Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Ann Am Thorac Soc ; 20(8): 1077-1087, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37526479

RESUMO

Rationale: To identify barriers and opportunities for Ph.D., basic and translational scientists to be fully integrated into clinical units. Objectives: In 2022, an ad hoc committee of the American Thoracic Society developed a project proposal and workshop to identify opportunities and barriers for scientists who do not practice medicine to develop successful careers and achieve tenure-track faculty positions in clinical departments and divisions within academic medical centers (AMCs) in the United States. Methods: This document focuses on results from a survey of adult and pediatric pulmonary, critical care, and sleep medicine division chiefs as well as a survey of workshop participants, including faculty in departmental and school leadership roles in both basic science and clinical units within U.S. AMCs. Results: We conclude that full integration of non-clinically practicing basic and translational scientists into the clinical units, in addition to their traditional placements in basic science units, best serves the tripartite mission of AMCs to provide care, perform research, and educate the next generation. Evidence suggests clinical units do employ Ph.D. scientists in large numbers, but these faculty are often hired into non-tenure track positions, which do not provide the salary support, start-up funds, research independence, or space often associated with hiring in basic science units within the same institution. These barriers to success of Ph.D. faculty in clinical units are largely financial. Conclusions: Our recommendation is for AMCs to consider and explore some of our proposed strategies to accomplish the goal of integrating basic and translational scientists into clinical units in a meaningful way.


Assuntos
Centros Médicos Acadêmicos , Médicos , Adulto , Estados Unidos , Humanos , Criança , Seleção de Pessoal , Liderança , Docentes de Medicina
3.
J Allergy Clin Immunol ; 147(6): 1983-1995, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34092351

RESUMO

The airway smooth muscle (ASM) cell plays a central role in the pathogenesis of asthma and constitutes an important target for treatment. These cells control muscle tone and thus regulate the opening of the airway lumen and air passage. Evidence indicates that ASM cells participate in the airway hyperresponsiveness as well as the inflammatory and remodeling processes observed in asthmatic subjects. Therapeutic approaches require a comprehensive understanding of the structure and function of the ASM in both the normal and disease states. This review updates current knowledge about ASM and its effects on airway narrowing, remodeling, and inflammation in asthma.


Assuntos
Asma/etiologia , Asma/metabolismo , Suscetibilidade a Doenças , Músculo Liso/metabolismo , Remodelação das Vias Aéreas/genética , Remodelação das Vias Aéreas/imunologia , Animais , Biomarcadores , Broncoconstrição/genética , Broncoconstrição/imunologia , Regulação da Expressão Gênica , Humanos , Músculo Liso/fisiopatologia , Miócitos de Músculo Liso/metabolismo
5.
J Allergy Clin Immunol ; 144(6): 1735-1736, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31564471
7.
Am J Respir Crit Care Med ; 195(2): e4-e19, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28084822

RESUMO

BACKGROUND: Airway remodeling (AR) is a prominent feature of asthma and other obstructive lung diseases that is minimally affected by current treatments. The goals of this Official American Thoracic Society (ATS) Research Statement are to discuss the scientific, technological, economic, and regulatory issues that deter progress of AR research and development of therapeutics targeting AR and to propose approaches and solutions to these specific problems. This Statement is not intended to provide clinical practice recommendations on any disease in which AR is observed and/or plays a role. METHODS: An international multidisciplinary group from within academia, industry, and the National Institutes of Health, with expertise in multimodal approaches to the study of airway structure and function, pulmonary research and clinical practice in obstructive lung disease, and drug discovery platforms was invited to participate in one internet-based and one face-to-face meeting to address the above-stated goals. Although the majority of the analysis related to AR was in asthma, AR in other diseases was also discussed and considered in the recommendations. A literature search of PubMed was performed to support conclusions. The search was not a systematic review of the evidence. RESULTS: Multiple conceptual, logistical, economic, and regulatory deterrents were identified that limit the performance of AR research and impede accelerated, intensive development of AR-focused therapeutics. Complementary solutions that leverage expertise of academia and industry were proposed to address them. CONCLUSIONS: To date, numerous factors related to the intrinsic difficulty in performing AR research, and economic forces that are disincentives for the pursuit of AR treatments, have thwarted the ability to understand AR pathology and mechanisms and to address it clinically. This ATS Research Statement identifies potential solutions for each of these factors and emphasizes the importance of educating the global research community as to the extent of the problem as a critical first step in developing effective strategies for: (1) increasing the extent and impact of AR research and (2) developing, testing, and ultimately improving drugs targeting AR.


Assuntos
Remodelação das Vias Aéreas , Pesquisa Biomédica , Remodelação das Vias Aéreas/fisiologia , Animais , Asma/fisiopatologia , Asma/terapia , Modelos Animais de Doenças , Humanos , Pulmão/fisiopatologia , Doenças Respiratórias/dietoterapia , Doenças Respiratórias/fisiopatologia , Doenças Respiratórias/terapia
8.
PLoS One ; 10(6): e0131582, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26121686

RESUMO

Asthma is characterized by airway inflammation and airflow obstruction from human airway smooth muscle (HASM) constriction due to increased local bronchoconstrictive substances. We have recently found bitter taste receptors (TAS2Rs) on HASM, which increase [Ca2+]i and relax the muscle. We report here that some, but not all, TAS2R agonists decrease [Ca2+]i and relax HASM contracted by G-protein coupled receptors (GPCRs) that stimulate [Ca2+]i. This suggests both a second pathway by which TAS2Rs relax, and, a heterogeneity of the response phenotype. We utilized eight TAS2R agonists and five procontractile GPCR agonists in cultured HASM cells. We find that heterogeneity in the inhibitory response hinges on which procontractile GPCR is activated. For example, chloroquine inhibits [Ca2+]i increases from histamine, but failed to inhibit [Ca2+]i increases from endothelin-1. Conversely, aristolochic acid inhibited [Ca2+]i increases from endothelin-1 but not histamine. Other dichotomous responses were found when [Ca2+]i was stimulated by bradykinin, angiotensin, and acetylcholine. There was no association between [Ca2+]i inhibition and TAS2R subtype, nor whether [Ca2+]i was increased by Gq- or Gi-coupled GPCRs. Selected studies revealed a correlation between [Ca2+]i inhibition and HASM cell-membrane hyperpolarization. To demonstrate physiologic correlates, ferromagnetic beads were attached to HASM cells and cell stiffness measured by magnetic twisting cytometry. Consistent with the [Ca2+]i inhibition results, chloroquine abolished the cell stiffening response (contraction) evoked by histamine but not by endothelin-1, while aristolochic acid inhibited cell stiffening from endothelin-1, but not from histamine. In studies using intact human bronchi, these same differential responses were found. Those TAS2R agonists that decreased [Ca2+]i, promoted hyperpolarization, and decreased HASM stiffness, caused relaxation of human airways. Thus TAS2Rs relax HASM in two ways: a low-efficiency de novo [Ca2+]i stimulation, and, a high-efficiency inhibition of GPCR-stimulated [Ca2+]i. Furthermore, there is an interaction between TAS2Rs and some GPCRs that facilitates this [Ca2+]i inhibition limb.


Assuntos
Potenciais de Ação , Sinalização do Cálcio , Pleiotropia Genética , Pulmão/citologia , Relaxamento Muscular , Miócitos de Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Potenciais de Ação/efeitos dos fármacos , Ácidos Aristolóquicos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Cloroquina/farmacologia , Endotelina-1/metabolismo , Histamina/farmacologia , Humanos , Relaxamento Muscular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Receptores Histamínicos/metabolismo
9.
Am J Respir Cell Mol Biol ; 52(4): 438-47, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25180620

RESUMO

Cyclooxygenase-2 (COX-2) expression and PGE2 secretion from human airway smooth muscle cells (hASMCs) may contribute to ß2-adrenoceptor hyporesponsiveness, a clinical feature observed in some patients with asthma. hASMCs from patients with asthma exhibit elevated expression of cytokine-responsive genes, and in some instances this is attributable to an altered histone code and/or microRNA expression. We hypothesized that COX-2 expression and PGE2 secretion might be elevated in asthmatic hASMCs in response to proinflammatory signals in part due to altered histone acetylation and/or microRNA expression. hASMCs obtained from nonasthmatic and asthmatic human subjects were treated with cytomix (IL-1ß, TNF-α, and IFN-γ). A greater elevation of COX-2 mRNA, COX-2 protein, and PGE2 secretion was observed in the asthmatic cells. We investigated histone H3/H4-acetylation, transcription factor binding, mRNA stability, p38 mitogen-activated protein kinase signaling, and microRNA (miR)-155 expression as potential mechanisms responsible for the differential elevation of COX-2 expression. We found that histone H3/H4-acetylation and transcription factor binding to the COX-2 promoter were similar in both groups, and histone H3/H4-acetylation did not increase after cytomix treatment. Cytomix treatment elevated NF-κB and RNA polymerase II binding to similar levels in both groups. COX-2 mRNA stability was increased in asthmatic cells. MiR-155 expression was higher in cytomix-treated asthmatic cells, and we show it enhances COX-2 expression and PGE2 secretion in asthmatic and nonasthmatic hASMCs. Thus, miR-155 expression positively correlates with COX-2 expression in the asthmatic hASMCs and may contribute to the elevated expression observed in these cells. These findings may explain, at least in part, ß2-adrenoceptor hyporesponsiveness in patients with asthma.


Assuntos
Asma/enzimologia , Ciclo-Oxigenase 2/metabolismo , MicroRNAs/metabolismo , Miócitos de Músculo Liso/enzimologia , Adolescente , Adulto , Idoso , Asma/patologia , Estudos de Casos e Controles , Células Cultivadas , Criança , Ciclo-Oxigenase 2/genética , Feminino , Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Estabilidade de RNA , Sistema Respiratório/patologia , Regulação para Cima , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno
10.
Am J Physiol Lung Cell Mol Physiol ; 307(9): L727-34, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25217662

RESUMO

MicroRNA (miR)-146a and miR-146b are negative regulators of inflammatory gene expression in lung fibroblasts, epithelial cells, monocytes, and endothelial cells. The abundance of cyclooxygenase-2 (COX-2) and IL-1ß is negatively regulated by the miR-146 family, suggesting miR-146a and/or miR-146b might modulate inflammatory mediator expression in airway smooth muscle thereby contributing to pathogenesis of asthma. To test this idea we compared miR-146a and miR-146b expression in human airway smooth muscle cells (hASMCs) from nonasthmatic and asthmatic subjects treated with cytomix (IL-1ß, TNF-α, and IFNγ) and examined the miRNAs' effects on COX-2 and IL-1ß expression. We found that cytomix treatment elevated miR-146a and miR-146b abundance. Induction with cytomix was greater than induction with individual cytokines, and asthmatic cells exhibited higher levels of miR-146a expression following cytomix treatment than nonasthmatic cells. Transfection of miR-146a or miR-146b mimics reduced COX-2 and IL-1ß expression. A miR-146a inhibitor increased COX-2 and IL-1ß expression, but a miR-146b inhibitor was ineffective. Repression of COX-2 and IL-1ß expression by miR-146a correlated with reduced abundance of the RNA-binding protein human antigen R. These results demonstrate that miR-146a and miR-146b expression is inducible in hASMCs by proinflammatory cytokines and that miR-146a expression is greater in asthmatic cells. Both miR-146a and miR-146b can negatively regulate COX-2 and IL-1ß expression at pharmacological levels, but loss-of-function studies showed that only miR-146a is an endogenous negative regulator in hASMCs. The results suggest miR-146 mimics may be an attractive candidate for further preclinical studies as an anti-inflammatory treatment of asthma.


Assuntos
Asma/genética , Ciclo-Oxigenase 2/genética , Proteínas ELAV/genética , MicroRNAs/genética , Miócitos de Músculo Liso/metabolismo , Mucosa Respiratória/metabolismo , Asma/metabolismo , Asma/patologia , Ciclo-Oxigenase 2/metabolismo , Proteínas ELAV/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interferon gama/farmacologia , Interleucina-1beta/biossíntese , Interleucina-1beta/farmacologia , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/patologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia
11.
Am J Physiol Lung Cell Mol Physiol ; 307(10): L775-80, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25260754

RESUMO

G protein-coupled receptors are the most pervasive signaling superfamily in the body and act as receptors to endogenous agonists and drugs. For ß-agonist-mediated bronchodilation, the receptor-G protein-effector network consists of the ß2-adrenergic receptor (ß2AR), Gs, and adenylyl cyclase, expressed on airway smooth muscle (ASM). Using ASM-targeted transgenesis, we previously explored which of these three early signaling elements represents a limiting factor, or bottleneck, in transmission of the signal from agonist binding to ASM relaxation. Here we overexpressed Gαs in transgenic mice and found that agonist-promoted relaxation of airways was enhanced in direct proportion to the level of Gαs expression. Contraction of ASM from acetylcholine was not affected in Gαs transgenic mice, nor was relaxation by bitter taste receptors. Furthermore, agonist-promoted (but not basal) cAMP production in ASM cells from Gαs-transgenic mice was enhanced compared with ASM from nontransgenic littermates. Agonist-promoted inhibition of platelet-derived growth factor-stimulated ASM proliferation was also enhanced in Gαs mouse ASM. The enhanced maximal ß-agonist response was of similar magnitude for relaxation, cAMP production, and growth inhibition. Taken together, it appears that a limiting factor in ß-agonist responsiveness in ASM is the expression level of Gαs. Gene therapy or pharmacological means of increasing Gαs (or its coupling efficiency to ß2AR) thus represent an interface for development of novel therapeutic agents for improvement of ß-agonist therapy.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/biossíntese , Músculo Liso/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Sistema Respiratório/metabolismo , Transdução de Sinais , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Broncodilatadores/farmacologia , Linhagem Celular , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Camundongos , Camundongos Transgênicos , Músculo Liso/patologia , Receptores Adrenérgicos beta 2/genética , Sistema Respiratório/patologia
12.
Curr Opin Pharmacol ; 13(3): 324-30, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23639507

RESUMO

Significant advances in understanding the cell and molecular biology of inflammation and airway smooth muscle (ASM) contractility have identified several potential novel targets for therapies of asthma. New agents targeting G-protein coupled receptors (GPCRs) including bitter taste receptors (TAS2R) agonists and prostaglandin EP4 receptor agonists elicit ASM relaxation. The cAMP/PKA pathway continues to be a promising drug target with the emergence of new PDE inhibitors and a novel PKA target protein, HSP20, which mediates smooth muscle relaxation via actin depolymerization. Smooth muscle relaxation can also be elicited by inhibitors of the RhoA/Rho kinase pathway via inhibition of myosin light chain phosphorylation and actin depolymerization. Targeting epigenetic processes that control chromatin remodeling and RNA-induced gene silencing in airway cells also holds great potential for novel asthma therapy. Further investigation may identify agents that inhibit smooth muscle contraction and/or restrain or reverse obstructive remodeling of the airways.


Assuntos
Asma/metabolismo , Animais , Asma/tratamento farmacológico , Asma/fisiopatologia , Proteínas de Choque Térmico HSP20/metabolismo , Histona Desacetilases/metabolismo , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Receptores Acoplados a Proteínas G/agonistas , Receptores de Prostaglandina E Subtipo EP4/agonistas , Quinases Associadas a rho/metabolismo
13.
Pulm Pharmacol Ther ; 26(1): 64-74, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22634303

RESUMO

Airway smooth muscle (ASM) is the main regulator of bronchomotor tone. Extensive studies show that in addition to their physical property, human airway smooth muscle (ASM) cells can participate in inflammatory processes modulating the initiation, perpetuation, amplification, and perhaps resolution of airway inflammation. Upon stimulation or interaction with immune cells, ASM cells produce and secrete a variety of inflammatory cytokines and chemokines, cell adhesion molecules, and extracellular matrix (ECM) proteins. These released mediators can, in turn, contribute to the inflammatory state, airway hyperresponsiveness, and airway remodeling present in asthma. As our knowledge of ASM myocyte biology improves, novel bioactive factors are emerging as potentially important regulators of inflammation. This review provides an overview of our understanding of some of these molecules, identifies rising questions, and proposes future studies to better define their role in ASM cell modulation of inflammation and immunity in the lung and respiratory diseases.


Assuntos
Inflamação/patologia , Miócitos de Músculo Liso/metabolismo , Doenças Respiratórias/fisiopatologia , Remodelação das Vias Aéreas/imunologia , Animais , Asma/imunologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/fisiopatologia , Humanos , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Pneumopatias/imunologia , Pneumopatias/fisiopatologia , Músculo Liso/citologia , Músculo Liso/imunologia , Músculo Liso/metabolismo , Miócitos de Músculo Liso/imunologia , Doenças Respiratórias/imunologia
14.
Am J Respir Cell Mol Biol ; 48(3): 346-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23239497

RESUMO

In asthma, basic fibroblast growth factor (FGF-2) plays an important (patho)physiological role. This study examines the effects of FGF-2 on the transforming growth factor-ß (TGF-ß)-stimulated differentiation of airway smooth muscle (ASM) cells in vitro. The differentiation of human ASM cells after incubation with TGF-ß (100 pM) and/or FGF-2 (300 pM) for 48 hours was assessed by increases in contractile protein expression, actin-cytoskeleton reorganization, enhancements in cell stiffness, and collagen remodeling. FGF-2 inhibited TGF-ß-stimulated increases in transgelin (SM22) and calponin gene expression (n = 15, P < 0.01) in an extracellular signal-regulated kinase 1/2 (ERK1/2) signal transduction-dependent manner. The abundance of ordered α-smooth muscle actin (α-SMA) filaments formed in the presence of TGF-ß were also reduced by FGF-2, as was the ratio of F-actin to G-actin (n = 8, P < 0.01). Furthermore, FGF-2 attenuated TGF-ß-stimulated increases in ASM cell stiffness and the ASM-mediated contraction of lattices, composed of collagen fibrils (n = 5, P < 0.01). However, the TGF-ß-stimulated production of IL-6 was not influenced by FGF-2 (n = 4, P > 0.05), suggesting that FGF-2 antagonism is selective for the regulation of ASM cell contractile protein expression, organization, and function. Another mitogen, thrombin (0.3 U ml(-1)), exerted no effect on TGF-ß-regulated contractile protein expression (n = 8, P > 0.05), α-SMA organization, or the ratio of F-actin to G-actin (n = 4, P > 0.05), suggesting that the inhibitory effect of FGF-2 is dissociated from its mitogenic actions. The addition of FGF-2, 24 hours after TGF-ß treatment, still reduced contractile protein expression, even when the TGF-ß-receptor kinase inhibitor, SB431542 (10 µM), was added 1 hour before FGF-2. We conclude that the ASM cell differentiation promoted by TGF-ß is antagonized by FGF-2. A better understanding of the mechanism of action for FGF-2 is necessary to develop a strategy for therapeutic exploitation in the treatment of asthma.


Assuntos
Diferenciação Celular/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Músculo Liso/citologia , Miócitos de Músculo Liso/citologia , Sistema Respiratório/citologia , Sistema Respiratório/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Actinas/genética , Actinas/metabolismo , Asma/genética , Asma/metabolismo , Asma/patologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Proteínas Elk-1 do Domínio ets/genética , Proteínas Elk-1 do Domínio ets/metabolismo , Calponinas
15.
Biochem J ; 446(1): 89-98, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22625849

RESUMO

Silencing of GATA5 gene expression as a result of promoter hypermethylation has been observed in lung, gastrointestinal and ovarian cancers. However, the regulation of GATA5 gene expression has been poorly understood. In the present study, we have demonstrated that an E (enhancer)-box in the GATA5 promoter (bp -118 to -113 in mice; bp -164 to -159 in humans) positively regulates GATA5 transcription by binding USF1 (upstream stimulatory factor 1). Using site-directed mutagenesis, EMSA (electrophoretic mobility-shift analysis) and affinity chromatography, we found that USF1 specifically binds to the E-box sequence (5'-CACGTG-3'), but not to a mutated E-box. CpG methylation of this E-box significantly diminished its binding of transcription factors. Mutation of the E-box within a GATA5 promoter fragment significantly decreased promoter activity in a luciferase reporter assay. Chromatin immunoprecipitation identified that USF1 physiologically interacts with the GATA5 promoter E-box in mouse intestinal mucosa, which has the highest GATA5 gene expression in mouse. Co-transfection with a USF1 expression plasmid significantly increased GATA5 promoter-driven luciferase transcription. Furthermore, real-time and RT (reverse transcription)-PCR analyses confirmed that overexpression of USF1 activates endogenous GATA5 gene expression in human bronchial epithelial cells. The present study provides the first evidence that USF1 activates GATA5 gene expression through the E-box motif and suggests a potential mechanism (disruption of the E-box) by which GATA5 promoter methylation reduces GATA5 expression in cancer.


Assuntos
Elementos E-Box , Fator de Transcrição GATA5/genética , Regiões Promotoras Genéticas , Fatores Estimuladores Upstream/metabolismo , Animais , Sítios de Ligação , Brônquios/citologia , Células Epiteliais/metabolismo , Fator de Transcrição GATA5/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Sequências Reguladoras de Ácido Nucleico , Fatores Estimuladores Upstream/genética
16.
J Allergy Ther ; 3(Suppl 1)2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25984393

RESUMO

Airway inflammation, lung remodeling, and Airway Hyperresponsiveness (AHR) are major features of asthma and Chronic Obstructive Pulmonary Disease (COPD). The inflammatory response to allergens, air pollutants, and other insults is likely to play a key role in promoting structural changes in the lung including the overabundance of Airway Smooth Muscle (ASM) seen in asthmatics. These alterations or remodeling could, in turn, impact the immunmodulatory actions of the ASM, the ASM's contractile properties, and the development of AHR. New evidences suggest that airway inflammation and AHR are not tightly related to each other and that the structural component of the airway, mainly the ASM, is a chief driver of AHR. Members of the S100/calgranulins family have been implicated in the regulation of inflammation and cell apoptosis in various systems. S100A12 is highly expressed in neutrophils and is one of the most abundant proteins in the lungs of patients with asthma or COPD. Studies with genetic engineered mice with smooth muscle cell-targeted expression of human S100A12 revealed that S100A12 reduces airway smooth muscle amounts and dampens airway inflammation and airway hyperreactivity in a model of allergic lung inflammation. Thus, targeting airway smooth muscle for instance through delivery of pro-apoptotic S100A12 could represent an attractive means to promote ASM apoptosis and to reduce ASM abundance in asthmatics.

17.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L701-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21378028

RESUMO

Airway smooth muscle (ASM) hypertrophy is a cardinal feature of severe asthma, but the underlying molecular mechanisms remain uncertain. Forced protein kinase B/Akt 1 activation is known to induce myocyte hypertrophy in other muscle types, and, since a number of mediators present in asthmatic airways can activate Akt signaling, we hypothesized that Akt activation could contribute to ASM hypertrophy in asthma. To test this hypothesis, we evaluated whether Akt activation occurs naturally within airway myocytes in situ, whether Akt1 activation is sufficient to cause hypertrophy of normal airway myocytes, and whether such hypertrophy is accompanied by excessive accumulation of contractile apparatus proteins (contractile phenotype maturation). Immunostains of human airway sections revealed concordant activation of Akt (reflected in Ser(473) phosphorylation) and of its downstream effector p70(S6Kinase) (reflected in Thr(389) phosphorylation) within airway muscle bundles, but there was no phosphorylation of the alternative Akt downstream target glycogen synthase kinase (GSK) 3ß. Artificial overexpression of constitutively active Akt1 (by plasmid transduction or lentiviral infection) caused a progressive increase in size and protein content of cultured canine tracheal myocytes and increased p70(S6Kinase) phosphorylation but not GSK3ß phosphorylation; however, constitutively active Akt1 did not cause disproportionate overaccumulation of smooth muscle (sm) α-actin and SM22. Furthermore, mRNAs encoding sm-α-actin and SM22 were reduced. These results indicate that forced Akt1 signaling causes hypertrophy of cultured airway myocytes without inducing further contractile phenotypic maturation, possibly because of opposing effects on contractile protein gene transcription and translation, and suggest that natural activation of Akt1 plays a similar role in asthmatic ASM.


Assuntos
Músculo Liso/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traqueia/patologia , Actinas/metabolismo , Animais , Asma/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteínas Contráteis/metabolismo , Cães , Ativação Enzimática , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Hipertrofia , Contração Muscular/fisiologia , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosforilação , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Traqueia/metabolismo
18.
Am J Respir Cell Mol Biol ; 45(3): 453-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21131446

RESUMO

We have previously shown that the transcription-promoting activity of serum response factor (SRF) is partially regulated by its extranuclear redistribution. In this study, we examined the cellular mechanisms that facilitate SRF nuclear entry in canine tracheal smooth muscle cells. We used in vitro pull-down assays to determine which karyopherin proteins bound SRF and found that SRF binds KPNA1 and KPNB1 through its nuclear localization sequence. Immunoprecipitation studies also demonstrated direct SRF-KPNA1 interaction in HEK293 cells. Import assays demonstrated that KPNA1 and KPNB1 together were sufficient to mediate rapid nuclear import of SRF-GFP. Our studies also suggest that SRF is able to gain nuclear entry through an auxiliary, nuclear localization sequence-independent mechanism.


Assuntos
Transporte Ativo do Núcleo Celular , Músculo Liso/citologia , Fator de Resposta Sérica/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Dimerização , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Microscopia de Fluorescência/métodos , Modelos Biológicos , Mutação , Ligação Proteica , Proteínas Recombinantes de Fusão/química , alfa Carioferinas/metabolismo
19.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1214-22, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19779014

RESUMO

GATA5 is a member of the GATA zinc finger transcription factor family involved in tissue-specific transcriptional regulation during cell differentiation and embryogenesis. Previous reports indicate that null mutation of the zebrafish GATA5 gene results in embryonic lethality, whereas deletion of exon 1 from the mouse GATA5 gene causes only derangement of female urogenital development. Here, we have identified an alternate promoter within intron 1 of the mouse GATA5 gene that transcribes a 2.5-kb mRNA that lacks exon 1 entirely but includes 82 bp from intron 1 and all of exons 2-6. The alternative promoter was active during transient transfection in cultured airway myocytes and bronchial epithelial cells, and it drove reporter gene expression in gastric epithelial cells in transgenic mice. The 2.5-kb alternative transcript encodes an NH(2)-terminally truncated "short GATA5" comprising aa 226-404 with a single zinc finger, which retains ability to transactivate the atrial natriuretic factor promoter (albeit less efficiently than full-length GATA5). Another new GATA5 transcript contains all of exons 1-5 and the 5' portion of exon 6 but lacks the terminal 1143 bp of the 3'-untranslated region from exon 6. These findings extend current understanding of the tissue distribution of GATA5 expression and suggests that GATA5 expression and function are more complex than previously appreciated.


Assuntos
Processamento Alternativo , Fator de Transcrição GATA5/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Região 5'-Flanqueadora , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Northern Blotting , Cães , Células Epiteliais/metabolismo , Éxons , Fator de Transcrição GATA5/metabolismo , Mucosa Gástrica/metabolismo , Genes Reporter , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos de Músculo Liso/metabolismo , Células NIH 3T3 , Isoformas de Proteínas , Técnica de Amplificação ao Acaso de DNA Polimórfico , Mucosa Respiratória/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sítio de Iniciação de Transcrição , Ativação Transcricional , Transfecção
20.
Transl Res ; 154(4): 165-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19766960

RESUMO

Asthma is a complex respiratory disease whose incidence has increased worldwide in the last decade. Currently there is no cure for asthma. Although bronchodilator and anti-inflammatory medications are effective medicines in some asthmatic patients, it is clear that an unmet therapeutic need persists for a subpopulation of individuals with severe asthma. This chronic lung disease is characterized by airflow limitation, lung inflammation, and remodeling that includes increased airway smooth muscle (ASM) mass. In addition to its contractile properties, the ASM also contributes to the inflammatory process by producing active mediators, which modify the extracellular matrix composition and interact with inflammatory cells. These undesirable functions make interventions aimed at reducing ASM abundance an attractive strategy for novel asthma therapies. The following three mechanisms could limit the accumulation of smooth muscle: decreased cell proliferation, augmented cell apoptosis, and reduced cell migration into the smooth muscle layer. Inhibitors of the mevalonate pathway or statins hold promise for asthma treatment, because they exhibit anti-inflammatory, antimigratory, and antiproliferative effects in preclinical and clinical studies, and they can target the smooth muscle. This review will discuss current knowledge of ASM biology and identify gaps in the field to stimulate future investigations of the cellular mechanisms that control ASM overabundance in asthma. Targeting ASM has the potential to be an innovative venue of treatment for patients with asthma.


Assuntos
Antiasmáticos/uso terapêutico , Asma/fisiopatologia , Asma/terapia , Músculo Liso/fisiopatologia , Obstrução das Vias Respiratórias/fisiopatologia , Asma/tratamento farmacológico , Asma/patologia , Técnicas de Cultura de Células/métodos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Músculo Liso/patologia , Ensaios Clínicos Controlados Aleatórios como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA