Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Diabetes Metab Syndr Obes ; 16: 1731-1743, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37323855

RESUMEN

Background: Profilin-1 (PFN1) regulates the dynamic balance of actin and plays an important role in cell functions as a hub protein in signaling molecule interaction networks. Dysregulation of PFN1 is related to pathologic kidney diseases. Diabetic nephropathy (DN) was recently reported as an inflammatory disorder, however, the molecular mechanisms of PFN1 in DN remain unclear. Therefore, the present study was conducted to explore the molecular and bioinformatic characteristics of PFN1 in DN. Methods: Bioinformatics analyses were performed on the chip of database in DN kidney tissues. A cellular model of DN was established in human renal tubular epithelial cells (HK-2) induced by high glucose. The PFN1 gene was overexpressed or knocked-down to investigate its function in DN. Flow cytometry was used to detect cell proliferation and apoptosis. PFN1 and proteins in the related signaling pathways were evaluated by Western blotting. Results: The expression of PFN1 was significantly increased in DN kidney tissues (P < 0.001) and was correlated with a high apoptosis-associated score (Pearson's correlation = 0.664) and cellular senescence-associated score (Pearson's correlation = 0.703). PFN1 protein was mainly located in cytoplasm. Overexpression of PFN1 promoted apoptosis and blocked the proliferation of HK-2 cells treated with high levels of glucose. Knockdown of PFN1 led to the opposite effects. Additionally, we found that PFN1 was correlated with the inactivation of the Hedgehog signaling pathway in HK-2 cells treated with high levels of glucose. Conclusion: PFN1 might play an integral role in the regulation of cell proliferation and apoptosis during DN development by activating the Hedgehog signaling pathway. This study provided molecular and bioinformatic characterizations of PFN1, and contributed to the understanding of the molecular mechanisms leading to DN.

2.
Adipocyte ; 11(1): 420-433, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35975941

RESUMEN

Strong links have been reported among trimethylamine N-oxide (TMAO), visceral white adipose tissue (vWAT), and cardiometabolic diseases. However, the effects of TMAO on vWAT in hypertension remained incompletely explored. The impact of a chronic 22-week-long treatment with 1 g/L TMAO on vWAT, and its transcriptional and metabolic changes in spontaneously hypertensive rats (SHRs) were evaluated by serum cytokine measurements, histological analysis, fatty acid determinations, and co-expression network analyses. TMAO increased the serum interleukin-6 levels and insulin secretion in SHRs. The adipocyte size was diminished in the SHR 1 g/L TMAO group. In addition, one kind of monounsaturated fatty acids (cis-15-tetracosenoate) and four kinds of polyunsaturated fatty acids (cis-11,14,17-eicosatrienoic acid, docosatetraenoate, docosapentaenoate n-3, and docosapentaenoate n-6) were elevated by TMAO treatment. Three co-expression modules significantly related to TMAO treatment were identified and pathway enrichment analyses indicated that phagosome, lysosome, fatty acid metabolism, valine, leucine, and isoleucine degradation and metabolic pathways were the most significantly altered biological pathways. This study shed new light on the metabolic roles of TMAO on the vWAT of SHRs. TMAO regulated the metabolic status of vWAT, including reduced lipogenesis and an improved specific fatty acid composition. The mechanisms underlying these effects likely involve phagosome and lysosome pathways.


Asunto(s)
Tejido Adiposo Blanco , Metilaminas , Tejido Adiposo Blanco/metabolismo , Animales , Ácidos Grasos , Metilaminas/metabolismo , Ratas , Ratas Endogámicas SHR
3.
Cell Death Dis ; 13(2): 141, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35145073

RESUMEN

Metastatic tumors are mainly composed of neoplastic cells escaping from the primary tumor and inflammatory cells egressing from bone marrow. Cancer cell and inflammatory cell are remained in the state of immaturity during migration to distant organs. Here, we show that ADRB3 is crucial in cell mobilization and differentiation. Immunohistochemistry revealed ADRB3 expression is significantly more frequent in breast cancer tissues than in adjacent noncancerous tissues (92.1% vs. 31.5%). Expression of ADRB3 correlated with malignant degree, TNM stage and poor prognosis. Moreover, ADRB3 expression was markedly high in activated disseminated tumor cells, myeloid-derived suppressor cells (MDSCs), lymphocytes and neutrophil extracellular traps of patients. Importantly, ADRB3 promoted the expansion of MDSC through stimulation of bone marrow mobilization and inhibiting of the differentiation of immature myeloid cells. Furthermore, ADRB3 promoted MCF-7 cells proliferation and inhibited transdifferentiation into adipocyte-like cell by activating mTOR pathway. Ultimately, the MDSC-deficient phenotype of ADRB3 -/- PyMT mice was associated with impairment of mammary tumorigenesis and reduction in pulmonary metastasis. Collectively, ADRB3 promotes metastasis by inducing mobilization and inhibiting differentiation of both breast cancer cells and MDSCs.


Asunto(s)
Neoplasias de la Mama , Células Supresoras de Origen Mieloide , Receptores Adrenérgicos beta 3 , Animales , Neoplasias de la Mama/patología , Diferenciación Celular , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , Células Mieloides/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo
4.
Stem Cells Int ; 2021: 5555590, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484348

RESUMEN

METHODS: Hypoxia in hBMSCs was induced for 0, 4, and 12 hours, and cellular senescence was evaluated by senescence-associated ß-galactosidase (SA-ß-gal) staining. Tandem mass tag (TMT) labeling was combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) for differential proteomic analysis of hypoxia in hBMSCs. Parallel reaction monitoring (PRM) analysis was used to validate the candidate proteins. Verifications of signaling pathways were evaluated by western blotting. Cell apoptosis was evaluated using Annexin V/7-AAD staining by flow cytometry. The production of reactive oxygen species (ROS) was detected by the fluorescent probe 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA). RESULTS: Cell senescence detected by SA-ß-gal activity was higher in the 12-hour hypoxia-induced group. TMT analysis of 12-hour hypoxia-induced cells identified over 6000 proteins, including 686 differentially expressed proteins. Based on biological pathway analysis, we found that the senescence-associated proteins were predominantly enriched in the cancer pathways, PI3K-Akt pathway, and cellular senescence signaling pathways. CDK1, CDK2, and CCND1 were important nodes in PPI analyses. Moreover, the CCND1, UQCRH, and COX7C expressions were verified by PRM. Hypoxia induction for 12 hours in hBMSCs reduced CCND1 expression but promoted ROS production and cell apoptosis. Such effects were markedly reduced by the PI3K agonist, 740 Y-P, and attenuated by LY294002. CONCLUSIONS: Hypoxia of hBMSCs inhibited CCND1 expression but promoted ROS production and cell apoptosis through activating the PI3K-dependent signaling pathway. These findings provided a detailed characterization of the proteomic profiles related to hypoxia-induced senescence of hBMSCs and facilitated our understanding of the molecular mechanisms leading to stem cell senescence.

5.
Cancer Immunol Immunother ; 69(11): 2345-2355, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32514619

RESUMEN

The cross-talk between cancer cells and monocyte-derived alveolar macrophages (Mo-AMs) promotes non-small cell lung carcinoma (NSCLC) progression. In this study, we report that both cancer cells and Mo-AMs robustly express beta 3-adrenergic receptor (ADRB3) in NSCLC. ADRB3 supports lung cancer cells proliferation and promotes chronic inflammation. Genetic and pharmacologic inhibition of ADRB3 reverses tumor growth and inflammation in mouse. Furthermore, we demonstrate that M5D1, a novel anti-ADRB3 monoclonal antibody, inhibits human lung cancer cells proliferation and inflammation via affecting the intracellular mTOR pathway and activating p53. In NSCLC patients, we confirmed that upregulation of ADRB3 expression correlates with tumor progression and poor prognosis. Altogether, these results shed light on the role of ADRB3 in NSCLC and suggest that M5D1 could become powerful antitumor weapons.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Macrófagos Alveolares/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Pronóstico
6.
Oncol Lett ; 18(2): 1786-1792, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31423246

RESUMEN

Cervical cancer (CC) remains a highly prevalent cancer and cause of mortality amongst women worldwide. miR-758 has been demonstrated to be associated with tumorigenesis by controlling the expression of oncogenic or tumor suppressor genes. However, the function and mechanisms of miR-758 in CC have not been well illustrated. The present study aimed to dissect the effect of miR-758 on the proliferation, migration and invasion of CC cells and determine the potential underlying molecular mechanism of these effects. qPCR results revealed that the expression of miR-758 was significantly decreased in CC tissues and cell lines compared with that in normal tissues and normal cells. Results of CCK-8, colony formation and Transwell assays revealed that miR-758 overexpression markedly decreased cell viability, proliferation, invasion and migration. However, miR-758 inhibitors significantly increased viability, proliferation, invasion and migration. In the mechanism study, we demonstrated that high mobility group box 3 (HMGB3) was a direct target of miR-758, and HMGB3 overexpression rescued the viability, proliferation, invasion and migration of HeLa cells reduced by an miR-758 mimic. It was demonstrated that HMGB3 regulated the WNT/ß-catenin signaling pathway under miR-758 regulation. In summary, these observations suggested that miR-758 is a tumor suppressor gene that can inhibit the metastatic phenotype of CC cells by negatively regulating HMGB3, which may present a path to novel therapeutic stratagems for CC therapy.

7.
Lancet Diabetes Endocrinol ; 6(8): 627-636, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29735394

RESUMEN

BACKGROUND: Glucokinase acts as a glucose sensor in the pancreas and a glucose processor in the liver, and has a central role in glucose homoeostasis. Dorzagliatin is a new, dual-acting, allosteric glucokinase activator that targets both pancreatic and hepatic glucokinases. Dorzagliatin has good pharmacokinetic and pharmacodynamic properties in humans, and provides effective 24-h glycaemic control and improves glucose sensitivity in patients with type 2 diabetes. We aimed to assess the efficacy and safety of dorzagliatin monotherapy at different doses in Chinese patients with type 2 diabetes. METHODS: In this multicentre, randomised, double-blind, placebo-controlled, phase 2 study, we randomly assigned (1:1:1:1:1) patients to receive oral placebo or one of four doses of oral dorzagliatin (75 mg once a day, 100 mg once a day, 50 mg twice a day, or 75 mg twice a day) using permuted-block randomisation, with a block size of ten and without stratification. Eligible patients were men or non-fertile women (aged 40-75 years) with type 2 diabetes who had a BMI of 19·0-30·0 kg/m2, were on a diet and exercise regimen, and were previously untreated or treated with metformin or α-glucosidase inhibitor monotherapy. The study started with a 4-week placebo run-in period followed by a 12-week treatment period. The primary endpoint was the change in HbA1c from baseline to week 12, which was assessed in all patients who received at least one dose of study drug and had both baseline and at least one post-baseline HbA1c value. Safety was assessed in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT02561338. FINDINGS: Between Sept 29, 2015, and Aug 17, 2016, we randomly assigned 258 patients to one of the five study groups. At the end of 12 weeks, the least squares mean change in HbA1c from baseline was -0·35% (95% CI -0·60 to -0·10) in the placebo group, -0·39% (-0·64 to -0·14) in the 75 mg once daily group, -0·65% (-0·92 to -0·38) in the 100 mg once daily group, -0·79% (-1·06 to -0·52) in the 50 mg twice daily group, and -1·12% (-1·39 to -0·86) in the 75 mg twice daily group. Compared with the placebo group, the change in HbA1c between baseline and 12 weeks was significant in the 50 mg twice daily (p=0·0104) and the 75 mg twice daily (p<0·0001) groups. The number of adverse events was similar between the treatment groups and the placebo group. There were no reports of drug-related serious adverse events or severe hypoglycaemia. INTERPRETATION: Dorzagliatin had a beneficial effect on glycaemic control and was safe and well tolerated over 12 weeks in Chinese patients with type 2 diabetes. FUNDING: Hua Medicine, National Major Scientific and Technological Special Project for Significant New Drugs Development, Shanghai Science and Technology Innovation Action Project, Shanghai Pudong District Science and Technology Innovation Action Project, and Shanghai Municipal Commission of Economy and Informatisation Innovation Action Project.


Asunto(s)
Biomarcadores/análisis , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Pirazoles/uso terapéutico , Adulto , Anciano , Pueblo Asiatico , Glucemia/análisis , Método Doble Ciego , Activadores de Enzimas/uso terapéutico , Femenino , Estudios de Seguimiento , Hemoglobina Glucada/análisis , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
8.
PLoS One ; 10(12): e0143802, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26625141

RESUMEN

Fibrosis, the hallmark of human injuries and diseases such as serious burns, is characterized by excessive collagen synthesis and myofibroblast accumulation. Transforming growth factor-ß (TGF-ß), a potent inducer of collagen synthesis, has been implicated in fibrosis in animals. In addition to TGF-ß, fibroblast growth factor-inducible molecule 14 (Fn14) has been reported to play an important role in fibrotic diseases, such as cardiac fibrosis. However, the function and detailed regulatory mechanism of Fn14 in fibrosis are unclear. Here, we investigated the effect of Fn14 on the activation of human dermal fibroblasts. In normal dermal fibroblasts, TGF-ß signaling increased collagen production and Fn14 expression. Furthermore, Fn14 siRNA blocked extracellular matrix gene expression; even when TGF-ß signaling was activated by TGF-ß1, fibroblast activation remained blocked in the presence of Fn14 siRNA. Overexpressing Fn14 increased extracellular matrix gene expression. In determining the molecular regulatory mechanism, we discovered that SMAD4, an important TGF-ß signaling co-mediator, bound to the Fn14 promoter and activated Fn14 transcription. Taken together, these results indicate that the TGF-ß signaling pathway activates Fn14 expression through the transcription factor SMAD4 and that activated Fn14 expression increases extracellular matrix synthesis and fibroblast activation. Therefore, Fn14 may represent a promising approach to preventing the excessive accumulation of collagen or ECM in skin fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal , Factor de Crecimiento Transformador beta/fisiología , Adulto , Colágeno/biosíntesis , Matriz Extracelular/metabolismo , Humanos , Persona de Mediana Edad , Receptores del Factor de Necrosis Tumoral/genética , Piel/citología , Proteína Smad4/metabolismo , Receptor de TWEAK , Activación Transcripcional , Adulto Joven
9.
J Biol Chem ; 287(1): 474-483, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22081605

RESUMEN

NOTCH signaling induced by Delta1 (DLL1) and Jagged1 (JAG1) NOTCH ligands is modulated by the ß3N-acetylglucosaminyl transferase Fringe. LFNG (Lunatic Fringe) and MFNG (Manic Fringe) transfer N-acetylglucosamine (GlcNAc) to O-fucose attached to EGF-like repeats of NOTCH receptors. In co-culture NOTCH signaling assays, LFNG generally enhances DLL1-induced, but inhibits JAG1-induced, NOTCH signaling. In mutant Chinese hamster ovary (CHO) cells that do not add galactose (Gal) to the GlcNAc transferred by Fringe, JAG1-induced NOTCH signaling is not inhibited by LFNG or MFNG. In mouse embryos lacking B4galt1, NOTCH signaling is subtly reduced during somitogenesis. Here we show that DLL1-induced NOTCH signaling in CHO cells was enhanced by LFNG, but this did not occur in either Lec8 or Lec20 CHO mutants lacking Gal on O-fucose glycans. Lec20 mutants corrected with a B4galt1 cDNA became responsive to LFNG. By contrast, MFNG promoted DLL1-induced NOTCH signaling better in the absence of Gal than in its presence. This effect was reversed in Lec8 cells corrected by expression of a UDP-Gal transporter cDNA. The MFNG effect was abolished by a DDD to DDA mutation that inactivates MFNG GlcNAc transferase activity. The binding of soluble NOTCH ligands and NOTCH1/EGF1-36 generally reflected changes in NOTCH signaling caused by LFNG and MFNG. Therefore, the presence of Gal on O-fucose glycans differentially affects DLL1-induced NOTCH signaling modulated by LFNG versus MFNG. Gal enhances the effect of LFNG but inhibits the effect of MFNG on DLL1-induced NOTCH signaling, with functional consequences for regulating the strength of NOTCH signaling.


Asunto(s)
Galactosa/metabolismo , Glicosiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Células CHO , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Cricetinae , Cricetulus , Factor de Crecimiento Epidérmico/metabolismo , Espacio Extracelular/metabolismo , Glucosiltransferasas , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/química , Ratones , Polisacáridos/metabolismo , Estructura Terciaria de Proteína , Receptores Notch/química , Proteínas Serrate-Jagged , Solubilidad
10.
J Biol Chem ; 285(46): 36245-54, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20837470

RESUMEN

Mammalian Notch receptors require modification by fucose on epidermal growth factor-like (EGF) repeats of their extracellular domain to respond optimally to signal induction by canonical Notch ligands. Inactivation of the Golgi GDP-fucose transporter Slc35c1 in mouse or human does not cause marked defects in Notch signaling during development, and shows milder fucosylation defects than those observed in mice unable to synthesize GDP-fucose, indicating the existence of another mechanism for GDP-fucose transport into the secretory pathway. We show here that fibroblasts from mice or humans lacking Slc35c1 exhibit robust Notch signaling in co-culture signaling assays. A potential candidate for a second GDP-fucose transporter is the related gene Slc35c2. Overexpression of Slc35c2 reduces expression of the fucosylated epitopes Lewis X and sialylated Lewis X in CHO cells, indicating competition with Slc35c1. The fucosylation of a Notch1 EGF repeat fragment that occurs in the endoplasmic reticulum was increased in CHO transfectants overexpressing Slc35c2. In CHO cells with low levels of Slc35c2, both Delta1- and Jagged1-induced Notch signaling were reduced, and the fucosylation of a Notch1 fragment was also decreased. Immunofluorescence microscopy of rat intestinal epithelial cells and HeLa cells, and analysis of rat liver membrane fractions showed that Slc35c2 is primarily colocalized with markers of the cis-Golgi network and endoplasmic reticulum-Golgi intermediate compartment (ERGIC). The combined results suggest that Slc35c2 is either a GDP-fucose transporter that competes with Slc35c1 for GDP-fucose, or a factor that otherwise enhances the fucosylation of Notch and is required for optimal Notch signaling in mammalian cells.


Asunto(s)
Proteínas de Neoplasias/metabolismo , Proteínas de Transporte de Nucleótidos/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Animales , Western Blotting , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Embrión de Mamíferos/citología , Retículo Endoplásmico/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Fucosa/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Ligandos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Fluorescente , Proteínas de Transporte de Monosacáridos , Mutación , Proteínas de Neoplasias/genética , Proteínas de Transporte de Nucleótidos/genética , Unión Proteica , Interferencia de ARN , Ratas , Receptor Notch1/genética
11.
BMC Dev Biol ; 8: 48, 2008 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-18445292

RESUMEN

BACKGROUND: Notch signaling is highly conserved in the metazoa and is critical for many cell fate decisions. Notch activation occurs following ligand binding to Notch extracellular domain. In vitro binding assays have identified epidermal growth factor (EGF) repeats 11 and 12 as the ligand binding domain of Drosophila Notch. Here we show that an internal deletion in mouse Notch1 of EGF repeats 8-12, including the putative ligand binding domain (lbd), is an inactivating mutation in vivo. We also show that maternal and zygotic Notch1(lbd/lbd) mutant embryos develop through gastrulation to mid-gestation. RESULTS: Notch1(lbd/lbd) embryos died at mid-gestation with a phenotype indistinguishable from Notch1 null mutants. In embryonic stem (ES) cells, Notch1(lbd) was expressed on the cell surface at levels equivalent to wild type Notch1, but Delta1 binding was reduced to the same level as in Notch1 null cells. In an ES cell co-culture assay, Notch signaling induced by Jagged1 or Delta1 was reduced to a similar level in Notch1(lbd) and Notch1 null cells. However, the Notch1(lbd/lbd) allele was expressed similarly to wild type Notch1 in Notch1(lbd/lbd) ES cells and embryos at E8.75, indicating that Notch1 signaling is not essential for the Notch1 gene to be expressed. In addition, maternal and zygotic Notch1 mutant blastocysts developed through gastrulation. CONCLUSION: Mouse Notch1 lacking the ligand binding domain is expressed at the cell surface but does not signal in response to the canonical Notch ligands Delta1 and Jagged1. Homozygous Notch1(lbd/lbd) mutant embryos die at approximately E10 similar to Notch1 null embryos. While Notch1 is expressed in oocytes and blastocysts, Notch1 signaling via canonical ligands is dispensable during oogenesis, blastogenesis, implantation and gastrulation.


Asunto(s)
Células Madre Embrionarias/metabolismo , Factor de Crecimiento Epidérmico/genética , Regulación del Desarrollo de la Expresión Génica , Receptor Notch1/genética , Eliminación de Secuencia , Animales , Sitios de Unión , Proteínas de Unión al ADN/genética , Ligandos , Ratones , Mutación Puntual , Unión Proteica , Secuencias Repetitivas de Aminoácido , Secuencias Repetitivas de Ácidos Nucleicos , Transducción de Señal
12.
J Biol Chem ; 282(28): 20133-41, 2007 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-17504756

RESUMEN

Cripto is a membrane-bound co-receptor for Nodal, a member of the transforming growth factor-beta superfamily. Mouse embryos lacking either Cripto or Nodal have the same lethal phenotype at embryonic day 7.5. Previous studies suggest that O-fucosylation of the epidermal growth factor-like (EGF) repeat in Cripto is essential for the facilitation of Nodal signaling. Substitution of Ala for the Thr to which O-fucose is attached led to functional inactivation of both human and mouse Cripto. However, embryos null for protein O-fucosyltransferase 1, the enzyme that adds O-fucose to EGF repeats, do not exhibit a Cripto null phenotype and die at about embryonic day 9.5. This suggested that the loss of O-fucose from the EGF repeat may not have led to the inactivation of Cripto in previous studies. Here we investigate this hypothesis and show the following: 1) protein O-fucosyltransferase 1 is indeed the enzyme that adds O-fucose to Cripto; 2) Pofut1(-/-) embryonic stem cells behave the same as Pofut1(+/+) embryonic stem cells in a Nodal signaling assay; 3) Pofut1(-/-) and Pofut1(+/+) embryoid bodies are indistinguishable in their ability to differentiate into cardiomyocytes; and 4) none of 10 amino acid substitutions at Thr(72), including Ser which acquires O-fucose, rescues the activity of mouse Cripto in Nodal signaling assays. Therefore, the Thr to which O-fucose is linked in Cripto plays a key functional role, but O-fucose at Thr(72) is not required for Cripto to function in cell-based signaling assays or in vivo. By contrast, we show that O-fucose, and not the Thr to which it is attached, is required in the ligand-binding domain of Notch1 for Notch1 signaling.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Modificación Traduccional de las Proteínas/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Sustitución de Aminoácidos , Animales , Diferenciación Celular/fisiología , Línea Celular , Células Madre Embrionarias/metabolismo , Factor de Crecimiento Epidérmico/genética , Fucosa/genética , Fucosa/metabolismo , Fucosiltransferasas/deficiencia , Fucosiltransferasas/metabolismo , Proteínas Ligadas a GPI , Humanos , Péptidos y Proteínas de Señalización Intercelular , Glicoproteínas de Membrana/genética , Ratones , Mutación Missense , Miocitos Cardíacos/metabolismo , Proteínas de Neoplasias/genética , Proteína Nodal , Receptor Notch1/genética , Receptor Notch1/metabolismo , Treonina/genética , Treonina/metabolismo , Factor de Crecimiento Transformador beta/genética
13.
Lung Cancer ; 54(2): 143-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16979789

RESUMEN

Bombesin receptor subtype-3 (BRS-3) is an orphan G protein-coupled receptor having sequence homologies to gastrin-releasing peptide and neuromedin B receptors. [d-Phe6, beta-Ala11, Phe13, Nle14]bombesin(6-14) is known to act as a synthetic receptor agonist for BRS-3. To characterize BRS-3-mediated biological responses, we examined the effect of BRS-3 activation by [d-Phe6, beta-Ala11, Phe13, Nle14]Bn(6-14) on the adhesion of the small cell lung cancer NCI-N417 cells that express native BRS-3. We found that the BRS-3 agonist stimulated adhesion of NCI-N417 cells in laminin-coated culture wells. The adhesion of the cells to laminin induced by BRS-3 activation was accompanied by an increase in vinculin-like immunoreactivity and diminished in the presence of an anti-beta1 integrin antibody, suggesting that the receptor activation stimulates focal adhesion formation. We suggest that BRS-3 may be involved in invasion and metastasis of certain cancer cells, like small cell lung cancer cells, upon attachment to laminin.


Asunto(s)
Carcinoma de Células Pequeñas/metabolismo , Adhesión Celular , Adhesiones Focales/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores de Bombesina/metabolismo , Bombesina/análogos & derivados , Bombesina/farmacología , Carcinoma de Células Pequeñas/química , Línea Celular Tumoral , Adhesiones Focales/efectos de los fármacos , Humanos , Integrina beta1/análisis , Laminina/metabolismo , Neoplasias Pulmonares/química , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Péptidos Cíclicos , Somatostatina/análogos & derivados , Somatostatina/farmacología , Vinculina/análisis
14.
Nat Cell Biol ; 8(9): 1025-31, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16892051

RESUMEN

The nicotinamide adenine dinucleotide (NAD)-dependent deacetylase Sir2 (silent information regulator 2) regulates gene silencing in yeast and promotes lifespan extension during caloric restriction. The mammalian homologue of Sir2 (SirT1) regulates p53, NF-kappaB and Forkhead transcription factors, and is implicated in stress response. This report shows that the cell-cycle and apoptosis regulator E2F1 induces SirT1 expression at the transcriptional level. Furthermore, SirT1 binds to E2F1 and inhibits E2F1 activities, forming a negative feedback loop. Knockdown of SirT1 by small interference RNA (siRNA) increases E2F1 transcriptional and apoptotic functions. DNA damage by etoposide causes E2F1-dependent induction of SirT1 expression and knockdown of SirT1 increases sensitivity to etoposide. These results reveal a mutual regulation between E2F1 and SirT1 that affects cellular sensitivity to DNA damage.


Asunto(s)
Apoptosis , Daño del ADN , Factor de Transcripción E2F1/metabolismo , Sirtuinas/metabolismo , Línea Celular , Línea Celular Tumoral , Etopósido/toxicidad , Retroalimentación Fisiológica , Humanos , Mutación , Unión Proteica , ARN Interferente Pequeño/genética , Sirtuina 1 , Sirtuinas/genética
15.
Regul Pept ; 132(1-3): 27-32, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16137778

RESUMEN

The novel 36-amino acid peptide, apelin, is the endogenous ligand for the orphan receptor APJ. Apelin may play important roles in the regulation of the cardiovascular system and the hypothalamic-pituitary axis. It is a potent hypotensive agent and one of the most potent stimulators of cardiac contractility. In this study, we investigated the roles of apelin derived from adipocytes in the regulation of cardiovascular homeostasis. We found that both apelin and APJ mRNAs were expressed in isolated mouse adipocytes and that apelin mRNA levels increased during the differentiation of 3T3-L1 cells to adipocytes. We also found that the administration of insulin (1 nM-100 nM) increased, while that of dexamethasone (0.1 nM-100 nM) decreased the apelin mRNA levels in 3T3-L1 adipocytes in a dose-dependent manner, suggesting that insulin and glucocorticoids regulate apelin gene expression in adipocytes. We speculate that high glucocorticoid levels suppress apelin production and stimulate angiotensin II production in adipocyte, decreasing the counter-regulatory activity of apelin against the pressor action of angiotensin II, which might partly be involved in the mechanism underlying the development of obesity-related hypertension.


Asunto(s)
Adipocitos/metabolismo , Obesidad/metabolismo , ARN/metabolismo , Células 3T3-L1/metabolismo , Adipoquinas , Animales , Apelina , Northern Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Dexametasona/farmacología , Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Glucocorticoides/fisiología , Homeostasis , Insulina/farmacología , Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Endogámicos C57BL , ARN/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
16.
J Biol Chem ; 280(13): 12339-43, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15713665

RESUMEN

The E2F1 transcription factor is a critical regulator of cell cycle due to its ability to promote S phase entry. However, E2F1 overexpression also sensitizes cells to apoptosis and E2F1-null mice are predisposed to tumor development, suggesting that it also has properties of a growth suppressor. E2F1 transcription function is regulated by interaction with hypophosphorylated pRb. Cdk inhibitors such as p16INK4a and p27Kip1 inhibit pRb phosphorylation by the cyclin D/Cdk4 and cyclin E/Cdk2 complexes, thus keeping E2F1 in an inactive state. We found that E2F1 binds to the p27 promoter in vivo and activates p27 mRNA and protein expression. Depletion of endogenous E2F1 by siRNA causes a reduction in basal p27 expression level. Inhibition of endogenous p27 expression by siRNA increases E2F1 transcriptional activity and permits accelerated cell cycle progression by exogenous E2F1. These observations suggest that induction of p27 acts as a negative feedback mechanism for E2F1 and may also contribute to other functions of E2F1.


Asunto(s)
Bioquímica/métodos , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Apoptosis , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Cromatina/metabolismo , Ciclina D , Ciclina E/metabolismo , Quinasa 4 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Genes Reporteros , Humanos , Inmunoprecipitación , Luciferasas/metabolismo , Ratones , Microscopía Fluorescente , Modelos Biológicos , Células 3T3 NIH , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fase S , Transcripción Genética , Transfección , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA