Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(13): e2116470119, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35333648

RESUMO

Thermogenesis and adipogenesis are tightly regulated mechanisms that maintain lipid homeostasis and energy balance; dysfunction of these critical processes underpins obesity and contributes to cardiometabolic disease. C-type natriuretic peptide (CNP) fulfills a multimodal protective role in the cardiovascular system governing local blood flow, angiogenesis, cardiac function, and immune cell reactivity. Herein, we investigated a parallel, preservative function for CNP in coordinating metabolic homeostasis. Global inducible CNP knockout mice exhibited reduced body weight, higher temperature, lower adiposity, and greater energy expenditure in vivo. This thermogenic phenotype was associated with increased expression of uncoupling protein-1 and preferential lipid utilization by mitochondria, a switch corroborated by a corresponding diminution of insulin secretion and glucose clearance. Complementary studies in isolated murine and human adipocytes revealed that CNP exerts these metabolic regulatory actions by inhibiting sympathetic thermogenic programming via Gi-coupled natriuretic peptide receptor (NPR)-C and reducing peroxisome proliferator-activated receptor-γ coactivator-1α expression, while concomitantly driving adipogenesis via NPR-B/protein kinase-G. Finally, we identified an association between CNP/NPR-C expression and obesity in patient samples. These findings establish a pivotal physiological role for CNP as a metabolic switch to balance energy homeostasis. Pharmacological targeting of these receptors may offer therapeutic utility in the metabolic syndrome and related cardiovascular disorders.


Assuntos
Homeostase , Peptídeo Natriurético Tipo C , Termogênese , Animais , Fator Natriurético Atrial , Doenças Cardiovasculares/metabolismo , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Knockout , Peptídeo Natriurético Tipo C/genética , Peptídeo Natriurético Tipo C/fisiologia , Receptores do Fator Natriurético Atrial/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(31): E7428-E7437, 2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-30012589

RESUMO

Heart failure (HF) is a shared manifestation of several cardiovascular pathologies, including hypertension and myocardial infarction, and a limited repertoire of treatment modalities entails that the associated morbidity and mortality remain high. Impaired nitric oxide (NO)/guanylyl cyclase (GC)/cyclic guanosine-3',5'-monophosphate (cGMP) signaling, underpinned, in part, by up-regulation of cyclic nucleotide-hydrolyzing phosphodiesterase (PDE) isozymes, contributes to the pathogenesis of HF, and interventions targeted to enhancing cGMP have proven effective in preclinical models and patients. Numerous PDE isozymes coordinate the regulation of cardiac cGMP in the context of HF; PDE2 expression and activity are up-regulated in experimental and human HF, but a well-defined role for this isoform in pathogenesis has yet to be established, certainly in terms of cGMP signaling. Herein, using a selective pharmacological inhibitor of PDE2, BAY 60-7550, and transgenic mice lacking either NO-sensitive GC-1α (GC-1α-/-) or natriuretic peptide-responsive GC-A (GC-A-/-), we demonstrate that the blockade of PDE2 promotes cGMP signaling to offset the pathogenesis of experimental HF (induced by pressure overload or sympathetic hyperactivation), reversing the development of left ventricular hypertrophy, compromised contractility, and cardiac fibrosis. Moreover, we show that this beneficial pharmacodynamic profile is maintained in GC-A-/- mice but is absent in animals null for GC-1α or treated with a NO synthase inhibitor, revealing that PDE2 inhibition preferentially enhances NO/GC/cGMP signaling in the setting of HF to exert wide-ranging protection to preserve cardiac structure and function. These data substantiate the targeting of PDE2 in HF as a tangible approach to maximize myocardial cGMP signaling and enhancing therapy.


Assuntos
GMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/fisiologia , Guanilato Ciclase/fisiologia , Insuficiência Cardíaca/tratamento farmacológico , Óxido Nítrico/fisiologia , Inibidores de Fosfodiesterase/farmacologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , GMP Cíclico/análise , Masculino , Camundongos
3.
Eur Heart J ; 41(9): 1006-1020, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30903134

RESUMO

AIMS: C-type natriuretic peptide (CNP) is an essential endothelium-derived signalling species that governs vascular homoeostasis; CNP is also expressed in the heart but an intrinsic role for the peptide in cardiac function is not established. Herein, we employ unique transgenic strains with cell-specific deletion of CNP to define a central (patho)physiological capacity of CNP in maintaining heart morphology and contractility. METHODS AND RESULTS: Cardiac structure and function were explored in wild type (WT), cardiomyocyte (cmCNP-/-), endothelium (ecCNP-/-), and fibroblast (fbCNP-/-)-specific CNP knockout mice, and global natriuretic peptide receptor (NPR)-B-/-, and NPR-C-/- animals at baseline and in experimental models of myocardial infarction and heart failure (HF). Endothelium-specific deletion of CNP resulted in impaired coronary responsiveness to endothelium-dependent- and flow-mediated-dilatation; changes mirrored in NPR-C-/- mice. Ex vivo, global ischaemia resulted in larger infarcts and diminished functional recovery in cmCNP-/- and NPR-C-/-, but not ecCNP-/-, vs. WT. The cardiac phenotype of cmCNP-/-, fbCNP-/-, and NPR-C-/- (but not ecCNP-/- or NPR-B-/-) mice was more severe in pressure overload- and sympathetic hyperactivation-induced HF compared with WT; these adverse effects were rescued by pharmacological CNP administration in WT, but not NPR-C-/-, mice. At a molecular level, CNP/NPR-C signalling is impaired in human HF but attenuates activation of well-validated pro-hypertrophic and pro-fibrotic pathways. CONCLUSION: C-type natriuretic peptide of cardiomyocyte, endothelial and fibroblast origins co-ordinates and preserves cardiac structure, function, and coronary vasoreactivity via activation of NPR-C. Targeting NPR-C may prove an innovative approach to treating HF and ischaemic cardiovascular disorders.


Assuntos
Insuficiência Cardíaca , Peptídeo Natriurético Tipo C , Animais , Fator Natriurético Atrial , Camundongos , Camundongos Knockout , Miócitos Cardíacos , Peptídeo Natriurético Tipo C/genética , Transdução de Sinais
4.
Circulation ; 139(13): 1612-1628, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30586761

RESUMO

BACKGROUND: Angiogenesis and vascular remodeling are complementary, innate responses to ischemic cardiovascular events, including peripheral artery disease and myocardial infarction, which restore tissue blood supply and oxygenation; the endothelium plays a critical function in these intrinsic protective processes. C-type natriuretic peptide (CNP) is a fundamental endothelial signaling species that coordinates vascular homeostasis. Herein, we sought to delineate a central role for CNP in angiogenesis and vascular remodeling in response to ischemia. METHODS: The in vitro angiogenic capacity of CNP was examined in pulmonary microvascular endothelial cells and aortic rings isolated from wild-type, endothelium-specific CNP-/-, global natriuretic peptide receptor (NPR)-B-/- and NPR-C-/- animals, and human umbilical vein endothelial cells. These studies were complemented by in vivo investigation of neovascularization and vascular remodeling after ischemia or vessel injury, and CNP/NPR-C expression and localization in tissue from patients with peripheral artery disease. RESULTS: Clinical vascular ischemia is associated with reduced levels of CNP and its cognate NPR-C. Moreover, genetic or pharmacological inhibition of CNP and NPR-C, but not NPR-B, reduces the angiogenic potential of pulmonary microvascular endothelial cells, human umbilical vein endothelial cells, and isolated vessels ex vivo. Angiogenesis and remodeling are impaired in vivo in endothelium-specific CNP-/- and NPR-C-/-, but not NPR-B-/-, mice; the detrimental phenotype caused by genetic deletion of endothelial CNP, but not NPR-C, can be rescued by pharmacological administration of CNP. The proangiogenic effect of CNP/NPR-C is dependent on activation of Gi, ERK1/2, and phosphoinositide 3-kinase γ/Akt at a molecular level. CONCLUSIONS: These data define a central (patho)physiological role for CNP in angiogenesis and vascular remodeling in response to ischemia and provide the rationale for pharmacological activation of NPR-C as an innovative approach to treating peripheral artery disease and ischemic cardiovascular disorders.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Peptídeo Natriurético Tipo C/metabolismo , Neovascularização Fisiológica , Transdução de Sinais , Animais , Hipóxia Celular , Humanos , Camundongos , Camundongos Knockout , Peptídeo Natriurético Tipo C/genética , Remodelação Vascular
5.
FASEB J ; 33(2): 1578-1594, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30204499

RESUMO

Psoriasis is characterized by keratinocyte hyperproliferation, erythema, as well as a form of pruritus, involving cutaneous discomfort. There is evidence from both clinical and murine models of psoriasis that chemical or surgical depletion of small-diameter sensory nerves/nociceptors benefits the condition, but the mechanisms are unclear. Hence, we aimed to understand the involvement of sensory nerve mediators with a murine model of psoriasis and associated spontaneous behaviors, indicative of cutaneous discomfort. We have established an Aldara model of psoriasis in mice and chemically depleted the small-diameter nociceptors in a selective manner. The spontaneous behaviors, in addition to the erythema and skin pathology, were markedly improved. Attenuated inflammation was associated with reduced dermal macrophage influx and production of reactive oxygen/nitrogen species (peroxynitrite and protein nitrosylation). Subsequently, this directly influenced observed behavioral responses. However, the blockade of common sensory neurogenic mechanisms for transient receptor potential (TRP)V1, TRPA1, and neuropeptides (substance P and calcitonin gene-related peptide) using genetic and pharmacological approaches inhibited the behaviors but not the inflammation. Thus, a critical role of the established sensory TRP-neuropeptide pathway in influencing cutaneous discomfort is revealed, indicating the therapeutic potential of agents that block that pathway. The ongoing inflammation is mediated by a distinct sensory pathway involving macrophage activation.-Kodji, X., Arkless, K. L., Kee, Z., Cleary, S. J., Aubdool, A. A., Evans, E., Caton, P., Pitchford, S. C., Brain, S. D. Sensory nerves mediate spontaneous behaviors in addition to inflammation in a murine model of psoriasis.


Assuntos
Inflamação/patologia , Psoríase/patologia , Células Receptoras Sensoriais/patologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Denervação , Modelos Animais de Doenças , Diterpenos/farmacologia , Imiquimode/farmacologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Psoríase/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células Receptoras Sensoriais/metabolismo , Pele/irrigação sanguínea , Pele/patologia , Substância P/metabolismo , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/metabolismo
6.
Circulation ; 136(4): 367-383, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28446517

RESUMO

BACKGROUND: Research into the therapeutic potential of α-calcitonin gene-related peptide (α-CGRP) has been limited because of its peptide nature and short half-life. Here, we evaluate whether a novel potent and long-lasting (t½ ≥7 hours) acylated α-CGRP analogue (αAnalogue) could alleviate and reverse cardiovascular disease in 2 distinct murine models of hypertension and heart failure in vivo. METHODS: The ability of the αAnalogue to act selectively via the CGRP pathway was shown in skin by using a CGRP receptor antagonist. The effect of the αAnalogue on angiotensin II-induced hypertension was investigated over 14 days. Blood pressure was measured by radiotelemetry. The ability of the αAnalogue to modulate heart failure was studied in an abdominal aortic constriction model of murine cardiac hypertrophy and heart failure over 5 weeks. Extensive ex vivo analysis was performed via RNA analysis, Western blot, and histology. RESULTS: The angiotensin II-induced hypertension was attenuated by cotreatment with the αAnalogue (50 nmol·kg-1·d-1, SC, at a dose selected for lack of long-term hypotensive effects at baseline). The αAnalogue protected against vascular, renal, and cardiac dysfunction, characterized by reduced hypertrophy and biomarkers of fibrosis, remodeling, inflammation, and oxidative stress. In a separate study, the αAnalogue reversed angiotensin II-induced hypertension and associated vascular and cardiac damage. The αAnalogue was effective over 5 weeks in a murine model of cardiac hypertrophy and heart failure. It preserved heart function, assessed by echocardiography, while protecting against adverse cardiac remodeling and apoptosis. Moreover, treatment with the αAnalogue was well tolerated with neither signs of desensitization nor behavioral changes. CONCLUSIONS: These findings, in 2 distinct models, provide the first evidence for the therapeutic potential of a stabilized αAnalogue, by mediating (1) antihypertensive effects, (2) attenuating cardiac remodeling, and (3) increasing angiogenesis and cell survival to protect against and limit damage associated with the progression of cardiovascular diseases. This indicates the therapeutic potential of the CGRP pathway and the possibility that this injectable CGRP analogue may be effective in cardiac disease.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/análogos & derivados , Peptídeo Relacionado com Gene de Calcitonina/uso terapêutico , Cardiomegalia/tratamento farmacológico , Cardiotônicos/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Hipertensão/tratamento farmacológico , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiotônicos/farmacologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Hipertensão/metabolismo , Hipertensão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/patologia , Insuficiência de Múltiplos Órgãos/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
7.
Ann Rheum Dis ; 76(1): 252-260, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27165180

RESUMO

OBJECTIVE: Transient receptor potential canonical 5 (TRPC5) is functionally expressed on a range of cells including fibroblast-like synoviocytes, which play an important role in arthritis. A role for TRPC5 in inflammation has not been previously shown in vivo. We investigated the contribution of TRPC5 in arthritis. METHODS: Male wild-type and TRPC5 knockout (KO) mice were used in a complete Freund's adjuvant (CFA)-induced unilateral arthritis model, assessed over 14 days. Arthritis was determined by measurement of knee joint diameter, hindlimb weightbearing asymmetry and pain behaviour. Separate studies involved chronic pharmacological antagonism of TRPC5 channels. Synovium from human postmortem control and inflammatory arthritis samples were investigated for TRPC5 gene expression. RESULTS: At baseline, no differences were observed. CFA-induced arthritis resulted in increased synovitis in TRPC5 KO mice assessed by histology. Additionally, TRPC5 KO mice demonstrated reduced ispilateral weightbearing and nociceptive thresholds (thermal and mechanical) following CFA-induced arthritis. This was associated with increased mRNA expression of inflammatory mediators in the ipsilateral synovium and increased concentration of cytokines in synovial lavage fluid. Chronic treatment with ML204, a TRPC5 antagonist, augmented weightbearing asymmetry, secondary hyperalgesia and cytokine concentrations in the synovial lavage fluid. Synovia from human inflammatory arthritis demonstrated a reduction in TRPC5 mRNA expression. CONCLUSIONS: Genetic deletion or pharmacological blockade of TRPC5 results in an enhancement in joint inflammation and hyperalgesia. Our results suggest that activation of TRPC5 may be associated with an endogenous anti-inflammatory/analgesic pathway in inflammatory joint conditions.


Assuntos
Artrite Experimental/metabolismo , Hiperalgesia/metabolismo , Sinovite/metabolismo , Canais de Cátion TRPC/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Artrite Experimental/complicações , Artrite Experimental/patologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Comportamento Animal , Edema/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Hiperalgesia/etiologia , Hiperalgesia/patologia , Indóis/farmacologia , Masculino , Camundongos Knockout , Pessoa de Meia-Idade , Neovascularização Patológica/metabolismo , Osteoartrite/metabolismo , Osteoartrite/patologia , Piperidinas/farmacologia , RNA Mensageiro/genética , Membrana Sinovial/irrigação sanguínea , Sinovite/etiologia , Sinovite/patologia , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/biossíntese , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética
8.
FASEB J ; 29(10): 4285-98, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26136480

RESUMO

Transient receptor potential vanilloid 1 (TRPV1) is involved in sensory nerve nociceptive signaling. Recently, it has been discovered that TRPV1 receptors also regulate basal body temperature in multiple species from mice to humans. In the present study, we investigated whether TRPV1 modulates basal sympathetic nervous system (SNS) activity. C57BL6/J wild-type (WT) mice and TRPV1 knockout (KO) mice were implanted with radiotelemetry probes for measurement of core body temperature. AMG9810 (50 mg/kg) or vehicle (2% DMSO/5% Tween 80/10 ml/kg saline) was injected intraperitoneally. Adrenoceptor antagonists or vehicle (5 ml/kg saline) was injected subcutaneously. In WT mice, the TRPV1 antagonist, AMG9810, caused significant hyperthermia, associated with increased noradrenaline concentrations in brown adipose tissue. The hyperthermia was significantly attenuated by the ß-adrenoceptor antagonist propranolol, the mixed α-/ß-adrenoceptor antagonist labetalol, and the α1-adrenoceptor antagonist prazosin. TRPV1 KO mice have a normal basal body temperature, indicative of developmental compensation. d-Amphetamine (potent sympathomimetic) caused hyperthermia in WT mice, which was reduced in TRPV1 KO mice, suggesting a decreased sympathetic drive in KOs. This study provides new evidence that TRPV1 controls thermoregulation upstream of the SNS, providing a potential therapeutic target for sympathetic hyperactivity thermoregulatory disorders.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Temperatura Corporal/fisiologia , Sistema Nervoso Simpático/fisiologia , Canais de Cátion TRPV/fisiologia , Acrilamidas/administração & dosagem , Acrilamidas/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas Adrenérgicos beta/administração & dosagem , Antagonistas Adrenérgicos beta/farmacologia , Animais , Temperatura Corporal/genética , Regulação da Temperatura Corporal/genética , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Febre/genética , Febre/fisiopatologia , Humanos , Injeções Intraperitoneais , Injeções Subcutâneas , Labetalol/administração & dosagem , Labetalol/farmacologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prazosina/administração & dosagem , Prazosina/farmacologia , Propranolol/administração & dosagem , Propranolol/farmacologia , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos beta/fisiologia , Sistema Nervoso Simpático/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética , Telemetria/métodos
10.
FASEB J ; 27(4): 1664-73, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23271050

RESUMO

The underlying mechanisms of itch are poorly understood. We have investigated a model involving the chemoattractant leukotriene B4 (LTB4) that is up-regulated in common skin diseases. Intradermal injection of LTB4 (0.1 nmol/site) into female CD1 mice induced significant scratching movements (used as an itch index) compared with vehicle-injected (0.1% bovine serum albumin-saline) mice. Intraperitoneal transient receptor potential (TRP) channel antagonist treatment significantly inhibited itch as follows: TRP vanilloid 1 (TRPV1) antagonist SB366791 (0.5 mg/kg, by 97%) and the TRP ankyrin 1 (TRPA1) antagonists TCS 5861528 (10 mg/kg; 82%) and HC-030031 (100 mg/kg; 76%). Leukotriene B4 receptor 2 antagonism by LY255283 (5 mg/kg i.p.; 62%) reduced itch. Neither TRPV1-knockout (TRPV1-KO) nor TRPA1-knockout (TRPA1-KO mice exhibited LTB4-induced itch compared with their wild-type counterparts. The reactive oxygen species scavengers N-acetylcysteine (NAC; 204 mg/kg i.p.; 86%) or superoxide dismutase (SOD; 10 mg/kg i.p.; 83%) also inhibited itch. LTB4-induced superoxide release was attenuated by TCS 5861528 (56%) and HC-030031 (66%), NAC (58%), SOD (50%), and LY255283 (59%) but not by the leukotriene B4 receptor 1 antagonist U-75302 (9 nmol/site) or SB366791. Itch, superoxide, and myeloperoxidase generation were inhibited by the leukocyte migration inhibitor fucoidan (10 mg/kg i.v.) by 80, 61, and 34%, respectively. Myeloperoxidase activity was also reduced by SB366791 (35%) and SOD (28%). TRPV1-KO mice showed impaired myeloperoxidase release, whereas TRPA1-KO mice exhibited diminished production of superoxide. This result provides novel evidence that TRPA1 and TRPV1 contribute to itch via distinct mechanisms.


Assuntos
Leucócitos/metabolismo , Leucotrieno B4/farmacologia , Superóxidos/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Anquirinas/farmacologia , Feminino , Leucócitos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Prurido/tratamento farmacológico , Prurido/metabolismo , Receptores do Leucotrieno B4/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
11.
Biomolecules ; 14(4)2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38672459

RESUMO

TRPC5 is a non-selective cation channel that is expressed in cardiomyocytes, but there is a lack of knowledge of its (patho)physiological role in vivo. Here, we examine the role of TRPC5 on cardiac function under basal conditions and during cardiac hypertrophy. Cardiovascular parameters were assessed in wild-type (WT) and global TRPC5 knockout (KO) mice. Despite no difference in blood pressure or activity, heart rate was significantly reduced in TRPC5 KO mice. Echocardiography imaging revealed an increase in stroke volume, but cardiac contractility was unaffected. The reduced heart rate persisted in isolated TRPC5 KO hearts, suggesting changes in basal cardiac pacing. Heart rate was further investigated by evaluating the reflex change following drug-induced pressure changes. The reflex bradycardic response following phenylephrine was greater in TRPC5 KO mice but the tachycardic response to SNP was unchanged, indicating an enhancement in the parasympathetic control of the heart rate. Moreover, the reduction in heart rate to carbachol was greater in isolated TRPC5 KO hearts. To evaluate the role of TRPC5 in cardiac pathology, mice were subjected to abdominal aortic banding (AAB). An exaggerated cardiac hypertrophy response to AAB was observed in TRPC5 KO mice, with an increased expression of hypertrophy markers, fibrosis, reactive oxygen species, and angiogenesis. This study provides novel evidence for a direct effect of TRPC5 on cardiac function. We propose that (1) TRPC5 is required for maintaining heart rate by regulating basal cardiac pacing and in response to pressure lowering, and (2) TRPC5 protects against pathological cardiac hypertrophy.


Assuntos
Cardiomegalia , Frequência Cardíaca , Camundongos Knockout , Canais de Cátion TRPC , Animais , Canais de Cátion TRPC/metabolismo , Canais de Cátion TRPC/genética , Cardiomegalia/metabolismo , Camundongos , Masculino , Miócitos Cardíacos/metabolismo , Camundongos Endogâmicos C57BL , Pressão Sanguínea
12.
Br J Pharmacol ; 179(11): 2443-2459, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34131904

RESUMO

BACKGROUND AND PURPOSE: cGMP underpins the bioactivity of NO and natriuretic peptides and is key to cardiovascular homeostasis. cGMP-driven responses are terminated primarily by PDEs, but cellular efflux via multidrug resistance proteins (MRPs) might contribute. Herein, the effect of pharmacological blockade of MRPs on cGMP signalling in the heart and vasculature was investigated in vitro and in vivo. EXPERIMENTAL APPROACH: Proliferation of human coronary artery smooth muscle cells (hCASMCs), vasorelaxation of murine aorta and reductions in mean arterial BP (MABP) in response to NO donors or natriuretic peptides were determined in the absence and presence of the MRP inhibitor MK571. The ability of MRP inhibition to reverse morphological and contractile deficits in a murine model of pressure overload-induced heart failure was also explored. KEY RESULTS: MK571 attenuated hCASMC growth and enhanced the anti-proliferative effects of NO and atrial natriuretic peptide (ANP). MRP blockade caused concentration-dependent relaxations of murine aorta and augmented responses to ANP (and to a lesser extent NO). MK571 did not decrease MABP per se but enhanced the hypotensive actions of ANP and improved structural and functional indices of disease severity in experimental heart failure. These beneficial actions of MRP inhibition were associated with a greater intracellular:extracellular cGMP ratio in vitro and in vivo. CONCLUSIONS AND IMPLICATIONS: MRP blockade promotes the cardiovascular functions of natriuretic peptides in vitro and in vivo, with more modest effects on NO. MRP inhibition may have therapeutic utility in cardiovascular diseases triggered by dysfunctional cGMP signalling, particularly those associated with altered natriuretic peptide bioactivity. LINKED ARTICLES: This article is part of a themed issue on cGMP Signalling in Cell Growth and Survival. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.11/issuetoc.


Assuntos
Fator Natriurético Atrial , Insuficiência Cardíaca , Subfamília B de Transportador de Cassetes de Ligação de ATP , Animais , Fator Natriurético Atrial/metabolismo , Fator Natriurético Atrial/farmacologia , GMP Cíclico/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Camundongos , Peptídeos Natriuréticos/metabolismo , Vasodilatadores
13.
J Pharmacol Exp Ther ; 337(1): 117-24, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21205926

RESUMO

This study explores the in vivo effects of the proposed transient receptor potential ankyrin 1 (TRPA1) agonist 4-oxo-2-nonenal (4-ONE). Pharmacological inhibitors and genetically modified mice were used to investigate the ability of 4-ONE to act via TRPA1 receptors and possible mechanisms involving transient receptor potential vanilloid 1 (TRPV1). We hypothesized that 4-ONE activates sensory nerves, via TRPA1 or possibly TRPV1, and thus triggers mechanical hyperalgesia, edema formation, and vasodilatation in mice. An automated dynamic plantar aesthesiometer was used to determine hind paw withdrawal thresholds, and a laser Doppler flowmeter was used to measure skin blood flow. Edema formation was determined by measuring paw weights and thickness. 4-ONE (10 nmol) triggers unilateral mechanical hyperalgesia, edema formation, and vasodilatation in mice and is shown here to exhibit TRPA1-dependent and -independent effects. Neurogenic vasodilatation and mechanical hyperalgesia at 0.5 h postinjection were significantly greater in TRPA1 wild-type (WT) mice compared with TRPA1 knockout (KO) mice. Edema formation throughout the time course as well as mechanical hyperalgesia from 1 to 4 h postinjection were similar in WT and TRPA1 KO mice. Studies involving TRPV1 KO mice revealed no evidence of TRPV1 involvement or interactions between TRPA1 and TRPV1 in mediating the in vivo effects of 4-ONE. Previously, 4-ONE was shown to be a potent TRPA1 agonist in vitro. We demonstrate its ability to mediate vasodilatation and certain nociceptive effects in vivo. These data indicate the potential of TRPA1 as an oxidant sensor for vasodilator responses in vivo. However, 4-ONE also triggers TRPA1-independent effects that relate to edema formation and pain.


Assuntos
Aldeídos/farmacologia , Medição da Dor/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , Aldeídos/toxicidade , Animais , Feminino , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxidantes/agonistas , Oxidantes/fisiologia , Medição da Dor/métodos , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/deficiência , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/fisiologia
14.
J Investig Dermatol Symp Proc ; 15(1): 33-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22076325

RESUMO

Neurogenic inflammation is involved in skin inflammation. It is hypothesized that it is involved in the pathogenesis of the common chronic cutaneous vascular disorder rosacea, but the exact mechanism of action is currently unknown. Transient receptor potential vanilloid 1 (TRPV1) and ankyrin 1 (TRPA1) are widely expressed on primary sensory neuron endings and non-neuronal cells such as keratinocytes. Here we describe the potential for TRPV1 and TRPA1 receptors to be involved in the pathophysiology of rosacea due to their polymodal activation, including cold and hot temperature, pungent products from vegetable and spices, reactive oxygen species, and mechanical stimuli. We discuss the role of both receptors and the sensory neuropeptides that they release in inflammation and pain sensation and evidence suggesting that both TRPV1 and TRPA1 receptors may be promising therapeutic targets for the treatment of the inflammatory symptoms of rosacea.


Assuntos
Dermatite/fisiopatologia , Inflamação Neurogênica/fisiopatologia , Rosácea/fisiopatologia , Pele/irrigação sanguínea , Pele/inervação , Animais , Anquirinas/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Cães , Feminino , Haplorrinos , Humanos , Masculino , Camundongos , Dor/fisiopatologia , Peptídeo Hidrolases/fisiologia , Ratos , Fatores Sexuais , Substância P/fisiologia , Canais de Cátion TRPV/fisiologia
15.
Bioorg Med Chem Lett ; 21(16): 4857-9, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21741838

RESUMO

TrpA1 is an ion channel involved in nociceptive and inflammatory pain. It is implicated in the detection of chemical irritants through covalent binding to a cysteine-rich intracellular region of the protein. While performing an HTS of the Pfizer chemical collection, a class of pyrimidines emerged as a non-reactive, non-covalently binding family of agonists of the rat and human TrpA1 channel. Given the issues identified with the reference agonist Mustard Oil (MO) in screening, a new, non-covalently binding agonist was optimized and proved to be a superior agent to MO for screening purposes. Compound 16a (PF-4840154) is a potent, selective agonist of the rat and human TrpA1 channel and elicited TrpA1-mediated nocifensive behaviour in mouse.


Assuntos
Anquirinas/agonistas , Desenho de Fármacos , Proteínas do Tecido Nervoso/agonistas , Piperazinas/farmacologia , Pirimidinas/farmacologia , Canais de Potencial de Receptor Transitório/agonistas , Animais , Canais de Cálcio , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Edema/tratamento farmacológico , Edema/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Estrutura Molecular , Dor/tratamento farmacológico , Dor/fisiopatologia , Piperazinas/síntese química , Piperazinas/química , Pirimidinas/síntese química , Pirimidinas/química , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Canal de Cátion TRPA1 , Canais de Cátion TRPC
16.
Hypertension ; 71(6): 1091-1100, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29686000

RESUMO

Voltage-gated Kv7.4 channels have been implicated in vascular smooth muscle cells' activity because they modulate basal arterial contractility, mediate responses to endogenous vasorelaxants, and are downregulated in several arterial beds in different models of hypertension. Angiotensin II (Ang II) is a key player in hypertension that affects the expression of several classes of ion channels. In this study, we evaluated the effects of Ang II on the expression and function of vascular Kv7.4. Western blot and quantitative polymerase chain reaction revealed that in whole rat mesenteric artery, Ang II incubation for 1 to 7 hours decreased Kv7.4 protein expression without reducing transcript levels. Moreover, Ang II decreased XE991 (Kv7)-sensitive currents and attenuated membrane potential hyperpolarization and relaxation induced by the Kv7 activator ML213. Ang II also reduced Kv7.4 staining at the plasma membrane of vascular smooth muscle cells. Proteasome inhibition with MG132 prevented Ang II-induced decrease of Kv7.4 levels and counteracted the functional impairment of ML213-induced relaxation in myography experiments. Proximity ligation assays showed that Ang II impaired the interaction of Kv7.4 with the molecular chaperone HSP90 (heat shock protein 90), enhanced the interaction of Kv7.4 with the E3 ubiquitin ligase CHIP (C terminus of Hsp70-interacting protein), and increased Kv7.4 ubiquitination. Similar alterations were found in mesenteric vascular smooth muscle cells isolated from Ang II-infused mice. The effect of Ang II was emulated by 17-AAG (17-demethoxy-17-(2-propenylamino) geldanamycin) that inhibits HSP90 interactions with client proteins. These results show that Ang II downregulates Kv7.4 by altering protein stability through a decrease of its interaction with HSP90. This leads to the recruitment of CHIP and Kv7.4 ubiquitination and degradation via the proteasome.


Assuntos
Angiotensina II/farmacologia , Regulação para Baixo , Proteínas de Choque Térmico HSP90/metabolismo , Hipertensão/genética , Canais de Potássio KCNQ/genética , Músculo Liso Vascular/metabolismo , Vasodilatação/fisiologia , Animais , Western Blotting , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Canais de Potássio KCNQ/biossíntese , Masculino , Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Estresse Oxidativo , Ratos , Ratos Wistar
17.
J Am Heart Assoc ; 6(6)2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28655735

RESUMO

BACKGROUND: Reliable measurement of blood pressure in conscious mice is essential in cardiovascular research. Telemetry, the "gold-standard" technique, is invasive and expensive and therefore tail-cuff, a noninvasive alternative, is widely used. However, tail-cuff requires handling and restraint during measurement, which may cause stress affecting blood pressure and undermining reliability of the results. METHODS AND RESULTS: C57Bl/6J mice were implanted with radio-telemetry probes to investigate the effects of the steps of the tail-cuff technique on central blood pressure, heart rate, and temperature. This included comparison of handling techniques, operator's sex, habituation, and influence of hypertension induced by angiotensin II. Direct comparison of measurements obtained by telemetry and tail-cuff were made in the same mouse. The results revealed significant increases in central blood pressure, heart rate, and core body temperature from baseline following handling interventions without significant difference among the different handling technique, habituation, or sex of the investigator. Restraint induced the largest and sustained increase in cardiovascular parameters and temperature. The tail-cuff readings significantly underestimated those from simultaneous telemetry recordings; however, "nonsimultaneous" telemetry, obtained in undisturbed mice, were similar to tail-cuff readings obtained in undisturbed mice on the same day. CONCLUSIONS: This study reveals that the tail-cuff technique underestimates the core blood pressure changes that occur simultaneously during the restraint and measurement phases. However, the measurements between the 2 techniques are similar when tail-cuff readings are compared with telemetry readings in the nondisturbed mice. The differences between the simultaneous recordings by the 2 techniques should be recognized by researchers.


Assuntos
Determinação da Pressão Arterial/instrumentação , Pressão Sanguínea , Hipertensão/diagnóstico , Cauda/irrigação sanguínea , Telemetria , Angiotensina II , Animais , Comportamento Animal , Determinação da Pressão Arterial/efeitos adversos , Determinação da Pressão Arterial/métodos , Regulação da Temperatura Corporal , Modelos Animais de Doenças , Feminino , Habituação Psicofisiológica , Manobra Psicológica , Frequência Cardíaca , Humanos , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Hipertensão/psicologia , Masculino , Camundongos Endogâmicos C57BL , Pletismografia/instrumentação , Valor Preditivo dos Testes , Fluxo Sanguíneo Regional , Reprodutibilidade dos Testes , Restrição Física/efeitos adversos , Estresse Psicológico/etiologia , Estresse Psicológico/fisiopatologia , Fatores de Tempo
18.
Sci Rep ; 6(1): 7, 2016 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-28442712

RESUMO

Cu(II)ATSM was developed as a hypoxia sensitive positron emission tomography agent. Recent reports have highlighted the neuroprotective properties of Cu(II)ATSM, yet there are no reports that it confers cardioprotection. We demonstrate that Cu(II)ATSM activates the redox-sensitive transcription factor Nrf2 in human coronary artery smooth muscle cells (HCASMC) and cardiac myocytes (HCM), leading to upregulation of antioxidant defense enzymes. Oral delivery of Cu(II)ATSM in mice induced expression of the Nrf2-regulated enzymes in the heart and aorta. In HCASMC, Cu(II)ATSM increased expression of the Nrf2 stabilizer DJ-1, and knockdown of Nrf2 or DJ-1 attenuated Cu(II)ATSM-mediated heme oxygenase-1 and NADPH quinone oxidoreductase-1 induction. Pre-treatment of HCASMC with Cu(II)ATSM protected against the pro-oxidant effects of angiotensin II (Ang II) by attenuating superoxide generation, apoptosis, proliferation and increases in intracellular calcium. Notably, Cu(II)ATSM-mediated protection against Ang II-induced HCASMC apoptosis was diminished by Nrf2 knockdown. Acute treatment with Cu(II)ATSM enhanced the association of DJ-1 with superoxide dismutase-1 (SOD1), paralleled by significant increases in intracellular Cu(II) levels and SOD1 activity. We describe a novel mechanism by which Cu(II)ATSM induces Nrf2-regulated antioxidant enzymes and protects against Ang II-mediated HCASMC dysfunction via activation of the Nrf2/DJ-1 axis. Cu(II)ATSM may provide a therapeutic strategy for cardioprotection via upregulation of antioxidant defenses.


Assuntos
Cardiotônicos , Miócitos Cardíacos , Miócitos de Músculo Liso , Fator 2 Relacionado a NF-E2 , Compostos Organometálicos , Proteína Desglicase DJ-1 , Tiossemicarbazonas , Animais , Humanos , Masculino , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação , Vasos Coronários/citologia , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Compostos Organometálicos/farmacologia , Proteína Desglicase DJ-1/metabolismo , Transdução de Sinais , Tiossemicarbazonas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo
19.
Br J Pharmacol ; 173(15): 2419-33, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27189253

RESUMO

BACKGROUND AND PURPOSE: Transient receptor potential ankyrin-1 (TRPA1) activation is known to mediate neurogenic vasodilatation. We investigated the mechanisms involved in TRPA1-mediated peripheral vasodilatation in vivo using the TRPA1 agonist cinnamaldehyde. EXPERIMENTAL APPROACH: Changes in vascular ear blood flow were measured in anaesthetized mice using laser Doppler flowmetry. KEY RESULTS: Topical application of cinnamaldehyde to the mouse ear caused a significant increase in blood flow in the skin of anaesthetized wild-type (WT) mice but not in TRPA1 knockout (KO) mice. Cinnamaldehyde-induced vasodilatation was inhibited by the pharmacological blockade of the potent microvascular vasodilator neuropeptide CGRP and neuronal NOS-derived NO pathways. Cinnamaldehyde-mediated vasodilatation was significantly reduced by treatment with reactive oxygen nitrogen species (RONS) scavenger such as catalase and the SOD mimetic TEMPOL, supporting a role of RONS in the downstream vasodilator TRPA1-mediated response. Co-treatment with a non-selective NOS inhibitor L-NAME and antioxidant apocynin further inhibited the TRPA1-mediated vasodilatation. Cinnamaldehyde treatment induced the generation of peroxynitrite that was blocked by the peroxynitrite scavenger FeTPPS and shown to be dependent on TRPA1, as reflected by an increase in protein tyrosine nitration in the skin of WT, but not in TRPA1 KO mice. CONCLUSION AND IMPLICATIONS: This study provides in vivo evidence that TRPA1-induced vasodilatation mediated by cinnamaldehyde requires neuronal NOS-derived NO, in addition to the traditional neuropeptide component. A novel role of peroxynitrite is revealed, which is generated downstream of TRPA1 activation by cinnamaldehyde. This mechanistic pathway underlying TRPA1-mediated vasodilatation may be important in understanding the role of TRPA1 in pathophysiological situations.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Neurogênese , Óxidos de Nitrogênio/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Vasodilatação , Acroleína/análogos & derivados , Acroleína/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/efeitos dos fármacos , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/deficiência , Vasodilatação/efeitos dos fármacos
20.
J Mol Neurosci ; 60(2): 157-70, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27525636

RESUMO

It is supposed that TRPA1 receptor can be activated by hydrogen sulphide (H2S). Here, we have investigated the role of TRPA1 receptor in H2S-induced [Ca(2+)]i increase in trigeminal ganglia (TRG) neurons, and the involvement of capsaicin-sensitive sensory nerves in H2S-evoked cutaneous vasodilatation. [Ca(2+)]i was measured with ratiometric technique on TRG neurons of TRPA1(+/+) and TRPA1(-/-) mice after NaHS, Na2S, allylisothiocyanate (AITC) or KCl treatment. Microcirculatory changes in the ear were detected by laser Doppler imaging in response to topical NaHS, AITC, NaOH, NaSO3 or NaCl. Mice were either treated with resiniferatoxin (RTX), or CGRP antagonist BIBN4096, or NK1 receptor antagonist CP99994, or K(+) ATP channel blocker glibenclamide. Alpha-CGRP(-/-) and NK1 (-/-) mice were also investigated. NaHS and Na2S increased [Ca(2+)]i in TRG neurons derived from TRPA(+/+) but not from TRPA1(-/-) mice. NaHS increased cutaneous blood flow, while NaOH, NaSO3 and NaCl did not cause significant changes. NaHS-induced vasodilatation was reduced in RTX-treated animals, as well as by pre-treatment with BIBN4096 or CP99994 alone or in combination. NaHS-induced vasodilatation was significantly smaller in alpha-CGRP(-/-) or NK1 (-/-) mice compared to wild-types. H2S activates capsaicin-sensitive sensory nerves through TRPA1 receptors and the resultant vasodilatation is mediated by the release of vasoactive sensory neuropeptides CGRP and substance P.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Sulfeto de Hidrogênio/farmacologia , Células Receptoras Sensoriais/metabolismo , Substância P/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Vasodilatação , Animais , Capsaicina/farmacologia , Linhagem Celular , Células Cultivadas , Dipeptídeos/farmacologia , Diterpenos/farmacologia , Feminino , Glibureto/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Cloreto de Potássio/farmacologia , Quinazolinas/farmacologia , Receptores da Neurocinina-1/genética , Receptores da Neurocinina-1/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/genética , Gânglio Trigeminal/citologia , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA