressure study. In IR, both the proteins administered intravenously at reperfusion reduced infarct size (IS) by 57% for VEGF and 52% for ICPP (P < 0.01). Pretreatment with a selective VEGFR2 receptor antagonist abolished the reduction in IS. VEGF and ICPP induced ERK phosphorylation in the myocardium. IR triggered mitochondrial pore opening and impaired mitochondrial respiratory function. These effects of IR were prevented by VEGF or ICPP, which increased mitochondrial calcium retention capacity by 37% compared with saline (P < 0.05) and improved mitochondrial respiratory function (by 71% and 65%, respectively for state 3, and 51% and 38% for state 4, P < 0.01 for VEGF). Thus, intravenous administration of VEGF or ICPP at reperfusion largely reduces IS in IR, through stimulation of VEGFR2 receptors. This effect is mediated, at least in part, by improvement of IR-induced mitochondrial dysfunction.


Assuntos
Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Proteínas/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Venenos de Víboras/química , Animais , Cálcio/metabolismo , Cardiotônicos/administração & dosagem , Cardiotônicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/patologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Proteínas/administração & dosagem , Proteínas/química , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
J Pharmacol Exp Ther ; 346(1): 23-30, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23591995

RESUMO

Cardiac ischemia is a leading cause of death, especially in diabetic patients. The diabetic ischemic heart is resistant experimentally to established cardioprotective treatments. New pharmacological approaches to cardiac protection are warranted. The kallikrein-kinin system is involved in myocardial protection in ischemia. Respective roles of B1 (B1R) and B2 (B2R) receptors remain controversial. We tested whether pharmacological activation of kinin receptors may have therapeutic effect in cardiac ischemia-reperfusion in nondiabetic (NDiab) and diabetic (Diab) mice. We assessed effect on infarct size (IS) and signaling pathways involved in myocardial protection of potent selective pharmacological agonists of B1R or B2R given at reperfusion. In NDiab mice, a B2R agonist reduced IS significantly by 47%, similarly to ramiprilat or ischemic postconditioning, via activation of phosphoinositide 3 kinase/Akt pathway leading to inhibition of glycogen synthase kinase-3ß (GSK-3ß). B1R agonist had no effect on IS. In contrast, in Diab mice, the B2R agonist, ramiprilat, or ischemic postconditioning failed to reduce IS but a B1R agonist significantly reduced IS by 44% via activation of phosphoinositide 3 kinase/Akt and extracellular signal-regulated kinase 1/2, both leading to GSK-3ß inhibition. Differential effect of B2R or B1R agonists in NDiab and Diab mice can be linked to inactivation of B2R signaling and induction of B1R in heart of Diab mice. Thus, a pharmacological B2R agonist is cardioprotective in acute ischemia in nondiabetic animals. B1R agonist overcomes resistance of diabetic heart to cardioprotective treatments. Pharmacological activation of B1R and B2R may become a treatment for diabetic and nondiabetic patients, respectively, in acute coronary syndromes.


Assuntos
Bradicinina/análogos & derivados , Cardiotônicos/uso terapêutico , Diabetes Mellitus Tipo 1/complicações , Isquemia Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Receptor B1 da Bradicinina/agonistas , Receptor B2 da Bradicinina/agonistas , Animais , Bradicinina/administração & dosagem , Bradicinina/efeitos adversos , Bradicinina/uso terapêutico , Cardiotônicos/administração & dosagem , Cardiotônicos/efeitos adversos , Diabetes Mellitus Tipo 1/metabolismo , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/complicações , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptor B1 da Bradicinina/genética , Receptor B1 da Bradicinina/metabolismo , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
Biol Chem ; 394(3): 329-33, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324381

RESUMO

Tissue kallikrein has been suggested to be involved in blood pressure regulation and in protection against hypertension. However, this hypothesis remains debated. Recently, murine genetic models of kallikrein deficiency have been engineered and partial genetic deficiency in kallikrein activity has been characterized in humans. Studies in kallikrein-deficient mice indicate that kallikrein indeed influences blood pressure in the setting of mineralocorticoid excess and salt retention but not in normotensive animals and in high renin hypertension. These observations suggest that kallikrein can have antihypertensive function in physiological situations where sodium retention can trigger blood pressure elevation.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão/fisiopatologia , Calicreínas Teciduais/deficiência , Calicreínas Teciduais/metabolismo , Animais , Humanos , Camundongos , Camundongos Transgênicos , Calicreínas Teciduais/genética
12.
Circ Res ; 108(3): 284-93, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21164105

RESUMO

RATIONALE: Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. OBJECTIVE: To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. METHODS AND RESULTS: Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow-derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B(2) receptor (B(2)R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B(2)R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B(2)R expression rescued the diabetic phenotype. CONCLUSIONS: This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor-dependent mechanisms.


Assuntos
Movimento Celular/fisiologia , Calicreínas/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Neovascularização Fisiológica/fisiologia , Adulto , Idoso , Animais , Estudos de Casos e Controles , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Regulação para Baixo , Feminino , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/metabolismo , Calicreínas/genética , Cininas/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Modelos Animais , Óxido Nítrico Sintase/metabolismo , RNA Mensageiro/metabolismo , Receptor B2 da Bradicinina/metabolismo , Transdução de Sinais/fisiologia
13.
Endocrinology ; 163(9)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35863039

RESUMO

Sex differences in the control of prolactin secretion are well documented. Sex-related differences in intrapituitary factors regulating lactotroph function have recently attracted attention. Sex differences in prolactinoma development are well documented in clinic, prolactinomas being more frequent in women but more aggressive in men, for poorly understood reasons. Kallikrein, the enzyme releasing kinins has been found in the pituitary, but there is no information on pituitary kinin receptors and their function. In the present work, we characterized pituitary bradykinin receptors (BRs) at the messenger RNA and protein levels in 2 mouse models of prolactinoma, Drd2 receptor gene inactivation and hCGß gene overexpression, in both males and females, wild type or genomically altered. BR B2 (B2R) accounted for 97% or more of total pituitary BRs in both models, regardless of genotype, and was present in lactotrophs, somatotrophs, and gonadotrophs. Male pituitaries displayed higher level of B2R than females, regardless of genotype. Pituitary B2R gene expression was downregulated by estrogen in both males and females but only in females by dopamine. Activation of B1R or B2R by selective pharmacological agonists induced prolactin release in male pituitaries but inhibited prolactin secretion in female pituitaries. Increased B2R content was observed in pituitaries of mutated animals developing prolactinomas, compared to their respective wild-type controls. The present study documents a novel sex-related difference in the control of prolactin secretion and suggests that kinins are involved, through B2R activation, in lactotroph function and prolactinoma development.


Assuntos
Neoplasias Hipofisárias , Prolactinoma , Animais , Feminino , Humanos , Cininas , Masculino , Camundongos , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Prolactina/metabolismo , Prolactinoma/genética , Prolactinoma/metabolismo , Receptor B2 da Bradicinina/agonistas , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo , Receptores da Bradicinina
14.
Diabetes Care ; 45(2): 407-415, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34853028

RESUMO

OBJECTIVE: The ACE insertion/deletion (I/D) polymorphism has been widely studied in people with diabetes, albeit not with regard to lower-limb amputation (LLA). We examined associations among this polymorphism, plasma ACE concentration, and LLA in people with type 1 diabetes. RESEARCH DESIGN AND METHODS: ACE I/D genotype and plasma ACE were assessed in three prospective cohorts of participants with type 1 diabetes. LLA was defined as minor (below-the-ankle amputation consisting of at least one ray metatarsal resection) or major (transtibial or transfemoral) amputation. Linear, logistic, and Cox regression models were computed to evaluate the likelihood of prevalent and incident LLA by ACE genotype (XD [ID or ID] vs. II) and plasma ACE, after adjusting for confounders. RESULTS: Among 1,301 participants (male 54%, age 41 ± 13 years), 90 (6.9%) had a baseline history of LLA. Baseline LLA was more prevalent in XD (7.4%) than in II genotype (4.5%, odds ratio [OR] 2.17 [95 %CI 1.03-4.60]). Incident LLA occurred in 53 individuals during the 14-year follow-up and was higher in XD versus II carriers (hazard ratio 3.26 [95% CI 1.16-13.67]). This association was driven by excess risk of minor, but not major, LLA. The D allele was associated with increased prevalent LLA at the end of follow-up (OR 2.48 [1.33-4.65]). LLA was associated with higher mean (95% CI) ACE levels in II (449 [360, 539] vs. 354 [286, 423] ng/mL), but not XD (512 [454, 570] vs. 537 [488, 586]), carriers. CONCLUSIONS: This report is the first of an independent association between ACE D allele and excess LLA risk, mainly minor amputations, in patients with type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1 , Peptidil Dipeptidase A , Adulto , Amputação Cirúrgica , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/cirurgia , Genótipo , Humanos , Extremidade Inferior/cirurgia , Masculino , Pessoa de Meia-Idade , Peptidil Dipeptidase A/genética , Polimorfismo Genético/genética , Estudos Prospectivos
15.
Microvasc Res ; 81(2): 206-15, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21167844

RESUMO

Angiotensin I-converting enzyme (kininase II, ACE, CD143) availability is a determinant of local angiotensin and kinin concentrations and physiological actions. Limited information is available on ACE synthesis in peripheral vascular beds. We studied the distribution of ACE along the human and rat vascular tree, and determined whether the enzyme was uniformly distributed in all endothelial cells (EC) or if differences occurred among vessels and organs. The distribution of ACE was assessed by using a panel of anti-human ACE monoclonal antibodies and serial sections of the entire vascular tree of humans. Comparison was made with other EC markers. EC of small muscular arteries and arterioles displayed high ACE immunoreactivity in all organs studied except the kidney, while EC of large arteries and of veins were poorly reactive or completely negative. Only 20% on average of capillary EC in each organ, including the heart, stained for ACE, with the remarkable exception of the lung and kidney. In the lung all capillary EC were labeled intensively for ACE, whereas in the kidney the entire vasculature was devoid of detectable enzyme. In contrast to the man, the rat showed homogeneous endothelial expression of ACE in all large and middle-sized arteries, and in veins, but in renal vessels ACE expression was reduced. This study documents a vessel, organ and species specific pattern of distribution of endothelial ACE. The markedly reduced ACE content of the renal vasculature may protect the renal circulation against excess angiotensin II formation and kinin depletion, and maintain high renal blood flow.


Assuntos
Estruturas Animais/metabolismo , Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Especificidade de Órgãos/fisiologia , Peptidil Dipeptidase A/metabolismo , Adulto , Idoso , Estruturas Animais/irrigação sanguínea , Animais , Anticorpos Monoclonais/imunologia , Artérias/metabolismo , Vasos Coronários/metabolismo , Humanos , Imuno-Histoquímica , Rim/irrigação sanguínea , Rim/fisiologia , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Microscopia Imunoeletrônica , Microvasos/metabolismo , Pessoa de Meia-Idade , Peptidil Dipeptidase A/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Mudanças Depois da Morte , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Especificidade da Espécie , Veias/metabolismo
16.
FASEB J ; 24(12): 4691-700, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20667972

RESUMO

Angiotensin I-converting enzyme (ACE; kininase II) levels in humans are genetically determined. ACE levels have been linked to risk of myocardial infarction, but the association has been inconsistent, and the causality underlying it remains undocumented. We tested the hypothesis that genetic variation in ACE levels influences myocardial tolerance to ischemia. We studied ischemia-reperfusion injury in mice bearing 1 (ACE1c), 2 (ACE2c, wild type), or 3 (ACE3c) functional copies of the ACE gene and displaying an ACE level range similar to humans. Infarct size in ACE1c was 29% lower than in ACE2c (P<0.05). Pretreatment with a kinin B2 receptor antagonist suppressed this reduction. In ACE3c, infarct size was the same as in ACE2c. But ischemic preconditioning, which reduced infarct size in ACE2c (-63%, P<0.001) and ACE1c (-52%, P<0.05), was not efficient in ACE3c (-2%, NS, P<0.01 vs. ACE2c). In ACE3c, ischemic preconditioning did not decrease myocardial inflammation or cardiomyocyte apoptosis. Pretreatment with a renin inhibitor had no cardioprotective effect in ACE2c, but in ACE3c partially restored (38%) the cardioprotection of ischemic preconditioning. Thus, a modest genetic increase in ACE impairs myocardial tolerance to ischemia. ACE level plays a critical role in cardiac ischemia, through both kinin and angiotensin mediated mechanisms.


Assuntos
Coração/efeitos dos fármacos , Infarto do Miocárdio/enzimologia , Isquemia Miocárdica/enzimologia , Miocárdio/enzimologia , Peptidil Dipeptidase A/metabolismo , Traumatismo por Reperfusão/genética , Amidas/farmacologia , Angiotensina I/farmacologia , Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Bradicinina/análogos & derivados , Bradicinina/farmacologia , Antagonistas dos Receptores da Bradicinina , Fumaratos/farmacologia , Cininas/farmacologia , Pulmão/enzimologia , Camundongos , Camundongos Mutantes , Infarto do Miocárdio/genética , Isquemia Miocárdica/genética , Peptidil Dipeptidase A/genética , Renina/antagonistas & inibidores
17.
Pharmaceuticals (Basel) ; 14(3)2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800422

RESUMO

This review addresses the physiological role of the kallikrein-kinin system in arteries, heart and kidney and the consequences of kallikrein and kinin actions in diseases affecting these organs, especially ischemic and diabetic diseases. Emphasis is put on pharmacological and genetic studies targeting kallikrein; ACE/kininase II; and the two kinin receptors, B1 (B1R) and B2 (B2R), distinguished through the work of Domenico Regoli and his collaborators. Potential therapeutic interest and limitations of the pharmacological manipulation of B1R or B2R activity in cardiovascular and renal diseases are discussed. This discussion addresses either the activation or inhibition of these receptors, based on recent clinical and experimental studies.

18.
Diabetes Care ; 44(6): 1377-1384, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33827803

RESUMO

OBJECTIVE: The deletion (D) allele of the ACE insertion/deletion (I/D) polymorphism is a risk factor for diabetic kidney disease. We assessed its contribution to long-term kidney outcomes and all-cause death in patients with long-standing type 1 diabetes. RESEARCH DESIGN AND METHODS: A total of 1,155 participants from three French and Belgian cohorts were monitored for a median duration of 14 (interquartile range 13) years. The primary outcome was the occurrence of end-stage kidney disease (ESKD) or a 40% drop in the estimated glomerular filtration rate (eGFR). Secondary outcomes were the individual components of the primary outcome, rapid decline in eGFR (steeper than -3 mL/min/1.73 m2 per year), incident albuminuria, all-cause death, and a composite ESKD or all-cause death. Hazard ratios (HRs) for XD versus II genotype and for baseline plasma ACE levels were computed by Cox analysis. Genotype performance in stratifying the primary outcome was tested. RESULTS: Genotype distribution was 954 XD and 201 II. The primary outcome occurred in 20% of XD and 13% of II carriers: adjusted HR 2.07 (95% CI 1.32-3.40; P = 0.001). Significant associations were also observed for rapid decline in eGFR, incident albuminuria, ESKD, all-cause death, and ESKD or all-cause death. Baseline plasma ACE levels were higher in XD carriers and significantly associated with an increased risk of the primary outcome. The ACE genotype enhanced net reclassification improvement (0.154, 95% CI 0.007-0.279; P = 0.04) and integrated discrimination improvement (0.012, 95%CI 0.001-0.021; P = 0.02) for primary outcome stratification. CONCLUSIONS: The D-allele of the ACE I/D polymorphism was associated with an increased risk of major kidney events and all-cause death in patients with long-standing type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1 , Nefropatias Diabéticas , Peptidil Dipeptidase A , Albuminúria/genética , Diabetes Mellitus Tipo 1/genética , Nefropatias Diabéticas/genética , Taxa de Filtração Glomerular , Humanos , Rim , Peptidil Dipeptidase A/sangue , Peptidil Dipeptidase A/genética
19.
Arterioscler Thromb Vasc Biol ; 29(5): 657-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19164804

RESUMO

OBJECTIVE: Human Tissue Kallikrein (hKLK1) overexpression promotes an enduring neovascularization of ischemic tissue, yet the cellular mechanisms of hKLK1-induced arteriogenesis remain unknown. Furthermore, no previous study has compared the angiogenic potency of hKLK1, with its loss of function polymorphic variant, rs5515 (R53H), which possesses reduced kinin-forming activity. METHODS AND RESULTS: Here, we demonstrate that tissue kallikrein knockout mice (KLK1-/-) show impaired muscle neovascularization in response to hindlimb ischemia. Gene-transfer of wild-type Ad.hKLK1 but not Ad.R53H-hKLK1 was able to rescue this defect. Similarly, in the rat mesenteric assay, Ad.hKLK1 induced a mature neovasculature with increased vessel diameter through kinin-B2 receptor-mediated recruitment of pericytes and vascular smooth muscle cells, whereas Ad.R53H-hKLK1 was ineffective. Moreover, hKLK1 but not R53H-hKLK1 overexpression in the zebrafish induced endothelial precursor cell migration and vascular remodeling. Furthermore, Ad.hKLK1 activates metalloproteinase (MMP) activity in normoperfused muscle and fails to promote reparative neovascularization in ischemic MMP9-/- mice, whereas its proarteriogenic action was preserved in ApoE-/- mice, an atherosclerotic model of impaired angiogenesis. CONCLUSIONS: These results demonstrate the fundamental role of endogenous Tissue Kallikrein in vascular repair and provide novel information on the cellular and molecular mechanisms responsible for the robust arterialization induced by hKLK1 overexpression.


Assuntos
Membro Posterior/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Circulação Esplâncnica/fisiologia , Calicreínas Teciduais/fisiologia , Animais , Humanos , Isquemia/fisiopatologia , Sistema Calicreína-Cinina/fisiologia , Masculino , Metaloproteinase 9 da Matriz/fisiologia , Camundongos , Camundongos Knockout , Ratos , Cicatrização/fisiologia , Peixe-Zebra
20.
Kidney Int ; 76(4): 395-403, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19516248

RESUMO

Tissue kallikrein is the main kinin-forming enzyme in mammals, and differences in kinin levels are thought to be a contributing factor to diabetic nephropathy. Here, we determined the role of the kallikrein-kinin system in the pathogenesis of streptozotocin-induced diabetic nephropathy in wild-type and tissue kallikrein-knockout mice. All diabetic mice developed similar hyperglycemia, but the knockout mice had a significant two-fold increase in albuminuria compared to the wild-type mice before and after blood pressure elevation. Ezrin mRNA, a podocyte protein potentially implicated in albuminuria, was downregulated in the kidney of knockout mice. One month after induction of diabetes, the mRNAs of kininogen, tissue kallikrein, kinin B1, and B2 receptors were all increased up to two-fold in the kidney in both genotypes. Diabetes caused a 50% decrease in renal angiotensin-converting enzyme expression and a 20-fold increase in kidney injury molecule-1 reflecting tubular dysfunction, but there was no genotype difference. Our study found an early activation of the kallikrein-kinin system in the kidney and that this has a protective role against the development of diabetic nephropathy. The effect of tissue kallikrein deficiency on microalbuminuria in diabetic mice is similar to the effect of genetically high angiotensin-converting enzyme levels, suggesting that both observations, in part, result from a deficiency in kinins.


Assuntos
Albuminúria/etiologia , Nefropatias Diabéticas/complicações , Calicreínas Teciduais/fisiologia , Animais , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/complicações , Feminino , Rim/metabolismo , Camundongos , Camundongos Knockout , Estreptozocina , Calicreínas Teciduais/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA