Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Genes (Basel) ; 12(10)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34680931

RESUMO

Smads are involved in a variety of biological activities by mediating bone morphogenetic protein (BMP) signals. The full-length coding sequences (CDSs) of buffalo Smads 1, 4, and 5 were isolated and identified through RT-PCR in this study. Their lengths are 1398 bp, 1662 bp, and 1398 bp, respectively. In silico analysis showed that their transcriptional region structures, as well as their amino acid sequences, physicochemical characteristics, motifs, conserved domains, and three-dimensional structures of their encoded proteins are highly consistent with their counterparts in the species of Bovidae. The three Smad proteins are all hydrophilic without the signal peptides and transmembrane regions. Each of them has an MH1 domain and an MH2 domain. A nuclear localization sequence was found in the MH1 domain of buffalo Smads 1 and 5. Prediction showed that the function of the three Smads is mainly protein binding, and they can interact with BMPs and their receptors. The three genes were expressed in all 10 buffalo tissues assayed, and their expression in the mammary gland, gonad, and spleen was relatively high. The results here indicate that the three buffalo Smads may be involved in the transcriptional regulation of genes in a variety of tissues.


Assuntos
Búfalos/genética , Proteínas Smad/genética , Animais , Sequência Conservada , Feminino , Gônadas/metabolismo , Humanos , Masculino , Glândulas Mamárias Humanas/metabolismo , Ligação Proteica , Domínios Proteicos , Proteínas Smad/química , Proteínas Smad/metabolismo , Baço/metabolismo
2.
Eur J Med Chem ; 223: 113660, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34246853

RESUMO

TGFß is crucial for the homeostasis of epithelial and neural tissues, wound repair, and regulating immune responses. Its dysregulation is associated with a vast number of diseases, of which modifying the tumor microenvironment is one of vital clinical interest. Despite various attempts, there is still no FDA-approved therapy to inhibit the TGFß pathway. Major mainstream approaches involve impairment of the TGFß pathway via inhibition of the TGFßRI kinase. With the purpose to identify non-receptor kinase-based inhibitors to impair TGFß signaling, an in-house chemical library was enriched, through a computational study, to eliminate TGFßRI kinase activity. Selected compounds were screened against a cell line engineered with a firefly luciferase gene under TGFß-Smad-dependent transcriptional control. Results indicated moderate potency for a molecule with phthalazine core against TGFß-Smad signaling. A series of phthalazine compounds were synthesized and evaluated for potency. The most promising compound (10p) exhibited an IC50 of 0.11 ± 0.02 µM and was confirmed to be non-cytotoxic up to 12 µM, with a selectivity index of approximately 112-fold. Simultaneously, 10p was confirmed to reduce the Smad phosphorylation using Western blot without exhibiting inhibition on the TGFßRI enzyme. This study identified a novel small-molecule scaffold that targets the TGFß pathway via a non-receptor-kinase mechanism.


Assuntos
Ftalazinas/química , Fator de Crescimento Transformador beta/antagonistas & inibidores , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Fosforilação/efeitos dos fármacos , Ftalazinas/metabolismo , Ftalazinas/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/química , Proteínas Smad/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade , Fator de Crescimento Transformador beta/metabolismo
3.
Front Immunol ; 12: 613438, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054795

RESUMO

Transforming growth factor-ß (TGF-ß) isoforms are secreted as inactive complexes formed through non-covalent interactions between bioactive TGF-ß entities and their N-terminal pro-domains called latency-associated peptides (LAP). Extracellular activation of latent TGF-ß within this complex is a crucial step in the regulation of TGF-ß activity for tissue homeostasis and immune cell function. We previously showed that the matrix glycoprotein Tenascin-X (TN-X) interacted with the small latent TGF-ß complex and triggered the activation of the latent cytokine into a bioactive TGF-ß. This activation most likely occurs through a conformational change within the latent TGF-ß complex and requires the C-terminal fibrinogen-like (FBG) domain of the glycoprotein. As the FBG-like domain is highly conserved among the Tenascin family members, we hypothesized that Tenascin-C (TN-C), Tenascin-R (TN-R) and Tenascin-W (TN-W) might share with TN-X the ability to regulate TGF-ß bioavailability through their C-terminal domain. Here, we demonstrate that purified recombinant full-length Tenascins associate with the small latent TGF-ß complex through their FBG-like domains. This association promotes activation of the latent cytokine and subsequent TGF-ß cell responses in mammary epithelial cells, such as cytostasis and epithelial-to-mesenchymal transition (EMT). Considering the pleiotropic role of TGF-ß in numerous physiological and pathological contexts, our data indicate a novel common function for the Tenascin family in the regulation of tissue homeostasis under healthy and pathological conditions.


Assuntos
Tenascina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Homeostase , Humanos , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Proteínas Smad/química , Proteínas Smad/metabolismo , Relação Estrutura-Atividade , Tenascina/química , Tenascina/genética , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/genética
4.
Parasit Vectors ; 12(1): 609, 2019 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-31881930

RESUMO

BACKGROUND: The Smad proteins function in TGF-ß signalling transduction. In the model nematode Caenorhabditis elegans, the co-Smad, DAF-3 mediates R-Smads and performs a central role in DAF-7 signal transduction, regulating dauer formation and reproductive processes. Considering the divergent evolutionary patterns of the DAF-7 signalling pathway in parasitic nematodes, it is meaningful to explore the structure and function of DAF-3 in parasitic nematodes, such as Haemonchus contortus. METHODS: A daf-3 gene (Hc-daf-3) and its predicted product (Hc-DAF-3) were identified from H. contortus and characterised using integrated genomic and genetic approaches. In addition to immunohistochemistry employed to localise Hc-DAF-3 within adult worm sections, real-time PCR was conducted to assess the transcriptional profiles in different developmental stages of H. contortus and RNA interference (RNAi) was performed in vitro to assess the functional importance of Hc-daf-3 in the development of H. contortus. RESULTS: Hc-DAF-3 sequences predicted from Hc-daf-3 displayed typical features of the co-Smad subfamily. The native Hc-DAF-3 was localised to the gonad and cuticle of adult parasites. In addition, Hc-daf-3 was transcribed in all developmental stages studied, with a higher level in the third-stage larvae (L3) and adult females. Moreover, silencing Hc-daf-3 by RNAi retarded L4 development. CONCLUSION: The findings of the present study demonstrated an important role of Hc-DAF-3 in the development of H. contortus larvae.


Assuntos
Haemonchus/crescimento & desenvolvimento , Haemonchus/metabolismo , Proteínas de Helminto/metabolismo , Proteínas Smad/metabolismo , Sequência de Aminoácidos , Animais , Evolução Molecular , Feminino , Haemonchus/genética , Proteínas de Helminto/química , Proteínas de Helminto/genética , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Masculino , Filogenia , Alinhamento de Sequência , Transdução de Sinais , Proteínas Smad/química , Proteínas Smad/genética
5.
Database (Oxford) ; 20192019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30943284

RESUMO

Since proteins evolve by divergent evolution, proteins with distant homology to each other may or may not bear similar functions. Improved computational approaches are required to recognize distant homologues that are functionally similar. One of the methods of assigning function to sequences is to use profiles derived from sequences of known structure. We describe an update of the Genomic Distribution of protein structural domain Superfamilies (GenDiS) database, namely GenDiS+, which provides a projection of SCOP superfamily members on the sequence space (NR database, NCBI). The sequences are validated using structure-based sequence alignment profiles and domain and full-length sequence alignments. GenDiS+ is a `tour de force' for detecting homologues within around 160 000 taxonomic identifiers, starting from nearly 11 000 domains of known structure. Features, like full-sequence alignment and phylogeny, domain sequence alignment and phylogeny, list of associated structural and sequence domains with strength of interactions, links to databases like Pfam, UniProt and ModBase and list of sequences with a PDB structure, are provided.


Assuntos
Bases de Dados de Proteínas , Família Multigênica , Homologia de Sequência de Aminoácidos , Sequência de Aminoácidos , Animais , Camundongos , Domínios Proteicos , Proteínas Smad/química
6.
Molecules ; 23(8)2018 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-30103395

RESUMO

Hepatic fibrosis is the wound-healing process of chronic hepatic disease that leads to the end-stage of hepatocellular carcinoma and demolition of hepatic structures. Epithelial⁻mesenchymal transition (EMT) has been identified to phenotypic conversion of the epithelium to mesenchymal phenotype that occurred during fibrosis. Smad decoy oligodeoxynucleotide (ODN) is a synthetic DNA fragment containing a complementary sequence of Smad transcription factor. Thus, this study evaluated the antifibrotic effects of Smad decoy ODN on carbon tetrachloride (CCl4)-induced hepatic fibrosis in mice. As shown in histological results, CCl4 treatment triggered hepatic fibrosis and increased Smad expression. On the contrary, Smad decoy ODN administration suppressed fibrogenesis and EMT process. The expression of Smad signaling and EMT-associated protein was markedly decreased in Smad decoy ODN-treated mice compared with CCl4-injured mice. In conclusion, these data indicate the practicability of Smad decoy ODN administration for preventing hepatic fibrosis and EMT processes.


Assuntos
Cirrose Hepática/patologia , Oligodesoxirribonucleotídeos/farmacologia , Proteínas Smad/genética , Animais , Sequência de Bases , Tetracloreto de Carbono/efeitos adversos , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Matriz Extracelular/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Masculino , Camundongos , Oligodesoxirribonucleotídeos/síntese química , Oligodesoxirribonucleotídeos/química , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/química , Proteínas Smad/metabolismo , Transfecção
7.
Proc Natl Acad Sci U S A ; 114(50): 13206-13211, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29180412

RESUMO

TGF-ß/BMP (bone morphogenetic protein) signaling pathways play conserved roles in controlling embryonic development, tissue homeostasis, and stem cell regulation. Inhibitory Smads (I-Smads) have been shown to negatively regulate TGF-ß/BMP signaling by primarily targeting the type I receptors for ubiquitination and turnover. However, little is known about how I-Smads access the membrane to execute their functions. Here we show that Dad, the Drosophila I-Smad, associates with the cellular membrane via palmitoylation, thereby targeting the BMP type I receptor for ubiquitination. By performing systematic biochemistry assays, we characterized the specific cysteine (Cys556) essential for Dad palmitoylation and membrane association. Moreover, we demonstrate that dHIP14, a Drosophila palmitoyl acyl-transferase, catalyzes Dad palmitoylation, thereby inhibiting efficient BMP signaling. Thus, our findings uncover a modification of the inhibitory Smads that controls TGF-ß/BMP signaling activity.


Assuntos
Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Proteínas Smad/metabolismo , Aciltransferases/metabolismo , Animais , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/metabolismo , Drosophila , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Lipoilação , Ligação Proteica , Transporte Proteico , Proteínas Smad/química , Proteínas Smad/genética , Fator de Crescimento Transformador beta/metabolismo
8.
Open Biol ; 7(8)2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28814648

RESUMO

Bone morphogenetic proteins (BMPs) play vital roles in regulating stem cell maintenance, differentiation and embryonic development. Intracellularly, BMP signalling is mediated by Smad proteins, which are regulated post-transcriptionally through reversible phosphorylation and ubiquitination. ZC4H2 is a small nuclear protein associated with intellectual disability and neural development in humans. Here, we report that ZC4H2 is highly expressed in the developing neural system and is involved in neural patterning and BMP signalling in Xenopus Knockdown of ZC4H2 led to expansion of the expression of the pan neural plate marker Sox2 in Xenopus embryos. In mammalian cells, ZC4H2 promotes BMP signalling and is involved in BMP regulated myogenic and osteogenic differentiation of mouse myoblast cells. Mechanistically, ZC4H2 binds and stabilizes Smad1 and Smad5 proteins through reducing their association with the Smurf ubiquitin ligases and thus their ubiquitination. We also found that a group of ZC4H2 mutations, which have been isolated in patients with intellectual disorders, showed weaker Smad-stabilizing activity, suggesting that the ZC4H2-Smad interaction might contribute to proper neural development in humans.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Smad/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/crescimento & desenvolvimento , Animais , Padronização Corporal , Proteínas de Transporte/genética , Diferenciação Celular , Linhagem Celular , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Desenvolvimento Muscular , Proteínas Nucleares/genética , Osteogênese , Estabilidade Proteica , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Proteínas Smad/química , Proteína Smad1/química , Proteína Smad1/metabolismo , Proteína Smad5/química , Proteína Smad5/metabolismo , Xenopus/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
9.
Sci Rep ; 7(1): 8778, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-28821740

RESUMO

Elaborate regulatory networks of the Bone Morphogenetic Protein (BMP) pathways ensure precise signalling outcome during cell differentiation and tissue homeostasis. Here, we identified IRS4 as a novel regulator of BMP signal transduction and provide molecular insights how it integrates into the signalling pathway. We found that IRS4 interacts with the BMP receptor BMPRII and specifically targets Smad1 for proteasomal degradation consequently leading to repressed BMP/Smad signalling in C2C12 myoblasts while concomitantly activating the PI3K/Akt axis. IRS4 is present in human and primary mouse myoblasts, the expression increases during myogenic differentiation but is downregulated upon final commitment coinciding with Myogenin expression. Functionally, IRS4 promotes myogenesis in C2C12 cells, while IRS4 knockdown inhibits differentiation of myoblasts. We propose that IRS4 is particularly critical in the myoblast stage to serve as a molecular switch between BMP/Smad and Akt signalling and to thereby control cell commitment. These findings provide profound understanding of the role of BMP signalling in early myogenic differentiation and open new ways for targeting the BMP pathway in muscle regeneration.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/genética , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Animais , Sítios de Ligação , Biomarcadores , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/química , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Proteínas Morfogenéticas Ósseas/química , Linhagem Celular , Membrana Celular/metabolismo , Técnicas de Silenciamento de Genes , Proteínas Substratos do Receptor de Insulina/química , Ligantes , Camundongos , Modelos Biológicos , Desenvolvimento Muscular , Mioblastos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteólise , Proteínas Proto-Oncogênicas c-akt/química , Ratos , Proteínas Smad/química , Ubiquitinação
10.
Fish Shellfish Immunol ; 67: 129-140, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28546027

RESUMO

The proteins of Smad family are critical components of the TGF-ß superfamily signal pathway. In this paper, we cloned two intracellular mediators of TGF-ß signaling, Smad3 and Smad5, from the pearl mussel Hyriopsis cumingii. The full length cDNA of HcSmad3 and HcSmad5 were 2052 bp and 1908 bp and encoded two polypeptides of 418 and 461amino acid residues, respectively. The deduced amino acid of HcSmad3 and HcSmad5 possessed two putative conserved domains, MH1 and MH2, a conserved phosphorylation motif SSXS at the carboxyl-terminal. The two Smad genes were detected muscle, mantle, hepatopancreas and gill, but with a very low level in heamocytes. The transcripts of Smad3 and Smad5 were up-regulated in hemocytes and hepatopancreas after A. hydrophila and PGN stimulation. However, the expression of Smad3 and Smad5 were only up-regulated in hepatopancreas after A. hydrophila stimulation. The transcripts of Smad3 and Smad5 had a slight change in hepatopancreas after PGN stimulation. The transcripts of HcSmad3 showed very little increase and HcSmad5 mRNA significantly up-regulated after wounding.


Assuntos
Imunidade Inata/genética , Proteínas Smad/genética , Proteínas Smad/imunologia , Unionidae/genética , Unionidae/imunologia , Cicatrização/imunologia , Aeromonas hydrophila/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Filogenia , Alinhamento de Sequência , Proteínas Smad/química
11.
Artigo em Inglês | MEDLINE | ID: mdl-27920040

RESUMO

Inhibitory Smads (I-Smads) have conserved carboxy-terminal MH2 domains but highly divergent amino-terminal regions when compared with receptor-regulated Smads (R-Smads) and common-partner Smads (co-Smads). Smad6 preferentially inhibits Smad signaling initiated by the bone morphogenetic protein (BMP) type I receptors ALK-3 and ALK-6, whereas Smad7 inhibits both transforming growth factor ß (TGF-ß)- and BMP-induced Smad signaling. I-Smads also regulate some non-Smad signaling pathways. Here, we discuss the vertebrate I-Smads, their roles as inhibitors of Smad activation and regulators of receptor stability, as scaffolds for non-Smad signaling, and their possible roles in the nucleus. We also discuss the posttranslational modification of I-Smads, including phosphorylation, ubiquitylation, acetylation, and methylation.


Assuntos
Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Conformação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Smad/química
12.
Sci Rep ; 6: 35483, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27759037

RESUMO

Transforming growth factor ß (TGF-ß) signaling facilitates tumor development during the advanced stages of tumorigenesis, but induces cell-cycle arrest for tumor suppression during the early stages. However, the mechanism of functional switching of TGF-ß is still unknown, and it is unclear whether inhibition of TGF-ß signaling results amelioration or exacerbation of cancers. Here we show that the tumor suppressor p53 cooperates with Smad proteins, which are TGF-ß signal transducers, to selectively activate plasminogen activator inhibitor type-1 (PAI-1) transcription. p53 forms a complex with Smad2/3 in the PAI-1 promoter to recruit histone acetyltransferase CREB-binding protein (CBP) and enhance histone H3 acetylation, resulting in transcriptional activation of the PAI-1 gene. Importantly, p53 is required for TGF-ß-induced cytostasis and PAI-1 is involved in the cytostatic activity of TGF-ß in several cell lines. Our results suggest that p53 enhances TGF-ß-induced cytostatic effects by activating PAI-1 transcription, and the functional switching of TGF-ß is partially caused by p53 mutation or p53 inactivation during cancer progression. It is expected that these findings will contribute to optimization of TGF-ß-targeting therapies for cancer.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas , Proteínas Smad/metabolismo , Ativação Transcricional , Fator de Crescimento Transformador beta/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular , Humanos , Complexos Multiproteicos/metabolismo , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Elementos de Resposta , Sialoglicoproteínas/metabolismo , Transdução de Sinais , Proteínas Smad/química , Proteína Supressora de Tumor p53/química
13.
Int J Cardiol ; 214: 246-53, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27077543

RESUMO

BACKGROUND: CXXC-type zinc-finger protein CXXC5 has been reported to be associated with the development of cardiovascular disease. Recently, through signaling pathway screening we found that CXXC5 activated Tgfß and myocardial differentiation signaling pathways simultaneously. Although the physiological and pathological function of CXXC5 in many organs has been well elucidated, its function in heart remains unclear. METHODS AND RESULTS: Here, we found that zebrafish CXXC5 and SMAD were interacting through ZF-CXXC and MH1 domain. Over-expression of CXXC5 in cardiomyocyte increased the luciferase report activity of Tgfß signaling pathway. Spatiotemporal expression profile of cxxc5 showed that it consistently expressed during cardiogenesis. Knockdown of cxxc5 in zebrafish displayed looping defects, cardiac dysplasia, pericardial edema, and decreased contraction ability, accompanied with down-regulation of members referring to cardiac looping downstream genes of Tgfß signaling pathway, such as nkx2.5, hand2, and has2. Co-injection of hand2 mRNA with cxxc5 morpholino rescued the cardiac looping detects. CONCLUSION: Our study is the first to provide an in vivo evidence for cxxc5 regulating heart development and cardiac looping via Tgfß related signaling pathway. This finding suggested that CXXC5 may serve as a possible marker that has potential diagnostic and prognostic value in fetus with congenital heart disease.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA/química , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Ligação Proteica , Ratos , Transdução de Sinais , Proteínas Smad/química , Proteínas de Peixe-Zebra/química
14.
J Pept Sci ; 21(11): 826-32, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26435515

RESUMO

The transforming growth factor-ß/bone morphogenic protein/Smad signaling pathway has been raised as a new and promising therapeutic target of heterotopic ossification, which is mediated by recruitment of transcription coactivator Yes-associated protein (YAP) to Smad. Here, we described a successful integration of computational modeling and experimental assay to rationally design novel peptide aptamers to disrupt YAP-Smad interaction by targeting YAP WW1 domain. In the protocol, a computational genetic evolution strategy was used to improve a population of potential YAP WW1-binding peptides generated from the YAP-recognition site in Smad protein, from which several promising peptides were selected and their affinities toward YAP WW1 domain were determined using binding assay. In addition, a high-activity peptide was further optimized based on its complex structure with YAP WW1 domain to derive a number of derivative peptides with higher binding potency to the domain. We also found that a strong YAP WW1 binder should have a negatively charged N-terminus, a positively charged C-terminus and a nonpolar core to match the electrostatic distribution pattern in peptide-binding pocket of YAP WW1 domain, which may also form additional nonbonded interactions such as hydrogen bond, salt bridge and π-π stacking to confer stability and specificity for the domain-peptide recognition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Desenho de Fármacos , Modelos Moleculares , Oligopeptídeos/química , Fosfoproteínas/antagonistas & inibidores , Proteínas Smad/química , Fator de Crescimento Transformador beta/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/química , Proteínas Morfogenéticas Ósseas/metabolismo , Biologia Computacional , Transferência de Energia , Evolução Molecular , Humanos , Cinética , Simulação de Dinâmica Molecular , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Quantitativa Estrutura-Atividade , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/genética , Proteínas Smad/metabolismo , Proteínas Smad/farmacologia , Eletricidade Estática , Fatores de Transcrição , Fator de Crescimento Transformador beta/química , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Sinalização YAP
15.
Trends Biochem Sci ; 40(6): 296-308, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25935112

RESUMO

Smad transcription factors are central to the signal transduction pathway that mediates the numerous effects of the transforming growth factor ß (TGF-ß) superfamily of cytokines in metazoan embryo development as well as in adult tissue regeneration and homeostasis. Although Smad proteins are conserved, recent genome-sequencing projects have revealed their sequence variation in metazoan evolution, human polymorphisms, and cancer. Structural studies of Smads bound to partner proteins and target DNA provide a framework for understanding the significance of these evolutionary and pathologic sequence variations. We synthesize the extant mutational and structural data to suggest how genetic variation in Smads may affect the structure, regulation, and function of these proteins. We also present a web application that compares Smad sequences and displays Smad protein structures and their disease-associated variants.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas Smad/química , Transcrição Gênica , Fator de Crescimento Transformador beta/química , Proteínas de Ligação a DNA/química , Desenvolvimento Embrionário/genética , Humanos , Mutação , Regeneração , Transdução de Sinais , Proteínas Smad/genética , Relação Estrutura-Atividade , Fator de Crescimento Transformador beta/genética
16.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 844-53, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849395

RESUMO

Gene-expression changes observed in Drosophila embryos after inducing the transcription factor Tramtrack led to the identification of the protein Expansion. Expansion contains an N-terminal domain similar in sequence to the MH2 domain characteristic of Smad proteins, which are the central mediators of the effects of the TGF-ß signalling pathway. Apart from Smads and Expansion, no other type of protein belonging to the known kingdoms of life contains MH2 domains. To compare the Expansion and Smad MH2 domains, the crystal structure of the Expansion domain was determined at 1.6 Šresolution, the first structure of a non-Smad MH2 domain to be characterized to date. The structure displays the main features of the canonical MH2 fold with two main differences: the addition of an α-helical region and the remodelling of a protein-interaction site that is conserved in the MH2 domain of Smads. Owing to these differences, to the new domain was referred to as Nα-MH2. Despite the presence of the Nα-MH2 domain, Expansion does not participate in TGF-ß signalling; instead, it is required for other activities specific to the protostome phyla. Based on the structural similarities to the MH2 fold, it is proposed that the Nα-MH2 domain should be classified as a new member of the Smad/FHA superfamily.


Assuntos
Proteínas de Drosophila/química , Drosophila/química , Proteínas Smad/química , Proteína Smad2/química , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mapas de Interação de Proteínas , Estrutura Terciária de Proteína , Alinhamento de Sequência , Transdução de Sinais , Proteínas Smad/metabolismo , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
17.
Mol Biol Rep ; 42(1): 289-302, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25280542

RESUMO

Elevation of egg performance is vital to goose farming. Many poultry scientists are seeking for efficient molecular genetic markers associated with egg yield. In this study, mRNA differential display was adopted to investigate gene expression profiling in the follicular development of goose. For the first time, a novel SMAD family protein SMAD9 (EST CJ111007) was found to be involved in follicular initiation and used to be a candidate gene. Functional regions analysis of Smad9 indicated that SMAD9 protein is highly conserved in MH1 and MH2 domains, and the connection area is highly variable region. 6 pairs of primers (p1-p6) were designed to detect the SNPs of Smad9 by PCR-SSCP method. The results revealed that polymorphisms were discovered in the PCR products amplified with P1 primers in exon1 and P3 primers in intron2. In Smad9 exon1, 5 genotypes were found: FK, FF, JJ, JK and KK, including 2 SNPs: 243 bp G → A, 309 bp T → G, the mutations did not result in amino acid mutations; In intron2, 3 genotypes were found: AA, BB and AB, only 1 SNP (C → T). The annual egg yield of FK genotype geese or allele gene A in intron2 are significantly more than those of other genotypes on the average (p < 0.05). Taken together, it is suggested that Smad9 is a promising candidate gene affecting egg performance in goose.


Assuntos
Éxons/genética , Gansos/genética , Mutação/genética , Folículo Ovariano/crescimento & desenvolvimento , Óvulo/metabolismo , Proteínas Smad/genética , Alelos , Motivos de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Eletroforese em Gel de Ágar , Feminino , Perfilação da Expressão Gênica , Desequilíbrio de Ligação/genética , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único/genética , Polimorfismo Conformacional de Fita Simples , Estrutura Secundária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Proteínas Smad/química
19.
J Biol Chem ; 288(1): 79-88, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23150675

RESUMO

Bone morphogenetic proteins (BMPs) signaling essentially regulates a wide range of biological responses. Although multiple regulators at different layers of the receptor-effectors axis have been identified, the mechanisms of homeostatic BMP signaling remain vague. Herein we demonstrated that myotubularin-related protein 4 (MTMR4), a FYVE domain-containing dual-specificity protein phosphatase (DUSP), preferentially associated with and dephosphorylated the activated R-Smads in cytoplasm, which is a critical checkpoint in BMP signal transduction. Therefore, transcriptional activation by BMPs was tightly controlled by the expression level and the intrinsic phosphatase activity of MTMR4. More profoundly, ectopic expression of MTMR4 or its Drosophila homolog CG3632 genetically interacted with BMP/Dpp signaling axis in regulation of the vein development of Drosophila wings. By doing so, MTMR4 could interact with and dephosphorylate Mothers against Decapentaplegic (Mad), the sole R-Smad in Drosophila BMP pathway, and hence affected the target genes expression of Mad. In conclusion, this study has suggested that MTMR4 is a necessary negative modulator for the homeostasis of BMP/Dpp signaling.


Assuntos
Drosophila melanogaster/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Smad/química , Animais , Cruzamentos Genéticos , Citoplasma/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Feminino , Genótipo , Células HEK293 , Células HeLa , Células Hep G2 , Homeostase , Humanos , Masculino , Fosforilação , Ligação Proteica , Interferência de RNA , Transdução de Sinais , Proteínas Smad/metabolismo
20.
Interdiscip Sci ; 4(2): 103-15, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22843233

RESUMO

Functional alteration in SMAD proteins leads to dis-regulation of its mechanism results in possibilities of high risk diseases like fibrosis, cancer, juvenile polyposis etc. Studying single nucleotide polymorphism (SNP) in SMAD genes helps understand the malfunction of these proteins. In this study, we focused on deleterious effects of nsSNPs in both structural and functional level using publically available bioinformatics tools. We have mainly focused on identifying deleterious nsSNPs in both structural and functional level in SMAD genes by using SIFT, PolyPhen, SNPs&GO, I-Mutant 3.0, MUpro and PANTHER. Structure analysis was carried out with the major mutation that occurred in the native protein coded by SMAD genes and its amino acid positions (R358W, K306S, R310G, S433R and R361C). SRide was used to check the stability of the native and mutant modelled proteins. In addition, we used MAPPER to identify SNPs present in transcription factor binding sites. These findings demonstrate that the in silico approaches can be used efficiently to identify potential candidate SNPs in large scale analysis.


Assuntos
Biologia Computacional/métodos , Família Multigênica/genética , Mutação Puntual/genética , Proteínas Smad/genética , Substituição de Aminoácidos/genética , Sítios de Ligação , Bases de Dados Genéticas , Bases de Dados de Proteínas , Humanos , Modelos Moleculares , Anotação de Sequência Molecular , Proteínas Mutantes/química , Polimorfismo de Nucleotídeo Único/genética , Estrutura Secundária de Proteína , Proteínas Smad/química , Software , Termodinâmica , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...