Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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.

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.
Front Cell Dev Biol ; 9: 764727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712672

RESUMEN

Transforming Growth Factor-ß (TGF-ß) is a key regulator of embryonic development, adult tissue homeostasis, and lesion repair. In tumors, TGF-ß is a potent inhibitor of early stage tumorigenesis and promotes late stage tumor progression and metastasis. Here, we review the roles of TGF-ß as well as components of its signaling pathways in tumorigenesis. We will discuss how a core property of TGF-ß, namely its ability to change cell differentiation, leads to the transition of epithelial cells, endothelial cells and fibroblasts to a myofibroblastoid phenotype, changes differentiation and polarization of immune cells, and induces metabolic reprogramming of cells, all of which contribute to the progression of epithelial tumors.

7.
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
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
9.
Cell Death Dis ; 11(3): 184, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32170115

RESUMEN

Prevailing insulin resistance and the resultant hyperglycemia elicits a compensatory response from pancreatic islet beta cells (ß-cells) that involves increases in ß-cell function and ß-cell mass. However, the sustained metabolic stress eventually leads to ß-cell failure characterized by severe ß-cell dysfunction and progressive loss of ß-cell mass. Whereas, ß-cell dysfunction is relatively well understood at the mechanistic level, the avenues leading to loss of ß-cell mass are less clear with reduced proliferation, dedifferentiation, and apoptosis all potential mechanisms. Butler and colleagues documented increased ß-cell apoptosis in pancreas from lean and obese human Type 2 diabetes (T2D) subjects, with no changes in rates of ß-cell replication or neogenesis, strongly suggesting a role for apoptosis in ß-cell failure. Here, we describe a permissive role for TGF-ß/Smad3 in ß-cell apoptosis. Human islets undergoing ß-cell apoptosis release increased levels of TGF-ß1 ligand and phosphorylation levels of TGF-ß's chief transcription factor, Smad3, are increased in human T2D islets suggestive of an autocrine role for TGF-ß/Smad3 signaling in ß-cell apoptosis. Smad3 phosphorylation is similarly increased in diabetic mouse islets undergoing ß-cell apoptosis. In mice, ß-cell-specific activation of Smad3 promotes apoptosis and loss of ß-cell mass in association with ß-cell dysfunction, glucose intolerance, and diabetes. In contrast, inactive Smad3 protects from apoptosis and preserves ß-cell mass while improving ß-cell function and glucose tolerance. At the molecular level, Smad3 associates with Foxo1 to propagate TGF-ß-dependent ß-cell apoptosis. Indeed, genetic or pharmacologic inhibition of TGF-ß/Smad3 signals or knocking down Foxo1 protects from ß-cell apoptosis. These findings reveal the importance of TGF-ß/Smad3 in promoting ß-cell apoptosis and demonstrate the therapeutic potential of TGF-ß/Smad3 antagonism to restore ß-cell mass lost in diabetes.


Asunto(s)
Apoptosis/genética , Linfocitos B/metabolismo , Proteína smad3/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Transducción de Señal , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/genética
10.
Sci Rep ; 9(1): 16992, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31740700

RESUMEN

Insulin signaling governs many processes including glucose homeostasis and metabolism, and is therapeutically used to treat hyperglycemia in diabetes. We demonstrated that insulin-induced Akt activation enhances the sensitivity to TGF-ß by directing an increase in cell surface TGF-ß receptors from a pool of intracellular TGF-ß receptors. Consequently, increased autocrine TGF-ß signaling in response to insulin participates in insulin-induced angiogenic responses of endothelial cells. With TGF-ß signaling controlling many cell responses, including differentiation and extracellular matrix deposition, and pathologically promoting fibrosis and cancer cell dissemination, we addressed to which extent autocrine TGF-ß signaling participates in insulin-induced gene responses of human endothelial cells. Transcriptome analyses of the insulin response, in the absence or presence of a TGF-ß receptor kinase inhibitor, revealed substantial positive and negative contributions of autocrine TGF-ß signaling in insulin-responsive gene responses. Furthermore, insulin-induced responses of many genes depended on or resulted from autocrine TGF-ß signaling. Our analyses also highlight extensive contributions of autocrine TGF-ß signaling to basal gene expression in the absence of insulin, and identified many novel TGF-ß-responsive genes. This data resource may aid in the appreciation of the roles of autocrine TGF-ß signaling in normal physiological responses to insulin, and implications of therapeutic insulin usage.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Insulina/farmacología , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteínas Smad/genética , Factor de Crecimiento Transformador beta/farmacología , Benzamidas/farmacología , Células Cultivadas , Dioxoles/farmacología , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hipoglucemiantes/farmacología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas Smad/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo
11.
Cells ; 8(10)2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31652618

RESUMEN

Non-Small Cell Lung Cancer (NSCLC) is a common malignancy and leading cause of death by cancer. Metastasis and drug resistance are serious clinical problems encountered in NSCLC therapy. Aberrant activation of the Transforming Growth Factor beta (TGFß) and Hedgehog (Hh) signal transduction cascades often associate with poor prognosis and aggressive disease progression in NSCLC, as these signals can drive cell proliferation, angiogenesis, metastasis, immune evasion and emergence of drug resistance. Therefore, simultaneous inhibition of TGFß and Hh signaling, by a single agent, or in combination with other drugs, could yield therapeutic benefits in NSCLC and other cancers. In the current study, we report on the biological and pharmacological evaluation of Oxy210, an oxysterol-based dual inhibitor of TGFß and Hh signaling. In NSCLC cells, Oxy210 inhibits proliferation, epithelial-mesenchymal transition (EMT) and invasive activity. Combining Oxy210 with Carboplatin (CP) increases the anti-proliferative response to CP and inhibits TGFß-induced resistance to CP in A549 NSCLC cells. In addition, Oxy210 displays encouraging drug-like properties, including chemical scalability, metabolic stability and oral bioavailability in mice. Unlike other known inhibitors, Oxy210 antagonizes TGFß and Hh signaling independently of TGFß receptor kinase inhibition and downstream of Smoothened, respectively.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas Hedgehog/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Oxiesteroles/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Células A549 , Animales , Carboplatino/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Células 3T3 NIH
12.
Oncogene ; 38(17): 3185-3200, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30626936

RESUMEN

Transforming growth factor-ß (TGF-ß) is major inducer of epithelial-to-mesenchymal transition (EMT), which associates with cancer cell metastasis and resistance to chemotherapy and targeted drugs, through both transcriptional and non-transcriptional mechanisms. We previously reported that, in cancer cells, heightened mitogenic signaling allows TGF-ß-activated Smad3 to interact with poly(RC) binding protein 1 (PCBP1) and together they regulate many alternative splicing events that favors expression of protein isoforms essential for EMT, cytoskeletal rearrangement, and adherens junction signaling. Here we show that the exclusion of TGF-ß-activated kinase 1 (TAK1) variable exon 12 requires another RNA-binding protein, Fox-1 homolog 2 (Rbfox2), which binds intronic sequences in front of exon 12 independently of the Smad3-PCBP1 complex. Functionally, exon 12-excluded TAK1∆E12 and full-length TAK1FL are distinct. The short isoform TAK1∆E12 is constitutively active and supports TGF-ß-induced EMT and nuclear factor kappa B (NF-κB) signaling, whereas the full-length isoform TAK1FL promotes TGF-ß-induced apoptosis. These observations offer a harmonious explanation for how a single TAK1 kinase can mediate the opposing responses of cell survival and apoptosis in response to TGF-ß. They also reveal a propensity of the alternatively spliced TAK1 isoform TAK1∆E12 to cause drug resistance due to its activity in supporting EMT and NF-κB survival signaling.


Asunto(s)
Empalme Alternativo/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Quinasas Quinasa Quinasa PAM/genética , Factor de Crecimiento Transformador beta/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Humanos , Ratones , Fosforilación/genética , Isoformas de Proteínas/genética , Proteínas Represoras/genética , Transducción de Señal/genética , Proteína smad3/genética , Transcripción Genética/genética
13.
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
14.
Int J Biol Sci ; 14(5): 542-548, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29805305

RESUMEN

Smad ubiquitin regulatory factor 2 (Smurf2) is a HECT domain-containing E3 ubiquitin ligase. Together with its closely related homolog Smurf1, Smurf2 was initially recognized as a negative regulator of transforming growth factor-ß (TGF-ß) and bone morphogenetic protein (BMP) signaling, but subsequent studies have expanded its function to regulate many different signaling pathways and play important roles in genomic stability, cell polarity, tissue homeostasis and carcinogenesis. Mice with conventional knockout of Smurf1 or Smurf2 alleles are viable, but conventional Smurf1 and Smurf2 double knockout mice were early embryonic lethal. In order to study the physiological function of Smurfs during late stage of embryonic development or in adult animals, we generated Smurf2flox/flox mice carrying a targeted mutation for conditional Smurf2 gene inactivation. We demonstrated that Cre-mediated recombination using Alb-Cre, a Cre line expressed in hepatocyte, results in specific deletion of the gene in liver tissue. We also showed that Cre-mediated recombination in mouse embryonic fibroblasts (MEFs) with Smurf2flox/flox genotype resulted in generation of Smurf2 knockout MEFs, and Smurf2 deficiency affects multiple signaling pathways. Therefore, this animal model will be useful to study the distinct roles of Smurf2 in different tissues at different ages.


Asunto(s)
Recombinación Genética , Ubiquitina-Proteína Ligasas/genética , Alelos , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Femenino , Fibroblastos/metabolismo , Genotipo , Heterocigoto , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Transducción de Señal , Distribución Tisular , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas/fisiología
15.
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
16.
Curr Opin Cell Biol ; 51: 1-7, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29149681

RESUMEN

Transforming growth factor-ß (TGF-ß) controls a wide range of cellular functions by activating both SMADs and non-SMAD pathways. In different tissue or physiological environment, cellular responses to TGF-ß can be diverse, even opposite. Complex regulations at the level of ligand mobilization, receptor presentation, and the network of intracellular signal transducers afford the TGF-ß pathway with versatile means to induce precise cellular responses in accordance to specific contextual demands. This article summarizes recent development in how cells manage their responses to TGF-ß through ligand activation, receptor abundance, as well as SMAD-dependent and SMAD-independent mechanisms.


Asunto(s)
Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Humanos , Transducción de Señal
17.
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
18.
Mol Cell Oncol ; 4(1): e1265699, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28197539

RESUMEN

Transforming growth factor ß (TGF-ß) is a well-known growth inhibitor of normal epithelial cells, but it is also secreted by solid tumors to promote cancer progression. Our recent discovery of SMAD3-PCBP1 complex with direct RNA-binding properties has shed light on how this conversion is implemented by controlling pre-mRNA splicing patterns.

19.
Artículo en Inglés | MEDLINE | ID: mdl-27864313

RESUMEN

Transforming growth factor ß (TGF-ß) and structurally related factors use several intracellular signaling pathways in addition to Smad signaling to regulate a wide array of cellular functions. These non-Smad signaling pathways are activated directly by ligand-occupied receptors to reinforce, attenuate, or otherwise modulate downstream cellular responses. This review summarizes the current knowledge of the mechanisms by which non-Smad signaling pathways are directly activated in response to ligand binding, how activation of these pathways impinges on Smads and non-Smad targets, and how final cellular responses are affected in response to these noncanonical signaling modes.


Asunto(s)
Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , GTP Fosfohidrolasas/metabolismo , Humanos , Fosforilación , Proteínas Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...