Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 299(2): 102820, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36549646

RESUMEN

In mammalian cells, Smad2 and Smad3, two receptor-regulated Smad proteins, play crucial roles in the signal transmission of transforming growth factor-ß (TGF-ß) and are involved in various cell regulatory processes, including epithelial-mesenchymal transition-associated cell responses, that is, cell morphological changes, E-cadherin downregulation, stress fiber formation, and cell motility enhancement. Smad2 contains an additional exon encoding 30 amino acid residues compared with Smad3, leading to distinct Smad2 and Smad3 functional properties. Intriguingly, Smad2 also has an alternatively spliced isoform termed Smad2Δexon3 (also known as Smad2ß) lacking the additional exon and behaving similarly to Smad3. However, Smad2Δexon3 and Smad3 signaling properties have not yet been compared in detail. In this study, we reveal that Smad2Δexon3 rescues multiple TGF-ß-induced in vitro cellular responses that would become defective upon SMAD3 KO but does not rescue cell motility enhancement. Using Smad2Δexon3/Smad3 chimeric proteins, we identified that residues Arg-104 and Asn-210 in Smad3, which are not conserved in Smad2Δexon3, are key for TGF-ß-enhanced cell motility. Moreover, we discovered that Smad2Δexon3 fails to rescue the enhanced cell motility as it does not mediate TGF-ß signals to downregulate transcription of ARHGAP24, a GTPase-activating protein that targets Rac1. This study reports for the first time distinct signaling properties of Smad2Δexon3 and Smad3.


Asunto(s)
Movimiento Celular , Exones , Eliminación de Secuencia , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Factor de Crecimiento Transformador beta , Animales , Mamíferos/metabolismo , Proteína Smad2/química , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/deficiencia , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Exones/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo
2.
BMC Genomics ; 23(Suppl 1): 525, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35858839

RESUMEN

BACKGROUND: The transforming growth factor beta-1 (TGF ß-1) cytokine exerts both pro-tumor and anti-tumor effects in carcinogenesis. An increasing body of literature suggests that TGF ß-1 signaling outcome is partially dependent on the regulatory targets of downstream receptor-regulated Smad (R-Smad) proteins Smad2 and Smad3. However, the lack of Smad-specific antibodies for ChIP-seq hinders convenient identification of Smad-specific binding sites. RESULTS: In this study, we use localization and affinity purification (LAP) tags to identify Smad-specific binding sites in a cancer cell line. Using ChIP-seq data obtained from LAP-tagged Smad proteins, we develop a convolutional neural network with long-short term memory (CNN-LSTM) as a deep learning approach to classify a pool of Smad-bound sites as being Smad2- or Smad3-bound. Our data showed that this approach is able to accurately classify Smad2- versus Smad3-bound sites. We use our model to dissect the role of each R-Smad in the progression of breast cancer using a previously published dataset. CONCLUSIONS: Our results suggests that deep learning approaches can be used to dissect binding site specificity of closely related transcription factors.


Asunto(s)
Aprendizaje Profundo , Sitios de Unión , Línea Celular , Transducción de Señal , Proteína Smad2/química , Proteína Smad2/metabolismo , Proteína smad3/química , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
J Food Biochem ; 46(3): e13882, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34312884

RESUMEN

Atherosclerosis, the major underlying pathology of cardiovascular disease, commences with the binding and trapping of lipids on modified proteoglycans, with hyperelongated glycosaminoglycan chains. Transforming growth factor (TGF)-ß stimulates glycosaminoglycan elongation in vascular smooth muscle cells. We have recently shown that this TGF-ß signaling pathway involves reactive oxygen species (ROS). YY-11 is a dodecapeptide derived from camel milk and it has antioxidant activity. We have investigated the role of YY-11 in blocking ROS signaling and downstream atherogenic responses. YY-11 inhibited TGF-ß stimulated ROS production and inhibited the expression of genes for glycosaminoglycan chain elongation as a component of an in vitro model of atherosclerosis. This study provides a biochemical mechanism for the role of camel milk as a potential nutritional product to contribute to the worldwide amelioration of cardiovascular disease. PRACTICAL APPLICATIONS: The identification of readily accessible foods with antioxidant properties would provide a convenient and cost-effective approach community wide reducing oxidative stress induced pathologies such as atherosclerosis. We demonstrate that camel milk-derived peptide is an antioxidant that can inhibit growth factor-mediated proteoglycan modification in vitro. As proteoglycan modification is being recognized as one of the earliest atherogenic responses, these data support the notion of camel milk as a suitable nutritional product to contribute to the prevention of early stage of atherosclerosis development.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Aterosclerosis/tratamiento farmacológico , Camelus/metabolismo , Enfermedades Cardiovasculares/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Leche/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Fosforilación , Proteoglicanos/química , Proteoglicanos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína Smad2/química , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
J Orthop Surg Res ; 16(1): 564, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526039

RESUMEN

BACKGROUND: The tumor necrosis factor-stimulated gene-6 (TSG-6) has been confirmed to inhibit inflammation. It is now generally accepted that local inflammatory stimulation around shoulder capsule causes proliferative fibrosis. This study aims to investigate the mechanism of recombinant TSG-6 protein inhibiting the growth of capsule fibroblasts in frozen shoulder via the TGF-ß/Smad2 signal pathway. METHODS: Human frozen shoulder capsule tissue was taken for primary and passage culture, and the 3rd generation fibroblasts from pathological frozen shoulder capsule were treated with different concentrations of recombinant TSG-6 protein, or with TGF-ß1 agonist SRI-011381. Immunoconfocal analysis was used to identify the isolated fibroblasts, and MTT assay, colony formation assay, and flow cytometry were used to detect the viability, proliferation, and apoptosis rate of fibroblast. The contents of fibrosis and inflammation indexes COL1A1, TNF-α, IL-6, and IL-1ß in the cell supernatant were detected using ELISA and then further examined by qRT-PCR. The expression of Bax, Bcl-2, and proteins related to TGF-ß/Smad2 pathway were detected by Western Blot. RESULTS: Compared with the blank control group, fibroblasts intervened with TSG-6 (2 µg and 5 µg) showed significantly decreased viability and proliferation ability and enhanced cell apoptosis, concurrent with the reductions in Bcl-2 expression; COL1A1, TNF-α, IL-6, and IL-1ß levels; and the expression of TGF-ß1 and phosphorylated Smad22, and an increase in Bax expression, while SRI-011381 treatment would reverse the effect of recombinant TSG-6 protein. CONCLUSION: Recombinant TSG-6 protein inhibited the growth of primary fibroblasts from human frozen shoulder capsule by suppressing the TGF-ß/Smad2 signaling pathway.


Asunto(s)
Bursitis , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1 , Proteína X Asociada a bcl-2/metabolismo , Fibroblastos/metabolismo , Fibrosis , Humanos , Inflamación , Interleucina-6 , Transducción de Señal , Proteína Smad2/química , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/química
5.
Cells ; 10(7)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34359912

RESUMEN

Transforming growth factor-ß (TGF-ß) has been identified as an inducer of hepatocyte epithelial-mesenchymal transition (EMT), which triggers liver fibrosis. Death-associated protein 6 (Daxx) is known to be associated with the TGF-ß-induced apoptotic pathway, but the function of Daxx in liver fibrosis remains unknown. This study aimed to elucidate the role of Daxx in liver fibrosis. We used liver fibrosis tissues from humans and mice to assess Daxx expression. EMT properties and TGF-ß signaling pathway activation were investigated in the Daxx-overexpressing FL83B cell line. The therapeutic effect of Daxx was investigated in a mouse model of liver fibrosis by the hydrodynamic injection of plasmids. The expression of Daxx was markedly decreased in hepatocytes from fibrotic human and mouse livers, as well as in hepatocytes treated with TGF-ß in vitro. The overexpression of Daxx inhibited the EMT process by interfering with the TGF-ß-induced phosphorylation of Smad2. Coimmunoprecipitation analysis confirmed that Daxx reduced the transcriptional activity of Smad2 by binding to its MH1 domain and interfering with Smad2 acetylation. In addition, the therapeutic delivery of Daxx alleviated liver fibrosis in a thioacetamide-induced fibrosis mouse model. Overall, our results indicate that Daxx could be a potential therapeutic target to modulate fibrogenesis, as well as a useful biomarker for liver fibrosis.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Cirrosis Hepática/metabolismo , Chaperonas Moleculares/metabolismo , Proteína Smad2/metabolismo , Acetilación , Animales , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Células HEK293 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Dominios Proteicos , Transducción de Señal , Proteína Smad2/química , Tioacetamida , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
6.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34360888

RESUMEN

Osteoarthritis (OA) is a degenerative joint disease characterized by irreversible cartilage damage, inflammation and altered chondrocyte phenotype. Transforming growth factor-ß (TGF-ß) signaling via SMAD2/3 is crucial for blocking hypertrophy. The post-translational modifications of these SMAD proteins in the linker domain regulate their function and these can be triggered by inflammation through the activation of kinases or phosphatases. Therefore, we investigated if OA-related inflammation affects TGF-ß signaling via SMAD2/3 linker-modifications in chondrocytes. We found that both Interleukin (IL)-1ß and OA-synovium conditioned medium negated SMAD2/3 transcriptional activity in chondrocytes. This inhibition of TGF-ß signaling was enhanced if SMAD3 could not be phosphorylated on Ser213 in the linker region and the inhibition by IL-1ß was less if the SMAD3 linker could not be phosphorylated at Ser204. Our study shows evidence that inflammation inhibits SMAD2/3 signaling in chondrocytes via SMAD linker (de)-phosphorylation. The involvement of linker region modifications may represent a new therapeutic target for OA.


Asunto(s)
Condrocitos/metabolismo , Condrocitos/patología , Osteoartritis/metabolismo , Transducción de Señal/genética , Proteína Smad2/química , Proteína Smad2/metabolismo , Proteína smad3/química , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Adulto , Animales , Bovinos , Línea Celular Tumoral , Humanos , Hipertrofia/metabolismo , Inflamación/metabolismo , Interleucina-1beta/farmacología , Osteoartritis/genética , Osteoartritis/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Dominios Proteicos/efectos de los fármacos , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Proteína Smad2/genética , Proteína smad3/genética , Membrana Sinovial/metabolismo , Transfección , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/farmacología
7.
Nucleic Acids Res ; 49(2): 601-620, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33406242

RESUMEN

It is a well-known and intensively studied phenomenon that the levels of many miRNAs are differentiated in cancer. miRNA biogenesis and functional expression are complex processes orchestrated by many proteins cumulatively called miRNA biogenesis proteins. To characterize cancer somatic mutations in the miRNA biogenesis genes and investigate their potential impact on the levels of miRNAs, we analyzed whole-exome sequencing datasets of over 10 000 cancer/normal sample pairs deposited within the TCGA repository. We identified and characterized over 3600 somatic mutations in 29 miRNA biogenesis genes and showed that some of the genes are overmutated in specific cancers and/or have recurrent hotspot mutations (e.g. SMAD4 in PAAD, COAD and READ; DICER1 in UCEC; PRKRA in OV and LIN28B in SKCM). We identified a list of miRNAs whose level is affected by particular types of mutations in either SMAD4, SMAD2 or DICER1 and showed that hotspot mutations in the RNase domains in DICER1 not only decrease the level of 5p-miRNAs but also increase the level of 3p-miRNAs, including many well-known cancer-related miRNAs. We also showed an association of the mutations with patient survival. Eventually, we created an atlas/compendium of miRNA biogenesis alterations providing a useful resource for different aspects of biomedical research.


Asunto(s)
ARN Helicasas DEAD-box/genética , MicroARNs/biosíntesis , Mutación , Proteínas de Neoplasias/genética , Neoplasias/genética , ARN Neoplásico/biosíntesis , Ribonucleasa III/genética , Proteína Smad2/genética , Proteína Smad4/genética , ARN Helicasas DEAD-box/metabolismo , Conjuntos de Datos como Asunto , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Estudio de Asociación del Genoma Completo , Humanos , MicroARNs/genética , Modelos Moleculares , Mutación Missense , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/mortalidad , Conformación Proteica , ARN Neoplásico/genética , Ribonucleasa III/metabolismo , Proteína Smad2/química , Proteína Smad2/metabolismo , Proteína Smad4/química , Proteína Smad4/metabolismo
8.
Sci Signal ; 13(662)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33323411

RESUMEN

Transforming growth factor-ß (TGF-ß) proteins regulate multiple cellular functions, including cell proliferation, apoptosis, and extracellular matrix formation. The dysregulation of TGF-ß signaling causes diseases such as cancer and fibrosis, and therefore, understanding the biochemical basis of TGF-ß signal transduction is important for elucidating pathogenic mechanisms in these diseases. SMAD proteins are transcription factors that mediate TGF-ß signaling-dependent gene expression. The transcriptional coactivator CBP directly interacts with the MH2 domains of SMAD2 to activate SMAD complex-dependent gene expression. Here, we report the structural basis for CBP recognition by SMAD2. The crystal structures of the SMAD2 MH2 domain in complex with the SMAD2-binding region of CBP showed that CBP forms an amphiphilic helix on the hydrophobic surface of SMAD2. The expression of a mutated CBP peptide that showed increased SMAD2 binding repressed SMAD2-dependent gene expression in response to TGF-ß signaling in cultured cells. Disrupting the interaction between SMAD2 and CBP may therefore be a promising strategy for suppressing SMAD-dependent gene expression.


Asunto(s)
Fragmentos de Péptidos/química , Sialoglicoproteínas/química , Transducción de Señal , Proteína Smad2/química , Factor de Crecimiento Transformador beta/química , Humanos , Fragmentos de Péptidos/metabolismo , Dominios Proteicos , Sialoglicoproteínas/metabolismo , Proteína Smad2/metabolismo , Relación Estructura-Actividad , Factor de Crecimiento Transformador beta/metabolismo
9.
ACS Chem Biol ; 15(4): 862-870, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32176847

RESUMEN

The transforming growth factor beta-receptor I/activin receptor-like kinase 5 (TGFBR1/ALK5) and its close homologue ALK4 are receptor protein kinases associated with the development of diverse diseases, including cancer, fibrosis, heart diseases, and dysfunctional immune response. Therefore, ALK4/5 are among the most studied kinases, and several inhibitors have been developed. However, current commercially available inhibitors either lack selectivity or have not been comprehensively characterized, limiting their value for studying ALK4/5 function in cellular systems. To this end, we report the characterization of the 2-oxo-imidazopyridine, TP-008, a potent chemical probe with dual activity for ALK4 and ALK5 as well as the development of a matching negative control compound. TP-008 has excellent cellular potency and strongly abrogates phosphorylation of the substrate SMAD2 (mothers against decapentaplegic homologue 2). Thus, this chemical probe offers an excellent tool for mechanistic studies on the ALK4/5 signaling pathway and the contribution of these targets to disease.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Animales , Sitios de Unión , Células HEK293 , Humanos , Imidazoles/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Piridinas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/química , Transducción de Señal/efectos de los fármacos , Proteína Smad2/química , Proteína Smad2/metabolismo
10.
Genes Dev ; 33(21-22): 1506-1524, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31582430

RESUMEN

TGF-ß receptors phosphorylate SMAD2 and SMAD3 transcription factors, which then form heterotrimeric complexes with SMAD4 and cooperate with context-specific transcription factors to activate target genes. Here we provide biochemical and structural evidence showing that binding of SMAD2 to DNA depends on the conformation of the E3 insert, a structural element unique to SMAD2 and previously thought to render SMAD2 unable to bind DNA. Based on this finding, we further delineate TGF-ß signal transduction by defining distinct roles for SMAD2 and SMAD3 with the forkhead pioneer factor FOXH1 as a partner in the regulation of differentiation genes in mouse mesendoderm precursors. FOXH1 is prebound to target sites in these loci and recruits SMAD3 independently of TGF-ß signals, whereas SMAD2 remains predominantly cytoplasmic in the basal state and set to bind SMAD4 and join SMAD3:FOXH1 at target promoters in response to Nodal TGF-ß signals. The results support a model in which signal-independent binding of SMAD3 and FOXH1 prime mesendoderm differentiation gene promoters for activation, and signal-driven SMAD2:SMAD4 binds to promoters that are preloaded with SMAD3:FOXH1 to activate transcription.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Modelos Moleculares , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Factor de Crecimiento Transformador beta/metabolismo , Animales , Embrión de Mamíferos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Estructura Terciaria de Proteína , Proteína Smad2/química , Proteína Smad2/metabolismo , Proteína smad3/química , Proteína smad3/metabolismo
11.
J Biol Chem ; 294(42): 15466-15479, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31481467

RESUMEN

Smad proteins are transcriptional regulators activated by TGF-ß. They are known to bind to two distinct Smad-responsive motifs, namely the Smad-binding element (SBE) (5'-GTCTAGAC-3') and CAGA motifs (5'-AGCCAGACA-3' or 5'-TGTCTGGCT-3'). However, the mechanisms by which these motifs promote Smad activity are not fully elucidated. In this study, we performed DNA CASTing, binding assays, ChIP sequencing, and quantitative RT-PCR to dissect the details of Smad binding and function of the SBE and CAGA motifs. We observed a preference for Smad3 to bind CAGA motifs and Smad4 to bind SBE, and that either one SBE or a triple-CAGA motif forms a cis-acting functional half-unit for Smad-dependent transcription activation; combining two half-units allows efficient activation. Unexpectedly, the extent of Smad binding did not directly correlate with the abilities of Smad-binding sequences to induce gene expression. We found that Smad proteins are more tolerant of single bp mutations in the context of the CAGA motifs, with any mutation in the SBE disrupting function. CAGA and CAGA-like motifs but not SBE are widely distributed among stimulus-dependent Smad2/3-binding sites in normal murine mammary gland epithelial cells, and the number of CAGA and CAGA-like motifs correlates with fold-induction of target gene expression by TGF-ß. These data, demonstrating Smad responsiveness can be tuned by both sequence and number of repeats, provide a compelling explanation for why CAGA motifs are predominantly used for Smad-dependent transcription activation in vivo.


Asunto(s)
Proteína smad3/química , Proteína smad3/metabolismo , Proteína Smad4/química , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Secuencias de Aminoácidos , Secuencia de Bases , Sitios de Unión , Humanos , Unión Proteica , Elementos de Respuesta , Proteína Smad2/química , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína Smad4/genética , Activación Transcripcional
12.
Mol Med Rep ; 19(6): 5203-5210, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31059039

RESUMEN

The aim of the current study was to investigate the expression and role of microRNA­486­5p (miR­486­5p) in hypertrophic scar (HS) formation, and to examine the associated mechanisms. First, miR­486­5p expression was detected in HS tissues and human hypertrophic scar fibroblasts (hHSFs) by reverse transcription­quantitative polymerase chain reaction. Target genes of miR­486­5p were predicted using TargetScan and verified by dual­luciferase reporter assays. To investigate the role of miR­486­5p in HS formation, miR­486­5p was overexpressed in hHSFs through transfection with miR­486­5p mimics. MTT, cell apoptosis and cell cycle assays were preformed to investigate the proliferation, cell apoptosis and cell cycle distribution of hHSFs, respectively. Additionally, protein expression was measured by western blot analysis. The results demonstrated that miR­486­5p expression was significantly decreased in HS tissues and cells. Mothers against decapentaplegic homolog (Smad)2 was a target gene of miR­486­5p, and it was negatively regulated by miR­486­5p. It was also found that Smad2 expression was significantly increased in HS tissues and cells. Further analysis indicated that miR­486­5p mimic transfection inhibited the proliferation, induced cell apoptosis and increased G1/S phase arrest in hHSFs. Furthermore, the expression of cyclin­dependent kinase (CDK)2, CDK4 and apoptosis regulator Bcl­2 was repressed, while apoptosis regulator BAX expression was enhanced by miR­486­5p mimic transfection. Notably, the effects of miR­486­5p mimic on hHSFs were significantly eliminated by Smad2 plasmid transfection. Taken together, these results demonstrated that miR­486­5p inhibited the proliferation, induced apoptosis and increased G1/S phase arrest of hHSFs by targeting Smad2. miR­486­5p may be a promising therapeutic target for HS management.


Asunto(s)
Cicatriz Hipertrófica/patología , MicroARNs/metabolismo , Proteína Smad2/metabolismo , Regiones no Traducidas 3' , Adulto , Antagomirs/metabolismo , Apoptosis , Proliferación Celular , Cicatriz Hipertrófica/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína Smad2/química , Proteína Smad2/genética , Proteína X Asociada a bcl-2/metabolismo
13.
Cell Signal ; 53: 365-373, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30423352

RESUMEN

Growth factors such as thrombin and transforming growth factor (TGF)-ß facilitate glycosaminoglycan (GAG) chain hyperelongation on proteoglycans, a phenomenon that increases lipoprotein binding in the vessel wall and the development of atherosclerosis. TGF-ß signals via canonical carboxy terminal phosphorylation of R-Smads and also non-canonical linker region phosphorylation of R-Smads. The G protein coupled receptor agonist, thrombin, can transactivate the TGF-ß receptor leading to both canonical and non-canonical Smad signalling. Linker region phosphorylation drives the expression of genes for the synthesis of the proteoglycan, biglycan. Proteoglycan synthesis involves core protein synthesis, the initiation of GAG chains and the subsequent elongation of GAG chains. We have explored the relationship between the thrombin stimulated phosphorylation of individual serine and threonine sites in the linker region of Smad2 and the expression of GAG initiation xylosyltransferase-1 (XT-1) and GAG elongation chondroitin 4-sulfotransferase-1 (C4ST-1) and chondroitin synthase-1 (CHSY-1) genes. Thrombin stimulated the phosphorylation of all four target residues (Thr220, Ser245, Ser250 and Ser255 residues) with a similar temporal pattern - phosphorylation was maximal at 15 min (the earliest time point studied) and the level of the phospho-proteins declined thereafter over the following 4 h. Jnk, p38 and PI3K, selectively mediated the phosphorylation of the Thr220 residue whereas the serine residues were variously phosphorylated by multiple kinases. Thrombin stimulated the expression of all three genes - XT-1, C4ST-1 and CHSY-1. The three pathways mediating Thr220 phosphorylation were also involved in the expression of XT-1. The target pathways (excluding Jnk) were involved in the expression of the GAG elongation genes (C4ST-1 and CHSY-1). These findings support the contention that individual Smad linker region phosphorylation sites are linked to the expression of genes for the initiation and elongation of GAG chains on proteoglycans. The context of this work is that a specific inhibitor of GAG elongation represents a potential therapeutic agent for preventing GAG elongation and lipid binding and the results indicate that the specificity of the pathways is such that it might be therapeutically feasible to specifically target GAG elongation without interfering with other physiological processes with which proteoglycans are involved.


Asunto(s)
Glicosaminoglicanos/genética , Proteoglicanos/genética , Proteína Smad2/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Fosforilación , Proteína Smad2/química , Trombina/metabolismo
14.
Nucleic Acids Res ; 46(22): 12139-12153, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30321401

RESUMEN

Receptor-regulated SMAD (R-SMAD: SMAD1, SMAD2, SMAD3, SMAD5 and SMAD8) proteins are key transcription factors of the transforming growth factor-ß (TGF-ß) superfamily of cytokines. MAN1, an integral protein of the inner nuclear membrane, is a SMAD cofactor that terminates TGF-ß superfamily signals. Heterozygous loss-of-function mutations in MAN1 result in osteopoikilosis, Buschke-Ollendorff syndrome and melorheostosis. MAN1 interacts with MAD homology 2 (MH2) domains of R-SMAD proteins using its C-terminal U2AF homology motif (UHM) domain and UHM ligand motif (ULM) and facilitates R-SMAD dephosphorylation. Here, we report the structural basis for R-SMAD recognition by MAN1. The SMAD2-MAN1 and SMAD1-MAN1 complex structures show that an intramolecular UHM-ULM interaction of MAN1 forms a hydrophobic surface that interacts with a hydrophobic surface among the H2 helix, the strands ß8 and ß9, and the L3 loop of the MH2 domains of R-SMAD proteins. The complex structures also show the mechanism by which SMAD cofactors distinguish R-SMAD proteins that possess a highly conserved molecular surface.


Asunto(s)
Proteínas de la Membrana/química , Proteínas Nucleares/química , Proteína Smad1/química , Proteína Smad2/química , Secuencias de Aminoácidos , Animales , Simulación por Computador , Cristalografía por Rayos X , Citocinas/metabolismo , Análisis Mutacional de ADN , Proteínas de Unión al ADN , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Mutación , Membrana Nuclear/química , Fosforilación , Unión Proteica , Dominios Proteicos , Transducción de Señal , Proteína smad3/química , Factor de Crecimiento Transformador beta/metabolismo
15.
J Biol Chem ; 293(41): 15867-15886, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30108174

RESUMEN

Transforming growth factor-ß (TGFß) signaling through SMAD2/3 is an important driver of pathological fibrosis in multiple organ systems. TGFß signaling and extracellular matrix (ECM) stiffness form an unvirtuous pathological circuit in which matrix stiffness drives activation of latent TGFß, and TGFß signaling then drives cellular stress and ECM synthesis. Moreover, ECM stiffness also appears to sensitize cells to exogenously activated TGFß through unknown mechanisms. Here, using human fibroblasts, we explored the effect of ECM stiffness on a putative inner nuclear membrane protein, LEM domain-containing protein 3 (LEMD3), which is physically connected to the cell's actin cytoskeleton and inhibits TGFß signaling. We showed that LEMD3-SMAD2/3 interactions are inversely correlated with ECM stiffness and TGFß-driven luciferase activity and that LEMD3 expression is correlated with the mechanical response of the TGFß-driven luciferase reporter. We found that actin polymerization but not cellular stress or LEMD3-nuclear-cytoplasmic couplings were necessary for LEMD3-SMAD2/3 interactions. Intriguingly, LEMD3 and SMAD2/3 frequently interacted in the cytosol, and we discovered LEMD3 was proteolytically cleaved into protein fragments. We confirmed that a consensus C-terminal LEMD3 fragment binds SMAD2/3 in a stiffness-dependent manner throughout the cell and is sufficient for antagonizing SMAD2/3 signaling. Using human lung biopsies, we observed that these nuclear and cytosolic interactions are also present in tissue and found that fibrotic tissues exhibit locally diminished and cytoplasmically shifted LEMD3-SMAD2/3 interactions, as noted in vitro Our work reveals novel LEMD3 biology and stiffness-dependent regulation of TGFß by LEMD3, providing a novel target to antagonize pathological TGFß signaling.


Asunto(s)
Mecanotransducción Celular/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Actinas/metabolismo , Citosol/metabolismo , Proteínas de Unión al ADN , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Lámina Nuclear/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosforilación , Proteína Fosfatasa 2C/metabolismo , Proteína Smad2/antagonistas & inhibidores , Proteína Smad2/química , Proteína smad3/antagonistas & inhibidores , Proteína smad3/química , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
16.
Nucleic Acids Res ; 46(17): 9220-9235, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30060237

RESUMEN

TGIF1 is a multifunctional protein that represses TGF-ß-activated transcription by interacting with Smad2-Smad4 complexes. We found that the complex structure of TGIF1-HD bound to the TGACA motif revealed a combined binding mode that involves the HD core and the major groove, on the one hand, and the amino-terminal (N-term) arm and the minor groove of the DNA, on the other. We also show that TGIF1-HD interacts with the MH1 domain of Smad proteins, thereby indicating that TGIF1-HD is also a protein-binding domain. Moreover, the formation of the HD-MH1 complex partially hinders the DNA-binding site of the complex, preventing the efficient interaction of TGIF1-HD with DNA. We propose that the binding of the TGIF1 C-term to the Smad2-MH2 domain brings both the HD and MH1 domain into close proximity. This local proximity facilitates the interaction of these DNA-binding domains, thus strengthening the formation of the protein complex versus DNA binding. Once the protein complex has been formed, the TGIF1-Smad system would be released from promoters/enhancers, thereby illustrating one of the mechanisms used by TGIF1 to exert its function as an active repressor of Smad-induced TGF-ß signaling.


Asunto(s)
ADN/química , Proteínas de Homeodominio/química , Proteínas Represoras/química , Proteína Smad2/química , Proteína Smad4/química , Factor de Crecimiento Transformador beta/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , ADN/genética , ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Modelos Moleculares , Motivos de Nucleótidos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
17.
Life Sci ; 199: 10-15, 2018 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-29510197

RESUMEN

Smads (sma/mothers against decapentaplegic) are transcription factors, which can be phosphorylated in the carboxy terminal (pSmad2/3C) or in the structurally central linker region (pSmad2/3 L). Only receptor kinases such as Transforming Growth Factor (TGF)-ß receptor (TGFBR1) can mediate carboxy terminal phosphorylation but multiple receptors, including TGFBR1 itself, can activate cytosolic serine/threonine kinases and mediate serine/threonine (S/T) linker region phosphorylation of Smad2/3. One important class of agents that can mediate Smad phosphorylation are the G protein coupled receptors (GPCRs) and their ligands and these agents can meditate both carboxy terminal and linker region phosphorylation. Linker region phosphorylation arises due to activation of kinases including those downstream of the transactivation of the EGFR and carboxy terminal Smad phosphorylation can occur as a result of the recently described activity of GPCRs, notably protease activated receptors (PAR)-1, to transactivate TGFBR1 leading to direct carboxy terminal Smad phosphorylation. This review will summarize the effects of GPCR-mediated receptor transactivation pathways on the phosphorylation of Smad2 linker region, as a better understanding of these pathways may provide new approaches for the identification of novel therapeutic agents.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/metabolismo , Factores de Transcripción/metabolismo , Animales , Humanos , Fosforilación/fisiología , Receptores Acoplados a Proteínas G/química , Proteínas Smad Reguladas por Receptores/química , Proteína Smad2/química , Proteína Smad2/metabolismo , Factores de Transcripción/química
18.
Sci Signal ; 11(523)2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29588413

RESUMEN

The transforming growth factor-ß (TGF-ß) superfamily of cytokines regulates various biological processes, including cell proliferation, immune responses, autophagy, and senescence. Dysregulation of TGF-ß signaling causes various diseases, such as cancer and fibrosis. SMAD2 and SMAD3 are core transcription factors involved in TGF-ß signaling, and they form heterotrimeric complexes with SMAD4 (SMAD2-SMAD2-SMAD4, SMAD3-SMAD3-SMAD4, and SMAD2-SMAD3-SMAD4) in response to TGF-ß signaling. These heterotrimeric complexes interact with cofactors to control the expression of TGF-ß-dependent genes. SMAD2 and SMAD3 may promote or repress target genes depending on whether they form complexes with other transcription factors, coactivators, or corepressors; therefore, the selection of specific cofactors is critical for the appropriate activity of these transcription factors. To reveal the structural basis by which SMAD2 and SMAD3 select cofactors, we determined the crystal structures of SMAD3 in complex with the transcription factor FOXH1 and SMAD2 in complex with the transcriptional corepressor SKI. The structures of the complexes show that the MAD homology 2 (MH2) domains of SMAD2 and SMAD3 have multiple hydrophobic patches on their surfaces. The cofactors tether to various subsets of these patches to interact with SMAD2 and SMAD3 in a cooperative or competitive manner to control the output of TGF-ß signaling.


Asunto(s)
Dominios Proteicos , Proteína Smad2/química , Proteína smad3/química , Factores de Transcripción/química , Secuencia de Aminoácidos , Animales , Línea Celular , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Unión Proteica , Homología de Secuencia de Aminoácido , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
19.
J Pharmacol Exp Ther ; 365(1): 156-164, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29438988

RESUMEN

Transforming growth factor-ß (TGF-ß) is a pleiotropic growth factor implicated in the development of atherosclerosis for its role in mediating glycosaminoglycan (GAG) chain hyperelongation on the proteoglycan biglycan, a phenomenon that increases the binding of atherogenic lipoproteins in the vessel wall. Phosphorylation of the transcription factor Smad has emerged as a critical step in the signaling pathways that control the synthesis of biglycan, both the core protein and the GAG chains. We have used flavopiridol, a well-known cyclin-dependent kinase inhibitor, to study the role of linker region phosphorylation in the TGF-ß-stimulated synthesis of biglycan. We used radiosulfate incorporation and SDS-PAGE to assess proteoglycan synthesis, real-time polymerase chain reaction to assess gene expression, and chromatin immunoprecipitation to assess the binding of Smads to the promoter region of GAG Synthesizing genes. Flavopiridol blocked TGF-ß-stimulated synthesis of mRNA for the GAG synthesizing enzymes, and chondroitin 4-sulfotransferase (C4ST-1), chondroitin sulfate synthase-1 (ChSy-1) and TGF-ß-mediated proteoglycans synthesis as well as GAG hyperelongation. Flavopiridol blocked TGF-ß-stimulated Smad2 phosphorylation at both the serine triplet and the isolated threonine residue in the linker region. The binding of Smad to the promoter region of the C4ST-1 and ChSy-1 genes was stimulated by TGF-ß, and this response was blocked by flavopiridol, demonstrating that linker region phosphorylated Smad can pass to the nucleus and positively regulate transcription. These results demonstrate the validity of the kinases, which phosphorylate the Smad linker region as potential therapeutic target(s) for the development of an agent to prevent atherosclerosis.


Asunto(s)
Biglicano/biosíntesis , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Flavonoides/farmacología , Piperidinas/farmacología , Proteína Smad2/química , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Músculo Liso Vascular/citología , Fosforilación/efectos de los fármacos , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/efectos de los fármacos
20.
J Recept Signal Transduct Res ; 38(1): 27-30, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29199516

RESUMEN

Macrophages can be alternatively activated by TGF-ß1 and high-ambient glucose, in which the role of Smad2 and the crosstalk between ERK and Smad2 pathways are not fully understood. The activation of ERK and Smad2 pathways and the expression of arginase-1 were detected by Western blot. The role of Smad2 and the relationship between ERK and Smad2 pathways were investigated by using biochemical inhibitors. The protein of arginase-1 was significantly overexpressed in RAW264.7 cells stimulated by TGF-ß1 and high-ambient glucose, which can be partially blocked by not only U0126 (ERK inhibitor) but also SB431542 (Smad2 inhibitor). Furthermore, simply inhibiting one pathway had no effect on the other pathway. In conclusion, both ERK and Smad2 signal pathways are involved in the activation of macrophages induced by TGF-ß1 and high-ambient glucose, while there is no crosstalk shown in the process.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/metabolismo , Proteína Smad2/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Benzamidas/farmacología , Butadienos/farmacología , Dioxoles/farmacología , Glucosa/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Nitrilos/farmacología , Fosforilación , Células RAW 264.7 , Proteína Smad2/química , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...