Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Toxicol Appl Pharmacol ; 472: 116573, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37269932

RESUMEN

Arsenic is associated with the development of breast cancer. However, the molecular mechanisms of arsenic induction of breast cancer are not fully defined. Interaction with zinc finger (ZnF) motifs in proteins is one of the proposed mechanisms of arsenic toxicity. GATA3 is a transcription factor that regulates the transcription of genes associated with cell proliferation, cell differentiation and the epithelial-mesenchymal transition (EMT) in mammary luminal cells. Given that GATA3 possesses two ZnF motifs essential for the function of this protein and that arsenic could alter the function of GATA3 through interaction with these structural motifs, we evaluated the effect of sodium arsenite (NaAsO2) on GATA3 function and its relevance in the development of arsenic-induced breast cancer. Breast cell lines derived from normal mammary epithelium (MCF-10A), hormone receptor-positive and hormone receptor negative breast cancer cells (T-47D and MDA-MB-453, respectively) were used. We observed a reduction on GATA3 protein levels at non-cytotoxic concentrations of NaAsO2 in MCF-10A and T-47D, but not in MDA-MB-453 cells. This reduction was associated with an increase in cell proliferation and cell migration in MCF-10A, but not in T-47D or MDA-MB-453 cells. The evaluation of cell proliferation and EMT markers indicate that the reduction on GATA3 protein levels by arsenic, disrupts the function of this transcription factor. Our data indicate that GATA3 is a tumor suppressor in the normal mammary epithelium and that arsenic could act as an initiator of breast cancer by disrupting the function of GATA3.


Asunto(s)
Arsénico , Neoplasias de la Mama , Factor de Transcripción GATA3 , Femenino , Humanos , Arsénico/toxicidad , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Células Epiteliales/metabolismo , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/metabolismo , Factores de Transcripción
2.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 63-71, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33434281

RESUMEN

Myristica fragrans is a traditional herbal medicine and has been shown to alleviate the development of atherosclerosis. However, the anti-atherogenic mechanisms of M. fragrans are still to be addressed. In this study, we explored the effect of M. fragrans on lipid metabolism and inflammation and its mechanisms in THP-1-derived macrophages. The quantitative polymerase chain reaction and western blot analysis results showed that M. fragrans promotes cholesterol efflux from THP-1-derived macrophages and reduces intracellular total cholesterol, cholesterol ester, and free cholesterol contents in a dose- and a time-dependent manner. Further study found that liver X receptor alpha (LXRα) antagonist GGPP significantly blocked the upregulation of ABCA1 expression with M. fragrans treatment. In addition, chromatin immunoprecipitation assay confirmed that GATA binding protein 3 (GATA3) can bind to the LXRα promoter, and inhibition of GATA3 led to the downregulation of LXRα and ATP-binding cassette subfamily A member 1 expression. Furthermore, M. fragrans reduced lipid accumulation, followed by decreasing tumor necrosis factor-α, interleukin (IL)-6, and IL-1ß and increasing IL-10 produced by THP-1-derived macrophages. Therefore, M. fragrans is identified as a valuable therapeutic medicine for atherosclerotic cardiovascular disease.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transporte Biológico/efectos de los fármacos , Ésteres del Colesterol/metabolismo , Citocinas/metabolismo , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/análisis , Receptores X del Hígado/genética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Myristica , Regiones Promotoras Genéticas , Células THP-1/citología , Regulación hacia Arriba
3.
Exp Dermatol ; 29(7): 647-658, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32472715

RESUMEN

Psoriasis is a recrudescent chronic immune-mediated inflammatory dermatosis; the production and release of proinflammatory cytokines/chemokines such as TNF-α has been regarded as critical issues during psoriasis pathogenesis. Based on online microarray profiles, the expression of the transcription factor GATA3 was downregulated in psoriasis lesion tissues. In the present study, we searched for miRNAs that might be related to TNF-α and GATA3 to investigate an in-depth understanding of psoriasis pathogenesis. Herein, higher TNF-α and GATA3 protein levels were observed in psoriasis lesion tissues and that GATA3 overexpression significantly reverses TNF-α-induced increases within the production of IL-6 and CXCL8 in keratinocytes. TNF-α stimulation increases miR-155 expression dose-independently, and the miR-155 inhibitor significantly reverses TNF-α-induced suppression of GATA3 protein levels and increases IL-6 and CXCL8 production. miR-155 could suppress the expression of GATA3 by targeting its 3'UTR, while GATA3 could activate the transcription of IL37 by targeting its promoter region. miR-155 overexpression reduces IL37 protein and increases CXCL8 production; GATA3 overexpression might significantly attenuate the effects of miR-155 overexpression. In contrast to GATA3, miR-155 expression is significantly upregulated in psoriasis lesion tissue and is negatively correlated with GATA3 and IL37. In summary, the miR-155/GATA3/IL37 axis modulates the production of IL-6 and CXCL8 upon TNF-α stimulation to affect psoriasis development. Thus, miR-155/GATA3/IL37 may be potent targets for psoriasis treatment, which needs further in vivo and clinical investigation.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Interleucina-1/metabolismo , MicroARNs/metabolismo , Psoriasis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regiones no Traducidas 3' , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Células HaCaT , Humanos , Interleucina-1/genética , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Queratinocitos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Regiones Promotoras Genéticas , Psoriasis/genética , Piel/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología
4.
Microb Pathog ; 125: 313-317, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30278208

RESUMEN

The current study was aimed to investigate the effect of benzofuran on asthma neonatal rat model. Twenty-five neonatal rats were assigned into five groups; Normal control, untreated, 1 mg/kg, 8 mg/kg and 10 mg/kg treatment groups. Methacholine was administered orally to the rats of untreated and treatment groups. Animals in the normal control group were given PBS as a vehicle. FlexiVent system employing a computer-controlled mouse ventilator along with respiratory mechanics was used for the analysis of airway resistance in the rats. Cytokine level and IFN-γ in the rat serum samples was performed by ELISA in accordance with the instructions of manufacturer. Methacholine administration into the rats caused a marked increase in lung airway resistance. However, treatment with 8 and 10 mg/kg doses of benzofuran led to marked decrease in the airway resistance. Benzofuran treatment prevented accumulation of macrophages and inflammatory cells in the lung airways. Inhibition of inflammation in methacholine administered rats by benzofuran was also confirmed by hematoxylin & eosin-staining. Examination of the rat serum showed significantly higher level of Th2 cytokines (IL-4, -5 and -13) in the untreated rats. However, treatment of methacholine administered rats with benzofuran significantly inhibited Th2 cytokine expression. The level of IFN-γ was increased by benzofuran treatment in methacholine administered rats. In methacholine administered rats the level of IgE was markedly higher however treatment of asthma rats with benzofuran inhibited up-regulation of IgE significantly. The expression of T-bet is decreased and that of GATA-3 is increased by methacholine administration in the rat lungs. Benzofuran treatment of methacholine administered rats prevented reduction in T-bet and up-regulation of GATA-3 expression in the rat lungs. The effect of benzofuran was significant at the doses of 8 and 10 mg/kg and non-significant at 1 mg/kg. These finding suggest that benzofuran inhibits expression of dominant T-helper 2 cytokines through targeting GATA-binding protein 3 transcription factor. Thus benzofuran can be of therapeutic importance for the treatment of asthma.


Asunto(s)
Antiasmáticos/administración & dosificación , Asma/patología , Benzofuranos/administración & dosificación , Factor de Transcripción GATA3/antagonistas & inhibidores , Inflamación/prevención & control , Animales , Asma/inducido químicamente , Broncoconstrictores/administración & dosificación , Citocinas/sangre , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Histocitoquímica , Inflamación/patología , Pulmón/patología , Cloruro de Metacolina/administración & dosificación , Ratas , Suero/química , Resultado del Tratamiento
5.
Neurotoxicology ; 67: 296-304, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29966605

RESUMEN

Clioquinol was used in the mid-1900s as an amebicide to treat indigestion and diarrhea. However, it was withdrawn from the market in Japan because it was linked to subacute myelo-optic neuropathy (SMON). The pathogenesis of SMON has not yet been elucidated in detail. As reported previously, we performed a global analysis on human neuroblastoma cells using DNA chips. The global analysis and quantitative PCR demonstrated that the mRNA level of interleukin-8 (IL-8) was significantly increased when SH-SY5Y neuroblastoma cells were treated with clioquinol. An enzyme-linked immunosorbent assay also demonstrated that clioquinol induced the secretion of IL-8 into culture media. Promoter analyses on SH-SY5Y cells revealed that a region responsive to clioquinol exists between -152 and -144 of the human IL-8 gene, which contains a consensus GATA-binding site sequence. The introduction of mutations at this site or the activator protein (AP)-1 site sequence at -126/-120 significantly reduced clioquinol-induced transcriptional activation. Among the GATA transcription factors expressed in SH-SY5Y cells, GATA-2 and GATA-3 protein levels were significantly decreased by the addition of clioquinol. Electrophoresis mobility shift assays using a probe corresponding to -159/-113 of the human IL-8 gene revealed two major shifted bands, one of which was increased and the other was decreased by clioquinol. The introduction of mutations showed that the former corresponded to binding to the AP-1 site, and the latter to binding to the GATA site. Supershift analyses revealed that the binding of c-Jun and c-Fos was increased, whereas that of GATA-3 was decreased by clioquinol. Genome editing against GATA-2 or GATA-3, not GATA-4 significantly enhanced clioquinol-induced IL-8 mRNA expression. On the other hand, the stable expression of GATA-2 or GATA-3 attenuated clioquinol-induced IL-8 mRNA expression and IL-8 secretion. These results suggest that the clioquinol-induced suppression of GATA-2 and GATA-3 expression mediates the up-regulation of IL-8.


Asunto(s)
Clioquinol/farmacología , Regulación hacia Abajo/efectos de los fármacos , Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA3/metabolismo , Interleucina-8/biosíntesis , Secuencia de Bases , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/fisiología , Factor de Transcripción GATA2/antagonistas & inhibidores , Factor de Transcripción GATA3/antagonistas & inhibidores , Expresión Génica , Humanos , Interleucina-8/genética
6.
Int J Biochem Cell Biol ; 95: 100-107, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29275211

RESUMEN

Sleep apnea syndrome (SAS) is characterized by intermittent hypoxia (IH) during sleep. SAS and obesity are strongly related to each other. Here, we investigated the effect of IH on the expression of major appetite regulatory genes in human neuronal cells. We exposed NB-1, SH-SY5Y, and SK-N-SH human neuronal cells to IH (64 cycles of 5 min hypoxia and 10 min normoxia), normoxia, or sustained hypoxia for 24 h and measured the mRNA levels of proopiomelanocortin (POMC), cocaine- and amphetamine-regulated transcript (CART), galanin, galanin-like peptide, ghrelin, pyroglutamylated RFamide peptide, agouti-related peptide, neuropeptide Y, and melanocortin 4 receptor by real-time RT-PCR. IH significantly increased the mRNA levels of POMC and CART in all the neuronal cells. Deletion analysis revealed that the -705 to -686 promoter region of POMC and the -950 to -929 region of CART were essential for the IH-induced promoter activity. As possible GATA factor binding sequences were found in the two regions, we performed real-time RT-PCR to determine which GATA family members were expressed and found that GATA2 and GATA3 mRNAs were predominantly expressed. Therefore, we introduced siRNAs against GATA2 and GATA3 into NB-1 cells and found that GATA2 and GATA3 siRNAs abolished the IH-induced up-regulation of both POMC and CART mRNAs. These results indicate that IH stress up-regulates the mRNA levels of anorexigenic peptides, POMC and CART, in human neuronal cells via GATA2 and GATA3. IH can have an anorexigenic effect on SAS patients through the transcriptional activation of POMC and CART in the central nervous system.


Asunto(s)
Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Sitios de Unión , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Factor de Transcripción GATA2/antagonistas & inhibidores , Factor de Transcripción GATA2/química , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/química , Factor de Transcripción GATA3/genética , Eliminación de Gen , Genes Reporteros , Humanos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Proopiomelanocortina/química , Proopiomelanocortina/genética , Regiones Promotoras Genéticas , Síndromes de la Apnea del Sueño/metabolismo , Síndromes de la Apnea del Sueño/patología , Factores de Tiempo , Regulación hacia Arriba
7.
Drug Des Devel Ther ; 11: 1027-1033, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28408801

RESUMEN

Rheumatoid arthritis (RA) is the most common form of chronic inflammatory arthritis characterized by pain, swelling and destruction of joints, with a resultant disability. Disease-modifying anti-rheumatic drugs (DMARDs) and biological drugs can interfere with the disease process. In this study, the effect of ß-d-mannuronic acid (M2000) as a novel non-steroidal anti-inflammatory drug (NSAID) with immunosuppressive and anti-inflammatory effects together with antioxidant effects was evaluated on IL17, RORγt, IL4 and GATA3 gene expression in 12 RA patients. Previously, M2000 driven from sodium alginate (natural product; patented, DEU: 102016113018.4) has shown a notable efficacy in experimental models of multiple sclerosis, RA and nephrotic syndrome. This study was performed on 12 patients with RA who had an inadequate response to conventional treatments. During this trial, patients were permitted to continue the conventional therapy excluding NSAIDs. M2000 was administered orally at a dose of 500 mg twice daily for 12 weeks. The peripheral blood mononuclear cells (PBMCs) were collected before and after treatment to evaluate the expression levels of IL4, GATA3, IL17 and RORγt. The gene expression results showed that M2000 has a potent efficacy, so that it could not only significantly decrease IL17 and RORγt levels but also increase IL4 and GATA3 levels after 12 weeks of treatment. Moreover, the gene expression results were in accordance with the clinical and preclinical assessments. In conclusion, M2000 as a natural novel agent has therapeutic and immunosuppressive properties on RA patients (identifier: IRCT2014011213739N2).


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Ácidos Hexurónicos/uso terapéutico , Inmunosupresores/uso terapéutico , Administración Oral , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Artritis Reumatoide/genética , Femenino , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Perfilación de la Expresión Génica , Ácidos Hexurónicos/administración & dosificación , Ácidos Hexurónicos/farmacología , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-4/antagonistas & inhibidores , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Persona de Mediana Edad , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo
8.
Cell Death Differ ; 24(5): 809-818, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28282036

RESUMEN

Cell differentiation is affected by complex networks of transcription factors that co-ordinate re-organisation of the chromatin landscape. The hierarchies of these relationships can be difficult to dissect. During in vitro differentiation of normal human uro-epithelial cells, formaldehyde-assisted isolation of regulatory elements (FAIRE-seq) and RNA-seq was used to identify alterations in chromatin accessibility and gene expression changes following activation of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) as a differentiation-initiating event. Regions of chromatin identified by FAIRE-seq, as having altered accessibility during differentiation, were found to be enriched with sequence-specific binding motifs for transcription factors predicted to be involved in driving basal and differentiated urothelial cell phenotypes, including forkhead box A1 (FOXA1), P63, GRHL2, CTCF and GATA-binding protein 3 (GATA3). In addition, co-occurrence of GATA3 motifs was observed within subsets of differentiation-specific peaks containing P63 or FOXA1. Changes in abundance of GRHL2, GATA3 and P63 were observed in immunoblots of chromatin-enriched extracts. Transient siRNA knockdown of P63 revealed that P63 favoured a basal-like phenotype by inhibiting differentiation and promoting expression of basal marker genes. GATA3 siRNA prevented differentiation-associated downregulation of P63 protein and transcript, and demonstrated positive feedback of GATA3 on PPARG transcript, but showed no effect on FOXA1 transcript or protein expression. This approach indicates that as a transcriptionally regulated programme, urothelial differentiation operates as a heterarchy, wherein GATA3 is able to co-operate with FOXA1 to drive expression of luminal marker genes, but that P63 has potential to transrepress expression of the same genes.


Asunto(s)
Diferenciación Celular/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor de Transcripción GATA3/genética , Factor Nuclear 3-alfa del Hepatocito/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Línea Celular , Cromatina/química , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Formaldehído/química , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , PPAR gamma/genética , PPAR gamma/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Elementos Reguladores de la Transcripción , Análisis de Secuencia de ARN , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Urotelio/citología , Urotelio/metabolismo
9.
Gastroenterology ; 152(1): 176-192.e5, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27639807

RESUMEN

BACKGROUND & AIMS: GATA3 is a transcription factor that regulates T-cell production of cytokines. We investigated the role of GATA3 in development of colitis in mice. METHODS: We performed quantitative polymerase chain reaction and immunofluorescence analyses of colon tissues from patients with Crohn's disease (n = 61) or ulcerative colitis (UC, n = 74) or from patients without inflammatory bowel diseases (n = 22), to measure levels of GATA3. Colitis was induced by administration of oxazolone or 2,4,6-trinitrobenzenesulfonic acid to control mice, mice with T-cell-specific deletion of GATA3, and mice with deletion of tumor necrosis factor receptor (TNFR) 1 and TNFR2 (TNFR double knockouts); some mice were given a GATA3-specific DNAzyme (hgd40) or a control DNAzyme via intrarectal administration, or systemic injections of an antibody to TNF before or during sensitization and challenge phase of colitis induction. Colon tissues were collected and immunofluorescence and histochemical analyses were performed. Lamina propria mononuclear cells and T cells were isolated and analyzed by flow cytometry or cytokine assays. Colonic distribution of labeled DNAzyme and inflammation were monitored by in vivo imaging (endoscopy) of mice. RESULTS: Levels of GATA3 messenger RNA were higher in colon tissues from patients with UC, but not ileal Crohn's disease, than control tissues; levels of GATA3 correlated with levels of inflammatory cytokines (interleukin [IL] 9, IL17A, IL6, IL5, IL4, IL13, and TNF). We observed increased expression of GATA3 by lamina propria T cells from mice with colitis compared with controls. Mice with T-cell-specific deletion of GATA3 did not develop colitis and their colonic tissues did not produce inflammatory cytokines (IL6, IL9, or IL13). The DNAzyme hgd40 inhibited expression of GATA3 messenger RNA by unstimulated and stimulated T cells, and distributed throughout the inflamed colons of mice with colitis. Colon tissues from mice given hgd40 had reduced expression of GATA3 messenger RNA, compared with mice given a control DNAzyme. Mice given hgd40 did not develop colitis after administration of oxazolone or 2,4,6-trinitrobenzenesulfonic acid; lamina propria cells from these mice expressed lower levels of IL6, IL9, and IL13 than cells from mice given the control DNAzyme. Mini-endoscopic images revealed that hgd40 and anti-TNF reduced colon inflammation over 3 days; hgd40 reduced colitis in TNFR double-knockout mice. CONCLUSIONS: Levels of GATA3 are increased in patients with UC and correlate with production of inflammatory cytokines in mice and humans. A DNAzyme that prevents expression of GATA3 reduces colitis in mice, independently of TNF, and reduces levels of cytokines in the colon. This DNAzyme might be developed for treatment of patients with UC.


Asunto(s)
Colitis/metabolismo , Colitis/prevención & control , ADN Catalítico/administración & dosificación , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/metabolismo , ARN Mensajero/análisis , Administración Rectal , Adolescente , Adulto , Anciano , Animales , Estudios de Casos y Controles , Niño , Colitis/inducido químicamente , Colitis Ulcerosa/genética , Colitis Ulcerosa/metabolismo , Colon/química , Colon/metabolismo , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Factor de Transcripción GATA3/genética , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Oxazolona , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Linfocitos T/metabolismo , Ácido Trinitrobencenosulfónico , Adulto Joven
10.
Eksp Klin Farmakol ; 79(4): 35-44, 2016 Aug.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-29949702

RESUMEN

Asthma is among the most common chronic disorders of airways, which affects both children and adults. Asthma being a common disease among different segments of population, it has a high mortality rate and, in the absence of appropriate care, affects the quality of life and leads to economics losses. In a view of continuing growth in the incidence of asthma, it is important to find relevant biological targets for developing new approaches to astma therapy. Recent advances in molecular immunology, genetics, and bioinformatics allowed genes involved in the pathogenesis of asthma to be identified, which provided prerequisites for the development of new types of drugs that can regulate the activity of pathogenically significant genes. To date, a number of technologies for sequence-specific gene regulation (ASO, ribozymes, DNAzymes, EGS, DNA-decoys, U 1-adapters) are available, but RNA interference is the most promising approach in both terms of efficacy and financial cost. This review focuses on the generalization and analysis of experimental data regarding the use of RNA interference technology for the treatment of astma.


Asunto(s)
Asma/terapia , Factor de Transcripción GATA3/antagonistas & inhibidores , Terapia Genética/métodos , Interleucina-13/antagonistas & inhibidores , Interleucina-4/antagonistas & inhibidores , Interferencia de ARN , Adulto , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/metabolismo , Aptámeros de Nucleótidos/uso terapéutico , Asma/genética , Asma/inmunología , Asma/patología , Niño , Enfermedad Crónica , ADN Catalítico/genética , ADN Catalítico/metabolismo , ADN Catalítico/uso terapéutico , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Oligonucleótidos Antisentido/uso terapéutico , Calidad de Vida , ARN Catalítico/genética , ARN Catalítico/metabolismo , ARN Catalítico/uso terapéutico
11.
J Leukoc Biol ; 98(2): 185-94, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26026068

RESUMEN

IFN-α/ß was first described as a potent inhibitor of viral replication, but it is now appreciated that IFN signaling plays a pleiotropic role in regulating peripheral T cell functions. Recently, IFN-α/ß was shown to block human Th2 development by suppressing the transcription factor GATA3. This effect is consistent with the role for IFN-α/ß in suppressing allergic inflammatory processes by blocking granulocyte activation and IL-4-mediated B cell isotype switching to IgE. With the consideration of recent studies demonstrating a defect in IFN-α/ß secretion in DCs and epithelial cells from individuals with severe atopic diseases, there is an apparent reciprocal negative regulatory loop in atopic individuals, whereby the lack of IFN-α/ß secretion by innate cells contributes to the development of allergic Th2 cells. Is it possible to overcome these events by treating with IFN-α/ß or by inducing its secretion in vivo? In support of this approach, case studies have documented the therapeutic potential of IFN-α/ß in treating steroid-resistant allergic asthma and other atopic diseases. Additionally, individuals with asthma who are infected with HCV and respond to IFN therapy showed a reduction in symptoms and severity of asthma attacks. These findings support a model, whereby allergic and antiviral responses are able to cross-regulate each other, as IgER cross-linking of pDCs prevents IFN-α/ß production in response to viral infection. The clinical importance of upper-respiratory viruses in the context of allergic asthma supports the need to understand how these pathways intersect and to identify potential therapeutic targets.


Asunto(s)
Hipersensibilidad/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Virosis/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Linfocitos B/virología , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Regulación de la Expresión Génica , Granulocitos/efectos de los fármacos , Granulocitos/inmunología , Granulocitos/patología , Granulocitos/virología , Humanos , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/patología , Hipersensibilidad/virología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/biosíntesis , Inmunoglobulina E/genética , Interferón-alfa/genética , Interferón-alfa/uso terapéutico , Interferón beta/genética , Interferón beta/uso terapéutico , Interleucina-4/genética , Interleucina-4/inmunología , Transducción de Señal , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/patología , Células Th2/virología , Virosis/tratamiento farmacológico , Virosis/patología , Virosis/virología
13.
Mol Med Rep ; 12(1): 71-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25738969

RESUMEN

Asthma is a serious health problem causing significant mortality and morbidity globally. Persistent airway inflammation, airway hyperresponsiveness, increased immunoglobulin E (IgE) levels and mucus hypersecretion are key characteristics of the condition. Asthma is mediated via a dominant T-helper 2 (Th2) immune response, causing enhanced expression of Th2 cytokines. These cytokines are responsible for the various pathological changes associated with allergic asthma. To investigate the anti-asthmatic potential of afzelin, as well as the underlying mechanisms involved, its anti-asthmatic potential were investigated in a murine model of asthma. In the present study, BALB/c mice were systemically sensitized using ovalbumin (OVA) followed by aerosol allergen challenges. The effect of afzelin on airway hyperresponsiveness, eosinophilic infiltration, Th2 cytokine and OVA­specific IgE production in a mouse model of asthma were investigated. It was found that afzelin­treated groups suppressed eosinophil infiltration, allergic airway inflammation, airway hyperresponsiveness, OVA-specific IgE and Th2 cytokine secretion. The results of the present study suggested that the therapeutic mechanism by which afzelin effectively treats asthma is based on reduction of Th2 cytokine via inhibition of GATA-binding protein 3 transcription factor, which is the master regulator of Th2 cytokine differentiation and production.


Asunto(s)
Asma/tratamiento farmacológico , Hiperreactividad Bronquial/genética , Eosinófilos/metabolismo , Factor de Transcripción GATA3/biosíntesis , Manósidos/administración & dosificación , Proantocianidinas/administración & dosificación , Alérgenos/toxicidad , Animales , Asma/inducido químicamente , Asma/genética , Asma/inmunología , Hiperreactividad Bronquial/inmunología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/patología , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Regulación de la Expresión Génica , Humanos , Inmunoglobulina E/inmunología , Inmunoglobulina E/metabolismo , Ratones , Ovalbúmina/administración & dosificación , Células Th2/inmunología , Células Th2/metabolismo
14.
Inflammation ; 37(5): 1476-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24706026

RESUMEN

Curcumin, a natural product derived from the plant Curcuma longa, has been found to have anti-inflammatory, antineoplastic and antifibrosis effects. It has been reported that curcumin attenuates allergic airway inflammation in mice through inhibiting NF-κB and its downstream transcription factor GATA3. It also has been proved the antineoplastic effect of curcumin through down-regulating Notch1 receptor and its downstream nuclear transcription factor NF-κB levels. In this study, we aimed to investigate the anti-inflammatory effect of curcumin on acute allergic asthma and its underlying mechanisms. 36 male BALB/c mice were randomly divided into four groups (normal, asthma, asthma+budesonide and asthma+curcumin groups). BALF (bronchoalveolar lavage fluid) and lung tissues were analyzed for airway inflammation and the expression of Notch1, Notch2, Notch3, Notch4 and the downstream transcription factor GATA3. Our findings showed that the levels of Notch1 and Notch2 receptors were up-regulated in asthma group, accompanied by the increased expression of GATA3. But the expression of Notch2 receptor was lower than Notch1 receptor. Curcumin pretreatment improved the airway inflammatory cells infiltration and reversed the increasing levels of Notch1/2 receptors and GATA3. Notch3 receptor was not expressed in all of the four groups. Notch4 receptor protein and mRNA expression level in the four groups had no significant differences. The results of the present study suggested that Notch1 and Notch2 receptor, major Notch1 receptor, played an important role in the development of allergic airway inflammation and the inhibition of Notch1-GATA3 signaling pathway by curcumin can prevent the development and deterioration of the allergic airway inflammation. This may be a possible therapeutic option of allergic asthma.


Asunto(s)
Asma/prevención & control , Curcuma , Curcumina/uso terapéutico , Factor de Transcripción GATA3/antagonistas & inhibidores , Receptor Notch1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Curcumina/farmacología , Factor de Transcripción GATA3/biosíntesis , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Distribución Aleatoria , Receptor Notch1/biosíntesis , Transducción de Señal/fisiología
15.
Am J Hum Genet ; 92(4): 489-503, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23540573

RESUMEN

Analysis of 4,405 variants in 89,050 European subjects from 41 case-control studies identified three independent association signals for estrogen-receptor-positive tumors at 11q13. The strongest signal maps to a transcriptional enhancer element in which the G allele of the best candidate causative variant rs554219 increases risk of breast cancer, reduces both binding of ELK4 transcription factor and luciferase activity in reporter assays, and may be associated with low cyclin D1 protein levels in tumors. Another candidate variant, rs78540526, lies in the same enhancer element. Risk association signal 2, rs75915166, creates a GATA3 binding site within a silencer element. Chromatin conformation studies demonstrate that these enhancer and silencer elements interact with each other and with their likely target gene, CCND1.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 11/genética , Ciclina D1/genética , Elementos de Facilitación Genéticos/genética , Polimorfismo de Nucleótido Simple/genética , Sitios de Unión , Estudios de Casos y Controles , Línea Celular Tumoral , Cromatina/química , Cromatina/genética , Inmunoprecipitación de Cromatina , Ciclina D1/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Elementos Silenciadores Transcripcionales/genética , Proteína Elk-4 del Dominio ets/antagonistas & inhibidores , Proteína Elk-4 del Dominio ets/genética , Proteína Elk-4 del Dominio ets/metabolismo
16.
Anesthesiology ; 117(3): 580-91, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22846675

RESUMEN

BACKGROUND: Evidence suggests that nebulized lidocaine is beneficial in asthma therapy, but to what extent and the mechanisms underlying this effect remain poorly understood. The aim of this study was to assess the impact of lidocaine treatment using a murine model of allergic asthma characterized by expression of pivotal features of the disease: inflammation, mucus production, and lung remodeling. METHODS: A/J mice sensitized with ovalbumin were treated with inhaled lidocaine or vehicle immediately after ovalbumin intranasal challenges. Lung function, total and differential leukocytes in bronchoalveolar lavage fluid, peribronchial eosinophil density, interleukin (IL)-4, IL-5 and eotaxin-1 levels, epithelial mucus, collagen, extracellular-matrix deposition, matrix metalloproteinase-9 activity, and GATA-3 expression were evaluated. Between five and eight animals per group were used. RESULTS: Inhaled lidocaine inhibited ovalbumin-induced airway hyperreactivity to methacholine, and accumulation of lymphocytes, neutrophils, and eosinophils in bronchoalveolar lavage fluid 24 h after the last allergen provocation. Lidocaine administration also prevented other pathophysiological changes triggered by ovalbumin in lung tissue, including peribronchial eosinophil and neutrophil infiltration, subepithelial fibrosis, increased content of collagen and mucus, matrix metalloproteinase-9 activity, and increased levels of IL-4, IL-5, IL-13, and eotaxin-1. Furthermore, inhaled lidocaine inhibited lung tissue GATA-3 expression in ovalbumin-challenged mice. We also demonstrated that lidocaine inhibited the expression of GATA-3 in ovalbumin-stimulated T cells in vitro. CONCLUSIONS: Inhaled lidocaine prevents eosinophilic inflammation, overproduction of mucus, and peribronchial fibrosis in a murine model of asthma, and impaired airway hyperreactivity, possibly by inhibiting allergen-evoked GATA-3 expression and the subsequent up-regulation of proinflammatory cytokines and chemokines.


Asunto(s)
Anestésicos Locales/farmacología , Asma/tratamiento farmacológico , Bronquios/patología , Lidocaína/farmacología , Moco/metabolismo , Animales , Asma/inmunología , Asma/patología , Modelos Animales de Enfermedad , Fibrosis , Factor de Transcripción GATA3/análisis , Factor de Transcripción GATA3/antagonistas & inhibidores , Lidocaína/administración & dosificación , Pulmón/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Nebulizadores y Vaporizadores , Linfocitos T/efectos de los fármacos
17.
Immunity ; 35(5): 733-45, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22118525

RESUMEN

The regulation of memory CD4(+) helper T (Th) cell function, such as polarized cytokine production, remains unclear. Here we show that memory T helper 2 (Th2) cells are divided into four subpopulations by CD62L and CXCR3 expression. All four subpopulations produced interleukin-4 (IL-4) and IL-13, whereas only the CD62L(lo)CXCR3(lo) population produced IL-5 accompanied by increased H3-K4 methylation at the Il5 gene locus. The transcription factor Eomesodermin (encoded by Eomes) was highly expressed in memory Th2 cells, whereas its expression was selectively downregulated in the IL-5-producing cells. Il5 expression was enhanced in Eomes-deficient cells, and Eomesodermin was shown to interact with the transcription factor GATA3, preventing GATA3 binding to the Il5 promoter. Memory Th2 cell-dependent airway inflammation was attenuated in the absence of the CD62L(lo)CXCR3(lo) population but was enhanced by Eomes-deficient memory Th2 cells. Thus, IL-5 production in memory Th2 cells is regulated by Eomesodermin via the inhibition of GATA3 activity.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Memoria Inmunológica/inmunología , Interleucina-5/biosíntesis , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Animales , Células Cultivadas , Factor de Transcripción GATA3/antagonistas & inhibidores , Expresión Génica , Inflamación/inmunología , Selectina L/metabolismo , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptores CXCR3/metabolismo , Sistema Respiratorio/inmunología , Proteínas de Dominio T Box/genética , Células Th2/metabolismo , Transcripción Genética
18.
EMBO J ; 29(19): 3421-33, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20808283

RESUMEN

Allergic airway inflammation is a disease in which T helper 2 (Th2) cells have a critical function. The molecular mechanisms controlling Th2 differentiation and function are of paramount importance in biology and immunology. Recently, a network of PB1-containing adapters and kinases has been shown to be essential in this process owing to its function in regulating cell polarity and the activation of critical transcription factors. Here, we show in vivo data showing that T-cell-specific NBR1-deficient mice show impaired lung inflammation and have defective Th2 differentiation ex vivo with alterations in T-cell polarity and the selective inhibition of Gata3 and nuclear factor of activated T c1 activation. These results establish NBR1 as a novel PB1 adapter in Th2 differentiation and asthma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular/inmunología , Proteínas/metabolismo , Hipersensibilidad Respiratoria/inmunología , Transducción de Señal/fisiología , Células Th2/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Compuestos de Alumbre/toxicidad , Animales , Western Blotting , Polaridad Celular/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor de Transcripción GATA3/antagonistas & inhibidores , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Ovalbúmina/toxicidad , Reacción en Cadena de la Polimerasa , Proteínas/genética , Proteínas/inmunología , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/genética , Células Th2/fisiología
19.
Cell Cycle ; 9(14): 2830-5, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20647758

RESUMEN

The generation of specific T lymphocyte subsets is under the strict control of specific transcription factors, as this has been shown by knockout experiments in mice. Here, we show that siRNAs that specifically target the transcription factor Gata3 (which is required for the development of T helper 1 cells) or T-Bet (which is required for the development of T helper 2 cells) can be effective in vivo. Thus, the intraperitoneal injection of siRNAs specific for Gata3 or t-Bet leads to the specific depletion of their target gene products in vivo, in the spleen and in the lymph nodes of mice. The immunomodulatory action of these siRNAs was validated in a model of anti-tumor vaccination in which colorectal cancer cells that succumb to anthracyclin-induced immunogenic cell death were injected subcutaneously into one flank, in the absence of any adjuvant and live tumor cells were injected simultaneously in the opposite flank of immunocompetent mice. In this setting, the siRNA targeting t-Bet was able to accelerate tumor growth while the siRNA targeting Gata3 significantly reduced the proliferation of cancer cells in vivo. These effects were dependent on the immune response elicited by dying tumor cells because both siRNAs failed to modulate the growth of tumors in non-vaccinated mice. The immune response-dependent anticancer effect of the Gata3-specific siRNA was not due to the induction of class I interferons and could be fully abolished by co-injection of t-Bet-specific siRNA. These results demonstrate the possibility to use siRNAs for immunomodulaton in vivo and illustrate the antagonistic implication of distinct T helper populations in anti-cancer immune responses.


Asunto(s)
Factor de Transcripción GATA3/antagonistas & inhibidores , Interferencia de ARN , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Ratones , ARN Interferente Pequeño/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
20.
Blood ; 116(20): 4202-11, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-20660789

RESUMEN

The transcription factor network in Hodgkin lymphoma (HL) represents a unique composition of proteins found in no other hematopoietic cell. Among these factors, an aberrant expression of the T-cell transcription factor GATA3 is observed in B cell-derived Hodgkin and Reed/Sternberg (HRS) tumor cells. Herein, we elucidate the regulation and function of this factor in HL. We demonstrate binding of NFκB and Notch-1, 2 factors with deregulated activity in HL to GATA3 promoter elements. Interference with NFκB and Notch-1 activity led to decreased GATA3 expression, indicating a dependency of deregulated GATA3 expression on these transcription factors. Down-regulation of GATA3 in HL cell lines demonstrated its role in the regulation of IL-5, IL-13, STAT4, and other genes. A correlation between GATA3 and IL-13 expression was confirmed for HRS cells in HL tissues. Thus, GATA3 shapes the cytokine expression and signaling that is typical of HL. Conclusively, aberrant GATA3 expression in HRS cells is stimulated by the deregulated constitutive activity of NFκB and Notch-1, indicating a complex network of deregulated transcription factors in these cells. GATA3 activity significantly contributes to the typical cytokine secretion of and signaling in HRS cells, which presumably plays an essential role in HL pathogenesis.


Asunto(s)
Citocinas/genética , Factor de Transcripción GATA3/genética , Enfermedad de Hodgkin/genética , Células de Reed-Sternberg/inmunología , Apoptosis , Línea Celular Tumoral , Citocinas/metabolismo , Regulación hacia Abajo/genética , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/metabolismo , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/patología , Humanos , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-15/genética , Interleucina-15/metabolismo , Lentivirus/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Interferente Pequeño/metabolismo , Receptor Notch1/metabolismo , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA