Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Am J Respir Cell Mol Biol ; 52(4): 438-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25180620

RESUMEN

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.


Asunto(s)
Asma/enzimología , Ciclooxigenasa 2/metabolismo , MicroARNs/metabolismo , Miocitos del Músculo Liso/enzimología , Adolescente , Adulto , Anciano , Asma/patología , Estudios de Casos y Controles , Células Cultivadas , Niño , Ciclooxigenasa 2/genética , Femenino , Expresión Génica , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Estabilidad del ARN , Sistema Respiratorio/patología , Regulación hacia Arriba , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos
2.
Am J Physiol Lung Cell Mol Physiol ; 307(9): L727-34, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25217662

RESUMEN

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.


Asunto(s)
Asma/genética , Ciclooxigenasa 2/genética , Proteínas ELAV/genética , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Mucosa Respiratoria/metabolismo , Asma/metabolismo , Asma/patología , Ciclooxigenasa 2/metabolismo , Proteínas ELAV/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/farmacología , Interleucina-1beta/biosíntesis , Interleucina-1beta/farmacología , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
3.
Biochem J ; 446(1): 89-98, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22625849

RESUMEN

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.


Asunto(s)
Elementos E-Box , Factor de Transcripción GATA5/genética , Regiones Promotoras Genéticas , Factores Estimuladores hacia 5'/metabolismo , Animales , Sitios de Unión , Bronquios/citología , Células Epiteliales/metabolismo , Factor de Transcripción GATA5/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Ratones , Secuencias Reguladoras de Ácidos Nucleicos , Factores Estimuladores hacia 5'/genética
4.
Am J Respir Cell Mol Biol ; 45(3): 453-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21131446

RESUMEN

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.


Asunto(s)
Transporte Activo de Núcleo Celular , Músculo Liso/citología , Factor de Respuesta Sérica/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Dimerización , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoprecipitación , Microscopía Fluorescente/métodos , Modelos Biológicos , Mutación , Unión Proteica , Proteínas Recombinantes de Fusión/química , alfa Carioferinas/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L701-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21378028

RESUMEN

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.


Asunto(s)
Músculo Liso/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tráquea/patología , Actinas/metabolismo , Animales , Asma/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas Contráctiles/metabolismo , Perros , Activación Enzimática , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipertrofia , Contracción Muscular/fisiología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosforilación , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Tráquea/metabolismo
6.
Am J Respir Crit Care Med ; 179(3): 194-204, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19011151

RESUMEN

RATIONALE: Airway smooth muscle (SM) of patients with asthma exhibits a greater velocity of shortening (Vmax) than that of normal subjects, and this is thought to contribute to airway hyperresponsiveness. A greater Vmax can result from increased myosin activation. This has been reported in sensitized human airway SM and in models of asthma. A faster Vmax can also result from the expression of specific contractile proteins that promote faster cross-bridge cycling. This possibility has never been addressed in asthma. OBJECTIVES: We tested the hypothesis that the expression of genes coding for SM contractile proteins is altered in asthmatic airways and contributes to their increased Vmax. METHODS: We quantified the expression of several genes that code for SM contractile proteins in mild allergic asthmatic and control human airway endobronchial biopsies. The function of these contractile proteins was tested using the in vitro motility assay. MEASUREMENTS AND MAIN RESULTS: We observed an increased expression of the fast myosin heavy chain isoform, transgelin, and myosin light chain kinase in patients with asthma. Immunohistochemistry demonstrated the expression of these genes at the protein level. To address the functional significance of this overexpression, we purified tracheal myosin from the hyperresponsive Fisher rats, which also overexpress the fast myosin heavy chain isoform as compared with the normoresponsive Lewis rats, and found a faster rate of actin filament propulsion. Conversely, transgelin did not alter the rate of actin filament propulsion. CONCLUSIONS: Selective overexpression of airway smooth muscle genes in asthmatic airways leads to increased Vmax, thus contributing to the airway hyperresponsiveness observed in asthma.


Asunto(s)
Asma/metabolismo , Expresión Génica , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso/metabolismo , Cadenas Pesadas de Miosina/genética , Quinasa de Cadena Ligera de Miosina/genética , ARN Mensajero/genética , Adolescente , Animales , Asma/genética , Asma/patología , Biopsia , Western Blotting , Broncoscopía , Electroforesis en Gel de Poliacrilamida , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Masculino , Proteínas de Microfilamentos/biosíntesis , Proteínas Musculares/biosíntesis , Músculo Liso/patología , Cadenas Pesadas de Miosina/biosíntesis , Quinasa de Cadena Ligera de Miosina/biosíntesis , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Proteínas de Xenopus
7.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1214-22, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19779014

RESUMEN

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.


Asunto(s)
Empalme Alternativo , Factor de Transcripción GATA5/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Región de Flanqueo 5' , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Northern Blotting , Perros , Células Epiteliales/metabolismo , Exones , Factor de Transcripción GATA5/metabolismo , Mucosa Gástrica/metabolismo , Genes Reporteros , Intrones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo , Células 3T3 NIH , Isoformas de Proteínas , Técnica del ADN Polimorfo Amplificado Aleatorio , Mucosa Respiratoria/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sitio de Iniciación de la Transcripción , Activación Transcripcional , Transfección
8.
Clin Rev Allergy Immunol ; 24(1): 73-84, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12644719

RESUMEN

Bronchial hyperresponsiveness (BHR), the occurrence of excessive bronchoconstriction in response to relatively small constrictor stimuli, is a cardinal feature of asthma. Here, we consider the role that airway smooth muscle might play in the generation of BHR. The weight of evidence suggests that smooth muscle isolated from asthmatic tissues exhibits normal sensitivity to constrictor agonists when studied during isometric contraction, but the increased muscle mass within asthmatic airways might generate more total force than the lesser amount of muscle found in normal bronchi. Another salient difference between asthmatic and normal individuals lies in the effect of deep inhalation (DI) on bronchoconstriction. DI often substantially reverses induced bronchoconstriction in normals, while it often has much less effect on spontaneous or induced bronchoconstriction in asthmatics. It has been proposed that abnormal dynamic aspects of airway smooth muscle contraction velocity of contraction or plasticity- elasticity balance might underlie the abnormal DI response in asthma. We suggest a speculative model in which abnormally long actin filaments might account for abnormally increased elasticity of contracted airway smooth muscle.


Asunto(s)
Asma/fisiopatología , Bronquios/fisiopatología , Hiperreactividad Bronquial/fisiopatología , Músculo Liso/fisiopatología , Actinas/fisiología , Animales , Bronquios/efectos de los fármacos , Broncoconstrictores/farmacología , Relación Dosis-Respuesta a Droga , Elasticidad , Histamina/farmacología , Humanos , Cloruro de Metacolina/farmacología , Respiración
10.
J Biol Chem ; 284(36): 24123-32, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19586906

RESUMEN

Lysophosphatidic acid (LPA), a bioactive phospholipid, induces a wide range of cellular effects, including gene expression, cytoskeletal rearrangement, and cell survival. We have previously shown that LPA stimulates secretion of pro- and anti-inflammatory cytokines in bronchial epithelial cells. This study provides evidence that LPA enhances pulmonary epithelial barrier integrity through protein kinase C (PKC) delta- and zeta-mediated E-cadherin accumulation at cell-cell junctions. Treatment of human bronchial epithelial cells (HBEpCs) with LPA increased transepithelial electrical resistance (TER) by approximately 2.0-fold and enhanced accumulation of E-cadherin to the cell-cell junctions through Galpha(i)-coupled LPA receptors. Knockdown of E-cadherin with E-cadherin small interfering RNA or pretreatment with EGTA (0.1 mm) prior to LPA (1 microm) treatment attenuated LPA-induced increases in TER in HBEpCs. Furthermore, LPA induced tyrosine phosphorylation of focal adhesion kinase (FAK) and overexpression of the FAK inhibitor, and FAK-related non-kinase-attenuated LPA induced increases in TER and E-cadherin accumulation at cell-cell junctions. Overexpression of dominant negative protein kinase delta and zeta attenuated LPA-induced phosphorylation of FAK, accumulation of E-cadherin at cell-cell junctions, and an increase in TER. Additionally, lipopolysaccharide decreased TER and induced E-cadherin relocalization from cell-cell junctions to cytoplasm in a dose-dependent fashion, which was restored by LPA post-treatment in HBEpCs. Intratracheal post-treatment with LPA (5 microm) reduced LPS-induced neutrophil influx, protein leak, and E-cadherin shedding in bronchoalveolar lavage fluids in a murine model of acute lung injury. These data suggest a protective role of LPA in airway inflammation and remodeling.


Asunto(s)
Células Epiteliales/metabolismo , Lipopolisacáridos/toxicidad , Lesión Pulmonar/metabolismo , Lesión Pulmonar/prevención & control , Lisofosfolípidos/farmacología , Mucosa Respiratoria/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Células Epiteliales/patología , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Mucosa Respiratoria/patología
11.
J Biol Chem ; 281(29): 20383-92, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16690609

RESUMEN

Transforming growth factor (TGF)-beta is present in large amounts in the airways of patients with asthma and with other diseases of the lung. We show here that TGFbeta treatment increased transcriptional activation of SM22alpha, a smooth muscle-specific promoter, in airway smooth muscle cells, and we demonstrate that this effect stems in part from TGFbeta-induced enhancement of serum response factor (SRF) DNA binding and transcription promoting activity. Overexpression of Smad7 inhibited TGFbeta-induced stimulation of SRF-dependent promoter function, and chromatin immunoprecipitation as well as co-immunoprecipitation assays established that endogenous or recombinant SRF interacts with Smad7 within the nucleus. The SRF binding domain of Smad7 mapped to the C-terminal half of the Smad7 molecule. TGFbeta treatment weakened Smad7 association with SRF, and conversely the Smad7-SRF interaction was increased by inhibition of the TGFbeta pathway through overexpression of a dominant negative mutant of TGFbeta receptor I or of Smad3 phosphorylation-deficient mutant. Our findings thus reveal that SRF-Smad7 interactions in part mediate TGFbeta regulation of gene transcription in airway smooth muscle. This offers potential targets for interventions in treating lung inflammation and asthma.


Asunto(s)
Músculo Liso/fisiología , Factor de Respuesta Sérica/fisiología , Proteína smad7/genética , Proteína smad7/metabolismo , Tráquea/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Sitios de Unión , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Perros , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Regiones Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Transcripción Genética/efectos de los fármacos , Transfección , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Repeticiones de Trinucleótidos
12.
Am J Hum Genet ; 76(2): 349-57, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15611928

RESUMEN

Asthma affects nearly 14 million people worldwide and has been steadily increasing in frequency for the past 50 years. Although environmental factors clearly influence the onset, progression, and severity of this disease, family and twin studies indicate that genetic variation also influences susceptibility. Linkage of asthma and related phenotypes to chromosome 6p21 has been reported in seven genome screens, making it the most replicated region of the genome. However, because many genes with individually small effects are likely to contribute to risk, identification of asthma susceptibility loci has been challenging. In this study, we present evidence from four independent samples in support of HLA-G as a novel asthma and bronchial hyperresponsiveness susceptibility gene in the human leukocyte antigen region on chromosome 6p21, and we speculate that this gene might contribute to risk for other inflammatory diseases that show linkage to this region.


Asunto(s)
Asma/genética , Mapeo Cromosómico , Cromosomas Humanos Par 6/genética , Predisposición Genética a la Enfermedad , Antígenos HLA/genética , Antígenos de Histocompatibilidad Clase I/genética , Adulto , Hiperreactividad Bronquial/genética , Niño , Femenino , Pruebas Genéticas , Antígenos HLA-G , Humanos , Masculino , Fenotipo
13.
J Allergy Clin Immunol ; 111(3): 527-32, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12642833

RESUMEN

BACKGROUND: Muscarinic acetylcholine receptors are members of the superfamily of G protein-coupled, 7 transmembrane- spanning proteins. They are important in the development of airway hyperresponsiveness. In the lung the M3 receptor, encoded by the cholinergic receptor muscarinic 3 gene, is present in airway smooth muscle and mediates smooth muscle contraction. OBJECTIVE: We considered the cholinergic receptor muscarinic 3 gene as a possible candidate gene for bronchial asthma and initiated studies to identify polymorphisms in the promoter region. METHOD: We identified 4 single-nucleotide polymorphisms (-708A/G, -627G/C, -513C/A, and -492C/T) and 2 short tandem repeat polymorphisms, a tetranucleotide (CTTT)12-20 and a dinucleotide (GT)6-19 repeat. RESULTS: None of the identified single nucleotide polymorphisms were significantly more frequent in asthmatic patients (n = 76) compared with in healthy control subjects (n = 81). Furthermore, there was no evidence for nonrandom transmission of short tandem repeat polymorphism haplotypes to individuals with asthma or bronchial hyperresponsiveness (P >.50) in a large Hutterite pedigree. However, there was significant nonrandom transmission of haplotypes to individuals with skin test reactivity to cockroach allergens (global transmission disequilibrium test: chi2 = 38.55, P =.013). CONCLUSIONS: These results suggest a possible role for this gene in atopic disorders.


Asunto(s)
Asma/genética , Variación Genética , Hipersensibilidad/genética , Regiones Promotoras Genéticas/genética , Receptores Muscarínicos/genética , Alérgenos/inmunología , Animales , Secuencia de Bases/genética , Cucarachas/inmunología , Frecuencia de los Genes , Haplotipos , Humanos , Hipersensibilidad/diagnóstico , Hipersensibilidad/inmunología , Linaje , Polimorfismo Genético , Polimorfismo de Nucleótido Simple , Receptor Muscarínico M3 , Valores de Referencia , Pruebas Cutáneas , Secuencias Repetidas en Tándem
14.
Am J Respir Cell Mol Biol ; 29(1): 39-47, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12600823

RESUMEN

RhoA and its downstream target Rho kinase regulate serum response factor (SRF)-dependent skeletal and smooth muscle gene expression. We previously reported that long-term serum deprivation reduces transcription of smooth muscle contractile apparatus encoding genes, by redistributing SRF out of the nucleus. Because serum components stimulate RhoA activity, these observations suggest the hypothesis that the RhoA/Rho kinase pathway regulates SRF-dependent smooth muscle gene transcription in part by controlling SRF subcellular localization. Our present results support this hypothesis: cotransfection of cultured airway myocytes with a plasmid expressing constitutively active RhoAV14 selectively enhanced transcription from the SM22 and smooth muscle myosin heavy chain promoters and from a purely SRF-dependent promoter, but had no effect on transcription from the MSV-LTR promoter or from an AP2-dependent promoter. Conversely, inhibition of the RhoA/Rho kinase pathway by cotransfection with a plasmid expressing dominant negative RhoAN19, by cotransfection with a plasmid expressing Clostridial C3 toxin, or by incubation with the Rho kinase inhibitor, Y-27632, all selectively reduced SRF-dependent smooth muscle promoter activity. Furthermore, treatment with Y-27632 selectively reduced binding of SRF from nuclear extracts to its consensus DNA target, selectively reduced nuclear SRF protein content, and partially redistributed SRF from nucleus to cytoplasm, as revealed by quantitative immunocytochemistry. Treatment of cultured airway myocytes with latrunculin B, which reduces actin polymerization, also caused partial redistribution of SRF into the cytoplasm. Together, these results demonstrate for the first time that the RhoA/Rho kinase pathway controls smooth muscle gene transcription in differentiated smooth muscle cells, in part by regulating the subcellular localization of SRF. It is conceivable that the RhoA/Rho kinase pathway influences SRF localization through its effect on actin polymerization dynamics.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Respuesta Sérica/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Amidas/farmacología , Animales , Toxinas Bacterianas/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Perros , Inhibidores Enzimáticos/farmacología , Péptidos y Proteínas de Señalización Intracelular , Células Musculares/citología , Células Musculares/metabolismo , Músculo Liso/fisiología , Cadenas Pesadas de Miosina/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Factor de Respuesta Sérica/efectos de los fármacos , Factor de Respuesta Sérica/genética , Transducción de Señal , Tiazoles/farmacología , Tiazolidinas , Tráquea/citología , Transcripción Genética , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/genética
15.
Am J Respir Cell Mol Biol ; 26(3): 298-305, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11867338

RESUMEN

We have isolated and characterized the human m3 muscarinic receptor gene and its promoter. Using 5' rapid amplification of cDNA ends (RACE), internal polymerase chain reaction (PCR), and homology searching to identify EST clones, we determined that the cDNA encoding the m3 receptor comprises 4,559 bp in 8 exons, which are alternatively spliced to exclude exons 2, 4, 6, and/or 7; the receptor coding sequence occurs within exon 8. Analysis of P1 artificial chromosome (PAC) and bacterial artificial chromosome (BAC) clones and of PCR- amplified genomic DNA, and homology searching of human chromosome 1 sequence provided from the Sanger Centre (Hinxton, Cambridge, UK) revealed that the m3 muscarinic receptor gene spans at least 285 kb. A promoter fragment containing bp -1240 to +101 (relative to the most 5' transcription start site) exhibited considerable transcriptional activity during transient transfection in cultured subconfluent, serum-fed canine tracheal myocytes, and 5' deletion analysis of promoter function revealed the presence of positive transcriptional regulatory elements between bp -526 and -269. Sequence analysis disclosed three potential AP-2 binding sites in this region; five more AP-2 consensus binding motifs occur between bp -269 and +101. Cotransfection with a plasmid expressing human AP-2alpha substantially increased transcription from m3 receptor promoter constructs containing 526 or 269 bp of 5' flanking DNA. Furthermore, m3 receptor promoter activity was enhanced by long-term serum deprivation of canine tracheal myocytes, a treatment that is known to increase AP-2 transcription-promoting activity in these cells. Together, these data suggest that expression of the human m3 muscarinic receptor gene is regulated in part by AP-2 in airway smooth muscle.


Asunto(s)
Genoma Humano , Regiones Promotoras Genéticas , Receptores Muscarínicos/genética , Empalme Alternativo , Animales , Secuencia de Bases , Células Cultivadas , ADN Complementario/análisis , ADN Complementario/genética , Perros , Exones/genética , Humanos , Datos de Secuencia Molecular , Receptor Muscarínico M3 , Alineación de Secuencia , Análisis de Secuencia de ADN , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA