Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 324(3): G219-G230, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36719093

RESUMEN

In cirrhosis, several molecular alterations such as resistance to apoptosis could accelerate carcinogenesis. Recently, mechanotransduction has been attracting attention as one of the causes of these disturbances. In patients with cirrhosis, the serum sodium levels progressively decrease in the later stage of cirrhosis, and hyponatremia leads to serum hypo-osmolality. Since serum sodium levels in patients with cirrhosis with liver cancer are inversely related to cancer's number, size, stage, and cumulative survival, we hypothesized that hypo-osmolality-induced mechanotransduction under cirrhotic conditions might contribute to oncogenesis and/or progression of hepatocellular carcinoma (HCC). In this study, we adjusted osmosis of culture medium by changing the sodium chloride concentration and investigated the influence of hypotonic conditions on the apoptosis resistance of an HCC cell line, HepG2, using a serum-deprivation-induced apoptosis model. By culturing the cells in a serum-free medium, the levels of an antiapoptotic protein Bcl-2 were downregulated. In contrast, the hypotonic conditions caused apoptosis resistance by upregulation of Bcl-2. Next, we examined which pathway was involved in the apoptosis resistance. Hypotonic conditions enhanced AKT signaling, and constitutive activation of AKT in HepG2 cells led to upregulation of Bcl-2. Moreover, we revealed that the enhancement of AKT signaling was caused by intracellular calcium influx via a mechanosensor, TRPV2. Our findings suggested that hyponatremia-induced serum hypotonic in patients with cirrhosis promoted the progression of hepatocellular carcinoma.NEW & NOTEWORTHY Our study first revealed that hypo-osmolarity-induced mechanotransduction enhanced calcium-mediated AKT signaling via TRPV2 activation, resulting in contributing to apoptosis resistance. The finding indicates a possible view that liver cirrhosis-induced hyponatremia promotes hepatocellular carcinogenesis.


Asunto(s)
Carcinoma Hepatocelular , Hiponatremia , Neoplasias Hepáticas , Humanos , Apoptosis , Calcio/metabolismo , Carcinogénesis , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Neoplasias Hepáticas/metabolismo , Mecanotransducción Celular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sodio/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(41): 20545-20555, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548395

RESUMEN

The extraordinarily thin alveolar type 1 (AT1) cell constitutes nearly the entire gas exchange surface and allows passive diffusion of oxygen into the blood stream. Despite such an essential role, the transcriptional network controlling AT1 cells remains unclear. Using cell-specific knockout mouse models, genomic profiling, and 3D imaging, we found that NK homeobox 2-1 (Nkx2-1) is expressed in AT1 cells and is required for the development and maintenance of AT1 cells. Without Nkx2-1, developing AT1 cells lose 3 defining features-molecular markers, expansive morphology, and cellular quiescence-leading to alveolar simplification and lethality. NKX2-1 is also cell-autonomously required for the same 3 defining features in mature AT1 cells. Intriguingly, Nkx2-1 mutant AT1 cells activate gastrointestinal (GI) genes and form dense microvilli-like structures apically. Single-cell RNA-seq supports a linear transformation of Nkx2-1 mutant AT1 cells toward a GI fate. Whole lung ChIP-seq shows NKX2-1 binding to 68% of genes that are down-regulated upon Nkx2-1 deletion, including 93% of known AT1 genes, but near-background binding to up-regulated genes. Our results place NKX2-1 at the top of the AT1 cell transcriptional hierarchy and demonstrate remarkable plasticity of an otherwise terminally differentiated cell type.


Asunto(s)
Células Epiteliales Alveolares/citología , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Pulmón/crecimiento & desarrollo , Mutación , Organogénesis , Factor Nuclear Tiroideo 1/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Diferenciación Celular , Pulmón/metabolismo , Ratones , Análisis de la Célula Individual , Factor Nuclear Tiroideo 1/antagonistas & inhibidores , Factor Nuclear Tiroideo 1/genética
3.
Development ; 145(5)2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29440304

RESUMEN

The entire lung epithelium arises from SRY box 9 (SOX9)-expressing progenitors that form the respiratory tree and differentiate into airway and alveolar cells. Despite progress in understanding their initial specification within the embryonic foregut, how these progenitors are subsequently maintained is less clear. Using inducible, progenitor-specific genetic mosaic mouse models, we showed that ß-catenin (CTNNB1) maintains lung progenitors by promoting a hierarchical lung progenitor gene signature, suppressing gastrointestinal (GI) genes, and regulating NK2 homeobox 1 (NKX2.1) and SRY box 2 (SOX2) in a developmental stage-dependent manner. At the early, but not later, stage post-lung specification, CTNNB1 cell-autonomously maintained normal NKX2.1 expression levels and suppressed ectopic SOX2 expression. Genetic epistasis analyses revealed that CTNNB1 is required for fibroblast growth factor (Fgf)/Kirsten rat sarcoma viral oncogene homolog (Kras)-mediated promotion of the progenitors. In silico screening of Eurexpress and translating ribosome affinity purification (TRAP)-RNAseq identified a progenitor gene signature, a subset of which depends on CTNNB1. Wnt signaling also maintained NKX2.1 expression and suppressed GI genes in cultured human lung progenitors derived from embryonic stem cells.


Asunto(s)
Linaje de la Célula/genética , Células Madre Embrionarias/metabolismo , Células Epiteliales/citología , Pulmón/embriología , Mucosa Respiratoria/citología , Mucosa Respiratoria/embriología , beta Catenina/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Pulmón/citología , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Embarazo , Mucosa Respiratoria/metabolismo , Transcriptoma , beta Catenina/genética
4.
Mol Cell ; 50(2): 185-99, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23523371

RESUMEN

Tissue-specific differentiation programs become dysregulated during cancer evolution. The transcription factor Nkx2-1 is a master regulator of pulmonary differentiation that is downregulated in poorly differentiated lung adenocarcinoma. Here we use conditional murine genetics to determine how the identity of lung epithelial cells changes upon loss of their master cell-fate regulator. Nkx2-1 deletion in normal and neoplastic lungs causes not only loss of pulmonary identity but also conversion to a gastric lineage. Nkx2-1 is likely to maintain pulmonary identity by recruiting transcription factors Foxa1 and Foxa2 to lung-specific loci, thus preventing them from binding gastrointestinal targets. Nkx2-1-negative murine lung tumors mimic mucinous human lung adenocarcinomas, which express gastric markers. Loss of the gastrointestinal transcription factor Hnf4α leads to derepression of the embryonal proto-oncogene Hmga2 in Nkx2-1-negative tumors. These observations suggest that loss of both active and latent differentiation programs is required for tumors to reach a primitive, poorly differentiated state.


Asunto(s)
Adenocarcinoma/metabolismo , Diferenciación Celular , Neoplasias Pulmonares/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Adenocarcinoma/patología , Animales , Sitios de Unión , Proliferación Celular , Transformación Celular Neoplásica , Mucosa Gástrica/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Hiperplasia/metabolismo , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Mutación Missense , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Especificidad de Órganos , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Estómago/patología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Activación Transcripcional , Transcriptoma , Carga Tumoral
5.
J Pharmacol Exp Ther ; 371(2): 360-374, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31420528

RESUMEN

Nonalcoholic steatohepatitis (NASH) is the progressive stage of nonalcoholic fatty liver disease that highly increases the risk of cirrhosis and liver cancer, and there are few therapeutic options available in the clinic. Withaferin A (WA), extracted from the ayurvedic medicine Withania somnifera, has a wide range of pharmacological activities; however, little is known about its effects on NASH. To explore the role of WA in treating NASH, two well defined NASH models were used, the methionine-choline-deficient diet and the 40 kcal% high-fat diet (HFD). In both NASH models, WA treatment or control vehicle was administered to evaluate its hepatoprotective effects. As assessed by biochemical and histologic analyses, WA prevented and therapeutically improved liver injury in both models, as revealed by lower serum aminotransaminases, hepatic steatosis, liver inflammation, and fibrosis. In the HFD-induced NASH model, both elevated serum ceramides and increased hepatic oxidative stress were decreased in the WA-treated group compared with the control vehicle-treated group. To further explore whether WA has an anti-NASH effect independent of its known action in leptin signaling associated with obesity, leptin signaling-deficient ob/ob mice maintained on an HFD were used to induce NASH. WA therapeutically reduced NASH in HFD-treated leptin-deficient ob/ob mice, thus demonstrating a leptin-independent hepatoprotective effect. This study revealed that WA treatment could be an option for NASH treatment.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Witanólidos/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología , Distribución Aleatoria , Witanólidos/farmacología
6.
Lab Invest ; 98(9): 1126-1132, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29946134

RESUMEN

While thyroid is considered to be a dormant organ, when required, it can regenerate through increased cell proliferation. However, the mechanism for regeneration remains unknown. Nkx2-1(fl/fl);TPO-cre mouse thyroids exhibit a very disorganized appearance because their thyroids continuously degenerate and regenerate. In mouse thyroids, a cluster of cells are found near the tracheal cartilage and muscle, which are positive for expression of NKX2-1, the master transcription factor governing thyroid development and function. In the present study, we propose that this cluster of NKX2-1-positive cells may be the precursor cells that mature to become thyroid follicular cells, forming thyroid follicles. We also found that phosphorylation of AKT is induced by NKX2-1 in the proposed thyroid progenitor-like side-population cell-derived thyroid cell line (SPTL) cells, suggesting the possibility that NKX2-1 plays a role in differentiation through the modulation of AKT signaling. This study revealed that Nkx2-1(fl/fl);TPO-cre mice provide a suitable model to study in vivo regeneration and folliculogenesis of the thyroid.


Asunto(s)
Modelos Biológicos , Regeneración/fisiología , Glándula Tiroides/fisiología , Factor Nuclear Tiroideo 1/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/crecimiento & desarrollo , Factor Nuclear Tiroideo 1/deficiencia , Factor Nuclear Tiroideo 1/genética
7.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30070361

RESUMEN

Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.


Asunto(s)
Receptores ErbB/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor de Insulina/deficiencia , Cáncer Papilar Tiroideo/metabolismo , Células Epiteliales Tiroideas/metabolismo , Neoplasias de la Tiroides/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal , Cáncer Papilar Tiroideo/patología , Células Epiteliales Tiroideas/patología , Neoplasias de la Tiroides/patología , Tirotropina/biosíntesis , Tirotropina/metabolismo
8.
Carcinogenesis ; 38(4): 391-401, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28186538

RESUMEN

Thyroid cancer originates from genetic and epigenetic changes that alter gene expression and cellular signaling pathways. Here, we report that altered expression of the nucleosome-binding protein HMGN4 potentiates thyroid tumorigenesis. Bioinformatics analyses reveal increased HMGN4 expression in thyroid cancer. We find that upregulation of HMGN4 expression in mouse and human cells, and in the thyroid of transgenic mice, alters the cellular transcription profile, downregulates the expression of the tumor suppressors Atm, Atrx and Brca2, and elevates the levels of the DNA damage marker γH2AX. Mouse and human cells overexpressing HMGN4 show increased tumorigenicity as measured by colony formation, by tumor generation in nude mice, and by the formation of preneoplastic lesions in the thyroid of transgenic mice. Our study identifies a novel epigenetic factor that potentiates thyroid oncogenesis and raises the possibility that HMGN4 may serve as an additional diagnostic marker, or therapeutic target in certain thyroid cancers.


Asunto(s)
Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Expresión Génica/genética , Proteínas HMGN/genética , Glándula Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Animales , Biomarcadores de Tumor/genética , Línea Celular , Línea Celular Tumoral , Daño del ADN/genética , Regulación hacia Abajo/genética , Epigénesis Genética/genética , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Transducción de Señal/genética , Transcripción Genética/genética , Regulación hacia Arriba/genética
9.
Biochem Biophys Res Commun ; 491(2): 271-276, 2017 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-28739257

RESUMEN

Peroxisome proliferator-activated receptor-γ (PPARγ) plays an important role in lipid and glucose metabolism. In this study, the function of PPARγ on lung development was investigated. Lung-specific Pparg conditional knockout mice (PpargΔLuEpC) were developed using Cre-Lox system. PpargΔLuEpC mice showed abnormal lung development with enlarged airspaces and followed by increase of apoptotic cells at E14.5 to E18.5. Gene analysis revealed that expression of Pmaip1, a gene related to apoptosis, was significantly increased while expression of Retnla, a gene related to anti-apoptosis, was dramatically decreased in the fetal lung (E14.5) of PpargΔLuEpC mice. In addition, expression of Pthlh, a gene phenotypically expressed in the congenital cystic adenomatoid malformation (CCAM), was increased at E14.5 to E18.5 in the lung of PpargΔLuEpC mice. Cell culture studies revealed that PPARγ could bind to promoter region of Pthlh gene as a repressor in the immortalized mouse lung epithelial cell line MLE-15. Surprisingly, phenotypic changes in MLE-15-shPparg cells, stably transfected with shPparg plasmid, were similar to the PpargΔLuEpC mice model. In addition, MLE-15-shPparg cells were easily detached from the cultured plate when cold phosphate buffered saline was applied. Furthermore, expression of Cdh1, a gene related to cell adhesion, was significantly reduced in the MLE-15-shPparg cells. Taken together, PPARγ may play an important role in fetal lung development via alveolar cell-to-cell adhesion system.


Asunto(s)
Malformación Adenomatoide Quística Congénita del Pulmón/genética , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , PPAR gamma/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Animales , Apoptosis , Sitios de Unión , Proteínas Cdh1/genética , Proteínas Cdh1/metabolismo , Adhesión Celular , Línea Celular Transformada , Malformación Adenomatoide Quística Congénita del Pulmón/metabolismo , Malformación Adenomatoide Quística Congénita del Pulmón/patología , Embrión de Mamíferos , Células Epiteliales/patología , Feto , Genes Reporteros , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , PPAR gamma/deficiencia , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal
10.
Gastroenterology ; 149(4): 1030-41.e6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26099526

RESUMEN

BACKGROUND & AIMS: Alcoholic steatohepatitis (ASH) is the progressive form of alcoholic liver disease and may lead to cirrhosis and hepatocellular carcinoma. We studied mouse models and human tissues to identify molecules associated with ASH progression and focused on the mouse fat-specific protein 27 (FSP-27)/human cell death-inducing DFF45-like effector C (CIDEC) protein, which is expressed in white adipose tissues and promotes formation of fat droplets. METHODS: C57BL/6N mice or mice with hepatocyte-specific disruption of Fsp27 (Fsp27(Hep-/-) mice) were fed the Lieber-Decarli ethanol liquid diet (5% ethanol) for 10 days to 12 weeks, followed by 1 or multiple binges of ethanol (5 or 6 g/kg) during the chronic feeding. Some mice were given an inhibitor (GW9662) of peroxisome proliferator-activated receptor γ (PPARG). Adenoviral vectors were used to express transgenes or small hairpin (sh) RNAs in cultured hepatocytes and in mice. Liver tissue samples were collected from ethanol-fed mice or from 31 patients with alcoholic hepatitis (AH) with biopsy-proved ASH and analyzed histologically and immunohistochemically and by transcriptome, immunoblotting, and real-time PCR analyses. RESULTS: Chronic-plus-binge ethanol feeding of mice, which mimics the drinking pattern of patients with AH, produced severe ASH and mild fibrosis. Microarray analyses revealed similar alterations in expression of many hepatic genes in ethanol-fed mice and humans with ASH, including up-regulation of mouse Fsp27 (also called Cidec) and human CIDEC. Fsp27(Hep-/-) mice and mice given injections of adenovirus-Fsp27shRNA had markedly reduced ASH following chronic-plus-binge ethanol feeding. Inhibition of PPARG and cyclic AMP-responsive element binding protein H (CREBH) prevented the increases in Fsp27α and FSP27ß mRNAs, respectively, and reduced liver injury in this chronic-plus-binge ethanol feeding model. Overexpression of FSP27 and ethanol exposure had synergistic effects in inducing production of mitochondrial reactive oxygen species and damage to hepatocytes in mice. Hepatic CIDEC mRNA expression was increased in patients with AH and correlated with the degree of hepatic steatosis and disease severity including mortality. CONCLUSIONS: In mice, chronic-plus-binge ethanol feeding induces ASH that mimics some histological and molecular features observed in patients with AH. Hepatic expression of FSP27/CIDEC is highly up-regulated in mice following chronic-plus-binge ethanol feeding and in patients with AH; this up-regulation contributes to alcohol-induced liver damage.


Asunto(s)
Hígado Graso Alcohólico/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Proteínas/metabolismo , Adulto , Animales , Proteínas Reguladoras de la Apoptosis , Consumo Excesivo de Bebidas Alcohólicas , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/prevención & control , Femenino , Perfilación de la Expresión Génica/métodos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mitocondrias Hepáticas/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Proteínas/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
11.
Int Arch Allergy Immunol ; 171(1): 36-44, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27820933

RESUMEN

BACKGROUND: Secretoglobin (SCGB) 3A2, a novel, lung-enriched, cytokine-like, secreted protein of small molecular weight, was demonstrated to exhibit various biological functions including anti-inflammatory, antifibrotic and growth-factor activities. Anti-inflammatory activity was uncovered using the ovalbumin-induced allergic airway inflammation model. However, further validation of this activity using knockout mice in a different allergic inflammation model is necessary in order to establish the antiallergic inflammatory role for this protein. METHODS: Scgb3a2-null (Scgb3a2-/-) mice were subjected to nasal inhalation of Dermatophagoides pteronyssinus extract for 5 days/week for 5 consecutive weeks; control mice received nasal inhalation of saline as a comparator. Airway inflammation was assessed by histological analysis, the number of inflammatory cells and various Th2-type cytokine levels in the lungs and bronchoalveolar lavage fluids by qRT-PCR and ELISA, respectively. RESULTS: Exacerbated inflammation was found in the airway of Scgb3a2-/- mice subjected to house dust mite (HDM)-induced allergic airway inflammation compared with saline-treated control groups. All the inflammation end points were increased in the Scgb3a2-/- mice. The Ccr4 and Ccl17 mRNA levels were higher in HDM-treated lungs of Scgb3a2-/- mice than wild-type mice or saline-treated Scgb3a2-/- mice, whereas no changes were observed for Ccr3 and Ccl11 mRNA levels. CONCLUSIONS: These results demonstrate that SCGB3A2 has an anti-inflammatory activity in the HDM-induced allergic airway inflammation model, in which SCGB3A2 may modulate the CCR4-CCL17 pathway. SCGB3A2 may provide a useful tool to treat allergic airway inflammation, and further studies on the levels and function of SCGB3A2 in asthmatic patients are warranted.


Asunto(s)
Alérgenos/inmunología , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Pyroglyphidae/inmunología , Secretoglobinas/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad/genética , Hipersensibilidad/patología , Ratones , Ratones Noqueados , Secretoglobinas/genética
12.
Lab Invest ; 95(11): 1269-77, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26146959

RESUMEN

KRAS(G12D) can cause lung cancer rapidly, but is not sufficient to induce thyroid cancer. It is not clear whether long-term serum thyroid stimulating hormone (TSH) stimulation can promote KRAS(G12D)-mediated thyroid follicular cell transformation. In the present study, we investigated the effect of long-term TSH stimulation in KRAS(G12D) knock-in mice and the role of Sprouty1 (SPRY1) in KRAS(G12D)-mediated signaling. We used TPO-KRAS(G12D) mice for thyroid-specific expression of KRAS(G12D) under the endogenous KRAS promoter. Twenty TPO-KRAS(G12D) mice were given anti-thyroid drug propylthiouracil (PTU, 0.1% w/v) in drinking water to induce serum TSH and 20 mice were without PTU treatment. Equal number of wild-type littermates (TPO-KRAS(WT)) was given the same treatment. The expression of SPRY1, a negative regulator of receptor tyrosine kinase (RTK) signaling, was analyzed in both KRAS(G12D)-and BRAF(V600E)-induced thyroid cancers. Without PTU treatment, only mild thyroid enlargement and hyperplasia were observed in TPO-KRAS(G12D) mice. With PTU treatment, significant thyroid enlargement and hyperplasia occurred in both TPO-KRAS(G12D) and TPO-KRAS(WT) littermates. Thyroids from TPO-KRAS(G12D) mice were six times larger than TPO-KRAS(WT) littermates. Distinct thyroid histology was found between TPO-KRAS(G12D) and TPO-KRAS(WT) mice: thyroid from TPO-KRAS(G12D) mice showed hyperplasia with well-maintained follicular architecture whereas in TPO-KRAS(WT) mice this structure was replaced by papillary hyperplasia. Among 10 TPO-KRAS(G12D) mice monitored for 14 months, two developed follicular thyroid cancer (FTC), one with pulmonary metastasis. Differential SPRY1 expression was demonstrated: increased in FTC and reduced in papillary thyroid cancer (PTC). The increased SPRY1 expression in FTC promoted TSH-RAS signaling through PI3K/AKT pathway whereas downregulation of SPRY1 by BRAF(V600E) in PTC resulted in both MAPK and PI3K/AKT activation. We conclude that chronic TSH stimulation can enhance KRAS(G12D)-mediated oncogenesis, leading to FTC. SPRY1 may function as a molecular switch to control MAPK signaling and its downregulation by BRAF(V600E) favors PTC development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Transformación Celular Neoplásica/efectos de los fármacos , Genes ras , Proteínas de la Membrana/fisiología , Fosfoproteínas/fisiología , Glándula Tiroides/citología , Neoplasias de la Tiroides/patología , Tirotropina/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Transformación Celular Neoplásica/genética , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/genética
13.
BMC Pulm Med ; 15: 72, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26178733

RESUMEN

BACKGROUND: Secretoglobin (SCGB) 3A2, a cytokine-like secretory protein of small molecular weight, is predominantly expressed in airway epithelial cells. While SCGB3A2 is known to have anti-inflammatory, growth factor, and anti-fibrotic activities, whether SCGB3A2 has any other roles, particularly in lung homeostasis and disease has not been demonstrated in vivo. The aim of this study was to address these questions in mice. METHODS: A transgenic mouse line that expresses SCGB3A2 in the lung using the human surfactant protein-C promoter was established. Detailed histological, immunohistochemical, physiological, and molecular characterization of the Scgb3a2-transgenic mouse lungs were carried out. Scgb3a2-transgenic and wild-type mice were subjected to bleomycin-induced pulmonary fibrosis model, and their lungs and bronchoalveolar lavage fluids were collected at various time points during 9 weeks post-bleomycin treatment for further analysis. RESULTS: Adult Scgb3a2-transgenic mouse lungs expressed approximately five-fold higher levels of SCGB3A2 protein in comparison to wild-type mice as determined by western blotting of lung tissues. Immunohistochemistry showed that expression was localized to alveolar type II cells in addition to airway epithelial cells, thus accurately reflecting the site of surfactant protein-C expression. Scgb3a2-transgenic mice showed normal lung development and histology, and no overt gross phenotypes. However, when subjected to a bleomycin-induced pulmonary fibrosis model, they initially exhibited exacerbated fibrosis at 3 weeks post-bleomycin administration that was more rapidly resolved by 6 weeks as compared with wild-type mice, as determined by lung histology, Masson Trichrome staining and hydroxyproline content, inflammatory cell numbers, expression of collagen genes, and proinflammatory cytokine levels. The decrease of fibrosis coincided with the increased expression of SCGB3A2 in Scgb3a2-transgenic lungs. CONCLUSIONS: These results demonstrate that SCGB3A2 is an anti-fibrotic agent, and suggest a possible therapeutic use of recombinant SCGB3A2 in the treatment of pulmonary fibrosis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Pulmón/metabolismo , Fibrosis Pulmonar/genética , ARN/genética , Secretoglobinas/genética , Animales , Bleomicina/toxicidad , Northern Blotting , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Pulmón/patología , Ratones , Ratones Transgénicos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Secretoglobinas/biosíntesis
14.
Proc Natl Acad Sci U S A ; 109(31): 12592-7, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22797898

RESUMEN

Clara cells of mammalian airways have multiple functions and are morphologically heterogeneous. Although Notch signaling is essential for the development of these cells, it is unclear how Notch influences Clara cell specification and if diversity is established among Clara cell precursors. Here we identify expression of the secretoglobin Scgb3a2 and Notch activation as early events in a program of secretory cell fate determination in developing murine airways. We show that Scgb3a2 expression in vivo is Notch-dependent at early stages and ectopically induced by constitutive Notch1 activation, and also that in vitro Notch signaling together with the pan-airway transcription factor Ttf1 (Nkx2.1) synergistically regulate secretoglobin gene transcription. Furthermore, we identified a subpopulation of secretory precursors juxtaposed to presumptive neuroepithelial bodies (NEBs), distinguished by their strong Scgb3a2 and uroplakin 3a (Upk3a) signals and reduced Ccsp (Scgb1a1) expression. Genetic ablation of Ascl1 prevented NEB formation and selectively interfered with the formation of this subpopulation of cells. Lineage labeling of Upk3a-expressing cells during development showed that these cells remain largely uncommitted during embryonic development and contribute to Clara and ciliated cells in the adult lung. Together, our findings suggest a role for Notch in the induction of a Clara cell-specific program of gene expression, and reveals that the NEB microenvironment in the developing airways is a niche for a distinct subset of Clara-like precursors.


Asunto(s)
Cuerpos Neuroepiteliales/metabolismo , Sistema Respiratorio/embriología , Nicho de Células Madre/fisiología , Células Madre/metabolismo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Cuerpos Neuroepiteliales/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Sistema Respiratorio/citología , Secretoglobinas/biosíntesis , Secretoglobinas/genética , Células Madre/citología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L10-22, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24213919

RESUMEN

Secretoglobin (SCGB) 3A2 is a member of the SCGB gene superfamily of small secreted proteins, predominantly expressed in lung airways. We hypothesize that human SCGB3A2 may exhibit anti-inflammatory, growth factor, and antifibrotic activities and be of clinical utility. Recombinant human SCGB3A2 was expressed, purified, and biochemically characterized as a first step to its development as a therapeutic agent in clinical settings. Human SCGB3A2, as well as mouse SCGB3A2, readily formed a dimer in solution and exhibited novel phospholipase A2 inhibitory activity. This is the first demonstration of any quantitative biochemical measurement for the evaluation of SCGB3A2 protein. In the mouse as an experimental animal, human SCGB3A2 exhibited growth factor activity by promoting embryonic lung development in both ex vivo and in vivo systems and antifibrotic activity in the bleomycin-induced lung fibrosis model. The results suggested that human SCGB3A2 can function as a growth factor and an antifibrotic agent in humans. When SCGB3A2 was administered to pregnant female mice through the tail vein, the protein was detected in the dam's serum and lung, as well as the placenta, amniotic fluids, and embryonic lungs at 10 min postadministration, suggesting that SCGB3A2 readily crosses the placenta. The results warrant further development of recombinant SCGB3A2 as a therapeutic agent in treating patients suffering from lung diseases or preterm infants with respiratory distress.


Asunto(s)
Pulmón/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Secretoglobinas/administración & dosificación , Animales , Disponibilidad Biológica , Bleomicina , Evaluación Preclínica de Medicamentos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Inhibidores de Fosfolipasa A2/administración & dosificación , Inhibidores de Fosfolipasa A2/química , Inhibidores de Fosfolipasa A2/farmacocinética , Fosfolipasas A2/química , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Secretoglobinas/química , Secretoglobinas/farmacocinética , Técnicas de Cultivo de Tejidos
16.
J Pharmacol Exp Ther ; 351(3): 559-67, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25277138

RESUMEN

The role of intestinal human pregnane X receptor (PXR) in colon cancer was determined through investigation of the chemopreventive role of rifaximin, a specific agonist of intestinal human PXR, toward azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced colon cancer. Rifaximin treatment significantly decreased the number of colon tumors induced by AOM/DSS treatment in PXR-humanized mice, but not wild-type or Pxr-null mice. Additionally, rifaximin treatment markedly increased the survival rate of PXR-humanized mice, but not wild-type or Pxr-null mice. These data indicated a human PXR-dependent therapeutic chemoprevention of rifaximin toward AOM/DSS-induced colon cancer. Nuclear factor κ-light-chain-enhancer of activated B cells-mediated inflammatory signaling was upregulated in AOM/DSS-treated mice, and inhibited by rifaximin in PXR-humanized mice. Cell proliferation and apoptosis were also modulated by rifaximin treatment in the AOM/DSS model. In vitro cell-based assays further revealed that rifaximin regulated cell apoptosis and cell cycle in a human PXR-dependent manner. These results suggested that specific activation of intestinal human PXR exhibited a chemopreventive role toward AOM/DSS-induced colon cancer by mediating anti-inflammation, antiproliferation, and proapoptotic events.


Asunto(s)
Azoximetano/toxicidad , Neoplasias del Colon/metabolismo , Neoplasias del Colon/prevención & control , Sulfato de Dextran/toxicidad , Receptores de Esteroides/metabolismo , Rifamicinas/uso terapéutico , Animales , Neoplasias del Colon/inducido químicamente , Células HT29 , Humanos , Masculino , Ratones , Ratones Noqueados , Receptor X de Pregnano , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Receptores de Esteroides/agonistas , Rifamicinas/farmacología , Rifaximina
17.
Cell Tissue Res ; 356(1): 253-60, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24514953

RESUMEN

Secretoglobin (SCGB) 3A2 was originally identified as a downstream target for the homeodomain transcription factor NKX2-1 in the lung. NKX2-1 plays a role in the genesis and expression of genes in the thyroid, lung and ventral forebrain; Nkx2-1-null mice have no thyroid and pituitary and severely hypoplastic lungs and hypothalamus. To demonstrate whether SCGB3A2 plays any role in pituitary hormone production, NKX2-1 and SCGB3A2 expression in the mouse pituitary gland was examined by immunohistochemical analysis and RT-PCR. NKX2-1 was localized in the posterior pituitary lobe, whereas SCGB3A2 was observed in both anterior and posterior lobes as shown by immunohistochemistry and RT-PCR. Expression of CCAAT-enhancer binding proteins (C/EBPs), which regulate mouse Scgb3a2 transcription, was also examined by RT-PCR. C/EBPß, γ, δ and ζ were expressed in the adult mouse pituitary gland. SCGB3A2 was expressed in the anterior and posterior lobes from postnatal days 1 and 5, respectively and the areas where SCGB3A2 expression was found coincided with the area where FSH-secreting cells were found. Double-staining for SCGB3A2 and pituitary hormones revealed that SCGB3A2 was mainly localized in gonadotrophs in 49 % of FSH-secreting cells and 47 % of LH-secreting cells. In addition, SCGB3A2 dramatically inhibited LH and FSH mRNA expression in rat pituitary primary cell cultures. These results suggest that SCGB3A2 regulates FSH/LH production in the anterior pituitary lobe and that transcription factors other than NKX2-1 may regulate SCGB3A2 expression.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo , Secretoglobinas/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Gonadotrofos/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Hipófisis/citología , Hormonas Hipofisarias/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Coloración y Etiquetado , Factor Nuclear Tiroideo 1 , Factores de Transcripción/metabolismo
18.
Gen Comp Endocrinol ; 198: 22-31, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24380675

RESUMEN

We have identified two distinct Pax8 (a and b) mRNAs from the thyroid gland of the rainbow trout (Oncorhynchus mykiss), which seemed to be generated by alternative splicing. Both Pax8a and Pax8b proteins were predicted to possess the paired domain, octapeptide, and partial homeodomain, while Pax8b lacked the carboxy-terminal portion due to an insertion in the coding region of the mRNA. RT-PCR analysis showed each of Pax8a and Pax8b mRNAs to be abundantly expressed in the thyroid and kidney. In situ hybridization histochemistry further detected the expression of Pax8 mRNA in the epithelial cells of the thyroid follicles of the adult trout and in the thyroid primordial cells of the embryo. The functional properties of Pax8a and Pax8b were investigated by dual luciferase assay. The transcriptional regulation by the rat thyroid peroxidase (TPO) promoter was found to be increased by Pax8a, but not by Pax8b. Pax8a further showed synergistic transcriptional activity with rat Nkx2-1 for the human TPO upstream region including the enhancer and promoter. On the other hand, Pax8b decreased the synergistic activity of Pax8a and Nkx2-1. Electrophoretic mobility shift assay additionally indicated that not only Pax8a but also Pax8b can bind to the TPO promoter and enhancer, implying that the inhibitory effect of Pax8b might result from the lack of the functional carboxy-terminal portion. Collectively, the results suggest that for the trout thyroid gland, Pax8a may directly increase TPO gene expression in cooperation with Nkx2-1 while Pax8b may work as a non-activating competitor for the TPO transcription.


Asunto(s)
Empalme Alternativo , Autoantígenos/genética , Proteínas de Peces/metabolismo , Regulación de la Expresión Génica/fisiología , Yoduro Peroxidasa/genética , Proteínas de Unión a Hierro/genética , Oncorhynchus mykiss/genética , Factores de Transcripción Paired Box/genética , Glándula Tiroides/metabolismo , Secuencia de Aminoácidos , Animales , Autoantígenos/metabolismo , Secuencia de Bases , Clonación Molecular , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Peces/genética , Hibridación in Situ , Yoduro Peroxidasa/metabolismo , Proteínas de Unión a Hierro/metabolismo , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oncorhynchus mykiss/crecimiento & desarrollo , Filogenia , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Glándula Tiroides/citología , Factor Nuclear Tiroideo 1 , Distribución Tisular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
19.
Mediators Inflamm ; 2014: 216465, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25242865

RESUMEN

Secretoglobin (SCGB) 3A2, a cytokine-like secretory protein of small molecular weight, which may play a role in lung inflammation, is predominantly expressed in airway epithelial cells. In order to understand the physiological role of SCGB3A2, Scgb3a2(-/-) mice were generated and characterized. Scgb3a2(-/-) mice did not exhibit any overt phenotypes. In ovalbumin- (OVA-) induced airway allergy inflammation model, Scgb3a2(-/-) mice in mixed background showed a decreased OVA-induced airway inflammation, while six times C57BL/6NCr backcrossed congenic Scgb3a2(-/-) mice showed a slight exacerbation of OVA-induced airway inflammation as compared to wild-type littermates. These results indicate that the loss of SCGB3A2 function was influenced by a modifier gene(s) in mixed genetic background and suggest that SCGB3A2 has anti-inflammatory property. The results further suggest the possible use of recombinant human SCGB3A2 as an anti-inflammatory agent.


Asunto(s)
Ovalbúmina/farmacología , Neumonía/inducido químicamente , Neumonía/metabolismo , Secretoglobinas/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Ratones , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Secretoglobinas/deficiencia , Secretoglobinas/genética
20.
Proc Natl Acad Sci U S A ; 108(4): 1615-20, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21220306

RESUMEN

Mutations of BRAF are found in ∼45% of papillary thyroid cancers and are enriched in tumors with more aggressive properties. We developed mice with a thyroid-specific knock-in of oncogenic Braf (LSL-Braf(V600E)/TPO-Cre) to explore the role of endogenous expression of this oncoprotein on tumor initiation and progression. In contrast to other Braf-induced mouse models of tumorigenesis (i.e., melanomas and lung), in which knock-in of Braf(V600E) induces mostly benign lesions, Braf-expressing thyrocytes become transformed and progress to invasive carcinomas with a very short latency, a process that is dampened by treatment with an allosteric MEK inhibitor. These mice also become profoundly hypothyroid due to deregulation of genes involved in thyroid hormone biosynthesis and consequently have high TSH levels. To determine whether TSH signaling cooperates with oncogenic Braf in this process, we first crossed LSL-Braf(V600E)/TPO-Cre with TshR knockout mice. Although oncogenic Braf was appropriately activated in thyroid follicular cells of these mice, they had a lower mitotic index and were not transformed. Thyroid-specific deletion of the Gsα gene in LSL-Braf(V600E)/TPO-Cre/Gnas-E1(fl/fl) mice also resulted in an attenuated cancer phenotype, indicating that the cooperation of TshR with oncogenic Braf is mediated in part by cAMP signaling. Once tumors were established in mice with wild-type TshR, suppression of TSH did not revert the phenotype. These data demonstrate the key role of TSH signaling in Braf-induced papillary thyroid cancer initiation and provide experimental support for recent observations in humans pointing to a strong association between TSH levels and thyroid cancer incidence.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptores de Tirotropina/metabolismo , Transducción de Señal , Neoplasias de la Tiroides/metabolismo , Animales , Carcinoma , Carcinoma Papilar , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas B-raf/genética , Radioinmunoensayo , Receptores de Tirotropina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cáncer Papilar Tiroideo , Glándula Tiroides/metabolismo , Glándula Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Tirotropina/sangre , Tirotropina/metabolismo , Tiroxina/sangre , Tiroxina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA