Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 5(4)2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-31961828

RESUMO

Pulmonary Langerhans cell histiocytosis (PLCH) is a rare smoking-related lung disease characterized by dendritic cell (DC) accumulation, bronchiolocentric nodule formation, and cystic lung remodeling. Approximately 50% of patients with PLCH harbor somatic BRAF-V600E mutations in cells of the myeloid/monocyte lineage. However, the rarity of the disease and lack of animal models have impeded the study of PLCH pathogenesis. Here, we establish a cigarette smoke-exposed (CS-exposed) BRAF-V600E-mutant mouse model that recapitulates many hallmark characteristics of PLCH. We show that CD11c-targeted expression of BRAF-V600E increases DC responsiveness to stimuli, including the chemokine CCL20, and that mutant cell accumulation in the lungs of CS-exposed mice is due to both increased cellular viability and enhanced recruitment. Moreover, we report that the chemokine CCL7 is secreted from DCs and human peripheral blood monocytes in a BRAF-V600E-dependent manner, suggesting a possible mechanism for recruitment of cells known to dominate PLCH lesions. Inflammatory lesions and airspace dilation in BRAF-V600E mice in response to CS are attenuated by transitioning animals to filtered air and treatment with a BRAF-V600E inhibitor, PLX4720. Collectively, this model provides mechanistic insights into the role of myelomonocytic cells and the BRAF-V600E mutation and CS exposure in PLCH pathogenesis and provides a platform to develop biomarkers and therapeutic targets.


Assuntos
Histiocitose de Células de Langerhans/etiologia , Pneumopatias/etiologia , Proteínas Quinases Ativadas por Mitógeno/genética , Mutação , Fumaça/efeitos adversos , Produtos do Tabaco , Animais , Antígeno CD11c/genética , Modelos Animais de Doenças , Camundongos , Proteínas Proto-Oncogênicas B-raf/genética
2.
Sci Transl Med ; 7(313): 313ra181, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26560359

RESUMO

Pulmonary alveolar microlithiasis (PAM) is a rare, autosomal recessive lung disorder associated with progressive accumulation of calcium phosphate microliths. Inactivating mutations in SLC34A2, which encodes the NPT2b sodium-dependent phosphate cotransporter, has been proposed as a cause of PAM. We show that epithelial deletion of Npt2b in mice results in a progressive pulmonary process characterized by diffuse alveolar microlith accumulation, radiographic opacification, restrictive physiology, inflammation, fibrosis, and an unexpected alveolar phospholipidosis. Cytokine and surfactant protein elevations in the alveolar lavage and serum of PAM mice and confirmed in serum from PAM patients identify serum MCP-1 (monocyte chemotactic protein 1) and SP-D (surfactant protein D) as potential biomarkers. Microliths introduced by adoptive transfer into the lungs of wild-type mice produce marked macrophage-rich inflammation and elevation of serum MCP-1 that peaks at 1 week and resolves at 1 month, concomitant with clearance of stones. Microliths isolated by bronchoalveolar lavage readily dissolve in EDTA, and therapeutic whole-lung EDTA lavage reduces the burden of stones in the lungs. A low-phosphate diet prevents microlith formation in young animals and reduces lung injury on the basis of reduction in serum SP-D. The burden of pulmonary calcium deposits in established PAM is also diminished within 4 weeks by a low-phosphate diet challenge. These data support a causative role for Npt2b in the pathogenesis of PAM and the use of the PAM mouse model as a preclinical platform for the development of biomarkers and therapeutic strategies.


Assuntos
Biomarcadores/sangue , Calcinose/etiologia , Calcinose/fisiopatologia , Calcinose/terapia , Doenças Genéticas Inatas/etiologia , Doenças Genéticas Inatas/fisiopatologia , Doenças Genéticas Inatas/terapia , Pneumopatias/etiologia , Pneumopatias/fisiopatologia , Pneumopatias/terapia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/deficiência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Animais , Dieta , Modelos Animais de Doenças , Epitélio/metabolismo , Epitélio/patologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Mutação , Fosfatos/metabolismo , Alvéolos Pulmonares/metabolismo
3.
J Invest Dermatol ; 132(9): 2255-62, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22572817

RESUMO

The melanocortin 1 receptor (MC1R), a G(s) protein-coupled receptor, has an important role in human pigmentation. We investigated the regulation of expression and activity of the MC1R in primary human melanocyte cultures. Human ß-defensin 3 (HBD3) acted as an antagonist for MC1R, inhibiting the α-melanocortin (α-melanocyte-stimulating hormone (α-MSH))-induced increase in the activities of adenylate cyclase and tyrosinase, the rate-limiting enzyme for melanogenesis. α-Melanocortin and forskolin, which activate adenylate cyclase, and 12-O-tetradecanoylphorbol-13-acetate, which activates protein kinase C, increased, whereas exposure to UV radiation reduced, MC1R gene and membrane protein expression. Brief treatment with α-MSH resulted in MC1R desensitization, whereas continuous treatment up to 3 hours caused a steady rise in cAMP, suggesting receptor recycling. Pretreatment with agouti signaling protein or HBD3 prohibited responsiveness to α-MSH, but not forskolin, suggesting receptor desensitization by these antagonists. Melanocytes from different donors expressed different levels of the G protein-coupled receptor kinases (GRKs) 2, 3, 5, and 6, as well as ß-arrestin 1. Therefore, in addition to the MC1R genotype, regulation of MC1R expression and activity is expected to affect human pigmentation and the responses to UV.


Assuntos
Proteína Agouti Sinalizadora/farmacologia , Melanocortinas/farmacologia , Melanócitos/efeitos dos fármacos , Receptor Tipo 1 de Melanocortina/agonistas , Receptor Tipo 1 de Melanocortina/antagonistas & inibidores , alfa-MSH/farmacologia , beta-Defensinas/farmacologia , Adenilil Ciclases/metabolismo , Arrestinas/biossíntese , Células Cultivadas , Colforsina/farmacologia , Quinases de Receptores Acoplados a Proteína G/biossíntese , Humanos , Melanócitos/metabolismo , Melanócitos/efeitos da radiação , Monofenol Mono-Oxigenase/metabolismo , Proteína Quinase C/metabolismo , Receptor Tipo 1 de Melanocortina/biossíntese , Pigmentação da Pele/efeitos dos fármacos , Pigmentação da Pele/fisiologia , Pigmentação da Pele/efeitos da radiação , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Raios Ultravioleta , beta-Arrestina 1 , beta-Arrestinas
4.
J Pharmacol Exp Ther ; 336(2): 440-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20974700

RESUMO

ß(2)-agonists are the most effective bronchodilators for the rapid relief of asthma symptoms, but for unclear reasons, their effectiveness may be decreased during severe exacerbations. Because peroxidase activity and nitrogen oxides are increased in the asthmatic airway, we examined whether salbutamol, a clinically important ß(2)-agonist, is subject to potentially inactivating nitration. When salbutamol was exposed to myeloperoxidase, eosinophil peroxidase or lactoperoxidase in the presence of hydrogen peroxide (H(2)O(2)) and nitrite (NO(2)(-)), both absorption spectroscopy and mass spectrometry indicated formation of a new metabolite with features expected for the nitrated drug. The new metabolites showed an absorption maximum at 410 nm and pK(a) of 6.6 of the phenolic hydroxyl group. In addition to nitrosalbutamol (m/z 285.14), a salbutamol-derived nitrophenol, formed by elimination of the formaldehyde group, was detected (m/z 255.13) by mass spectrometry. It is noteworthy that the latter metabolite was detected in exhaled breath condensates of asthma patients receiving salbutamol but not in unexposed control subjects, indicating the potential for ß(2)-agonist nitration to occur in the inflamed airway in vivo. Salbutamol nitration was inhibited in vitro by ascorbate, thiocyanate, and the pharmacological agents methimazole and dapsone. The efficacy of inhibition depended on the nitrating system, with the lactoperoxidase/H(2)O(2)/NO(2)(-) being the most affected. Functionally, nitrated salbutamol showed decreased affinity for ß(2)-adrenergic receptors and impaired cAMP synthesis in airway smooth muscle cells compared with the native drug. These results suggest that under inflammatory conditions associated with asthma, phenolic ß(2)-agonists may be subject to peroxidase-catalyzed nitration that could potentially diminish their therapeutic efficacy.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/metabolismo , Albuterol/metabolismo , Asma/tratamento farmacológico , Brônquios/enzimologia , Nitritos/metabolismo , Peroxidases/fisiologia , Albuterol/farmacologia , Ácido Ascórbico/farmacologia , Asma/metabolismo , Testes Respiratórios , Catálise , Criança , AMP Cíclico/biossíntese , Dapsona/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Espectrometria de Massas , Metimazol/farmacologia , Receptores Adrenérgicos beta 2/metabolismo , Tiocianatos/farmacologia
5.
J Am Soc Nephrol ; 21(10): 1745-55, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20798260

RESUMO

Proton receptors are G protein-coupled receptors that accept protons as ligands and function as pH sensors. One of the proton receptors, GPR4, is relatively abundant in the kidney, but its potential role in acid-base homeostasis is unknown. In this study, we examined the distribution of GPR4 in the kidney, its function in kidney epithelial cells, and the effects of its deletion on acid-base homeostasis. We observed GPR4 expression in the kidney cortex, in the outer and inner medulla, in isolated kidney collecting ducts, and in cultured outer and inner medullary collecting duct cells (mOMCD1 and mIMCD3). Cultured mOMCD1 cells exhibited pH-dependent accumulation of intracellular cAMP, characteristic of GPR4 activation; GPR4 knockdown attenuated this accumulation. In vivo, deletion of GPR4 decreased net acid secretion by the kidney and resulted in a nongap metabolic acidosis, indicating that GPR4 is required to maintain acid-base homeostasis. Collectively, these findings suggest that GPR4 is a pH sensor with an important role in regulating acid secretion in the kidney collecting duct.


Assuntos
Equilíbrio Ácido-Base , Túbulos Renais Coletores/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Acidose Tubular Renal/metabolismo , Ácidos/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética
6.
Chem Res Toxicol ; 22(6): 1137-50, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19462961

RESUMO

Phenolic beta(2)-adrenoreceptor agonists salbutamol, fenoterol, and terbutaline relax smooth muscle cells that relieve acute airway bronchospasm associated with asthma. Why their use sometimes fails to relieve bronchospasm and why the drugs appear to be less effective in patients with severe asthma exacerbations remains unclear. We show that in the presence of hydrogen peroxide, both myeloperoxidase, secreted by activated neutrophils present in inflamed airways, and lactoperoxidase, which is naturally present in the respiratory system, catalyze oxidation of these beta(2)-agonists. Azide, cyanide, thiocyanate, ascorbate, glutathione, and methimazole inhibited this process, while methionine was without effect. Inhibition by ascorbate and glutathione was associated with their oxidation to corresponding radical species by the agonists' derived phenoxyl radicals. Using electron paramagnetic resonance (EPR), we detected free radical metabolites from beta(2)-agonists by spin trapping with 2-methyl-2-nitrosopropane (MNP). Formation of these radicals was inhibited by pharmacologically relevant concentrations of methimazole and dapsone. In alkaline buffers, radicals from fenoterol and its structural analogue, metaproteronol, were detected by direct EPR. Analysis of these spectra suggests that oxidation of fenoterol and metaproterenol, but not terbutaline, causes their transformation through intramolecular cyclization by addition of their amino nitrogen to the aromatic ring. Together, these results indicate that phenolic beta(2)-agonists function as substrates for airway peroxidases and that the resulting products differ in their structural and functional properties from their parent compounds. They also suggest that these transformations can be modulated by pharmacological approaches using appropriate peroxidase inhibitors or alternative substrates. These processes may affect therapeutic efficacy and also play a role in adverse reactions of the beta(2)-agonists.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2 , Agonistas Adrenérgicos beta/metabolismo , Albuterol/metabolismo , Fenoterol/metabolismo , Agonistas Adrenérgicos beta/química , Albuterol/análogos & derivados , Albuterol/química , Espectroscopia de Ressonância de Spin Eletrônica , Fenoterol/análogos & derivados , Fenoterol/química , Radicais Livres/química , Radicais Livres/metabolismo , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Lactoperoxidase/química , Lactoperoxidase/metabolismo , Oxirredução
7.
J Clin Invest ; 119(3): 636-49, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19197141

RESUMO

Chronic obstructive pulmonary disease (COPD) is a lethal progressive lung disease culminating in permanent airway obstruction and alveolar enlargement. Previous studies suggest CTL involvement in COPD progression; however, their precise role remains unknown. Here, we investigated whether the CTL activation receptor NK cell group 2D (NKG2D) contributes to the development of COPD. Using primary murine lung epithelium isolated from mice chronically exposed to cigarette smoke and cultured epithelial cells exposed to cigarette smoke extract in vitro, we demonstrated induced expression of the NKG2D ligand retinoic acid early transcript 1 (RAET1) as well as NKG2D-mediated cytotoxicity. Furthermore, a genetic model of inducible RAET1 expression on mouse pulmonary epithelial cells yielded a severe emphysematous phenotype characterized by epithelial apoptosis and increased CTL activation, which was reversed by blocking NKG2D activation. We also assessed whether NKG2D ligand expression corresponded with pulmonary disease in human patients by staining airway and peripheral lung tissues from never smokers, smokers with normal lung function, and current and former smokers with COPD. NKG2D ligand expression was independent of NKG2D receptor expression in COPD patients, demonstrating that ligand expression is the limiting factor in CTL activation. These results demonstrate that aberrant, persistent NKG2D ligand expression in the pulmonary epithelium contributes to the development of COPD pathologies.


Assuntos
Pulmão/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Mucosa Respiratória/fisiopatologia , Fumaça/efeitos adversos , Fumar/efeitos adversos , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Enfisema/etiologia , Enfisema/imunologia , Regulação da Expressão Gênica , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Proteínas de Membrana/genética , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia
8.
J Clin Invest ; 117(5): 1391-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17415415

RESUMO

Receptor-mediated airway smooth muscle (ASM) contraction via G(alphaq), and relaxation via G(alphas), underlie the bronchospastic features of asthma and its treatment. Asthma models show increased ASM G(alphai) expression, considered the basis for the proasthmatic phenotypes of enhanced bronchial hyperreactivity to contraction mediated by M(3)-muscarinic receptors and diminished relaxation mediated by beta(2)-adrenergic receptors (beta(2)ARs). A causal effect between G(i) expression and phenotype has not been established, nor have mechanisms whereby G(i) modulates G(q)/G(s) signaling. To delineate isolated effects of altered G(i), transgenic mice were generated overexpressing G(alphai2) or a G(alphai2) peptide inhibitor in ASM. Unexpectedly, G(alphai2) overexpression decreased contractility to methacholine, while G(alphai2) inhibition enhanced contraction. These opposite phenotypes resulted from different crosstalk loci within the G(q) signaling network: decreased phospholipase C and increased PKCalpha, respectively. G(alphai2) overexpression decreased beta(2)AR-mediated airway relaxation, while G(alphai2) inhibition increased this response, consistent with physiologically relevant coupling of this receptor to both G(s) and G(i). IL-13 transgenic mice (a model of asthma), which developed increased ASM G(alphai), displayed marked increases in airway hyperresponsiveness when G(alphai) function was inhibited. Increased G(alphai) in asthma is therefore a double-edged sword: a compensatory event mitigating against bronchial hyperreactivity, but a mechanism that evokes beta-agonist resistance. By selective intervention within these multipronged signaling modules, advantageous G(s)/G(q) activities could provide new asthma therapies.


Assuntos
Hiper-Reatividade Brônquica/metabolismo , Espasmo Brônquico/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Relaxamento Muscular/fisiologia , Músculo Liso/fisiologia , Transdução de Sinais/fisiologia , Animais , Asma/genética , Asma/metabolismo , Asma/fisiopatologia , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/fisiopatologia , Espasmo Brônquico/genética , Espasmo Brônquico/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Relaxamento Muscular/genética , Coelhos , Receptor Cross-Talk/fisiologia , Transdução de Sinais/genética
9.
Physiol Genomics ; 27(2): 171-7, 2006 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-16849635

RESUMO

Beta(2)-Adrenergic receptors (beta(2)AR) are expressed on airway smooth muscle cells and act to relax the airway on activation by beta-agonists. These agents are utilized for treating asthma but are associated with adverse outcomes. To ascertain the effects of persistent beta(2)AR activation on gene expression, cultured airway smooth muscle cells derived from wild-type (WT) and transgenic mice overexpressing beta(2)AR were subjected to DNA microarray analysis; 319 genes were increased and 164 were decreased. Differential expression was observed in genes from 22 Gene Ontology Slim categories, including those associated with ion transport and calcium ion binding. A 60% decrease (P = 0.008) in phospholamban (PLN), an intracellular Ca(2+) concentration ([Ca(2+)]i)-handling protein that is at a signaling nodal point in cardiomyocytes, was observed in beta(2)AR-overexpressing cells and confirmed at the protein level. To isolate the physiological effect of decreased PLN in airway smooth muscle, airway contraction and relaxation responses were studied in WT and PLN(-/-) mice. PLN(-/-) mice had a markedly reduced constrictive response to methacholine. In contrast, the bronchodilatory effect of beta-agonist was not different between WT and PLN(-/-) mice. These results revealed an unanticipated therapeutic effect of beta-agonists, PLN downregulation, which acts to decrease airway hyperreactivity. Thus agents that inhibit PLN may act synergistically with the bronchodilating action of beta-agonists. A number of other genes related to [Ca(2+)]i are also differentially regulated by beta(2)AR activity, some of which may act to oppose, or augment, the efficacy of chronic beta-agonists. These genes or pathways may also represent additional targets in the treatment of asthma and related obstructive lung diseases.


Assuntos
Broncoconstrição/genética , Proteínas de Ligação ao Cálcio/biossíntese , Músculo Liso/efeitos dos fármacos , Receptores Adrenérgicos beta 2/fisiologia , Resistência das Vias Respiratórias/efeitos dos fármacos , Animais , Antiasmáticos/farmacologia , Broncoconstrição/efeitos dos fármacos , Broncoconstritores/farmacologia , Broncodilatadores/farmacologia , Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/fisiologia , Células Cultivadas , Perfilação da Expressão Gênica , Isoproterenol/farmacologia , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Músculo Liso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais/efeitos dos fármacos , Traqueia/citologia , Transcrição Gênica/efeitos dos fármacos
10.
J Clin Invest ; 116(5): 1400-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16670773

RESUMO

Multiple and paradoxical effects of airway smooth muscle (ASM) 7-transmembrane-spanning receptors activated during asthma, or by treatment with bronchodilators such as beta(2)-adrenergic receptor (beta(2)AR) agonists, indicate extensive receptor crosstalk. We examined the signaling of the prostanoid-EP(1) receptor, since its endogenous agonist prostaglandin E(2) is abundant in the airway, but its functional implications are poorly defined. Activation of EP(1) failed to elicit ASM contraction in mouse trachea via this G(alphaq)-coupled receptor. However, EP(1) activation markedly reduced the bronchodilatory function of beta(2)AR agonist, but not forskolin, indicating an early pathway interaction. Activation of EP(1) reduced beta(2)AR-stimulated cAMP in ASM but did not promote or augment beta(2)AR phosphorylation or alter beta(2)AR trafficking. Bioluminescence resonant energy transfer showed EP(1) and beta(2)AR formed heterodimers, which were further modified by EP(1) agonist. In cell membrane [(35)S]GTPgammaS binding studies, the presence of the EP(1) component of the dimer uncoupled beta(2)AR from G(alphas), an effect accentuated by EP(1) agonist activation. Thus alone, EP(1) does not appear to have a significant direct effect on airway tone but acts as a modulator of the beta(2)AR, altering G(alphas) coupling via steric interactions imposed by the EP(1):beta(2)AR heterodimeric signaling complex and ultimately affecting beta(2)AR-mediated bronchial relaxation. This mechanism may contribute to beta-agonist resistance found in asthma.


Assuntos
Músculo Liso/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , Receptores de Prostaglandina E/fisiologia , Traqueia/citologia , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Colforsina/metabolismo , Dimerização , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Camundongos , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Prostaglandina E Subtipo EP1 , Transdução de Sinais
11.
Proc Am Thorac Soc ; 2(4): 292-6; discussion 311-2, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16267351

RESUMO

It is now clear that the beta2-adrenergic receptor continuously oscillates between various conformations in the basal state, and that agonists act to stabilize one or more conformations. It is conceivable that synthetic agonists might be engineered to preferentially confine the receptor to certain conformations deemed clinically important while having a less stabilizing effect on unwanted conformations. In addition, studies of genetically engineered mice have revealed previously unrecognized cross-talk between the beta2-receptor and phospholipase C, such that removal of the primary dilating pathway results in downregulation of constrictive pathways and overactivity of the dilating pathway increases the contractile response. These results indicate a dynamic interaction between beta2-receptor activity and Gq-coupled receptors that constrict the airway. Potentially, then, during chronic beta-agonist therapy, expression of phospholipase C is increased, the functions of Gq-coupled constrictive receptors are enhanced, and there may be an increased tendency for clinical decompensation due to asthma and chronic obstructive pulmonary disease triggers. Antagonists to these receptors might be able to act synergistically with chronic beta-agonists to block the effect of phospholipase C. Alternatively, perhaps novel phospholipase C antagonists would provide the most efficacious approach to blocking the physiologic sequelae of cross-talk between the beta2-receptor and phospholipase C.


Assuntos
Receptores Adrenérgicos beta 2/fisiologia , Agonistas de Receptores Adrenérgicos beta 2 , Animais , Asma/fisiopatologia , Broncoconstrição/fisiologia , Humanos , Camundongos , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Receptor Cross-Talk , Transdução de Sinais , Fosfolipases Tipo C/metabolismo
12.
Circ Res ; 96(7): 756-66, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15746443

RESUMO

Abnormal calcium cycling, characteristic of experimental and human heart failure, is associated with impaired sarcoplasmic reticulum calcium uptake activity. This reflects decreases in the cAMP-pathway signaling and increases in type 1 phosphatase activity. The increased protein phosphatase 1 activity is partially due to dephosphorylation and inactivation of its inhibitor-1, promoting dephosphorylation of phospholamban and inhibition of the sarcoplasmic reticulum calcium-pump. Indeed, cardiac-specific expression of a constitutively active inhibitor-1 results in selective enhancement of phospholamban phosphorylation and augmented cardiac contractility at the cellular and intact animal levels. Furthermore, the beta-adrenergic response is enhanced in the transgenic hearts compared with wild types. On aortic constriction, the hypercontractile cardiac function is maintained, hypertrophy is attenuated and there is no decompensation in the transgenics compared with wild-type controls. Notably, acute adenoviral gene delivery of the active inhibitor-1, completely restores function and partially reverses remodeling, including normalization of the hyperactivated p38, in the setting of pre-existing heart failure. Thus, the inhibitor 1 of the type 1 phosphatase may represent an attractive new therapeutic target.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Insuficiência Cardíaca/prevenção & controle , Contração Miocárdica , Fosfoproteínas Fosfatases/antagonistas & inibidores , Proteínas/fisiologia , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/fisiologia , ATPases Transportadoras de Cálcio/fisiologia , Cardiomegalia/etiologia , Cardiomegalia/prevenção & controle , Terapia Genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Camundongos , Camundongos Transgênicos , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Proteína Fosfatase 1 , Proteínas/genética , Ratos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Remodelação Ventricular
13.
J Clin Invest ; 112(4): 619-26, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12925702

RESUMO

beta-adrenergic receptors (betaARs) relax airway smooth muscle and bronchodilate, but chronic beta-agonist treatment in asthma causes increased sensitivity to airway constriction (hyperreactivity) and is associated with exacerbations. This paradox was explored using mice with ablated betaAR genes (betaAR-/-) and transgenic mice overexpressing airway smooth muscle beta2AR (beta2AR-OE) representing two extremes: absence and persistent activity of airway betaAR. Unexpectedly, betaAR-/- mice, lacking these bronchodilating receptors, had markedly decreased bronchoconstrictive responses to methacholine and other Gq-coupled receptor agonists. In contrast, beta2AR-OE mice had enhanced constrictive responses. Contraction to permeabilization with beta-escin was unaltered by gene ablation or overexpression. Inositol phosphate accumulation by Gq-coupled M3-muscarinic, thromboxane-A2, and 5-HT2 receptors was desensitized in airway smooth muscle cells from betaAR-/- mice and sensitized in cells from beta2AR-OE mice. Thus, betaAR antithetically regulates constrictive signals, affecting bronchomotor tone/reactivity by additional means other than direct dilatation. Studies of signaling elements in these pathways revealed the nodal point of this cross talk as phospholipase C-beta1, whose expression was altered by betaAR in a direction and magnitude consistent with the physiologic and cellular responses. These results establish a mechanism of the beta-agonist paradox and identify a potential asthma modifier gene (phospholipase C-beta1), which may also be a therapeutic target in asthma when chronic beta-agonists are required.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Fosfolipases Tipo C/metabolismo , Animais , Western Blotting , Broncoconstritores/farmacologia , Relação Dose-Resposta a Droga , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Fosfatos de Inositol/metabolismo , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Contração Muscular , Músculo Liso/metabolismo , Fosfolipase C beta , Traqueia/patologia
14.
Annu Rev Pharmacol Toxicol ; 43: 381-411, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12540746

RESUMO

Adrenergic receptors are expressed on virtually every cell type in the body and are the receptors for epinephrine and norepinephrine within the sympathetic nervous system. They serve critical roles in maintaining homeostasis in normal physiologic settings as well as pathologic states. These receptors are also targets for therapeutically administered agonists and antagonists. Recent studies have shown that at least seven adrenergic receptor subtypes display variation in amino acid sequence in the human population due to common genetic polymorphisms. Variations in potential regulatory domains in noncoding sequence are also present. Here, we review the consequences of these polymorphisms in terms of signaling, human physiology and disease, and response to therapy.


Assuntos
Farmacogenética , Polimorfismo Genético/genética , Receptores Adrenérgicos/genética , Sequência de Bases , Humanos , Dados de Sequência Molecular , Receptores Adrenérgicos/efeitos dos fármacos , Transdução de Sinais
15.
J Allergy Clin Immunol ; 110(6 Suppl): S236-41, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12464930

RESUMO

Many, if not most, of the different cell types within the lung have been shown to express beta(2)-adrenergic receptors (beta(2)ARs) on the cell surface. beta(2)ARs have thus been implicated in the regulation of many aspects of lung function. However, the physiologic and therapeutic relevance for many of these cell-specific responses has been difficult to establish because of confounding effects that result from the simultaneous activation of receptors on multiple cell types in an in vivo environment. It has been recognized in in vitro models that overexpression of G-protein-coupled receptors such as the beta(2)ARs can increase the number of spontaneously active receptors (R*) and thereby promote autonomous signal transduction or enhanced agonist sensitivity. With transgenic techniques, high levels of receptor expression and activation can also be attained in vivo. By further using cell-specific promoters to direct expression, it is possible to express beta(2)ARs in select cell types of a heterogeneous organ such as the lung. In this manner, activation of receptor signaling is limited to the targeted cell. Promoter-directed transgenesis can therefore be a useful tool to delineate cell-specific beta(2)AR-mediated effects in the lung.


Assuntos
Pulmão/metabolismo , Receptores Adrenérgicos beta 2/genética , Transgenes , Animais , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Modelos Animais , Miócitos de Músculo Liso/fisiologia , Receptores Adrenérgicos beta 2/metabolismo , Mucosa Respiratória/fisiologia , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA