Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
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
2.
J Cell Biol ; 221(1)2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34860252

RESUMEN

Transforming growth factor ß (TGF-ß) signaling plays a fundamental role in metazoan development and tissue homeostasis. However, the molecular mechanisms concerning the ubiquitin-related dynamic regulation of TGF-ß signaling are not thoroughly understood. Using a combination of proteomics and an siRNA screen, we identify pVHL as an E3 ligase for SMAD3 ubiquitination. We show that pVHL directly interacts with conserved lysine and proline residues in the MH2 domain of SMAD3, triggering degradation. As a result, the level of pVHL expression negatively correlates with the expression and activity of SMAD3 in cells, Drosophila wing, and patient tissues. In Drosophila, loss of pVHL leads to the up-regulation of TGF-ß targets visible in a downward wing blade phenotype, which is rescued by inhibition of SMAD activity. Drosophila pVHL expression exhibited ectopic veinlets and reduced wing growth in a similar manner as upon loss of TGF-ß/SMAD signaling. Thus, our study demonstrates a conserved role of pVHL in the regulation of TGF-ß/SMAD3 signaling in human cells and Drosophila wing development.


Asunto(s)
Proteolisis , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Drosophila melanogaster/metabolismo , Células HCT116 , Células HeLa , Humanos , Unión Proteica , Dominios Proteicos , Proteína Smad2/metabolismo , Proteína smad3/química , Ubiquitina-Proteína Ligasas/metabolismo
3.
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
4.
Life Sci ; 274: 119299, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33675899

RESUMEN

AIMS: Cardiac fibrosis is a pathological hallmark of progressive heart diseases currently lacking effective treatment. Nicotinamide mononucleotide (NMN), a member of the vitamin B3 family, is a defined biosynthetic precursor of nicotinamide adenine dinucleotide (NAD+). Its beneficial effects on cardiac diseases are known, but its effects on cardiac fibrosis and the underlying mechanism remain unclear. We aimed to elucidate the protective effect of NMN against cardiac fibrosis and its underlying mechanisms of action. MATERIALS AND METHODS: Cardiac fibrosis was induced by isoproterenol (ISO) in mice. NMN was administered by intraperitoneal injection. In vitro, cardiac fibroblasts (CFs) were stimulated by transforming growth factor-beta (TGF-ß) with or without NMN and sirtinol, a SIRT1 inhibitor. Levels of cardiac fibrosis, NAD+/SIRT1 alteration, oxidative stress, and Smad3 acetylation were evaluated by real-time polymerase chain reaction, western blots, immunohistochemistry staining, immunoprecipitation, and assay kits. KEY FINDINGS: ISO treatment induced cardiac dysfunction, fibrosis, and hypertrophy in vivo, whereas NMN alleviated these changes. Additionally, NMN suppressed CFs activation stimulated by TGF-ß in vitro. Mechanistically, NMN restored the NAD+/SIRT1 axis and inhibited the oxidative stress and Smad3 acetylation induced by ISO or TGF-ß. However, the protective effects of NMN were partly antagonized by sirtinol in vitro. SIGNIFICANCE: NMN could attenuate cardiac fibrosis in vivo and fibroblast activation in vitro by suppressing oxidative stress and Smad3 acetylation in a NAD+/SIRT1-dependent manner.


Asunto(s)
Fibrosis/tratamiento farmacológico , Cardiopatías/tratamiento farmacológico , Isoproterenol/toxicidad , Mononucleótido de Nicotinamida/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteína smad3/metabolismo , Acetilación , Animales , Cardiotónicos/toxicidad , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Fibrosis/patología , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Cardiopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Proteína smad3/química
5.
ACS Synth Biol ; 10(3): 505-514, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33587591

RESUMEN

Quantifying the binding affinity of protein-protein interactions is important for elucidating connections within biochemical signaling pathways, as well as characterization of binding proteins isolated from combinatorial libraries. We describe a quantitative yeast-yeast two-hybrid (qYY2H) system that not only enables the discovery of specific protein-protein interactions but also efficient, quantitative estimation of their binding affinities (KD). In qYY2H, the bait and prey proteins are expressed as yeast cell surface fusions using yeast surface display. We developed a semiempirical framework for estimating the KD of monovalent bait-prey interactions, using measurements of bait-prey yeast-yeast binding, which is mediated by multivalent interactions between yeast-displayed bait and prey. Using qYY2H, we identified interaction partners of SMAD3 and the tandem WW domains of YAP from a cDNA library and characterized their binding affinities. Finally, we showed that qYY2H could also quantitatively evaluate binding interactions mediated by post-translational modifications on the bait protein.


Asunto(s)
Mapas de Interacción de Proteínas , Saccharomyces cerevisiae/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción/metabolismo , Biblioteca de Genes , Genes Reporteros , Unión Proteica , Dominios Proteicos , Saccharomyces cerevisiae/genética , Proteína smad3/química , Factores de Transcripción/química , Técnicas del Sistema de Dos Híbridos
6.
Mol Genet Genomic Med ; 8(9): e1378, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32597575

RESUMEN

BACKGROUND: Thoracic aortic aneurysm and dissection (TAA/D) represents a potentially lethal disease group characterized by an increased risk of dissection or rupture. Only a small percentage (approximately 30%) of individuals with nonsyndromic familial TAA/D have a pathogenic variant in one of the genes that have been found to be associated with the disease. METHODS: A targeted sequencing panel and direct sequencing approach were used to identify causative mutations in the index patients and other family members. RESULTS: In this study we report two apparently unrelated Cypriot families with nonsyndromic familial TAA/D. The proband A is a female patient diagnosed with TAA/D and intracranial aneurysm and opted for an elective intervention. The proband B is a male patient who was diagnosed with TAA/D and underwent cardiac surgery. Sequencing analysis identified a novel splice site variant (c.871+1G>A) in SMAD3 which is shown to be associated with the disease. Analysis of mRNA from the patient's tissue confirmed aberrant splicing and exon 6 skipping. CONCLUSION: Our findings expand the mutation spectrum of variants that have been shown to be associated with nonsyndromic familial TAA/D. This study demonstrates the importance of a comprehensive clinical and genetic evaluation aiming at early diagnosis and intervention.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Mutación , Proteína smad3/genética , Adulto , Anciano , Aneurisma de la Aorta Torácica/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Empalme del ARN , Proteína smad3/química
7.
Comput Biol Chem ; 83: 107159, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31743832

RESUMEN

The transforming growth factor ß (TGFß) plays an essential role in the regulation of cellular processes such as cell proliferation, migration, differentiation, and apoptosis by association with SMAD transcriptional factors that are regulated by the transcriptional regulator SnoN. The crystal structure of SnoN-SMAD4 reveals that SnoN can adopt two binding modes, the open and closed forms, at the interfaces of SMAD4 subunits. Accumulating evidence indicates that SnoN can interact with both SMAD3 and SMAD4 to form a ternary SnoN-SMAD3-SMAD4 complex in the TGFß signaling pathway. However, how the interaction of SnoN with the SMAD3 and SMAD4 remains unclear. Here, molecular dynamics (MD) simulations and molecular modeling methods were performed to figure out this issue. The simulations reveal that SnoNopen exists in two, open and semi-closed, conformations. Molecular modeling and MD simulation studies suggest that the SnoNclosed form interferes with the SMAD3-SMAD4 protein; in contract, the SnoNopen can form a stable SnoN-SMAD3-SMAD4 complex. These mechanistic mechanisms may help elucidate the detailed engagement of SnoN with two SMAD3 and SMAD4 transcriptional factors in the regulation of TGFß signaling pathway.


Asunto(s)
Simulación por Computador , Péptidos y Proteínas de Señalización Intracelular/química , Modelos Moleculares , Proteínas Proto-Oncogénicas/química , Proteína smad3/química , Proteína Smad4/química , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Conformación Proteica , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Proteína Smad4/metabolismo
8.
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
9.
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
10.
Cell Death Dis ; 10(6): 458, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31189885

RESUMEN

Hepatocyte apoptosis is a hallmark of nonalcoholic steatohepatitis (NASH) and contributes to liver injury, fibrosis, and inflammation. However, the molecular mechanisms underlying excessive hepatocyte apoptosis in NASH remain largely unknown. This study aimed to explore whether and how the v-ets avian erythroblastosis virus E26 oncogene homolog 1 (Ets-1) is involved in diet-induced hepatocyte apoptosis in mice. The study found that the expression level of hepatic Ets-1 was elevated in a NASH mouse model as a result of the activation of transforming growth factor beta1 (TGF-ß1) signaling. In the presence of TGF-ß1, phosphorylated mothers against decapentaplegic homolog 2/3 (p-Smad2/3) translocated to the binding sites of the Ets-1 promoter to upregulate the expression of Ets-1 in primary hepatocytes. In addition, Ets-1 bound directly to phosphorylated Smad3 (p-Smad3), thereby preventing the ubiquitination and proteasomal degradation of p-Smad3 and enhancing the activity of TGF-ß1/Smad3 signaling. Consequently, elevated Ets-1 stimulated TGF-ß1-induced hepatocyte apoptosis. However, Ets-1 knockdown alleviated diet-induced hepatocyte apoptosis and NASH with reduced liver injury, inflammation, and fibrosis. Taken together, Ets-1 had an adverse impact on hepatocyte survival under TGF-ß1 treatment and accelerated the development of NASH in mice.


Asunto(s)
Apoptosis/genética , Hepatocitos/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Dieta/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hepatocitos/efectos de los fármacos , Humanos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/genética , Fosforilación , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/química , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
11.
J Med Genet ; 56(4): 252-260, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30661052

RESUMEN

BACKGROUND: Pathogenic variants in SMAD3 cause thoracic aortic aneurysms and dissections, along with aneurysms and rupture of other arteries. Here, we examined differences in clinical presentation of aortic events (dissection or surgical repair of an aneurysm) with respect to age and variant type in an international cohort of individuals with SMAD3 variants. METHODS: Aortic status and events, vital status and clinical features were abstracted through retrospective review of medical records of 212 individuals with 51 unique SMAD3 variants, including haploinsufficiency (HI) and missense substitutions in the MH2 domain, as well as novel in-frame deletions and missense variants in the MH1 domain. RESULTS: Aortic events were documented in 37% of cases, with dissections accounting for 70% of events. The median age at first aortic event was significantly lower in individuals with SMAD3 MH2 missense variants than those with HI variants (42years vs 49 years; p=0.003), but there was no difference in frequency of aortic events by variant type. The cumulative risk of an aortic event was 50% at 54 years of age. No aortic events in childhood were observed. CONCLUSIONS: SMAD3 pathogenic variants cause thoracic aortic aneurysms and dissections in the majority of individuals with variable age of onset and reduced penetrance. Of the covariates examined, the type of underlying SMAD3 variant was responsible for some of this variation. Later onset of aortic events and the absence of aortic events in children associated with SMAD3 variants support gene-specific management of this disorder.


Asunto(s)
Aneurisma de la Aorta Torácica/diagnóstico , Aneurisma de la Aorta Torácica/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Fenotipo , Proteína smad3/genética , Adolescente , Adulto , Anciano , Alelos , Sustitución de Aminoácidos , Aneurisma de la Aorta Torácica/complicaciones , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , Dominios Proteicos/genética , Factores de Riesgo , Proteína smad3/química
12.
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
13.
Sci Transl Med ; 10(462)2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305452

RESUMEN

Transforming growth factor-ß (TGF-ß) is a well-established central mediator of renal fibrosis, a common outcome of almost all progressive chronic kidney diseases. Here, we identified a poorly conserved and kidney-enriched long noncoding RNA in TGF-ß1-stimulated human tubular epithelial cells and fibrotic kidneys, which we termed TGF-ß/Smad3-interacting long noncoding RNA (lnc-TSI). Lnc-TSI was transcriptionally regulated by Smad3 and specifically inhibited TGF-ß-induced Smad3 phosphorylation and downstream profibrotic gene expression. Lnc-TSI acted by binding with the MH2 domain of Smad3, blocking the interaction of Smad3 with TGF-ß receptor I independent of Smad7. Delivery of human lnc-TSI into unilateral ureteral obstruction (UUO) mice, a well-established model of renal fibrosis, inhibited phosphorylation of Smad3 in the kidney and attenuated renal fibrosis. In a cohort of 58 patients with biopsy-confirmed IgA nephropathy (IgAN), lnc-TSI renal expression negatively correlated with the renal fibrosis index (r = -0.56, P < 0.001) after adjusting for cofounders. In a longitudinal study, 32 IgAN patients with low expression of renal lnc-TSI at initial biopsy had more pronounced increases in their renal fibrosis index and experienced stronger declines in renal function at repeat biopsy at a mean of 48 months of follow-up. These data suggest that lnc-TSI reduced renal fibrogenesis through negative regulation of the TGF-ß/Smad pathway.


Asunto(s)
Riñón/patología , ARN Largo no Codificante/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Secuencia de Bases , Línea Celular , Células Epiteliales/metabolismo , Fibrosis , Humanos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Dominios Proteicos , ARN Largo no Codificante/genética , Proteína smad3/química , Regulación hacia Arriba , Obstrucción Ureteral/patología
14.
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
15.
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
16.
Nat Commun ; 8(1): 2070, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29234012

RESUMEN

Smad transcription factors activated by TGF-ß or by BMP receptors form trimeric complexes with Smad4 to target specific genes for cell fate regulation. The CAGAC motif has been considered as the main binding element for Smad2/3/4, whereas Smad1/5/8 have been thought to preferentially bind GC-rich elements. However, chromatin immunoprecipitation analysis in embryonic stem cells showed extensive binding of Smad2/3/4 to GC-rich cis-regulatory elements. Here, we present the structural basis for specific binding of Smad3 and Smad4 to GC-rich motifs in the goosecoid promoter, a nodal-regulated differentiation gene. The structures revealed a 5-bp consensus sequence GGC(GC)|(CG) as the binding site for both TGF-ß and BMP-activated Smads and for Smad4. These 5GC motifs are highly represented as clusters in Smad-bound regions genome-wide. Our results provide a basis for understanding the functional adaptability of Smads in different cellular contexts, and their dependence on lineage-determining transcription factors to target specific genes in TGF-ß and BMP pathways.


Asunto(s)
Secuencias de Aminoácidos , Proteína Goosecoide/genética , Proteína smad3/química , Proteína Smad4/química , Regulación Alostérica/genética , Animales , Sitios de Unión/genética , Proteínas Morfogenéticas Óseas/metabolismo , Sistemas CRISPR-Cas , Linaje de la Célula/genética , Cristalografía por Rayos X , Regulación del Desarrollo de la Expresión Génica , Espectroscopía de Resonancia Magnética , Ratones , Células Madre Embrionarias de Ratones , Regiones Promotoras Genéticas , Unión Proteica , Proteína smad3/genética , Proteína Smad4/genética , Factor de Crecimiento Transformador beta/metabolismo
17.
J Biol Chem ; 292(34): 14270-14278, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28659340

RESUMEN

Smooth muscle cell (SMC) differentiation is essential for vascular development, and TGF-ß signaling plays a critical role in this process. Although long non-coding RNAs (lncRNAs) regulate various cellular events, their functions in SMC differentiation remain largely unknown. Here, we demonstrate that the lncRNA growth arrest-specific 5 (GAS5) suppresses TGF-ß/Smad3 signaling in smooth muscle cell differentiation of mesenchymal progenitor cells. We found that forced expression of GAS5 blocked, but knockdown of GAS5 increased, the expression of SMC contractile proteins. Mechanistically, GAS5 competitively bound Smad3 protein via multiple RNA Smad-binding elements (rSBEs), which prevented Smad3 from binding to SBE DNA in TGF-ß-responsive SMC gene promoters, resulting in suppression of SMC marker gene transcription and, consequently, in inhibition of TGF-ß/Smad3-mediated SMC differentiation. Importantly, other lncRNAs or artificially synthesized RNA molecules that contained rSBEs also effectively inhibited TGF-ß/Smad3 signaling, suggesting that lncRNA-rSBE may be a general mechanism used by cells to fine-tune Smad3 activity in both basal and TGF-ß-stimulated states. Taken together, our results have uncovered an lncRNA-based mechanism that modulates TGF-ß/Smad3 signaling during SMC differentiation.


Asunto(s)
Músculo Liso Vascular/metabolismo , ARN Largo no Codificante/metabolismo , ARN/metabolismo , Elementos de Respuesta , Transducción de Señal , Proteína smad3/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Unión Competitiva , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Hibridación Fluorescente in Situ , Isoquinolinas/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Proteínas Musculares/agonistas , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Conformación de Ácido Nucleico , Piridinas/farmacología , Pirroles/farmacología , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/química , Elementos de Respuesta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína smad3/química , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/química , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
18.
Cell Physiol Biochem ; 42(1): 357-372, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28535533

RESUMEN

BACKGROUND/AIMS: Endothelial-to-mesenchymal transition (EndMT) plays significant roles under various pathological conditions including cardiovascular diseases, fibrosis, and cancer. EndMT of endothelial progenitor cells (EPCs) contributes to neointimal hyperplasia following cell therapy Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA (lncRNA) that promotes metastasis and cancer. MicroRNA-145 (miR-145) is a tumor suppressor that has been reported to inhibit SMAD3-mediated epithelial-to-mesenchymal transition (EMT) of cancer cells. In the present study, we investigated the role of MALAT1 and miR-145 in EndMT of human circulating EPCs induced by transforming growth factor beta1 (TGF-ß1). METHODS: Human circulating EPCs were isolated and characterized by fluorescence-activated cell sorting (FACS). Expression levels of EndMT markers were assessed by qRT-PCR and western blotting. Alpha-smooth muscle actin (α-SMA) expression was measured by cell immunofluorescence staining. The regulatory relationship between MALAT1 and miR-145 and its target genes, TGFBR2 (TGFß receptortype II) and SMAD3 (mothers against decapentaplegic homolog 3) was analyzed using the luciferase reporter assay. RESULTS: We found that EndMT of EPCs induced by TGF-ß1 is accompanied by increased MALAT1 expression and decreased miR-145 expression, and MALAT1 and miR-145 directly bind and reciprocally repress each other in these cells. Dual-Luciferase Reporter assay indicated that miR-145 inhibits TGF-ß1-induced EndMT by directly targeting TGFBR2 and SMAD3. CONCLUSIONS: MALAT1 modulates TGF-ß1-induced EndMT of EPCs through regulation of TGFBR2 and SMAD3 via miR-145. Thus, the MALAT1-miR-145-TGFBR2/SMAD3 signaling pathway plays a key role in TGF-ß1-induced EndMT.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regiones no Traducidas 3' , Antagomirs/metabolismo , Secuencia de Bases , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Células Progenitoras Endoteliales/citología , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Alineación de Secuencia , Transducción de Señal , Proteína smad3/química , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
19.
Sci Rep ; 6: 33116, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27604640

RESUMEN

Transforming growth factor ß-1 (TGFß-1)-induced phosphorylation of transcription factors Smad2 and Smad3 plays a crucial role in the pathogenesis of idiopathic pulmonary fibrosis (IPF). However, the molecular regulation of Smad2/Smad3 proteins stability remains a mystery. Here, we show that ubiquitin carboxyl-terminal hydrolase-L5 (UCHL5 or UCH37) de-ubiquitinates both Smad2 and Smad3, up-regulates their stability, and promotes TGFß-1-induced expression of profibrotic proteins, such as fibronectin (FN) and α-smooth muscle actin (α-SMA). Inhibition or down-regulation of UCHL5 reduced Smad2/Smad3 levels and TGFß-1-induced the expression of FN and α-SMA in human lung fibroblast. We demonstrate that Smad2 and Smad3 ubiquitination was diminished by over-expression of UCHL5, while it was enhanced by inhibition or down-regulation of UCHL5. UCHL5 is highly expressed in IPF lungs. UCHL5, Smad2, and Smad3 levels were increased in bleomycin-injured lungs. Administration of UCHL5 inhibitor, b-AP15, reduced the expression of FN, type I collagen, Smad2/Smad3, and the deposition of collagen in lung tissues in a bleomycin-induced model of pulmonary fibrosis. Our studies provide a molecular mechanism by which UCHL5 mitigates TGFß-1 signaling by stabilizing Smad2/Smad3. These data indicate that UCHL5 may contribute to the pathogenesis of IPF and may be a potential therapeutic target.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Bleomicina/toxicidad , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/etiología , Ratones , Ratones Endogámicos C57BL , Piperidonas/farmacología , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína Smad2/química , Proteína smad3/química , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitinación/efectos de los fármacos
20.
Biochem Pharmacol ; 116: 200-9, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27473774

RESUMEN

PURPOSE: Smad3 is a critical signaling protein in renal fibrosis. Proteolysis targeting chimeric molecules (PROTACs) are small molecules designed to degrade target proteins via ubiquitination. They have three components: (1) a recognition motif for E3 ligase; (2) a linker; and (3) a ligand for the target protein. We aimed to design a new PROTAC to prevent renal fibrosis by targeting Smad3 proteins and using hydroxylated pentapeptide of hypoxia-inducible factor-1α as the recognition motif for von Hippel-Lindau (VHL) ubiquitin ligase (E3). METHODS: Computer-aided drug design was used to find a specific ligand targeting Smad3. Surface plasmon resonance (SPR) was used to verify and optimize screening results. Synthesized PROTAC was validated by two-stage mass spectrometry. The PROTAC's specificity for VHL (E3 ligase) was proved with two human renal carcinoma cell lines, 786-0 (VHL(-)) and ACHN (VHL(+)), and its anti-fibrosis effect was tested in renal fibrosis cell models. RESULTS: Thirteen small molecular compounds (SMCs) were obtained from the Enamine library using GLIDE molecular docking program. SPR results showed that #8 SMC (EN300-72284) combined best with Smad3 (KD=4.547×10(-5)M). Mass spectrometry showed that synthesized PROTAC had the correct peptide molecular weights. Western blot showed Smad3 was degraded by PROTAC with whole-cell lysate of ACHN but not 786-0. Degradation, but not ubiquitination, of Smad3 was inhibited by proteasome inhibitor MG132. The upregulation of fibronectin and Collagen I induced by TGF-ß1 in both renal fibroblast and mesangial cells were inhibited by PROTAC. CONCLUSION: The new PROTAC might prevent renal fibrosis by targeting Smad3 for ubiquitination and degradation.


Asunto(s)
Diseño de Fármacos , Riñón/efectos de los fármacos , Modelos Moleculares , Proteínas Recombinantes de Fusión/uso terapéutico , Insuficiencia Renal Crónica/tratamiento farmacológico , Proteína smad3/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Benzofuranos/química , Benzofuranos/metabolismo , Benzofuranos/uso terapéutico , Sitios de Unión , Línea Celular Tumoral , Fibrosis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/uso terapéutico , Riñón/metabolismo , Riñón/patología , Ligandos , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/uso terapéutico , Dominios y Motivos de Interacción de Proteínas , Proteolisis/efectos de los fármacos , Piridinas/química , Piridinas/metabolismo , Piridinas/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Proteína smad3/química , Proteína smad3/metabolismo , Bibliotecas de Moléculas Pequeñas , Resonancia por Plasmón de Superficie , Ubiquitina-Proteína Ligasas/química , Ubiquitinación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...