Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 48(4): 745-759.e6, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669252

RESUMEN

It is unclear how quiescence is enforced in naive T cells, but activation by foreign antigens and self-antigens is allowed, despite the presence of inhibitory signals. We showed that active transforming growth factor ß (TGF-ß) signaling was present in naive T cells, and T cell receptor (TCR) engagement reduced TGF-ß signaling during T cell activation by downregulating TGF-ß type 1 receptor (TßRI) through activation of caspase recruitment domain-containing protein 11 (CARD11) and nuclear factor κB (NF-κB). TGF-ß prevented TCR-mediated TßRI downregulation, but this was abrogated by interleukin-6 (IL-6). Mitigation of TCR-mediated TßRI downregulation through overexpression of TßRI in naive and activated T cells rendered T cells less responsive and suppressed autoimmunity. Naive T cells in autoimmune patients exhibited reduced TßRI expression and increased TCR-driven proliferation compared to healthy subjects. Thus, TCR-mediated regulation of TßRI-TGF-ß signaling acts as a crucial criterion to determine T cell quiescence and activation.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Guanilato Ciclasa/metabolismo , Activación de Linfocitos/inmunología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Autoinmunidad/inmunología , Proteínas Adaptadoras de Señalización CARD/genética , Línea Celular , Proliferación Celular , Colitis/inmunología , Colitis/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Guanilato Ciclasa/genética , Células HEK293 , Humanos , Interleucina-6/inmunología , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/biosíntesis , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/biosíntesis
2.
Semin Cancer Biol ; 101: 1-11, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38614376

RESUMEN

Epithelial to mesenchymal transition (EMT) is a physiological process during development where epithelial cells transform to acquire mesenchymal characteristics, which allows them to migrate and colonize secondary tissues. Many cellular signaling pathways and master transcriptional factors exert a myriad of controls to fine tune this vital process to meet various developmental and physiological needs. Adding to the complexity of this network are post-transcriptional and post-translational regulations. Among them, alternative splicing has been shown to play important roles to drive EMT-associated phenotypic changes, including actin cytoskeleton remodeling, cell-cell junction changes, cell motility and invasiveness. In advanced cancers, transforming growth factor-ß (TGF-ß) is a major inducer of EMT and is associated with tumor cell metastasis, cancer stem cell self-renewal, and drug resistance. This review aims to provide an overview of recent discoveries regarding alternative splicing events and the involvement of splicing factors in the EMT and TGF-ß signaling. It will emphasize the importance of various splicing factors involved in EMT and explore their regulatory mechanisms.


Asunto(s)
Empalme Alternativo , Transición Epitelial-Mesenquimal , Neoplasias , Transducción de Señal , Factor de Crecimiento Transformador beta , Humanos , Transición Epitelial-Mesenquimal/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo , Animales , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica
3.
Mol Cell ; 64(3): 549-564, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27746021

RESUMEN

In advanced stages of cancers, TGF-ß promotes tumor progression in conjunction with inputs from receptor tyrosine kinase pathways. However, mechanisms that underpin the signaling cooperation and convert TGF-ß from a potent growth inhibitor to a tumor promoter are not fully understood. We report here that TGF-ß directly regulates alternative splicing of cancer stem cell marker CD44 through a phosphorylated T179 of SMAD3-mediated interaction with RNA-binding protein PCBP1. We show that TGF-ß and EGF respectively induce SMAD3 and PCBP1 to colocalize in SC35-positive nuclear speckles, and the two proteins interact in the variable exon region of CD44 pre-mRNA to inhibit spliceosome assembly in favor of expressing the mesenchymal isoform CD44s over the epithelial isoform CD44E. We further show that the SMAD3-mediated alternative splicing is essential to the tumor-promoting role of TGF-ß and has a global influence on protein products of genes instrumental to epithelial-to-mesenchymal transition and metastasis.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/genética , Receptores de Hialuranos/genética , Neoplasias Pulmonares/genética , Proteína smad3/genética , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Exones , Femenino , Perfilación de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Proteínas de Unión al ARN , Transducción de Señal , Proteína smad3/metabolismo , Treonina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
4.
Am J Pathol ; 192(12): 1699-1711, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36063900

RESUMEN

Wound healing is a highly conserved process that restores the integrity and functionality of injured tissues. Transforming growth factor (TGF)-ß is a master regulator of wound healing, whose signaling is attenuated by the E3 ubiquitin ligase Smurf2. Herein, the roles of Smurf2 in cutaneous wound healing were examined using a murine incisional cutaneous model. Loss of Smurf2 increased early inflammation in the wounds and led to narrower wounds with greater breaking strength. Loss of Smurf2 also led to more linearized collagen bundles in normal and wounded skin. Gene expression analyses by real-time quantitative PCR indicated that Smurf2-deficient fibroblasts had increased levels of TGF-ß/Smad3 signaling and changes in expression profile of genes related to matrix turnover. The effect of Smurf2 loss on wound healing and collagen bundling was attenuated by the heterozygous loss of Smad3. Together, these results show that Smurf2 affects inflammation and collagen processing in cutaneous wounds by down-regulating TGF-ß/Smad3 signaling.


Asunto(s)
Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta , Ratones , Animales , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Colágeno , Cicatrización de Heridas , Inflamación , Factores de Crecimiento Transformadores
5.
Int J Mol Sci ; 23(10)2022 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-35628290

RESUMEN

Inflammatory responses by the innate and adaptive immune systems protect against infections and are essential to health and survival. Many diseases including atherosclerosis, osteoarthritis, rheumatoid arthritis, psoriasis, and obesity involve persistent chronic inflammation. Currently available anti-inflammatory agents, including non-steroidal anti-inflammatory drugs, steroids, and biologics, are often unsafe for chronic use due to adverse effects. The development of effective non-toxic anti-inflammatory agents for chronic use remains an important research arena. We previously reported that oral administration of Oxy210, a semi-synthetic oxysterol, ameliorates non-alcoholic steatohepatitis (NASH) induced by a high-fat diet in APOE*3-Leiden.CETP humanized mouse model of NASH and inhibits expression of hepatic and circulating levels of inflammatory cytokines. Here, we show that Oxy210 also inhibits diet-induced white adipose tissue inflammation in APOE*3-Leiden.CETP mice, evidenced by the inhibition of adipose tissue expression of IL-6, MCP-1, and CD68 macrophage marker. Oxy210 and related analogs exhibit anti-inflammatory effects in macrophages treated with lipopolysaccharide in vitro, mediated through inhibition of toll-like receptor 4 (TLR4), TLR2, and AP-1 signaling, independent of cyclooxygenase enzymes or steroid receptors. The anti-inflammatory effects of Oxy210 are correlated with the inhibition of macrophage polarization. We propose that Oxy210 and its structural analogs may be attractive candidates for future therapeutic development for targeting inflammatory diseases.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Oxiesteroles , Animales , Apolipoproteínas E/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxiesteroles/metabolismo , Oxiesteroles/farmacología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
6.
J Biol Chem ; 295(52): 18485-18493, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33097595

RESUMEN

Timely repair of DNA double-strand breaks (DSBs) is essential to maintaining genomic integrity and preventing illnesses induced by genetic abnormalities. We previously demonstrated that the E3 ubiquitin ligase SMURF2 plays a critical tumor suppressing role via its interaction with RNF20 (ring finger protein 20) in shaping chromatin landscape and preserving genomic stability. However, the mechanism that mobilizes SMURF2 in response to DNA damage remains unclear. Using biochemical approaches and MS analysis, we show that upon the onset of the DNA-damage response, SMURF2 becomes phosphorylated at Ser384 by ataxia telangiectasia mutated (ATM) serine/threonine kinase, and this phosphorylation is required for its interaction with RNF20. We demonstrate that a SMURF2 mutant with an S384A substitution has reduced capacity to ubiquitinate RNF20 while promoting Smad3 ubiquitination unabatedly. More importantly, mouse embryonic fibroblasts expressing the SMURF2 S384A mutant show a weakened ability to sustain the DSB response compared with those expressing WT SMURF2 following etoposide treatment. These data indicate that SMURF2-mediated RNF20 ubiquitination and degradation controlled by ataxia telangiectasia mutated-induced phosphorylation at Ser384 constitutes a negative feedback loop that regulates DSB repair.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cromatina/metabolismo , Daño del ADN , Reparación del ADN , Retroalimentación Fisiológica , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Cromatina/genética , Inestabilidad Genómica , Humanos , Ratones , Fosforilación , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
7.
PLoS Biol ; 16(12): e3000091, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30566427

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is characterized by abnormal accumulation of triglycerides (TG) in the liver and other metabolic syndrome symptoms, but its molecular genetic causes are not completely understood. Here, we show that mice deficient for ubiquitin ligase (E3) Smad ubiquitin regulatory factor 1 (Smurf1) spontaneously develop hepatic steatosis as they age and exhibit the exacerbated phenotype under a high-fat diet (HFD). Our data indicate that loss of Smurf1 up-regulates the expression of peroxisome proliferator-activated receptor γ (PPARγ) and its target genes involved in lipid synthesis and fatty acid uptake. We further show that PPARγ is a direct substrate of Smurf1-mediated non-proteolytic lysine 63 (K63)-linked ubiquitin modification that suppresses its transcriptional activity, and treatment of Smurf1-deficient mice with a PPARγ antagonist, GW9662, completely reversed the lipid accumulation in the liver. Finally, we demonstrate an inverse correlation of low SMURF1 expression to high body mass index (BMI) values in human patients, thus revealing a new role of SMURF1 in NAFLD pathogenesis.


Asunto(s)
Hígado Graso/prevención & control , PPAR gamma/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Línea Celular , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Humanos , Hígado/metabolismo , Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Triglicéridos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
8.
Int J Mol Sci ; 21(11)2020 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-32527012

RESUMEN

Serine-threonine kinase receptor-associated protein (STRAP) functions as a regulator of both TGF-ß and p53 signaling that participates in the regulation of cell proliferation and cell death in response to various stresses. Here, we demonstrate that STRAP acetylation plays an important role in p53-mediated cell cycle arrest and apoptosis. STRAP is acetylated at lysines 147, 148, and 156 by the acetyltransferases CREB-binding protein (CBP) and that the acetylation is reversed by the deacetylase sirtuin7 (SIRT7). Hypo- or hyperacetylation mutations of STRAP at lysines 147, 148, and 156 (3KR or 3KQ) influence its activation and stabilization of p53. Moreover, following 5-fluorouracil (5-FU) treatment, STRAP is mobilized from the cytoplasm to the nucleus and promotes STRAP acetylation. Our finding on the regulation of STRAP links p53 with SIRT7 influencing p53 activity and stability.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Sirtuinas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Fluorouracilo/farmacología , Células HCT116 , Humanos , Lisina/metabolismo , Estabilidad Proteica , Proteínas de Unión al ARN/genética , Sirtuinas/genética , Proteína p53 Supresora de Tumor/genética
10.
J Biol Chem ; 292(10): 4302-4312, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28154170

RESUMEN

Transforming growth factor-ß (TGF-ß) signals through both SMAD and non-SMAD pathways to elicit a wide array of biological effects. Existing data have shown the association and coordination between STATs and SMADs in mediating TGF-ß functions in hepatic cells, but it is not clear how STATs are activated under these circumstances. Here, we report that JAK1 is a constitutive TGFßRI binding protein and is absolutely required for phosphorylation of STATs in a SMAD-independent manner within minutes of TGF-ß stimulation. Following the activation of SMADs, TGF-ß also induces a second phase of STAT phosphorylation that requires SMADs, de novo protein synthesis, and contribution from JAK1. Our global gene expression profiling indicates that the non-SMAD JAK1/STAT pathway is essential for the expression of a subset of TGF-ß target genes in hepatic stellate cells, and the cooperation between the JAK1-STAT3 and SMAD pathways is critical to the roles of TGF-ß in liver fibrosis.


Asunto(s)
Embrión de Mamíferos/patología , Células Estrelladas Hepáticas/patología , Janus Quinasa 1/metabolismo , Cirrosis Hepática/patología , Factor de Transcripción STAT3/metabolismo , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Células Cultivadas , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Janus Quinasa 1/genética , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos
11.
Nature ; 480(7375): 113-7, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22020285

RESUMEN

Selective autophagy involves the recognition and targeting of specific cargo, such as damaged organelles, misfolded proteins, or invading pathogens for lysosomal destruction. Yeast genetic screens have identified proteins required for different forms of selective autophagy, including cytoplasm-to-vacuole targeting, pexophagy and mitophagy, and mammalian genetic screens have identified proteins required for autophagy regulation. However, there have been no systematic approaches to identify molecular determinants of selective autophagy in mammalian cells. Here, to identify mammalian genes required for selective autophagy, we performed a high-content, image-based, genome-wide small interfering RNA screen to detect genes required for the colocalization of Sindbis virus capsid protein with autophagolysosomes. We identified 141 candidate genes required for viral autophagy, which were enriched for cellular pathways related to messenger RNA processing, interferon signalling, vesicle trafficking, cytoskeletal motor function and metabolism. Ninety-six of these genes were also required for Parkin-mediated mitophagy, indicating that common molecular determinants may be involved in autophagic targeting of viral nucleocapsids and autophagic targeting of damaged mitochondria. Murine embryonic fibroblasts lacking one of these gene products, the C2-domain containing protein, SMURF1, are deficient in the autophagosomal targeting of Sindbis and herpes simplex viruses and in the clearance of damaged mitochondria. Moreover, SMURF1-deficient mice accumulate damaged mitochondria in the heart, brain and liver. Thus, our study identifies candidate determinants of selective autophagy, and defines SMURF1 as a newly recognized mediator of both viral autophagy and mitophagy.


Asunto(s)
Autofagia/genética , Estudio de Asociación del Genoma Completo , ARN Interferente Pequeño/genética , Animales , Proteínas de la Cápside/metabolismo , Células HeLa , Humanos , Lisosomas/metabolismo , Ratones , Mitocondrias/metabolismo , Transporte de Proteínas/genética , Virus Sindbis/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
12.
Mol Cell ; 31(6): 918-24, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18922473

RESUMEN

In many physiological and disease processes, TGF-beta usurps branches of MAP kinase pathways in conjunction with Smads to induce apoptosis and epithelial-to-mesenchymal transition, but the detailed mechanism of how a MAP kinase cascade is activated by TGF-beta receptors is not clear. We report here that TRAF6 is specifically required for the Smad-independent activation of JNK and p38, and its carboxyl TRAF homology domain physically interacts with TGF-beta receptors. TGF-beta induces K63-linked ubiquitination of TRAF6 and promotes association between TRAF6 and TAK1. Our results indicate that TGF-beta activates JNK and p38 through a mechanism similar to that operating in the interleukin-1beta/Toll-like receptor pathway.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Activación Enzimática/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Humanos , Lisina/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Unión Proteica/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Ubiquitinación/efectos de los fármacos
13.
EMBO J ; 30(23): 4777-89, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045334

RESUMEN

TGF-ß signalling is regulated by post-translational modifications of Smad proteins to translate quantitative difference in ligand concentration into proportional transcriptional output. Previous studies in cell culture systems suggested that Smad ubiquitination regulatory factors (Smurfs) act in this regulation by targeting Smads for proteasomal degradation, but whether this mechanism operates under physiological conditions is not clear. Here, we generated mice harbouring a target-disrupted Smurf2 allele. Using primary mouse embryonic fibroblasts and dermal fibroblasts, we show that TGF-ß-mediated, Smad-dependent transcriptional responses are elevated in the absence of Smurf2. Instead of promoting poly-ubiquitination and degradation, we show that Smurf2 actually induces multiple mono-ubiquitination of Smad3 in vivo. Phosphorylation of T179, immediately upstream of the Smad3 PY motif, enhances Smurf2 and Smad3 interaction and Smad3 ubiquitination. We have mapped Smurf2-induced Smad3 ubiquitination sites to lysine residues at the MH2 domain, and demonstrate that Smad3 ubiquitination inhibits the formation of Smad3 complexes. Thus, our data support a model in which Smurf2 negatively regulates TGF-ß signalling by attenuating the activity of Smad3 rather than promoting its degradation.


Asunto(s)
Transducción de Señal/fisiología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinación/fisiología , Animales , Western Blotting , Fibroblastos/metabolismo , Ratones , Modelos Biológicos , Unión Proteica , Transcripción Genética/fisiología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
14.
J Cell Sci ; 126(Pt 21): 4809-13, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24172535

RESUMEN

The latest advances on the transforming growth factor ß (TGF-ß) and bone morphogenetic protein (BMP) signaling pathways were reported at the July 2013 FASEB Summer Research Conference 'The TGF-ß Superfamily: Development and Disease'. The meeting was held in Steamboat Springs, Colorado, USA at 6700 feet above sea level in the Rocky Mountains. This was the seventh biannual meeting in the series. In attendance were investigators from a broad range of disciplines with a common interest in the mechanics of TGF-ß and BMP signaling pathways, their normal developmental and homeostatic functions, and the diseases associated with pathway misregulation.


Asunto(s)
Familia de Multigenes , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Enfermedad/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Factor de Crecimiento Transformador beta/genética
15.
J Biol Chem ; 288(30): 21784-92, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23760265

RESUMEN

Smad ubiquitin regulatory factors (Smurfs) are HECT-domain ubiquitin E3 ligases that regulate diverse cellular processes, including normal and tumor cell migration. However, the underlying mechanism of the Smurfs' role in cell migration is not fully understood. Here we show that Smurf1 induces ubiquitination of tumor necrosis factor receptor-associated factor 4 (TRAF4) at K190. Using the K190R mutant of TRAF4, we demonstrate that Smurf1-induced ubiquitination is required for proper localization of TRAF4 to tight junctions in confluent epithelial cells. We further show that TRAF4 is essential for the migration of both normal mammary epithelial and breast cancer cells. The ability of TRAF4 to promote cell migration is also dependent on Smurf1-mediated ubiquitination, which is associated with Rac1 activation by TRAF4. These results reveal a new regulatory circuit for cell migration, consisting of Smurf1-mediated ubiquitination of TRAF4 and Rac1 activation.


Asunto(s)
Movimiento Celular , Células Epiteliales/metabolismo , Factor 4 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Células Epiteliales/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Lisina/genética , Lisina/metabolismo , Células MCF-7 , Glándulas Mamarias Humanas/citología , Ratones , Microscopía Confocal , Mutación , Interferencia de ARN , Factor 4 Asociado a Receptor de TNF/genética , Uniones Estrechas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteína de Unión al GTP rac1/metabolismo
16.
Cancer Cell ; 9(6): 445-57, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16766264

RESUMEN

In the liver, derangement of TGF-beta signaling is associated with an increased incidence of hepatocellular carcinoma (HCC), but the mechanism is not clear. We report here that forced expression of a major TGF-beta signaling transducer, Smad3, reduces susceptibility to HCC in a chemically induced murine model. This protection is conferred by Smad3's ability to promote apoptosis by repressing Bcl-2 transcription in vivo through a GC-rich element in the Bcl-2 promoter. We also show that the proapoptotic activity of Smad3 requires both input from TGF-beta signaling and activation of p38 MAPK, which occurs selectively in the liver tumor cells. Thus, Smad3 enables the tumor suppression function of TGF-beta by serving as a physiological mediator of TGF-beta-induced apoptosis.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular/metabolismo , Hepatocitos/patología , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteína smad3/fisiología , Animales , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/patología , Células Cultivadas , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Hepatocitos/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal , Proteína smad3/biosíntesis , Proteína smad3/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cell Biosci ; 12(1): 119, 2022 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-35908024

RESUMEN

BACKGROUND: Developmental signaling pathways such as those of Hedgehog (HH) and WNT play critical roles in cancer stem cell self-renewal, migration, and differentiation. They are often constitutively activated in many human malignancies, including non-small cell lung cancer (NSCLC). Previously, we reported that two oxysterol derivatives, Oxy186 and Oxy210, are potent inhibitors of HH/GLI signaling and NSCLC cancer cell growth. In addition, we also showed that Oxy210 is a potent inhibitor of TGF-ß/SMAD signaling. In this follow-up study, we further explore the mechanism of action by which these oxysterols control NSCLC cell proliferation and tumor growth. RESULTS: Using a GLI-responsive luciferase reporter assay, we show here that HH ligand could not mount a signaling response in the NSCLC cell line A549, even though Oxy186 and Oxy210 still inhibited non-canonical GLI activity and suppressed the proliferation of A549 cells. Further, we uncover an unexpected activity of these two oxysterols in inhibiting the WNT/ß-catenin signaling at the level of LRP5/6 membrane receptors. We also show that in a subcutaneous xenograft tumor model generated from A549 cells, Oxy186, but not Oxy210, exhibits strong inhibition of tumor growth. Subsequent RNA-seq analysis of the xenograft tumor tissue reveal that the WNT/ß-catenin pathway is the target of Oxy186 in vivo. CONCLUSION: The oxysterols Oxy186 and Oxy210 both possess inhibitory activity towards WNT/ß-catenin signaling, and Oxy186 is also a potent inhibitor of NSCLC tumor growth.

19.
Nat Commun ; 13(1): 6069, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36241625

RESUMEN

Interleukin-9 (IL-9)-producing CD4+ T helper cells (Th9) have been implicated in allergy/asthma and anti-tumor immunity, yet molecular insights on their differentiation from activated T cells, driven by IL-4 and transforming growth factor-beta (TGF-ß), is still lacking. Here we show opposing functions of two transcription factors, D-binding protein (DBP) and E2F8, in controlling Th9 differentiation. Specifically, TGF-ß and IL-4 signaling induces phosphorylation of the serine 213 site in the linker region of the Smad3 (pSmad3L-Ser213) via phosphorylated p38, which is necessary and sufficient for Il9 gene transcription. We identify DBP and E2F8 as an activator and repressor, respectively, for Il9 transcription by pSmad3L-Ser213. Notably, Th9 cells with siRNA-mediated knockdown for Dbp or E2f8 promote and suppress tumor growth, respectively, in mouse tumor models. Importantly, DBP and E2F8 also exhibit opposing functions in regulating human TH9 differentiation in vitro. Thus, our data uncover a molecular mechanism of Smad3 linker region-mediated, opposing functions of DBP and E2F8 in Th9 differentiation.


Asunto(s)
Interleucina-4 , Interleucina-9 , Animales , Humanos , Ratones , Diferenciación Celular/genética , Interleucina-4/metabolismo , Proteínas Represoras/genética , ARN Interferente Pequeño/metabolismo , Serina/metabolismo , Linfocitos T Colaboradores-Inductores , Factor de Crecimiento Transformador beta/metabolismo , Factores de Crecimiento Transformadores/metabolismo
20.
J Biol Chem ; 285(12): 8703-10, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20097766

RESUMEN

Our previous study demonstrated that transforming growth factor (TGF)-beta activates beta-catenin signaling through Smad3 interaction with beta-catenin in chondrocytes. In the present studies, we further investigated the detailed molecular mechanism of the cross-talk between TGF-beta/Smad3 and Wnt/beta-catenin signaling pathways. We found that C-terminal Smad3 interacted with both the N-terminal region and the middle region of beta-catenin protein in a TGF-beta-dependent manner. Both Smad3 and Smad4 were required for the interaction with beta-catenin and protected beta-catenin from an ubiquitin-proteasome-dependent degradation. In addition, the formation of the Smad3-Smad4-beta-catenin protein complex also mediated beta-catenin nuclear translocation. This Smad3-mediated regulatory mechanism of beta-catenin protein stability enhanced the activity of beta-catenin to activate downstream target genes during chondrogenesis. Our findings demonstrate a novel mechanism between TGF-beta and Wnt/beta-catenin signaling pathways during chondrocyte development.


Asunto(s)
Transporte Activo de Núcleo Celular , Condrocitos/metabolismo , Proteína smad3/metabolismo , beta Catenina/metabolismo , Animales , Células COS , Chlorocebus aethiops , Condrogénesis , Humanos , Modelos Biológicos , Unión Proteica , Ratas , Transducción de Señal , Proteína Smad4/metabolismo , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA