Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Recept Signal Transduct Res ; 41(4): 371-377, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32896205

RESUMEN

Transmembrane proteins (TMEMs), spanning the entire width of lipid bilayers and anchored to them permanently, exist in diverse cell types to implement a series of essential physiological functions. Recently, TMEM48, a member of the TMEM family, has been demonstrated to be closely associated with tumorigenesis. However, little is known about the specific role of TMEM48 in cervical cancer (CC). This study aimed to investigate the biological functions of TMEM48 in CC. The CCK-8 assay was performed to detect CC cell proliferation. The wound healing and transwell assays were conducted to measure cell migration and invasion, respectively. The levels of TMEM48, ß-catenin, T cell factor 1(TCF1) and axis formation inhibitor 2 (AXIN2) were examined by the western blot analysis. Xenograft models were established for the tumorigenesis assay in vivo. The results showed that TMEM48 was overexpressed in CC tissues and cell lines. Knockdown of TMEM48 significantly inhibited CC cell proliferation, migration and invasion in vitro and suppressed CC cell growth in vivo. In addition, the investigation on the molecular mechanisms indicated that TMEM48 down-regulation remarkably decreased the protein levels of ß-catenin, TCF1 and AXIN2 in CC cells and TMEM48 exerted its promoting effect on CC progression via activation of the Wnt/ß-catenin pathway. Taken together, our study suggested TMEM48 as a promising therapeutic target for CC treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Complejo Poro Nuclear/biosíntesis , Neoplasias del Cuello Uterino/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Proteína Axina/biosíntesis , Proteína Axina/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Células HeLa , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Inmunohistoquímica , Membrana Dobles de Lípidos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Cicatrización de Heridas
2.
Immunity ; 53(3): 658-671.e6, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937153

RESUMEN

Identifying signals in the tumor microenvironment (TME) that shape CD8+ T cell phenotype can inform novel therapeutic approaches for cancer. Here, we identified a gradient of increasing glucocorticoid receptor (GR) expression and signaling from naïve to dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). Conditional deletion of the GR in CD8+ TILs improved effector differentiation, reduced expression of the transcription factor TCF-1, and inhibited the dysfunctional phenotype, culminating in tumor growth inhibition. GR signaling transactivated the expression of multiple checkpoint receptors and promoted the induction of dysfunction-associated genes upon T cell activation. In the TME, monocyte-macrophage lineage cells produced glucocorticoids and genetic ablation of steroidogenesis in these cells as well as localized pharmacologic inhibition of glucocorticoid biosynthesis improved tumor growth control. Active glucocorticoid signaling associated with failure to respond to checkpoint blockade in both preclinical models and melanoma patients. Thus, endogenous steroid hormone signaling in CD8+ TILs promotes dysfunction, with important implications for cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glucocorticoides/metabolismo , Macrófagos/metabolismo , Melanoma Experimental/patología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Hematopoyesis/inmunología , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inhibidores de Puntos de Control Inmunológico , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/inmunología
3.
Acta Biochim Biophys Sin (Shanghai) ; 52(3): 241-250, 2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-32072180

RESUMEN

Pancreatic ductal adenocarcinoma is one of the deadliest malignant tumors, and many genes play important roles in its development. The hepatocyte nuclear factor-1a (HNF-1a) gene encodes HNF-1a, which is a transcriptional activator. HNF-1a regulates the tissue-specific expression of multiple genes, especially in pancreatic islet cells and in the liver. However, the role of the HNF-1a gene in the development of pancreatic cancer is still unclear. Here, we used immunohistochemical staining and real-time PCR to analyze HNF-1a expression in pancreatic cancer tissue. Stable cell lines with HNF-1a knockdown or overexpression were established to analyze the role of HNF-1a in pancreatic cancer cell proliferation and apoptosis by colony formation assay and flow cytometry. We also analyzed the L-type pyruvate kinase (PKLR) promoter sequence to identify the regulatory effect of HNF-1a on PKLR transcription and confirmed the HNF-1a binding site in the PKLR promoter via a chromatin immunoprecipitation assay. HNF-1a was found to be overexpressed in pancreatic cancer and promoted proliferation while inhibiting apoptosis in pancreatic cancer cells. PKLR was identified as the downstream target gene of HNF-1a and binding of HNF-1a at two sites in PKLR (-1931/-1926 and -966/-961) regulated PKLR transcription. In conclusion, HNF-1a is overexpressed in pancreatic cancer, and the transcription factor HNF-1a can promote pancreatic cancer growth and apoptosis resistance via its target gene PKLR.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito/metabolismo , Neoplasias Pancreáticas/metabolismo , Piruvato Quinasa/metabolismo , Apoptosis/efectos de los fármacos , Sitios de Unión , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Inmunohistoquímica/métodos , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional
4.
Sci Rep ; 9(1): 7693, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118464

RESUMEN

Genome-wide association studies (GWAS) have identified many genetic variants in genes related to lipid metabolism. However, how these variations affect lipid levels remains elusive. Long non-coding RNAs (lncRNAs) have been implicated in a variety of biological processes. We hypothesize lncRNAs are likely to be located within disease or trait-associated DNA regions to regulate lipid metabolism. The aim of this study was to investigate whether and how lncRNAs in lipid- associated DNA regions regulate cholesterol homeostasis in hepatocytes. In this study, we identified a novel long non-coding RNA in Lipid Associated Single nucleotide polymorphism gEne Region (LASER) by bioinformatic analysis. We report that LASER is highly expressed in both hepatocytes and peripheral mononuclear cells (PBMCs). Clinical studies showed that LASER expression is positively related with that of cholesterol containing apolipoprotein levels. In particular, we found that LASER is positively correlated with plasma PCSK9 levels in statin free patients. siRNAs mediated knock down of LASER dramatically reduces intracellular cholesterol levels and affects the expression of genes involved in cholesterol metabolism. Transcriptome analyses show that knockdown of LASER affects the expression of genes involved in metabolism pathways. We found that HNF-1α and PCSK9 were reduced after LASER knock-down. Interestingly, the reduction of PCSK9 can be blocked by the treatment of berberine, a natural cholesterol-lowering compound which functions as a HNF-1α antagonist. Mechanistically, we found that LASER binds to LSD1 (lysine-specific demethylase 1), a member of CoREST/REST complex, in nucleus. LASER knock-down enhance LSD1 targeting to genomic loci, resulting in decreased histone H3 lysine 4 mono-methylation at the promoter regions of HNF-1α gene. Conversely, LSD1 knock-down abolished the effect of LASER on HNF-1α and PCSK9 expressions. Finally, we found that statin treatment increased LASER expression, accompanied with increased PCSK9 expression, suggesting a feedback regulation of cholesterol on LASER expression. This observation may partly explain the statin escape during anti-cholesterol treatment. These findings identified a novel lncRNA in cholesterol homeostasis. Therapeutic targeting LASER might be an effective approach to augment the effect of statins on cholesterol levels in clinics.


Asunto(s)
Colesterol/metabolismo , Hepatocitos/metabolismo , Leucocitos Mononucleares/metabolismo , ARN Largo no Codificante/fisiología , Anticolesterolemiantes/farmacología , Anticolesterolemiantes/uso terapéutico , Apolipoproteínas/metabolismo , Atorvastatina/farmacología , Atorvastatina/uso terapéutico , Benzoatos/farmacología , Bencilaminas/farmacología , Berberina/farmacología , Cromosomas Humanos Par 11/genética , Enfermedad Coronaria/tratamiento farmacológico , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células Hep G2 , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Código de Histonas/efectos de los fármacos , Histona Demetilasas/metabolismo , Homeostasis/genética , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/metabolismo , Proproteína Convertasa 9/biosíntesis , Proproteína Convertasa 9/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Interferencia de ARN , ARN Largo no Codificante/genética , ARN Interferente Pequeño/genética
5.
J Pharmacol Exp Ther ; 368(3): 353-362, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30602592

RESUMEN

Expression of cytochrome P450s (P450s) is regulated by epigenetic factors, such as DNA methylation, histone modifications, and noncoding RNAs through different mechanisms. Among these factors, long noncoding RNAs (lncRNAs) have been shown to play important roles in the regulation of gene expression; however, little is known about the effects of lncRNAs on the regulation of P450 expression. The aim of this study was to explore the role of lncRNAs in the regulation of P450 expression by using human liver tissues and hepatoma Huh7 cells. Through lncRNA microarray analysis and quantitative polymerase chain reaction in human liver tissues, we found that the lncRNA hepatocyte nuclear factor 1 alpha antisense 1 (HNF1α-AS1), an antisense RNA of HNF1α, is positively correlated with the mRNA expression of CYP2C8, 2C9, 2C19, 2D6, 2E1, and 3A4 as well as pregnane X receptor (PXR) and constitutive androstane receptor (CAR). Gain- and loss-of-function studies in Huh7 cells transfected with small interfering RNAs or overexpression plasmids showed that HNF1α not only regulated the expression of HNF1α-AS1 and P450s, but also regulated the expression of CAR, PXR, and aryl hydrocarbon receptor (AhR). In turn, HNF1α-AS1 regulated the expression of PXR and most P450s without affecting the expression of HNF1α, AhR, and CAR. Moreover, the rifampicin-induced expression of P450s was also affected by HNF1α and HNF1α-AS1. In summary, the results of this study suggested that HNF1α-AS1 is involved in the HNF1α-mediated regulation of P450s in the liver at both basal and drug-induced levels.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Hígado/metabolismo , ARN Largo no Codificante/biosíntesis , Línea Celular Tumoral , Sistema Enzimático del Citocromo P-450/genética , Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , ARN Largo no Codificante/genética
6.
J Clin Invest ; 129(1): 246-251, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30507613

RESUMEN

Using an integrated approach to characterize the pancreatic tissue and isolated islets from a 33-year-old with 17 years of type 1 diabetes (T1D), we found that donor islets contained ß cells without insulitis and lacked glucose-stimulated insulin secretion despite a normal insulin response to cAMP-evoked stimulation. With these unexpected findings for T1D, we sequenced the donor DNA and found a pathogenic heterozygous variant in the gene encoding hepatocyte nuclear factor-1α (HNF1A). In one of the first studies of human pancreatic islets with a disease-causing HNF1A variant associated with the most common form of monogenic diabetes, we found that HNF1A dysfunction leads to insulin-insufficient diabetes reminiscent of T1D by impacting the regulatory processes critical for glucose-stimulated insulin secretion and suggest a rationale for a therapeutic alternative to current treatment.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Variación Genética , Factor Nuclear 1-alfa del Hepatocito , Células Secretoras de Insulina/metabolismo , Transcripción Genética , Adolescente , Adulto , Diabetes Mellitus Tipo 1/patología , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Heterocigoto , Humanos , Células Secretoras de Insulina/patología , Masculino
7.
Blood Adv ; 2(14): 1685-1690, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30021780

RESUMEN

Expression of the transcription factor T-cell factor 1 (TCF1) identifies antigen-experienced murine CD8+ T cells that retain potential for lymphoid recirculation and the ability to self-renew while producing more differentiated effector cells. We found that CD8+ T cells in the blood of both healthy and chronically infected humans expressed TCF1 at 3 distinct levels: high (TCF1-hi), intermediate (TCF1-int), and low (TCF1-lo). TCF1-hi cells could be found within both the naive and memory compartments and were characterized by relative quiescence and lack of immediate effector function. A substantial fraction of TCF1-int cells were found among memory cells, and TCF1-int cells exhibited robust immediate effector functions. TCF1-lo cells were most enriched in effector memory cells that expressed the senescence marker CD57. Following reactivation, TCF1-hi cells gave rise to TCF1-lo descendants while self-renewing the TCF1-hi progenitor. By contrast, reactivation of TCF1-lo cells produced more TCF1-lo cells without evidence of de-differentiating into TCF1-hi cells. Flow cytometric analyses of TCF1 expression from patient specimens may become a useful biomarker for adaptive immune function in response to vaccination, infection, autoimmunity, and cancer.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Memoria Inmunológica , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Femenino , Factor Nuclear 1-alfa del Hepatocito/inmunología , Humanos , Masculino
8.
Development ; 145(10)2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29712641

RESUMEN

Nephron progenitor cells (NPCs) are Six2-positive metanephric mesenchyme cells, which undergo self-renewal and differentiation to give rise to nephrons until the end of nephrogenesis. Histone deacetylases (HDACs) are a group of epigenetic regulators that control cell fate, but their role in balancing NPC renewal and differentiation is unknown. Here, we report that NPC-specific deletion of Hdac1 and Hdac2 genes in mice results in early postnatal lethality owing to renal hypodysplasia and loss of NPCs. HDAC1/2 interact with the NPC renewal regulators Six2, Osr1 and Sall1, and are co-bound along with Six2 on the Six2 enhancer. Although the mutant NPCs differentiate into renal vesicles (RVs), Hdac1/2 mutant kidneys lack nascent nephrons or mature glomeruli, a phenocopy of Lhx1 mutants. Transcriptional profiling and network analysis identified disrupted expression of Lhx1 and its downstream genes, Dll1 and Hnf1a/4a, as key mediators of the renal phenotype. Finally, although HDAC1/2-deficient NPCs and RVs overexpress hyperacetylated p53, Trp53 deletion failed to rescue the renal dysgenesis. We conclude that the epigenetic regulators HDAC1 and HDAC2 control nephrogenesis via interactions with the transcriptional programs of nephron progenitors and renal vesicles.


Asunto(s)
Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Nefronas/embriología , Organogénesis/genética , Células Madre/citología , Transcripción Genética/genética , Animales , Proteínas de Unión al Calcio , Línea Celular , Proliferación Celular/genética , Células HEK293 , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 4 del Hepatocito/biosíntesis , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Enfermedades Renales/genética , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Noqueados , Nefronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/genética
9.
Toxicol Lett ; 264: 1-11, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27818225

RESUMEN

Sirtuin 1 (SIRT1) is the most conserved mammalian NAD+-dependent protein deacetylase and is a member of the silent information regulator 2 (Sir2) families of proteins (also known as Sirtuins). In the liver, hepatic SIRT1 modulates bile acid metabolism through the regulation of farnesoid X receptor (FXR) expression. FXR is one of the most important nuclear receptors involved in the regulation of bile acid metabolism. SIRT1 modulates the FXR expression at multiple levels, including direct deacetylation of this transcription factor and transcriptional regulation through hepatocyte nuclear factor 1α (HNF1α). Therefore, hepatic SIRT1 is a vital regulator of the HNF1α/FXR signalling pathway and hepatic bile acid metabolism. However, whether SIRT1 is a suitable therapeutic target for the treatment of cholestasis is unknown. In the present study, we examined the protective effect of SRT1720, which is a specific activator of SIRT1, against 17α-ethinylestradiol (EE)-induced cholestasis in mice. Our data demonstrated that SRT1720 significantly prevented EE-induced changes in the serum levels of total bile acids (TBA), total bilirubin (TBIL), γ-glutamyltranspeptidase (γ-GGT) and alkaline phosphatase (ALP). SRT1720 also relieved EE-induced liver pathological injuries as indicated by haematoxylin and eosin (H&E) staining. SRT1720 treatment protected against EE-induced liver injury through the HNF1α/FXR signalling pathway, which up-regulated the expression of hepatic efflux transporter (Bsep and Mrp2) and hepatic uptake transporters (Ntcp and Oatp1b2). Moreover, SRT1720 significantly inhibited the TNF-α and IL-6 levels induced by EE. These findings indicate that SRT1720 exerts a dose-dependent protective effect on EE-induced cholestatic liver injury in mice and that the mechanism underlying this activity is related to the activation of the HNF1α/FXR signalling pathway and anti-inflammatory mechanisms.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Colestasis/tratamiento farmacológico , Etinilestradiol/toxicidad , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Sustancias Protectoras/uso terapéutico , Receptores Citoplasmáticos y Nucleares/biosíntesis , Animales , Ácidos y Sales Biliares/metabolismo , Colestasis/inducido químicamente , Expresión Génica/efectos de los fármacos , Factor Nuclear 1-alfa del Hepatocito/genética , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , ARN Interferente Pequeño/farmacología , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/efectos de los fármacos
10.
Cell Res ; 25(8): 930-45, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26169608

RESUMEN

Hepatocytes are critical for the maintenance of liver homeostasis, but its involvement in hepatic fibrogenesis remains elusive. Hepatocyte nuclear factor 1α (HNF1α) is a liver-enriched transcription factor that plays a key role in hepatocyte function. Our previous study revealed a significant inhibitory effect of HNF1α on hepatocellular carcinoma. In this study, we report that the expression of HNF1α is significantly repressed in both human and rat fibrotic liver. Knockdown of HNF1α in the liver significantly aggravates hepatic fibrogenesis in either dimethylnitrosamine (DMN) or bile duct ligation (BDL) model in rats. In contrast, forced expression of HNF1α markedly alleviates hepatic fibrosis. HNF1α regulates the transcriptional expression of SH2 domain-containing phosphatase-1 (SHP-1) via directly binding to SHP-1 promoter in hepatocytes. Inhibition of SHP-1 expression abrogates the anti-fibrotic effect of HNF1α in DMN-treated rats. Moreover, HNF1α repression in primary hepatocytes leads to the activation of NF-κB and JAK/STAT pathways and initiates an inflammatory feedback circuit consisting of HNF1α, SHP-1, STAT3, p65, miR-21 and miR-146a, which sustains the deregulation of HNF1α in hepatocytes. More interestingly, a coordinated crosstalk between hepatocytes and hepatic stellate cells (HSCs) participates in this positive feedback circuit and facilitates the progression of hepatocellular damage. Our findings demonstrate that impaired hepatocytes play an active role in hepatic fibrogenesis. Early intervention of HNF1α-regulated inflammatory feedback loop in hepatocytes may have beneficial effects in the treatment of chronic liver diseases.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Hepatocitos/metabolismo , Hígado/patología , Animales , Comunicación Celular , Retroalimentación Fisiológica , Fibrosis , Humanos , Quinasas Janus/metabolismo , Hígado/metabolismo , FN-kappa B/metabolismo , Ratas , Factores de Transcripción STAT/metabolismo
11.
J Immunol ; 195(3): 1071-81, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26085685

RESUMEN

The IL-6 cytokine family utilizes the common signal transduction molecule gp130, which can mediate a diverse range of outcomes. To clarify the role of gp130 signaling in vivo during acute viral infection, we infected Cd4-cre Il6st(fl/fl) mice, in which gp130 is conditionally ablated in T cells, with acute lymphocytic choriomeningitis virus. We found that by day 12, but not at day 8, after infection the number of virus-specific CD4(+) T cells was reduced in the absence of gp130, and this was sustained for up to 2 mo postinfection. Additionally, gp130-deficient T follicular helper cells had lower expression of Maf, IL-21, and ICOS, and this was accompanied by a reduction in the proportion of germinal center B cells and plasmablasts. Remarkably, at 2 mo postinfection the proportion of IgG2a/c(+) memory B cells and the systemic levels of lymphocytic choriomeningitis virus-specific IgG2 Abs were dramatically decreased, whereas there was a corresponding increase in IgG1(+) memory B cells and virus-specific IgG1 Abs. In the same animals gp130-deficient virus-specific CD8(+) T cells showed a reduced proportion of memory cells, which expressed lower levels of Tcf7, and displayed diminished recall responses on secondary infection. Mixed bone marrow chimeras revealed that the aforementioned gp130 effects on CD4(+) T cells were cell intrinsic. Overall, our data show that gp130 signaling in T cells influences the quantity and quality of long-lasting CD4(+) T cell responses as well as CD8(+) T cell- and Ab-mediated immunity after acute viral infection.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Receptor gp130 de Citocinas/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/citología , Receptor gp130 de Citocinas/genética , Centro Germinal/citología , Centro Germinal/inmunología , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inmunoglobulina G/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/biosíntesis , Interleucina-6/inmunología , Interleucinas/biosíntesis , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-maf/biosíntesis , Transducción de Señal/inmunología
12.
J Clin Invest ; 125(4): 1533-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25774505

RESUMEN

The cause of organ failure is enigmatic for many degenerative diseases, including end-stage liver disease. Here, using a CCl4-induced rat model of irreversible and fatal hepatic failure, which also exhibits terminal changes in the extracellular matrix, we demonstrated that chronic injury stably reprograms the critical balance of transcription factors and that diseased and dedifferentiated cells can be returned to normal function by re-expression of critical transcription factors, a process similar to the type of reprogramming that induces somatic cells to become pluripotent or to change their cell lineage. Forced re-expression of the transcription factor HNF4α induced expression of the other hepatocyte-expressed transcription factors; restored functionality in terminally diseased hepatocytes isolated from CCl4-treated rats; and rapidly reversed fatal liver failure in CCl4-treated animals by restoring diseased hepatocytes rather than replacing them with new hepatocytes or stem cells. Together, the results of our study indicate that disruption of the transcription factor network and cellular dedifferentiation likely mediate terminal liver failure and suggest reinstatement of this network has therapeutic potential for correcting organ failure without cell replacement.


Asunto(s)
Redes Reguladoras de Genes , Terapia Genética , Vectores Genéticos/uso terapéutico , Cirrosis Hepática Experimental/terapia , Fallo Hepático/terapia , Factores de Transcripción/fisiología , Animales , Proteína alfa Potenciadora de Unión a CCAAT/biosíntesis , Proteína alfa Potenciadora de Unión a CCAAT/genética , Intoxicación por Tetracloruro de Carbono/genética , Intoxicación por Tetracloruro de Carbono/terapia , Desdiferenciación Celular/genética , Células Cultivadas , Dependovirus/genética , Progresión de la Enfermedad , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/biosíntesis , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/biosíntesis , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/fisiología , Hepatocitos/metabolismo , Hepatocitos/patología , Cirrosis Hepática Experimental/complicaciones , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/patología , Fallo Hepático/etiología , Fallo Hepático/genética , Fallo Hepático/patología , Masculino , PPAR alfa/biosíntesis , PPAR alfa/genética , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes de Fusión/metabolismo , Transcriptoma , Transducción Genética
13.
Am J Physiol Renal Physiol ; 307(11): F1283-91, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25234312

RESUMEN

The human organic anion transporter 1 (OAT1) is crucial for the excretion of organic anions in renal proximal tubular cells and has been classified as a clinically relevant transporter in the kidneys. Our previous study indicated that renal male-predominant expression of rat Oat1 and Oat3 appears to be regulated by transcription factor B-cell CLL/lymphoma 6 (BCL6). The aim of this study was to characterize the effect of BCL6 on human OAT1 promoter and on the transcription of OAT1 mediated by hepatocyte nuclear factor-1α (HNF-1α). Luciferase assays were carried out in opossum kidney (OK) cells transiently transfected with promoter constructs of OAT1, expression vectors for BCL6 and HNF-1α, and the empty control vectors. BCL6 and HNF-1α binding on OAT1 promoter was analyzed using electrophoretic mobility shift assay (EMSA). Protein expression of HNF-1α was investigated by Western blot analysis. Site-directed mutagenesis was used to introduce mutations into BCL6 and HNF-1α binding sites within the OAT1 promoter. BCL6 enhanced the promoter activity of OAT1 independently of predicted BCL6 binding sites but was dependent on HNF-1α response element and HNF-1α protein. Coexpression of BCL6 and HNF-1α induced an additive effect on OAT1 promoter activation compared with BCL6 or HNF-1α alone. BCL6 does not bind directly or indirectly to OAT1 promoter but increases the protein expression of HNF-1α and thereby indirectly enhances OAT1 gene transcription. BCL6 constitutes a promising candidate gene for the regulation of human OAT1 transcription and other renal and/or hepatic drug transporters that have been already shown to be activated by HNF-1α.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Zarigüeyas/fisiología , Proteína 1 de Transporte de Anión Orgánico/biosíntesis , Animales , Línea Celular , ADN/metabolismo , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Riñón/metabolismo , Mutagénesis Sitio-Dirigida , Mutación/genética , Mutación/fisiología , Proteína 1 de Transporte de Anión Orgánico/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-bcl-6
14.
J Immunol ; 193(6): 2784-91, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25127860

RESUMEN

Protection against reinfection is mediated by Ag-specific memory CD8 T cells, which display stem cell-like function. Because canonical Wnt (Wingless/Int1) signals critically regulate renewal versus differentiation of adult stem cells, we evaluated Wnt signal transduction in CD8 T cells during an immune response to acute infection with lymphocytic choriomeningitis virus. Whereas naive CD8 T cells efficiently transduced Wnt signals, at the peak of the primary response to infection only a fraction of effector T cells retained signal transduction and the majority displayed strongly reduced Wnt activity. Reduced Wnt signaling was in part due to the downregulation of Tcf-1, one of the nuclear effectors of the pathway, and coincided with progress toward terminal differentiation. However, the correlation between low and high Wnt levels with short-lived and memory precursor effector cells, respectively, was incomplete. Adoptive transfer studies showed that low and high Wnt signaling did not influence cell survival but that Wnt high effectors yielded memory cells with enhanced proliferative potential and stronger protective capacity. Likewise, following adoptive transfer and rechallenge, memory cells with high Wnt levels displayed increased recall expansion, compared with memory cells with low Wnt signaling, which were preferentially effector-like memory cells, including tissue-resident memory cells. Thus, canonical Wnt signaling identifies CD8 T cells with enhanced proliferative potential in part independent of commonly used cell surface markers to discriminate effector and memory T cell subpopulations. Interventions that maintain Wnt signaling may thus improve the formation of functional CD8 T cell memory during vaccination.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Proteínas Wnt/inmunología , Vía de Señalización Wnt/inmunología , Traslado Adoptivo , Animales , Proteína Axina/biosíntesis , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular/inmunología , Proliferación Celular , Regulación hacia Abajo , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Memoria Inmunológica/inmunología , Lectinas Tipo C , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/biosíntesis , Subgrupos de Linfocitos T/inmunología , Vacunación
15.
Exp Clin Endocrinol Diabetes ; 121(6): 368-71, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23616187

RESUMEN

Hepatocyte nuclear factor 1 alpha (HNF1A) mutations cause maturity-onset diabetes of the young (MODY) type 3. Further extending the phenotypic spectrum, HNF1A mutations are associated with hepatic adenomatosis. A 20-year old lean, female patient with newly diagnosed diabetes mellitus was negative for diabetes-associated autoantibodies and had no relevant family history. Hepatic adenomatosis was diagnosed. Her HNF1A gene was examined and identified alterations further analysed.Sequencing of her HNF1A gene revealed a previously uncharacterised Q495X nonsense mutation, along with the known A98V polymorphism, both in the heterozygous state. The patient's father was also a carrier of both the mutation and the polymorphism. An oral glucose tolerance test (OGTT) revealed impaired glucose tolerance, whereas imaging of his liver was unremarkable. Wild type HNF1A and HNF1A carrying the Q495X mutation were co-transfected in reporter gene assays. The mutation causes a dominant-negative HNF1A protein variant which blocks HNF1A wild-type-mediated gene expression.The novel Q495X mutation is the likely cause of our patient's diabetes and hepatic adenomatosis. It may also cause her father's impaired glucose tolerance. More generally speaking, if non-autoimmune diabetes is suspected, examination of the liver may provide important diagnostic clues. Furthermore, patients with hepatic adenomatosis without known diabetes should be screened by OGTT. Relatives of patients with HNF1A mutations should also be screened by OGTT to detect potential early-stage diabetes.


Asunto(s)
Adenoma de Células Hepáticas , Codón sin Sentido , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito , Neoplasias Hepáticas , Adenoma de Células Hepáticas/diagnóstico por imagen , Adenoma de Células Hepáticas/genética , Adenoma de Células Hepáticas/metabolismo , Adulto , Edad de Inicio , Diabetes Mellitus , Femenino , Células HEK293 , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Polimorfismo de Nucleótido Simple , Radiografía
16.
Int J Dev Biol ; 57(1): 49-54, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23585352

RESUMEN

TCF1 belongs to the family of LEF1/TCF transcription factors that regulate gene expression downstream of Wnt/ß-catenin signaling, which is crucial for embryonic development and is involved in adult stem cell regulation and tumor growth. In early Xenopus embryos, tcf1 plays an important role in mesoderm induction and patterning. Foxd3 emerged as a potential tcf1 target gene in a microarray analysis of gastrula stage embryos. Because foxd3 and tcf1 are coexpressed during gastrulation, we investigated whether foxd3 is regulated by tcf1. By using morpholino-mediated knockdown, we show that during gastrulation foxd3 expression is dependent on tcf1. By chromatin immunoprecipitation, we also demonstrate direct interaction of ß-catenin/tcf complexes with the foxd3 gene locus. Hence, our results indicate that tcf1 acts as an essential activator of foxd3, which is critical for dorsal mesoderm formation in early embryos.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Gastrulación , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Factores de Transcripción Forkhead/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Mesodermo/embriología , Morfolinos , Transducción de Señal/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Proteínas de Xenopus/biosíntesis , Xenopus laevis/genética , Xenopus laevis/metabolismo , beta Catenina/metabolismo
17.
Toxicol Lett ; 211(3): 304-11, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22531820

RESUMEN

Prior research suggests that sex hormones and metabolic changes, such as obesity and hyperglycemia, can alter renal transporter expression in rodents. The purpose of this study was to characterize the expression of kidney efflux transporters and regulatory transcription factors in response to Type I diabetes and pregnancy. Female C57BL/6 mice were treated with multiple low doses of streptozotocin (STZ) to induce hyperglycemia and then mated with normoglycemic male mice. Transporter mRNA and protein expression were quantified in kidneys from vehicle- and STZ-treated non-pregnant and pregnant mice on gestation day 14. Pregnancy decreased the expression of Mdr1b, Mrp4, and 5 proteins and increased the mRNA and protein expression of Mrp3 by 50-60%. STZ treatment elevated Mrp1, 2, 4, and 5 and reduced Mrp3, 6, and Mdr1b mRNA and/or protein in non-pregnant mice. Pregnancy had little effect on STZ-mediated changes in renal efflux transporter expression. Transcriptional profiles of Hnf1α, PXR, AhR, and Nrf2 were altered in patterns similar to some efflux transporters suggesting potential involvement in their regulation. Taken together, these results suggest that renal drug efflux transporters and regulatory signaling pathways are altered by endocrine and metabolic changes that occur during pregnancy and Type I diabetes.


Asunto(s)
Proteínas Portadoras/biosíntesis , Diabetes Mellitus Tipo 1/metabolismo , Riñón/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Glucemia/metabolismo , Western Blotting , Receptor de Androstano Constitutivo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica/efectos de los fármacos , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/biosíntesis , Tamaño de los Órganos/efectos de los fármacos , PPAR alfa/biosíntesis , Páncreas/patología , Embarazo , Receptor X de Pregnano , ARN Mensajero/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Hidrocarburo de Aril/biosíntesis , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores de Esteroides/biosíntesis , Transducción de Señal/efectos de los fármacos
18.
Gut ; 61(8): 1187-96, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21948943

RESUMEN

OBJECTIVES: During pancreatitis, specific transcriptional programmes govern functional regeneration after injury. The objective of this study was to analyse the dynamic regulation of pancreatic genes and the role of transcriptional regulators during recovery from pancreatitis. DESIGN: Wild-type and genetically modified mice (Hnf1α(-/-) and Ptf1a(+/-)) were used. After caerulein or L-arginine induced pancreatitis, blood or pancreata were processed for enzymatic assays, ELISA, histology, immunohistochemistry, western blotting and quantitative reverse transcriptase-PCR. Nr5a2 promoter reporter and chromatin immunoprecipitation assays for Hnf1α were also performed. RESULTS: After caerulein pancreatic injury, expression of acinar and endocrine genes rapidly decreased, but eventually recovered, depicting distinct cell-type-specific patterns. Pdx1 and Hnf1α mRNAs underwent marked downregulation, matching endocrine/exocrine gene expression profiles. Ptf1a, Pdx1 and Hnf1α protein levels were also reduced and recovered gradually. These changes were associated with transient impairment of exocrine and endocrine function, including abnormal glucose tolerance. On l-arginine pancreatitis, changes in Ptf1a, Pdx1 and Hnf1α gene and protein expression were recapitulated. Reduced Hnf1α and Ptf1a levels after pancreatitis coincided with increased acinar cell proliferation, both in Hnf1α(-/-) and Ptf1a(+/-) mice. Moreover, Hnf1α(-/-) mice had reduced Ptf1a protein as well as transcripts for Ptf1a and digestive enzymes. Dispersed acini from Hnf1α(-/-) mice showed suboptimal secretory responses to caerulein. Bioinformatics analysis did not support a role for Hnf1α as a direct regulator of digestive enzyme genes. Instead, it was found that Hnf1α binds to, and regulates, the promoter of Nr5a2, coding an orphan nuclear receptor that regulates acinar gene expression. CONCLUSIONS: Dynamic changes in gene expression occur on pancreatitis induction, determining altered exocrine and endocrine function. This analysis uncovers roles for Hnf1α in the regulation of acinar cell determination and function. This effect may be mediated, in part, through direct regulation of Nr5a2.


Asunto(s)
Células Acinares/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Homeostasis/genética , Pancreatitis/genética , ARN Mensajero/genética , Células Acinares/patología , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Pancreatitis/metabolismo , Pancreatitis/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
PLoS One ; 4(11): e7855, 2009 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-19924231

RESUMEN

BACKGROUND: The HNF1A, HNF1B and HNF4A genes are part of an autoregulatory network in mammalian pancreas, liver, kidney and gut. The layout of this network appears to be similar in rodents and humans, but inactivation of HNF1A, HNF1B or HNF4A genes in animal models cause divergent phenotypes to those seen in man. We hypothesised that some differences may arise from variation in the expression profile of alternatively processed isoforms between species. METHODOLOGY/PRINCIPAL FINDINGS: We measured the expression of the major isoforms of the HNF1A, HNF1B and HNF4A genes in human and rodent pancreas, islet, liver and kidney by isoform-specific quantitative real-time PCR and compared their expression by the comparative Ct (DeltaDeltaCt) method. We found major changes in the expression profiles of the HNF genes between humans and rodents. The principal difference lies in the expression of the HNF1A gene, which exists as three isoforms in man, but as a single isoform only in rodents. More subtle changes were to the balance of HNF1B and HNF4A isoforms between species; the repressor isoform HNF1B(C) comprised only 6% in human islets compared with 24-26% in rodents (p = 0.006) whereas HNF4A9 comprised 22% of HNF4A expression in human pancreas but only 11% in rodents (p = 0.001). CONCLUSIONS/SIGNIFICANCE: The differences we note in the isoform-specific expression of the human and rodent HNF1A, HNF1B and HNF4A genes may impact on the absolute activity of these genes, and therefore on the activity of the pancreatic transcription factor network as a whole. We conclude that alterations to expression of HNF isoforms may underlie some of the phenotypic variation caused by mutations in these genes.


Asunto(s)
Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 1-beta del Hepatocito/biosíntesis , Factor Nuclear 4 del Hepatocito/biosíntesis , Adulto , Empalme Alternativo , Animales , Femenino , Perfilación de la Expresión Génica , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Ratas , Ratas Wistar , Especificidad de la Especie
20.
Arch Toxicol ; 82(9): 573-82, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18677463

RESUMEN

UDP-glucuronosyltransferases contribute to the detoxification of drugs by forming water soluble beta-D-glucopyranosiduronic acids. The human UGT1A3 protein catalyzes the glucuronidation of estrogens, bile acids and xenobiotics including non-steroidal anti-inflammatory drugs and lipid lowering drugs. Regulation of UGT1A3 by xenobiotic response elements is likely, but the responsible elements are yet uncharacterized. In addition, genetic promoter variants may affect UGT1A3 regulation and potential induction by xenobiotics. The UGT1A3 promoter was analyzed by mutagenesis, reporter gene, and mobility shift analyses. Three hundred and eighty-nine blood donors were genotyped for promoter single nucleotide polymorphisms (SNPs) showing an allelic frequency of 42% of variants at -66 (T to C) and -204 (A to G). A xenobiotic response element regulating aryl hydrocarbon receptor (AhR)-mediated UGT1A3 transcription was identified and characterized. UGT1A3 transcription was reduced in the presence of promoter SNPs. These data demonstrate xenobiotic induced regulation of the UGT1A3 gene by the AhR, which shows genetic variability.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Estrógenos/biosíntesis , Glucuronosiltransferasa/biosíntesis , Receptores de Hidrocarburo de Aril/fisiología , Regiones no Traducidas 5'/genética , Alelos , Cartilla de ADN , Regulación Enzimológica de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Genes Reporteros/genética , Genotipo , Glucuronosiltransferasa/genética , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Humanos , Hígado/enzimología , Luciferasas/genética , Mutagénesis , Polimorfismo de Nucleótido Simple/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Xenobióticos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...