Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 809: 130-140, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28483457

RESUMO

Peroxisome-proliferator activated receptors (PPAR) are members of the nuclear hormone receptor superfamily which regulate gene transcription. PPARα is a key regulator of lipid homeostasis and a negative regulator of inflammation. Under conditions of metabolic stress such as fasting or glucose deprivation, PPARα is upregulated in order to control gene expression necessary for processing alternate fuel sources (e.g. fatty acid oxidation) and thereby promote maintenance of cell viability. Clinically, PPARα expression is upregulated in diseased tissues such as melanoma, chronic lymphocytic leukemia, ovarian and prostate cancer. This may allow for cellular proliferation and metastasis. Importantly, genetic knockouts of PPARα have been shown to be protected against tumor growth in a variety of syngeneic tumors models. We hypothesized that a potent and selective PPARα antagonist could represent a novel cancer therapy. Early in our discovery research, we identified NXT629 (Bravo et al., 2014). Herein we describe the pharmacology of NXT629 and demonstrate that it is a potent and selective PPARα antagonist. We identify NXT629 as a valuable tool for use in in vivo assessment of PPARα due to its good systemic exposure following intraperitoneal injection. We explore the in vivo pharmacology of NXT629 and demonstrate that it is efficacious in pharmacodynamic models that are driven by PPARα. Finally, we probe the efficacy of NXT629 in disease models where PPARα knockouts have shown to be protected. We believe that PPARα antagonists will be beneficial in diseases such as ovarian cancer and melanoma where PPARα and fatty acid oxidation may be involved.


Assuntos
Aminopiridinas/farmacologia , PPAR alfa/antagonistas & inibidores , Sulfonamidas/farmacologia , Aminopiridinas/farmacocinética , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/sangue , Humanos , Camundongos , Metástase Neoplásica , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Sulfonamidas/farmacocinética
2.
Mol Cell Neurosci ; 68: 177-85, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26092702

RESUMO

The goal of this study was to define profiles of secreted neuropeptide and catecholamine neurotransmitters that undergo co-release from sympathoadrenal chromaffin cells upon stimulation by distinct secretagogues. Chromaffin cells of the adrenal medulla participate in the dynamic responses to stress, especially that of 'fight and flight', and, thus, analyses of the co-release of multiple neurotransmitters is necessary to gain knowledge of how the stress response regulates cell-cell communication among physiological systems. Results of this study demonstrated that six different secretagogues stimulated the co-release of the neuropeptides Met-enkephalin, galanin, NPY, and VIP with the catecholamines dopamine, norepinephrine, and epinephrine. Importantly, the quantitative profiles of the secreted neurotransmitters showed similarities and differences upon stimulation by the different secretagogues evaluated, composed of KCl depolarization, nicotine, carbachol, PACAP, bradykinin, and histamine. The rank-orders of the secreted profiles of the neurotransmitters were generally similar among these secretagogues, but differences in the secreted amounts of each neurotransmitter occurred with different secretagogues. Epinephrine among the catecholamines showed the highest level of secretion. (Met)enkephalin showed the largest levels of secretion compared to the other neuropeptides examined. Levels of secreted catecholamines were greater than that of the neuropeptides. These data support the hypothesis that profiles of secreted neuropeptide and catecholamine neurotransmitters show similarities and differences upon stimulation by distinct secretagogues. These results illustrate the co-release of concerted neurotransmitter profiles that participate in the stress response of the sympathoadrenal nervous system.


Assuntos
Catecolaminas/metabolismo , Células Cromafins/metabolismo , Neuropeptídeos/metabolismo , Medula Suprarrenal/citologia , Análise de Variância , Animais , Bradicinina/farmacologia , Carbacol/farmacologia , Bovinos , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Células Cromafins/efeitos dos fármacos , Histamina/farmacologia , Agonistas dos Receptores Histamínicos/farmacologia , Neurotransmissores/farmacologia , Nicotina/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Cloreto de Potássio/farmacologia , Vasodilatadores/farmacologia
3.
Bioorg Med Chem Lett ; 24(10): 2267-72, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24745969

RESUMO

The discovery and SAR of a novel series of potent and selective PPARα antagonists are herein described. Exploration of replacements for the labile acyl sulfonamide linker led to a biaryl sulfonamide series of which compound 33 proved to be suitable for further profiling in vivo. Compound 33 demonstrated excellent potency, selectivity against other nuclear hormone receptors, and good pharmacokinetics in mouse.


Assuntos
PPAR alfa/antagonistas & inibidores , Sulfonamidas/química , Sulfonamidas/farmacologia , Animais , Butiratos/química , Butiratos/farmacologia , Humanos , Camundongos , Estrutura Molecular , Oxazóis/química , Oxazóis/farmacologia , Compostos de Fenilureia/química , Compostos de Fenilureia/farmacologia , Propionatos/química , Propionatos/farmacologia , Relação Estrutura-Atividade , Triazóis/química , Triazóis/farmacologia , Tirosina/análogos & derivados , Tirosina/química , Tirosina/farmacologia
4.
J Biol Chem ; 285(17): 12504-11, 2010 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20157112

RESUMO

The intestinal epithelium is dynamic, with proliferation of undifferentiated crypt cells balanced by terminal differentiation and cell death at the colon surface or small intestinal villus tips. Cyclic AMP, induced by agonists such as prostaglandin E(2) and vasoactive intestinal polypeptide, promotes proliferation and ion secretion and suppresses apoptosis in intestinal epithelial cells. Here, we show that cell differentiation in a model intestinal epithelium leads to attenuation of cAMP production in response to G protein-coupled receptor and receptor-independent agonists. Concomitantly, key components of the cAMP cascade, the alpha subunit of the stimulatory G protein, G(s), and adenylyl cyclase (AC) isoforms 3, 4, 6, and 7 are down-regulated. By contrast, AC1, AC2, AC8, and AC9, and the receptors for prostaglandin E(2) and vasoactive intestinal polypeptide, are not expressed or not affected by differentiation. We confirmed key findings in normal murine colon epithelium, in which the major AC isoforms and G(s)alpha are markedly down-regulated in differentiated surface cells. Suppression of AC isoforms and G(s)alpha is functionally important, because their constitutive expression completely reverses differentiation-induced cAMP attenuation. Thus, down-regulation of AC isoforms and G(s)alpha is an integral part of the intestinal epithelial differentiation program, perhaps serving to release cells from cAMP-promoted anti-apoptosis as a prerequisite for cell death upon terminal differentiation.


Assuntos
Adenilil Ciclases/biossíntese , Diferenciação Celular/fisiologia , Colo/enzimologia , Células Epiteliais/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Mucosa Intestinal/enzimologia , Animais , Linhagem Celular , Dinoprostona/metabolismo , Regulação para Baixo/fisiologia , Humanos , Isoenzimas/biossíntese
5.
Neuroendocrinology ; 89(2): 210-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18840998

RESUMO

BACKGROUND/AIMS: The endopin serpin protease inhibitors have been identified by molecular studies as components of secretory vesicles that produce neuropeptides. Endopin 1 inhibits trypsin-like serine proteases, and endopin 2 inhibits cathepsin L that produces neuropeptides in secretory vesicles. To assess the secretory vesicle and neuroendocrine tissue distribution of these endopins, the goal of this study was to define specific antisera for each endopin isoform and to examine their localization with neuropeptides and in neuroendocrine tissues. METHODS: This study utilized methods consisting of Western blots, immunoelectron microscopy, and immunofluorescence microscopy for evaluation of the localization of endopin protease inhibitors in neuroendocrine tissues. RESULTS: Immunoelectron microscopy with these selective antisera demonstrated the localization of endopins 1 and 2 within secretory vesicles of adrenal medulla (bovine). Cellular immunofluorescence confocal microscopy illustrated the high level of colocalization of endopins 1 and 2 with enkephalin and NPY neuropeptides that are present in secretory vesicles of adrenal medullary chromaffin cells in primary culture. Tissue distribution studies (by Western blots) showed the expression of endopins 1 and 2 in bovine brain, pituitary, adrenal medulla, and other neuroendocrine tissues. CONCLUSIONS: These results implicate endopins 1 and 2 as endogenous protease inhibitors in neuropeptide-containing secretory vesicles and neuroendocrine tissues.


Assuntos
Medula Suprarrenal/química , Células Neuroendócrinas/química , Neuropeptídeos/análise , Sistemas Neurossecretores/química , Inibidores de Proteases/análise , Vesículas Secretórias/química , Serpinas/análise , Medula Suprarrenal/ultraestrutura , Animais , Bovinos , Células Neuroendócrinas/ultraestrutura , Vesículas Secretórias/ultraestrutura , Distribuição Tecidual
6.
Am J Physiol Renal Physiol ; 294(3): F638-44, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18199602

RESUMO

Activation of adenosine A(1) receptors (A(1)R) can inhibit arginine vasopressin (AVP)-induced cAMP formation in isolated cortical and medullary collecting ducts. To assess the in vivo consequences of the absence of A(1)R, we performed experiments in mice lacking A(1)R (A(1)R(-/-)). We assessed the effects of the vasopressin V(2) receptor (V(2)R) agonist 1-desamino-8-d-arginine vasopressin (dDAVP) on cAMP formation in isolated inner medullary collecting ducts (IMCD) and on water excretion in conscious water-loaded mice. dDAVP-induced cAMP formation in isolated IMCD was significantly greater ( approximately 2-fold) in A(1)R(-/-) compared with wild-type mice (WT) and, in contrast to WT, was not inhibited by the A(1)R agonist N6-cyclohexyladenosine. A(1)R(-/-) and WT had similar basal urinary excretion of vasopressin, expression of aquaporin-2 protein in renal cortex and medulla, and acute increases in urinary flow rate and electrolyte-free water clearance in response to the V(2)R antagonist SR121463 or acute water loading; the latter increased inner medullary A(1)R expression in WT. Dose dependence of dDAVP-induced antidiuresis after acute water loading was not different between the genotypes. However, A(1)R(-/-) had greater inner medullary expression of cyclooxygenase-1 under basal conditions and of the P2Y(2) and EP(3) receptor in response to water loading compared with WT mice. Thus vasopressin-induced cAMP formation is enhanced in isolated IMCD of mice lacking A(1)R, but the adenosine-A(1)R/V(2)R interaction demonstrated in vitro is likely compensated in vivo by multiple mechanisms, a number of which can be "uncovered" by water loading.


Assuntos
Arginina Vasopressina/metabolismo , Túbulos Renais Coletores/metabolismo , Receptor A1 de Adenosina/metabolismo , Água/metabolismo , Animais , AMP Cíclico/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Desamino Arginina Vasopressina/farmacologia , Endotelina-1/metabolismo , Técnicas In Vitro , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/metabolismo , Reação em Cadeia da Polimerase , Receptor A1 de Adenosina/genética , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP3 , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y2 , Receptores de Vasopressinas/agonistas
7.
FASEB J ; 21(13): 3717-26, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17575258

RESUMO

Extracellular nucleotides (e.g., ATP) regulate many physiological and pathophysiological processes through activation of nucleotide (P2) receptors in the plasma membrane. Here we report that gene-targeted (knockout) mice that lack P2Y2 receptors have salt-resistant arterial hypertension in association with an inverse relationship between salt intake and heart rate, indicating intact baroreceptor function. Knockout mice have multiple alterations in their handling of salt and water: these include suppressed plasma renin and aldosterone concentrations, lower renal expression of the aldosterone-induced epithelial sodium channel alpha-ENaC, greater medullary expression of the Na-K-2Cl-cotransporter NKCC2, and greater furosemide-sensitive Na+ reabsorption in association with greater renal medullary expression of aquaporin-2 and vasopressin-dependent renal cAMP formation and water reabsorption despite similar vasopressin levels compared with wild type. Of note, smaller increases in plasma aldosterone were required to adapt renal Na+ excretion to restricted intake in knockout mice, suggesting a facilitation in renal Na+ retention. The results thus identify a previously unrecognized role for P2Y2 receptors in blood pressure regulation that is linked to an inhibitory influence on renal Na+ and water reabsorption. Based on these findings in knockout mice, we propose that a blunting in P2Y2 receptor expression or activity is a new mechanism for salt-resistant arterial hypertension.


Assuntos
Hipertensão/genética , Rim/fisiopatologia , Receptores Purinérgicos P2/fisiologia , Sódio/metabolismo , Água/metabolismo , Animais , Hipertensão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y2
8.
Circulation ; 115(17): 2282-91, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17438153

RESUMO

BACKGROUND: Endothelial dysfunction predisposes to vascular injury in association with hypertension. Endothelin (ET-1) is a potent vasoactive peptide that is synthesized and released by the vascular endothelium and is a marker of endothelial function. Chromogranin A (CHGA) regulates the storage and release of catecholamines and may have direct actions on the microvasculature. CHGA, a candidate gene for intermediate phenotypes that contribute to hypertension, shows a pattern of single nucleotide polymorphism variations that alter the expression and function of this gene both in vivo and in vitro. METHODS AND RESULTS: In a study of twins (n=238 pairs), plasma ET-1 was 58+/-5% (P<0.0001) heritable. Plasma ET-1 was both correlated and associated with chromogranin fragment levels, and the 2 were influenced by shared genetic determination (pleiotropy [rhoG]; for the CHGA precursor, rhoG=0.318+/-0.105; P=0.0032). We therefore hypothesized that variation in the CHGA gene may influence ET-1 secretion. Carriers of the CHGA promoter -988G, -462A, and -89A minor alleles showed significantly higher mean plasma ET-1 than their major allele homozygote counterparts (P=0.02, P=0.006, P=0.03, respectively). Analysis of a linkage disequilibrium block that spans these 3 single nucleotide polymorphisms showed a significant association between the GATACA haplotype and plasma ET-1 (P=0.0075). In cultured human umbilical vein endothelial cells, CHGA caused dose-dependent secretion of ET-1 over a brief (<1 hour) time course at relatively low concentrations of CHGA (10 to 100 nmol/L) with a threshold concentration (10 nmol/L) in the range found circulating in humans in vivo. CONCLUSIONS: These results suggest that common, heritable variation in expression of the human CHGA gene influences endothelial ET-1 secretion in vivo, explained by a CHGA stimulus/ET-1 secretion coupling in endothelial cells in vitro. The findings document a previously unsuspected interaction between the sympathochromaffin system and the endothelium and suggest novel genetic and cell biological approaches to the prediction, diagnosis, and mechanism of endothelial dysfunction in human disease.


Assuntos
Cromogranina A/genética , Endotelina-1/metabolismo , Endotélio Vascular/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Adulto , Cromogranina A/sangue , Endotelina-1/sangue , Feminino , Variação Genética , Haplótipos , Humanos , Masculino , Fenótipo , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Fatores Sexuais , Sistema Nervoso Simpático/fisiologia
10.
J Biol Chem ; 281(42): 31317-25, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16916802

RESUMO

In C11-MDCK cells, which resemble intercalated cells from collecting ducts of the canine kidney, P2Y agonists promote transient activation of the Na+,K+,Cl- cotransporter (NKCC), followed by its sustained inhibition. We designed this study to identify P2Y receptor subtypes involved in dual regulation of this carrier. Real time polymerase chain reaction analysis demonstrated that C11-MDCK cells express abundant P2Y1 and P2Y2 mRNA compared with that of other P2Y receptor subtypes. The rank order of potency of agents (ATP approximately UTP >> 2-(methylthio)-ATP (2MeSATP); adenosine 5'-[beta-thio]diphosphate (ADPbetaS) inactive) indicated that P2Y2 rather than P2Y1 receptors mediate a 3-4-fold activation of NKCC within the first 5-10 min of nucleotide addition. NKCC activation in ATP-treated cells was abolished by the intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, calmodulin (CaM) antagonists trifluoroperazine and W-7, and KN-62, an inhibitor of Ca2+/CaM-dependent protein kinase II. By contrast with the transient activation, 30-min incubation with nucleotides produced up to 4-5-fold inhibition of NKCC, and this inhibition exhibited a rank order of potency (2MeSATP > ADPbetaS > ATP >> UTP) typical of P2Y1 receptors. Unlike the early response, delayed inhibition of NKCC occurred in 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-loaded cells and was completely abolished by the P2Y1 antagonists MRS2179 and MRS2500. Transient activation and delayed inhibition of NKCC in C11 cell monolayers were observed after the addition of ATP to mucosal and serosal solutions, respectively. NKCC inhibition triggered by basolateral application of ADPbetaS was abolished by MRS2500. Our results thus show that transient activation and delayed inhibition of NKCC in ATP-treated C11-MDCK cells is mediated by Ca2+/CaM-dependent protein kinase II- and Ca2+-independent signaling triggered by apical P2Y2 and basolateral P2Y1 receptors, respectively.


Assuntos
Trifosfato de Adenosina/farmacologia , Calmodulina/metabolismo , Receptores Purinérgicos P2/fisiologia , Simportadores de Cloreto de Sódio-Potássio/fisiologia , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Transporte Biológico , Cloro/química , Nucleotídeos de Desoxiadenina/farmacologia , Cães , Ativação Enzimática , Regulação da Expressão Gênica , Cinética , Potássio/metabolismo , Receptores Purinérgicos P2/metabolismo , Membro 2 da Família 12 de Carreador de Soluto
11.
Mol Pharmacol ; 70(5): 1700-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16885208

RESUMO

Overexpression of adenylyl cyclase (AC) has been proposed as a potential gene therapy strategy to increase cAMP formation in cardiomyocytes and cardiac function in vivo. The impact of AC overexpression on endothelial cells, which will be traversed by genes delivered in vivo, has not been examined. Hence, the goal of the current study was to determine the consequence of AC overexpression on vascular endothelial cells in terms of G-protein-coupled receptor (GPCR) signaling and endothelial barrier function. We demonstrate that adenoviral-mediated gene transfer of AC6 in human umbilical vein endothelial cells preferentially enhances prostacyclin receptor (versus other GPCR)-stimulated cAMP synthesis and, in parallel, inhibits thrombin-stimulated increases in endothelial cell barrier function. Using multiple strategies, including prostacyclin receptor-targeted small interfering RNA, we identify that the enhancement of endothelial barrier function by AC6 overexpression is dependent on an autocrine/paracrine feedback pathway involving the release of prostacyclin and activation of prostacyclin receptors. AC6 overexpression in endothelial cells may have use as a means to enhance prostacyclin function and reduce endothelial barrier permeability.


Assuntos
Adenilil Ciclases/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Células Endoteliais/metabolismo , Expressão Gênica , Receptores de Epoprostenol/metabolismo , Adenoviridae/metabolismo , Adenilil Ciclases/genética , Permeabilidade da Membrana Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Células Endoteliais/citologia , Epoprostenol/farmacologia , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Trombina/farmacologia
12.
Science ; 313(5783): 101-4, 2006 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-16825572

RESUMO

Natural populations of beach mice exhibit a characteristic color pattern, relative to their mainland conspecifics, driven by natural selection for crypsis. We identified a derived, charge-changing amino acid mutation in the melanocortin-1 receptor (Mc1r) in beach mice, which decreases receptor function. In genetic crosses, allelic variation at Mc1r explains 9.8% to 36.4% of the variation in seven pigmentation traits determining color pattern. The derived Mc1r allele is present in Florida's Gulf Coast beach mice but not in Atlantic coast mice with similar light coloration, suggesting that different molecular mechanisms are responsible for convergent phenotypic evolution. Here, we link a single mutation in the coding region of a pigmentation gene to adaptive quantitative variation in the wild.


Assuntos
Cor de Cabelo/genética , Mutação , Peromyscus/genética , Pigmentação/genética , Receptor Tipo 1 de Melanocortina/genética , Adaptação Biológica , Alelos , Substituição de Aminoácidos , Animais , Linhagem Celular , Cruzamentos Genéticos , AMP Cíclico/metabolismo , Feminino , Florida , Frequência do Gene , Genótipo , Cabelo , Humanos , Masculino , Dados de Sequência Molecular , Fenótipo , Polimorfismo de Nucleotídeo Único , Análise de Componente Principal , Receptor Tipo 1 de Melanocortina/química , Receptor Tipo 1 de Melanocortina/metabolismo , Análise de Sequência de DNA
13.
J Physiol ; 568(Pt 3): 789-801, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16109726

RESUMO

This study examines the mechanism of P 2Y-induced Cl- secretion in monolayers of C7-Madin-Darby canine kidney (MDCK) cells triggered by basolateral application of ATP and measured as transcellular short current (I(SC)). Both ATP-induced arachidonic acid (AA) synthesis and I(SC) in ATP-treated cells were abolished by the phosholipase A2 (PLA2) inhibitor, AACOCF3. The cyclo-oxygenase inhibitor indomethacin decreased I(SC) and cAMP production in ATP-treated cells with an IC50 of approximately 0.3 microm. ATP led to rapid activation of cAMP-dependent protein kinase A (PKA), as estimated by phosphorylation of a vasodilator-stimulated phosphoprotein. PKA activity and I(SC) evoked by ATP, as well as by prostaglandin E1 (PGE1), were diminished in the presence of the PKA inhibitor H-89 or an adenovirus-mediated expression of PKA-inhibitor protein, PKI. In contrast, indomethacin completely blocked the increment of PKA and I(SC) triggered by ATP and AA, but did not affect PKA activation and I(SC) detected with PGE1. The kinetics of [Ca2+]i elevation in ATP- and thapsigargin-treated cells were similar and suppressed by the Ca(2+)i chelator BAPTA. Neither baseline nor maximal increment of ATP-induced I(SC) was affected by thapsigargin and BAPTA. Real-time PCR showed that C7 cells express more mRNA for P 2Y1 and P 2Y2 than for other P 2Y receptor subtypes. The rank order of potency (2MeSATP > ATP > ADP >> UTP) indicates that P 2Y1 rather than P 2Y2 receptors contribute to PKA and I(SC) activation. Viewed collectively, these data show that Cl- secretion in C7-MDCK monolayers treated with basolateral ATP is triggered by P 2Y1 receptors and is mediated by subsequent [Ca2+]i-independent activation of PLA2 and PKA.


Assuntos
Trifosfato de Adenosina/farmacologia , Cloro/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Epiteliais/metabolismo , Rim/metabolismo , Fosfolipases A/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Linhagem Celular , Cães , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Rim/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Fosfolipases A2 , Receptores Purinérgicos P2Y1
14.
Biol Chem ; 385(6): 473-80, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15255178

RESUMO

Peptide neurotransmitters and hormones are synthesized as protein precursors that require proteolytic processing to generate smaller, biologically active peptides that are secreted to mediate neurotransmission and hormone actions. Neuropeptides within their precursors are typically flanked by pairs of basic residues, as well as by monobasic residues. In this review, evidence for secretory vesicle cathepsin L and Arg/Lys aminopeptidase as a distinct proteolytic pathway for processing the prohormone proenkephalin is presented. Cleavage of prohormone processing sites by secretory vesicle cathepsin L occurs at the NH2-terminal side of dibasic residues, as well as between the dibasic residues, resulting in peptide intermediates with Arg or Lys extensions at their NH2-termini. A subsequent Arg/Lys aminopeptidase step is then required to remove NH2-terminal basic residues to generate the final enkephalin neuropeptide. The cathepsin L and Arg/Lys aminopeptidase prohormone processing pathway is distinct from the proteolytic pathway mediated by the subtilisin-like prohormone convertases 1/3 and 2 (PC1/3 and PC2) with carboxypeptidase E/H. Differences in specific cleavage sites at paired basic residue sites distinguish these two pathways. These two proteolytic pathways demonstrate the increasing complexity of regulatory mechanisms for the production of peptide neurotransmitters and hormones.


Assuntos
Aminopeptidases/metabolismo , Catepsinas/metabolismo , Hormônios/biossíntese , Neurotransmissores/biossíntese , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Catepsina L , Cisteína Endopeptidases
15.
Sci STKE ; 2004(231): pe19, 2004 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-15126677

RESUMO

Soluble adenylyl cyclase can function in the nucleus, defining a nuclear microdomain of adenosine 3',5'-monophosphate (cAMP) signaling. Bundey and Insel discuss the evidence for discrete signaling microdomains of cAMP, including the nucleus and caveolae, and conclude that such microdomains may be defined by the localized, subcellular expression of adenylyl cyclase isoforms.


Assuntos
Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Sistemas do Segundo Mensageiro , Adenilil Ciclases/análise , Animais , Cavéolas/enzimologia , Núcleo Celular/enzimologia , Modelos Biológicos , Isoformas de Proteínas/metabolismo
16.
J Biol Chem ; 279(19): 19846-53, 2004 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-15007069

RESUMO

Several cell types, including cardiac myocytes and vascular endothelial cells, produce nitric oxide (NO) via both constitutive and inducible isoforms of NO synthase. NO attenuates cardiac contractility and contributes to contractile dysfunction in heart failure, although the precise molecular mechanisms for these effects are poorly defined. Adenylyl cyclase (AC) isoforms type 5 and 6, which are preferentially expressed in cardiac myocytes, may be inhibited via a direct nitrosylation by NO. Because endothelial NO synthase (eNOS and NOS3), beta-adrenergic (betaAR) receptors, and AC6 all can localize in lipid raft/caveolin-rich microdomains, we sought to understand the role of lipid rafts in organizing components of betaAR-G(s)-AC signal transduction together with eNOS. Using neonatal rat cardiac myocytes, we found that disruption of lipid rafts with beta-cyclodextrin inhibited forskolin-stimulated AC activity and cAMP production, eliminated caveolin-3-eNOS interaction, and increased NO production. betaAR- and G(s)-mediated activation of AC activity were inhibited by beta-cyclodextrin treatment, but prostanoid receptor-stimulated AC activity, which appears to occur outside caveolin-rich microdomains, was unaffected unless eNOS was overexpressed and lipid rafts were disrupted. An NO donor, SNAP, inhibited basal and forskolin-stimulated cAMP production in both native cardiac myocytes and cardiac myocytes and pulmonary artery endothelial cells engineered to overexpress AC6. These effects of SNAP were independent of guanylyl cyclase activity and were mimicked by overexpression of eNOS. The juxtaposition of eNOS with betaAR and AC types 5 and 6 results in selective regulation of betaAR by eNOS activity in lipid raft domains over other G(s)-coupled receptors localized in nonraft domains. Thus co-localization of multiple signaling components in lipid rafts provides key spatial regulation of AC activity.


Assuntos
Inibidores de Adenilil Ciclases , Adenilil Ciclases/metabolismo , Caveolinas/metabolismo , Microdomínios da Membrana/metabolismo , Óxido Nítrico/metabolismo , Penicilamina/análogos & derivados , beta-Ciclodextrinas , Animais , Animais Recém-Nascidos , Bovinos , Caveolina 1 , Caveolina 3 , Caveolinas/química , Membrana Celular/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Ciclodextrinas/química , Relação Dose-Resposta a Droga , Immunoblotting , Microdomínios da Membrana/química , Camundongos , Modelos Biológicos , Contração Miocárdica , Miocárdio/enzimologia , Óxido Nítrico Sintase/química , Penicilamina/farmacologia , Testes de Precipitina , Ligação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Transdução de Sinais
17.
Proc Natl Acad Sci U S A ; 100(16): 9590-5, 2003 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12869695

RESUMO

Multistep proteolytic mechanisms are essential for converting proprotein precursors into active peptide neurotransmitters and hormones. Cysteine proteases have been implicated in the processing of proenkephalin and other neuropeptide precursors. Although the papain family of cysteine proteases has been considered the primary proteases of the lysosomal degradation pathway, more recent studies indicate that functions of these enzymes are linked to specific biological processes. However, few protein substrates have been described for members of this family. We show here that secretory vesicle cathepsin L is the responsible cysteine protease of chromaffin granules for converting proenkephalin to the active enkephalin peptide neurotransmitter. The cysteine protease activity was identified as cathepsin L by affinity labeling with an activity-based probe for cysteine proteases followed by mass spectrometry for peptide sequencing. Production of [Met]enkephalin by cathepsin L occurred by proteolytic processing at dibasic and monobasic prohormone-processing sites. Cellular studies showed the colocalization of cathepsin L with [Met]enkephalin in secretory vesicles of neuroendocrine chromaffin cells by immunofluorescent confocal and immunoelectron microscopy. Functional localization of cathepsin L to the regulated secretory pathway was demonstrated by its cosecretion with [Met]enkephalin. Finally, in cathepsin L gene knockout mice, [Met]enkephalin levels in brain were reduced significantly; this occurred with an increase in the relative amounts of enkephalin precursor. These findings indicate a previously uncharacterized biological role for secretory vesicle cathepsin L in the production of [Met]enkephalin, an endogenous peptide neurotransmitter.


Assuntos
Catepsinas/fisiologia , Encefalinas/química , Hormônios/metabolismo , Neurotransmissores/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Catepsina L , Catepsinas/metabolismo , Bovinos , Células Cromafins/metabolismo , Cisteína Endopeptidases/metabolismo , Eletroforese em Gel Bidimensional , Espectrometria de Massas , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Modelos Químicos , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica
19.
Circ Res ; 92(6): 676-82, 2003 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-12623876

RESUMO

12-lipoxygenase (12-LO) has been shown to be a factor in acute ischemic preconditioning (IPC) in the isolated rat heart; however, no studies have been reported in delayed PC. We characterized the role of 12-LO in an intact rat model of delayed PC induced by a delta-opioid agonist SNC-121 (SNC). Rats were pretreated with SNC and allowed to recover for 24 hours. They were then treated with either baicalein or phenidone, 2 selective 12-LO inhibitors. In addition, SNC-pretreated rats had plasma samples isolated at different times after ischemia-reperfusion for liquid chromatographic-mass spectrometric analysis of the major metabolic product of 12-LO, 12-HETE. Similar studies were conducted with inhibitors. Gene array data showed a significant induction of 12-LO message (P<0.05) after opioid pretreatment. This induction in 12-LO mRNA was confirmed by real-time polymerase chain reaction, and 12-LO protein expression was enhanced by SNC pretreatment at 24 hours relative to vehicle treatment. Both baicalein and phenidone attenuated the protective effects of SNC pretreatment on infarct size (50+/-4% and 42+/-3% versus 29+/-2%, P<0.05, respectively). No significant differences were observed in 12-HETE concentrations between baseline control and SNC-treated rats. However, 12-HETE concentrations were increased significantly at both 15 minutes during ischemia and at 1 hour of reperfusion in the SNC-treated rats compared with controls. Baicalein and phenidone attenuated the increase in 12-HETE at 1 hour of reperfusion. These data suggest that SNC-121 appears to enhance message and subsequently the activity and expression of 12-LO protein during times of stress, resulting in delayed cardioprotection.


Assuntos
Araquidonato 12-Lipoxigenase/fisiologia , Benzamidas/farmacologia , Cardiotônicos/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Piperazinas/farmacologia , Receptores Opioides delta/agonistas , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/biossíntese , Animais , Araquidonato 12-Lipoxigenase/genética , Ácido Araquidônico/metabolismo , Western Blotting , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Hemodinâmica/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico , Cinética , Inibidores de Lipoxigenase , Masculino , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/enzimologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização por Electrospray
20.
Biochemistry ; 41(33): 10397-405, 2002 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-12173926

RESUMO

This study demonstrates that endopin 2 is a unique secretory vesicle serpin that displays cross-class inhibition of cysteine and serine proteases, indicated by effective inhibition of papain and elastase, respectively. Homology of the reactive site loop (RSL) domain of endopin 2, notably at P1-P1' residues, with other serpins that inhibit cysteine and serine proteases predicted that endopin 2 may inhibit similar proteases. Recombinant N-His-tagged endopin 2 inhibited papain and elastase with second-order rate constants (k(ass)) of 1.4 x 10(6) and 1.7 x 10(5) M(-1) s(-1), respectively. Endopin 2 formed SDS-stable complexes with papain and elastase, a characteristic property of serpins. Interactions of the RSL domain of endopin 2 with papain and elastase were indicated by cleavage of endopin 2 near the predicted P1-P1' residues by these proteases. Endopin 2 did not inhibit the cysteine protease cathepsin B, or the serine proteases chymotrypsin, trypsin, plasmin, and furin. Endopin 2 in neuroendocrine chromaffin cells was colocalized with the secretory vesicle component (Met)enkephalin by confocal immunonfluorescence microscopy, and was present in isolated secretory vesicles (chromaffin granules) from chromaffin cells as a glycoprotein of 72-73 kDa. Moreover, regulated secretion of endopin 2 from chromaffin cells was induced by nicotine and KCl depolarization. Overall, these results demonstrate that the serpin endopin 2 possesses dual specificity for inhibiting both papain-like cysteine and elastase-like serine proteases. These findings demonstrate that endopin 2 inhibitory functions may occur in the regulated secretory pathway.


Assuntos
Grânulos Cromafim/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Sistemas Neurossecretores/metabolismo , Elastase Pancreática/antagonistas & inibidores , Papaína/antagonistas & inibidores , Vesículas Secretórias/metabolismo , Inibidores de Serina Proteinase/farmacologia , Serpinas/fisiologia , Sequência de Aminoácidos , Animais , Bovinos , Grânulos Cromafim/enzimologia , Humanos , Cinética , Dados de Sequência Molecular , Sistemas Neurossecretores/enzimologia , Elastase Pancreática/metabolismo , Papaína/metabolismo , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Vesículas Secretórias/enzimologia , Serpinas/biossíntese , Serpinas/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...