Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(21)2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37787089

RESUMO

BMP signaling is crucial to blood vessel formation and function, but how pathway components regulate vascular development is not well-understood. Here, we find that inhibitory SMAD6 functions in endothelial cells to negatively regulate ALK1-mediated responses, and it is required to prevent vessel dysmorphogenesis and hemorrhage in the embryonic liver vasculature. Reduced Alk1 gene dosage rescued embryonic hepatic hemorrhage and microvascular capillarization induced by Smad6 deletion in endothelial cells in vivo. At the cellular level, co-depletion of Smad6 and Alk1 rescued the destabilized junctions and impaired barrier function of endothelial cells depleted for SMAD6 alone. Mechanistically, blockade of actomyosin contractility or increased PI3K signaling rescued endothelial junction defects induced by SMAD6 loss. Thus, SMAD6 normally modulates ALK1 function in endothelial cells to regulate PI3K signaling and contractility, and SMAD6 loss increases signaling through ALK1 that disrupts endothelial cell junctions. ALK1 loss-of-function also disrupts vascular development and function, indicating that balanced ALK1 signaling is crucial for proper vascular development and identifying ALK1 as a 'Goldilocks' pathway in vascular biology that requires a certain signaling amplitude, regulated by SMAD6, to function properly.


Assuntos
Junções Aderentes , Células Endoteliais , Humanos , Junções Aderentes/metabolismo , Células Endoteliais/metabolismo , Hemorragia/metabolismo , Fígado/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Smad6/metabolismo
2.
Development ; 150(11)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37272529

RESUMO

The mechanism of pattern formation during limb muscle development remains poorly understood. The canonical view holds that naïve limb muscle progenitor cells (MPCs) invade a pre-established pattern of muscle connective tissue, thereby forming individual muscles. Here, we show that early murine embryonic limb MPCs highly accumulate pSMAD1/5/9, demonstrating active signaling of bone morphogenetic proteins (BMP) in these cells. Overexpression of inhibitory human SMAD6 (huSMAD6) in limb MPCs abrogated BMP signaling, impaired their migration and proliferation, and accelerated myogenic lineage progression. Fewer primary myofibers developed, causing an aberrant proximodistal muscle pattern. Patterning was not disturbed when huSMAD6 was overexpressed in differentiated muscle, implying that the proximodistal muscle pattern depends on BMP-mediated expansion of MPCs before their differentiation. We show that limb MPCs differentially express Hox genes, and Hox-expressing MPCs displayed active BMP signaling. huSMAD6 overexpression caused loss of HOXA11 in early limb MPCs. In conclusion, our data show that BMP signaling controls expansion of embryonic limb MPCs as a prerequisite for establishing the proximodistal muscle pattern, a process that involves expression of Hox genes.


Assuntos
Proteínas Morfogenéticas Ósseas , Músculo Esquelético , Animais , Humanos , Camundongos , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/fisiologia , Genes Homeobox , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Proteína Smad6/metabolismo
3.
EMBO J ; 40(7): e106151, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33616251

RESUMO

Interleukin (IL)-33 cytokine plays a critical role in allergic diseases and cancer. IL-33 also has a nuclear localization signal. However, the nuclear function of IL-33 and its impact on cancer is unknown. Here, we demonstrate that nuclear IL-33-mediated activation of SMAD signaling pathway in epithelial cells is essential for cancer development in chronic inflammation. Using RNA and ChIP sequencing, we found that nuclear IL-33 repressed the expression of an inhibitory SMAD, Smad6, by interacting with its transcription factor, RUNX2. IL-33 was highly expressed in the skin and pancreatic epithelial cells in chronic inflammation, leading to a markedly repressed Smad6 expression as well as dramatically upregulated p-SMAD2/3 and p-SMAD1/5 in the epithelial cells. Blocking TGF-ß/SMAD signaling attenuated the IL-33-induced cell proliferation in vitro and inhibited IL-33-dependent epidermal hyperplasia and skin cancer development in vivo. IL-33 and SMAD signaling were upregulated in human skin cancer, pancreatitis, and pancreatitis-associated pancreatic cancer. Collectively, our findings reveal that nuclear IL-33/SMAD signaling is a cell-autonomous tumor-promoting axis in chronic inflammation, which can be targeted by small-molecule inhibitors for cancer treatment and prevention.


Assuntos
Carcinogênese/metabolismo , Interleucina-33/metabolismo , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Proteína Smad6/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta/metabolismo
4.
J Med Genet ; 61(4): 363-368, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38290823

RESUMO

BACKGROUND: SMAD6 encodes an intracellular inhibitor of the bone morphogenetic protein (BMP) signalling pathway. Until now, rare heterozygous loss-of-function variants in SMAD6 were demonstrated to increase the risk of disparate clinical disorders including cardiovascular disease, craniosynostosis and radioulnar synostosis. Only two unrelated patients harbouring biallelic SMAD6 variants presenting a complex cardiovascular phenotype and facial dysmorphism have been described. CASES: Here, we present the first two patients with craniosynostosis harbouring homozygous SMAD6 variants. The male probands, both born to healthy consanguineous parents, were diagnosed with metopic synostosis and bilateral or unilateral radioulnar synostosis. Additionally, one proband had global developmental delay. Echocardiographic evaluation did not reveal cardiac or outflow tract abnormalities. MOLECULAR ANALYSES: The novel missense (c.[584T>G];[584T>G], p.[(Val195Gly)];[(Val195Gly)]) and missense/splice-site variant (c.[817G>A];[817G>A], r.[(817g>a,817delins[a;817+2_817+228])];[(817g>a,817delins[a;817+2_817+228])], p.[(Glu273Lys,Glu273Serfs*72)];[(Glu273Lys,Glu273Serfs*72)]) both locate in the functional MH1 domain of the protein and have not been reported in gnomAD database. Functional analyses of the variants showed reduced inhibition of BMP signalling or abnormal splicing, respectively, consistent with a hypomorphic mechanism of action. CONCLUSION: Our data expand the spectrum of variants and phenotypic spectrum associated with homozygous variants of SMAD6 to include craniosynostosis.


Assuntos
Craniossinostoses , Rádio (Anatomia)/anormalidades , Sinostose , Ulna/anormalidades , Humanos , Masculino , Craniossinostoses/diagnóstico , Craniossinostoses/genética , Rádio (Anatomia)/metabolismo , Ulna/metabolismo , Mutação de Sentido Incorreto/genética , Proteína Smad6/genética , Proteína Smad6/metabolismo
5.
J Bone Miner Metab ; 42(1): 1-16, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38055109

RESUMO

INTRODUCTION: Osteoarthritis (OA) compromises patients' quality of life and requires further study. Although miR-92a-3p was reported to possess chondroprotective effects, the underlying mechanism requires further clarification. The objectives of this study were to elucidate the mechanism by which miR-92a-3p alleviates OA and to examine the efficacy of shRNA-92a-3p, which was designed based on mature miR-92a-3p. MATERIALS AND METHODS: TargetScan and luciferase reporter assay were used to predict the target of miR-92a-3p. Adipose-derived stem cells (ADSCs) were transfected with miR-92a-3p/miR-NC mimic for the analysis of chondrogenic biomarkers and SMAD proteins. ADSCs and osteoarthritic chondrocytes were transduced with shRNA-92a-3p for the analysis of chondrogenic biomarkers and SMAD proteins. OA was surgically induced in C57BL/6JJcl mice, and ADSCs with/without shRNA-92a-3p transduction were intra-articularly injected for the assessment of cartilage damage. RESULTS: SMAD6 and SMAD7 were predicted as direct targets of miR-92a-3p by TargetScan and luciferase reporter assay. Transfection of the miR-92a-3p mimic resulted in a decrease in SMAD6 and SMAD7 levels and an increase in phospho-SMAD2/3, phospho-SMAD1/5/9, SOX9, collagen type II, and aggrecan levels in ADSCs. Furthermore, shRNA-92a-3p decreased SMAD6 and SMAD7 levels, and increased phospho-SMAD2/3, phospho-SMAD1/5/9, SOX9, collagen type II, and aggrecan levels in ADSCs and osteoarthritic chondrocytes. Additionally, ADSC-shRNA-92a-3p-EVs reduced the rate of decrease of SOX9, collagen type II, and aggrecan in osteoarthritic chondrocytes. In mice with surgically induced OA, shRNA-92a-3p-treated ADSCs alleviated cartilage damage more effectively than nontreated ADSCs. CONCLUSIONS: miR-92a-3p and shRNA-92a-3p exhibit therapeutic effects in treating OA by targeting SMAD6 and SMAD7, thereby enhancing TGF-ß signaling.


Assuntos
MicroRNAs , Osteoartrite , Humanos , Animais , Camundongos , Condrócitos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Colágeno Tipo II/metabolismo , Agrecanas/metabolismo , Qualidade de Vida , Camundongos Endogâmicos C57BL , Osteoartrite/genética , Osteoartrite/terapia , Osteoartrite/metabolismo , Proteínas Smad/metabolismo , Biomarcadores/metabolismo , Luciferases/metabolismo , Luciferases/farmacologia , Proteína Smad6/metabolismo , Proteína Smad6/farmacologia
6.
Reproduction ; 165(3): 269-279, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36534533

RESUMO

In brief: Follicle selection is a key event in monovular species. In this manuscript, we demonstrate the role of SMAD6 in promoting decreased granulosa cell proliferation and follicle growth rate in carriers vs noncarriers of the Trio allele and after vs before follicle deviation. Abstract: Cattle are generally considered a monovular species; however, recently, a bovine high fecundity allele, termed the Trio allele, was discovered. Carriers of Trio have an elevated ovulation rate (3-5), while half-sibling noncarriers are monovular. Carriers of the Trio allele have overexpression in granulosa cells of SMAD6, an inhibitor of oocyte-derived regulators of granulosa cell proliferation and differentiation. In experiment 1, follicle size was tracked for each follicle during a follicular wave. Follicle growth rate was greater before vs after follicle deviation in both carriers and noncarriers. Additionally, follicle growth rate was consistently less in carriers vs noncarriers. In experiment 2, we collected granulosa cells from follicles before and after deviation for evaluation of granulosa cell gene expression. Granulosa cell proliferation was less in carriers vs noncarriers and after vs before follicle deviation (decreased expression of cell cycle genes CCNB1 and CCNA2). The decreased granulosa cell proliferation in noncarriers after deviation was associated with increased SMAD6 expression. Similarly, in experiment 3, decreased expression of SMAD6 in granulosa cells of noncarriers cultured in vitro for 60 h was associated with increased expression of cell cycle genes. This suggests that SMAD6 may not just be inhibiting follicle growth rate in carriers of Trio but may also play a role in the decreased follicle growth after deviation in noncarriers. The hypotheses were supported that (1) follicle growth and granulosa cell proliferation decrease after deviation in both carriers and noncarriers and that (2) granulosa cell proliferation is reduced in carriers compared to noncarriers.


Assuntos
Folículo Ovariano , Ovulação , Animais , Bovinos , Feminino , Alelos , Proliferação de Células , Células da Granulosa/metabolismo , Folículo Ovariano/metabolismo , Ovulação/genética , Proteína Smad6/metabolismo
7.
Oral Dis ; 29(8): 3447-3459, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35957556

RESUMO

OBJECTIVES: Increasing evidence indicated circRNAs were involved in stem cells osteogenesis differentiation. Herein, we aimed to clarify the role of hsa-circ-0107593 during the osteogenesis process of human adipose-derived stem cells (hADSCs) and the underlying mechanisms. METHODS: The ring structure of hsa-circ-0107593 was confirmed using RNase R treatment and Sanger sequencing. Nucleoplasmic separation and fluorescence in situ hybridization detected hsa-circ-0107593 distribution. Lentivirus and siRNA were used to modulate the expression of hsa-circ-0107593, and the binding relationship between hsa-circ-0107593 and miR-20a-5p was verified by luciferase assay and RNA immunoprecipitation. We detected the osteogenic activity of hADSCs through alkaline phosphatase staining, alizarin red S staining, real-time polymerase chain reaction (RT-PCR), western blot, and cellular immunofluorescence experiment. In vivo, micro-computed tomography was performed to analyze bone formation around skull defect. RESULTS: RT-PCR results exhibited that hsa-circ-0107593 was downregulated while miR-20a-5p was upregulated during hADSCs osteogenesis. In vivo and in vitro experiments results indicated that knocking down hsa-circ-0107593 promoted the osteogenic differentiation of hADSCs, while overexpression of hsa-circ-0107593 showed an inhibitory effect on hADSCs osteogenic differentiation. In vitro experiment results showed hsa-circ-0107593 acted as a hADSCs osteogenic differentiation negative factor for it inhibited the suppressing effect of miR-20a-5p on SMAD6. CONCLUSION: Knocking down hsa-circ-0107593 acts as a positive factor of the osteogenic differentiation of hADSCs via miR-20a-5p/SMAD6 signaling.


Assuntos
MicroRNAs , Osteogênese , Humanos , Osteogênese/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Regulação para Baixo , Hibridização in Situ Fluorescente , Microtomografia por Raio-X , Diferenciação Celular/genética , Proliferação de Células/genética , Proteína Smad6/genética , Proteína Smad6/metabolismo
8.
Oral Dis ; 29(8): 3433-3446, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35917232

RESUMO

OBJECTIVES: Chondrogenic differentiation of human dental pulp stem cells (hDPSCs) is highly promising for cartilage repair. The specific mechanism, however, still needs to be explicated. MATERIALS AND METHODS: In this study, we isolated hDPSCs and transfected cells with lentiviruses containing an over-expression, knock-down, or negative control of miR-20a-5p. Three-D pellet cultures of hDPSCs were used for the chondrogenic induction. Following the pellet culture period, chondrogenesis was assessed by histological and immunohistochemical analysis and expression of chondrogenic-related genes. Dual-luciferase report assay was performed to determine potential targeted genes of miR-20a-5p, and the phosphorylation levels of P65 and IκBα were explored. Animal experiments were performed to determine the effect of miR-20a-5p on cartilage regeneration. RESULTS: miR-20a-5p was showed to repress the expression of SMAD6 to inhibit the chondrogenic differentiation of hDPSCs. Accordingly, the knock-down of miR-20a-5p promoted cartilage regeneration in the osteochondral defects of rats. Mechanically, it is indicated that NF-κB signaling is the potential down-stream network of miR-20a-5p/Smad6 crosstalk during chondrogenic differentiation. CONCLUSIONS: miR-20a-5p could target SMAD6 to activate NF-κB signaling pathway, and thus inhibit chondrogenesis of hDPSCs, which provided promising therapeutic target for cartilage defects clinically.


Assuntos
MicroRNAs , Humanos , Ratos , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Condrogênese/genética , NF-kappa B/metabolismo , Diferenciação Celular/genética , Cartilagem/metabolismo , Proteína Smad6/metabolismo
9.
J Biol Chem ; 296: 100496, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33667543

RESUMO

Bone morphogenetic proteins (BMPs) secreted by a variety of cell types are known to play essential roles in cell differentiation and matrix formation in the bone, cartilage, muscle, blood vessel, and neuronal tissue. BMPs activate intracellular effectors via C-terminal phosphorylation of Smad1, Smad5, and Smad9, which relay the signaling by forming a complex with Smad4 and translocate to the nucleus for transcriptional activation. Smad6 inhibits BMP signaling through diverse mechanisms operative at the membrane, cytosolic, and nuclear levels. However, the mechanistic underpinnings of Smad6 functional diversity remain unclear. Here, using a biochemical approach and cell differentiation systems, we report a cytosolic mechanism of action for Smad6 that requires arginine methylation at arginine 81 (R81) and functions through association with Smad1 and interference with the formation of Smad1-Smad4 complexes. By mutating the methylated arginine residue, R81, and by silencing the expression of protein arginine methyltransferase 1, we show that protein arginine methyltransferase 1 catalyzes R81 methylation of Smad6 upon BMP treatment, R81 methylation subsequently facilitates Smad6 interaction with the phosphorylated active Smad1, and R81 methylation facilitates Smad6-mediated interruption of Smad1-Smad4 complex formation and nuclear translocation. Furthermore, Smad6 WT but not the methylation-deficient R81A mutant inhibited BMP-responsive transcription, attenuated BMP-mediated osteogenic differentiation, and antagonized BMP-mediated inhibition of cell invasion. Taken together, our results suggest that R81 methylation plays an essential role in Smad6-mediated inhibition of BMP responses.


Assuntos
Arginina/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Osteogênese/fisiologia , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Proteína Smad1/metabolismo , Proteína Smad4/metabolismo , Proteína Smad6/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Metilação , Proteína Smad1/antagonistas & inibidores , Proteína Smad4/antagonistas & inibidores , Proteína Smad6/química
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(5): 588-599, 2022 May 28.
Artigo em Inglês, Zh | MEDLINE | ID: mdl-35753729

RESUMO

OBJECTIVES: Patients with hepatocellular carcinoma (HCC) have poor prognosis due to lack of early diagnosis and effective treatment. Therefore, there is an urgent need to better understand the molecular mechanisms associated with HCC and to identify effective targets for early diagnosis and treatment. This study is to explore the expression and biological role of ceramide synthase 3 (CerS3) in HCC. METHODS: A total of 159 pairs of HCC tissues and adjacent non-tumor tissues were obtained from the patients underwent radical resection in Shenzhen People's Hospital, and the total RNA and proteins from HCC tissues and adjacent non-tumor tissues were obtained. The expression of CerS3 protein and mRNA in HCC was detected by immunohistochemistry, Western blotting and real-time PCR. In vitro experiments, Hep3B cells were divided into a control vector group and a CerS3 vector group, and the cells were transfected with retroviral vector containing control cDNA or CerS3 cDNA, respectively. HCCLM3 cells were divided into a normal control shRNA group and a CerS3 shRNA group, and the cells were transfected with lentiviral vectors containing normal control shRNA or CerS3 shRNA, respectively. MTT, EdU, Transwell and scratch method were used to detect cell proliferation, migration and invasion. RNA sequencing was performed to determine the downstream signal of CerS3. RESULTS: Compared with the corresponding adjacent tissues,the mRNA and protein levels of CerS3 were elevated in the HCC tissues, with significant difference (both P<0.05). The Univariate and multivariate analysis showed that the overall survival rate was significantly correlated with the presence of venous invasion (95% CI 1.8-9.2, P<0.01), TNM stage (95% CI 2.3-5.2, P<0.05), poor histological grade (95% CI 1.4-6.8, P<0.05), and CerS3 (95% CI 1.5-3.9, P<0.05). Furthermore, the high CerS3 expression levels in tumor tissues were significantly associated with shorter overall survival rates compared with the low CerS3 expression (P<0.05). Compared with the vector control group, the Hep3B cell viability, EdU positive cells, and migration and invasion cell numbers in the CerS3 vector group were significantly increased (all P<0.05). Compared with the shRNA normal control group, the HCCLM3 cell viability, EdU positive cells, and numbers of migrating and invasive cells in the CerS3 shRNA group were significantly lower (all P<0.05). The RNA sequencing confirmed that the small mothers against decapentaplegic family member 6 (SMAD6) gene as an oncogenic gene could promote the HCC metastasis. CONCLUSIONS: Clinically, the overexpression of CerS3 is closely related to poor clinical features and poor prognosis. Functionally, CerS3 participates in the proliferation, invasion and metastasis of liver cancer cells via activating SMAD6 gene.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Esfingosina N-Aciltransferase/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , DNA Complementar , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Invasividade Neoplásica/genética , Oxirredutases , RNA Mensageiro/genética , RNA Interferente Pequeno , Proteína Smad6/genética , Proteína Smad6/metabolismo
11.
Angiogenesis ; 24(2): 387-398, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33779885

RESUMO

Fluid shear stress provided by blood flow instigates a transition from active blood vessel network expansion during development, to vascular homeostasis and quiescence that is important for mature blood vessel function. Here we show that SMAD6 is required for endothelial cell flow-mediated responses leading to maintenance of vascular homeostasis. Concomitant manipulation of the mechanosensor Notch1 pathway and SMAD6 expression levels revealed that SMAD6 functions downstream of ligand-induced Notch signaling and transcription regulation. Mechanistically, full-length SMAD6 protein was needed to rescue Notch loss-induced flow misalignment. Endothelial cells depleted for SMAD6 had defective barrier function accompanied by upregulation of proliferation-associated genes and down regulation of junction-associated genes. The vascular protocadherin PCDH12 was upregulated by SMAD6 and required for proper flow-mediated endothelial cell alignment, placing it downstream of SMAD6. Thus, SMAD6 is a required transducer of flow-mediated signaling inputs downstream of Notch1 and upstream of PCDH12, as vessels transition from an angiogenic phenotype to maintenance of a homeostatic phenotype.


Assuntos
Homeostase , Células Endoteliais da Veia Umbilical Humana/metabolismo , Mecanotransdução Celular , Receptor Notch1/metabolismo , Proteína Smad6/metabolismo , Circulação Sanguínea , Regulação da Expressão Gênica , Humanos , Protocaderinas/biossíntese , Resistência ao Cisalhamento
12.
Biol Chem ; 402(4): 469-480, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33938174

RESUMO

TGFß signaling is a known pathway to be involved in colorectal cancer (CRC) progression and miRNAs play crucial roles by regulating different components of this pathway. Hence, finding the link between miRNAs and the pathway could be beneficial for CRC therapy. Array data indicated that miR-186-5p is a differentially expressed miRNA in colorectal Tumor/Normal tissues and bioinformatics tools predicted SMAD6/7 (inhibitory SMADs) as bona fide targets of this miRNA. Here, we intended to investigate the regulatory effect of the miR-186-5p expression on TGFß signaling in CRC. Firstly, the miR-186-5p overexpression in HCT116 cells resulted in a significant reduction of SMAD6/7 expression, measured through RT-qPCR. Then, the direct interactions of miR-186-5p with SMAD6/7 3'UTRs were supported through dual luciferase assay. Furthermore, miR-186-5p overexpression suppressed proliferation, cell viability, and migration while, it increased apoptosis in CRC cells, assessed by cell cycle, MTT, scratch and Annexin V/PI apoptosis assays. Consistently, miR-186-5p overexpression resulted in reduced CyclinD1 protein using western blot, and also resulted in increased P21 and decreased c-Myc expression. Overall, these results introduced miR-186-5p as a cell cycle suppressor through downregulation of SMAD6/7 expression. Thus, miR-186-5p might be served as a novel tumor suppressive biomarker and therapeutic target in CRC treatment.


Assuntos
Neoplasias Colorretais/metabolismo , MicroRNAs/metabolismo , Proteína Smad6/genética , Proteína Smad7/genética , Fator de Crescimento Transformador beta/metabolismo , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Neoplasias Colorretais/patologia , Biologia Computacional , Humanos , MicroRNAs/genética , Transdução de Sinais , Proteína Smad6/metabolismo , Proteína Smad7/metabolismo , Células Tumorais Cultivadas
13.
BMC Cancer ; 21(1): 453, 2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33892661

RESUMO

BACKGROUND: Long non-coding RNA (lncRNA) was a vital factor in the progression and initiation of human cancers. This study found a new lncRNA, FGD5-AS1, which can inhibit EMT process, proliferation, and metastasis in vitro and in vivo. METHODS: qRT-PCR was employed to test the expression of lncFGD5-AS1 in 30 gastric cancer patients' cancer tissue and para-cancer tissue. Overexpressed lncFGD5-AS1 cells shown sharply decrease of proliferation, migration, and epithelial-mesenchymal transition (EMT). miR-196a-5p/SMAD6 was confirmed as downstream molecular mechanism of lncFGD5-AS1 by expression correlation analysis and mechanism experiments. In vivo study illustrated overexpression of lncFGD5-AS1 suppression tumor growth. RESULTS: LncFGD5-AS1 served as a ceRNA of miR-196a-5p to release its inhibition on SMAD6, a conventional inhibitor on the BMP pathway. Comparing with normal gastric cancer cells, FGD5-AS1 overexpressed group had fewer migration cells, lower cell viability, and lower EMT transformation rate. Meanwhile, xenografts nude mice injecting with overexpressed-FGD5-AS1 cells also shown smaller tumor weight and volume. CONCLUSION: In conclusion, this research supported the first evidence that FGD5-AS1 suppressed proliferation and metastasis in gastric cancer by regulating miR-196a-5p/SMAD6/BMP axis and suggested a potential therapeutic candidate for gastric cancer.


Assuntos
Transição Epitelial-Mesenquimal , Fatores de Troca do Nucleotídeo Guanina/metabolismo , MicroRNAs/metabolismo , Proteína Smad6/metabolismo , Neoplasias Gástricas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Mucosa Gástrica/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , RNA Longo não Codificante/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia , Carga Tumoral , Ensaio Tumoral de Célula-Tronco
14.
Mol Cell ; 51(1): 5-19, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23747011

RESUMO

Kinase activation and substrate phosphorylation commonly form the backbone of signaling cascades. Bone morphogenetic proteins (BMPs), a subclass of TGF-ß family ligands, induce activation of their signaling effectors, the Smads, through C-terminal phosphorylation by transmembrane receptor kinases. However, the slow kinetics of Smad activation in response to BMP suggests a preceding step in the initiation of BMP signaling. We now show that arginine methylation, which is known to regulate gene expression, yet also modifies some signaling mediators, initiates BMP-induced Smad signaling. BMP-induced receptor complex formation promotes interaction of the methyltransferase PRMT1 with the inhibitory Smad6, resulting in Smad6 methylation and relocalization at the receptor, leading to activation of effector Smads through phosphorylation. PRMT1 is required for BMP-induced biological responses across species, as evidenced by the role of its ortholog Dart1 in BMP signaling during Drosophila wing development. Activation of signaling by arginine methylation may also apply to other signaling pathways.


Assuntos
Arginina/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Metilação , Fosforilação , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/fisiologia , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Proteína Smad6/análise , Proteína Smad6/química , Proteína Smad6/metabolismo
15.
J Struct Biol ; 212(3): 107661, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33166654

RESUMO

Smad6 and Smad7 are classified as inhibitory Smads (I-Smads). They are crucial in the fine-tuning of signals by cytokines of the transforming growth factor-ß (TGF-ß) family. They are negative feedback regulators and principally target the activated type I receptors as well as the activated Smad complexes, but with distinct specificities. Smad7 inhibits Smad signaling from all seven type I receptors of the TGF-ß family, whereas Smad6 preferentially inhibits Smad signaling from the bone morphogenetic protein (BMP) type I receptors, BMPR1A and BMPR1B. The target specificities are attributed to the C-terminal MH2 domain. Notably, Smad7 utilizes two alternative molecular surfaces for its inhibitory function against type I receptors. One is a basic groove composed of the first α-helix and the L3 loop, a structure that is shared with Smad6 and receptor-regulated Smads (R-Smads). The other is a three-finger-like structure (consisting of residues 331-361, 379-387, and the L3 loop) that is unique to Smad7. The underlying structural basis remains to be elucidated in detail. Here, we report the crystal structure of the MH2 domain of mouse Smad7 at 1.9 Å resolution. The three-finger-like structure is stabilized by a network of hydrogen bonds between residues 331-361 and 379-387, thus forming a molecular surface unique to Smad7. Furthermore, we discuss how Smad7 antagonizes the activated Smad complexes composed of R-Smad and Smad4, a common partner Smad.


Assuntos
Transdução de Sinais/fisiologia , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Ligação de Hidrogênio , Camundongos , Conformação Proteica em alfa-Hélice/fisiologia , Domínios Proteicos/fisiologia , Proteína Smad4/metabolismo , Proteína Smad6/metabolismo
16.
J Cell Mol Med ; 24(24): 14539-14548, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33174391

RESUMO

Prostate cancer is the most common malignancy in men in developed countries. In previous study, we identified HNF1B (Hepatocyte Nuclear Factor 1ß) as a downstream effector of Enhancer of zeste homolog 2 (EZH2). HNF1B suppresses EZH2-mediated migration of two prostate cancer cell lines via represses the EMT process by inhibiting SLUG expression. Besides, HNF1B expression inhibits cell proliferation through unknown mechanisms. Here, we demonstrated that HNF1B inhibited the proliferation rate of prostate cancer cells. Overexpression of HNF1B in prostate cancer cells led to the arrest of G1 cell cycle and decreased Cyclin D1 expression. In addition, we re-explored data from ChIP-sequencing (ChIP-seq) and RNA-sequencing (RNA-seq), and demonstrated that HNF1B repressed Cyclin D1 via direct suppression of SMAD6 expression. We also identified CDKN2A as a HNF1B-interacting protein that would contribute to HNF1B-mediated repression of SMAD6 expression. In summary, we provide the novel mechanisms and evidence in support HNF1B as a tumour suppressor gene for prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteína Smad6/genética , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Imuno-Histoquímica , Masculino , Neoplasias da Próstata/patologia , Ligação Proteica , Proteína Smad6/metabolismo
17.
J Cell Mol Med ; 24(5): 2802-2818, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32022446

RESUMO

The replacement of normal endometrial epithelium by fibrotic tissue is the pathological feature of intrauterine adhesion (IUA), which is caused by trauma to the basal layer of the endometrium. COL5A2 is a molecular subtype of collagen V that regulates collagen production in fibrotic tissue. Here, we investigated the roles of Foxf2 and Smad6 in regulating the transcription of COL5A2 and their involvement in the pathogenesis of IUA. Small interference-mediated Foxf2 (si-Foxf2) silencing and pcDNA3.1-mediated Smad6 (pcDNA3.1-Smad6) up-regulation were performed in a TGF-ß1-induced human endometrial stromal cell line (HESC) fibrosis model. Assessment of collagen expression by Western blotting, immunofluorescence and qRT-PCR showed that COL5A2, COL1A1 and FN were significantly down-regulated in response to si-Foxf2 and pcDNA3.1-Smad6. Transfection of lentivirus vector-Foxf2 (LV-Foxf2) and pcDNA3.1-Smad6 into HESCs and qRT-PCR showed that Foxf2 promoted COL5A2 expression and Smad6 inhibited Foxf2-induced COL5A2 expression. Co-immunoprecipitation, chromatin immunoprecipitation and dual-luciferase reporter assays to detect the interaction between Foxf2 and Smad6 and their role in COL5A2 transcription showed that Foxf2 interacted with Smad6 and bond the same promoter region of COL5A2. In a rat IUA model, injection of ADV2-Foxf2-1810 and ADV4-Smad6 into the uterine wall showed that Foxf2 down-regulation and Smad6 up-regulation decreased fibrosis and the expression of COL5A2 and COL1A1, as detected by haematoxylin/eosin, Masson trichrome staining and immunohistochemistry. Taken together, these results suggested that Foxf2 interacted with Smad6 and co-regulated COL5A2 transcription in the pathogenesis of IUA, whereas they played opposite roles in fibrosis.


Assuntos
Colágeno Tipo V/genética , Fatores de Transcrição Forkhead/metabolismo , Proteína Smad6/metabolismo , Aderências Teciduais/genética , Doenças Uterinas/genética , Animais , Ciclo Celular/genética , Linhagem Celular , Proliferação de Células/genética , Colágeno Tipo V/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/genética , Endométrio/metabolismo , Endométrio/patologia , Feminino , Fibrose , Fatores de Transcrição Forkhead/genética , Humanos , Ratos Sprague-Dawley , Proteína Smad6/genética , Células Estromais/metabolismo , Aderências Teciduais/patologia , Transcrição Gênica , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/genética , Doenças Uterinas/patologia
18.
Stroke ; 51(7): 2197-2207, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32486965

RESUMO

BACKGROUND AND PURPOSE: In unruptured brain arteriovenous malformations (bAVMs), microhemorrhage portends a higher risk of future rupture and may represent a transitional state along the continuum of destabilization. Exploration of the molecular and cellular mechanisms of microhemorrhage will provide a possible target for medical treatment to prevent bAVM bleeding. METHODS: We performed RNA sequencing analysis on 34 unruptured bAVM surgical samples. Functional pathway analysis was performed to identify potential signals associated with the microhemorrhagic phenotype. Candidate gene was then investigated in bAVM specimens by immunohistochemical staining. Several functional assays were used to investigate the effects of candidate genes on the phenotypic properties of cultured human umbilical vein endothelial cells. Then, Masson trichrome staining and immunofluorescence staining were used to evaluate the phenotypic and molecular changes in bAVM tissue. RESULTS: Via RNA sequencing, we identified differential gene expression between 18 microhemorrhagic bAVMs and 16 nonmicrohemorrhagic bAVMs. TGFß (transforming growth factor-beta)/BMP (bone morphogenetic protein) signaling was associated with the bAVM microhemorrhage group when SMAD6 (SMAD family member 6) was downregulated. Immunohistochemical staining showed that the vascular endothelium of microhemorrhagic bAVMs exhibited decreased SMAD6 expression. Functional assays revealed that SMAD6 downregulation promoted the formation of endothelial cell tubes with deficient cell-cell junctions and facilitated the acquisition of mesenchymal behavior by endothelial cells. Masson trichrome and immunofluorescence staining demonstrated that mesenchymal phenotype of endothelial cells is promoted in microhemorrhagic bAVMs. CONCLUSIONS: TGFß/BMP signaling mediated by SMAD6 in vascular endothelial cells is associated with microhemorrhagic bAVMs, and mesenchymal behavior of endothelial cells induced by SMAD6 downregulation is related with bAVM microhemorrhage.


Assuntos
Fístula Arteriovenosa/patologia , Hemorragia Cerebral/etiologia , Células Endoteliais/metabolismo , Malformações Arteriovenosas Intracranianas/patologia , Proteína Smad6/metabolismo , Adulto , Fístula Arteriovenosa/complicações , Fístula Arteriovenosa/metabolismo , Hemorragia Cerebral/metabolismo , Regulação para Baixo , Feminino , Humanos , Malformações Arteriovenosas Intracranianas/complicações , Malformações Arteriovenosas Intracranianas/metabolismo , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
19.
J Nanobiotechnology ; 18(1): 68, 2020 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-32375794

RESUMO

BACKGROUND: Enhancing angiogenesis is critical for accelerating wound healing. Application of different types of exosomes (Exos) to promote angiogenesis represents a novel strategy for enhanced wound repair. Saliva is known to accelerate wound healing, but the underlying mechanisms remain unclear. RESULTS: Our results have demonstrated that saliva-derived exosomes (saliva-Exos) induce human umbilical vein endothelial cells (HUVEC) proliferation, migration, and angiogenesis in vitro, and promote cutaneous wound healing in vivo. Further experiments documented that Ubiquitin-conjugating enzyme E2O (UBE2O) is one of the main mRNAs of saliva-Exos, and activation of UBE2O has effects similar to those of saliva-Exos, both in vitro and in vivo. Mechanistically, UBE2O decreases the level of SMAD family member 6 (SMAD6), thereby activating bone morphogenetic protein 2 (BMP2), which, in turn, induces angiogenesis. CONCLUSIONS: The present work suggests that administration of saliva-Exos and UBE2O represents a promising strategy for enhancing wound healing through promotion of angiogenesis.


Assuntos
Exossomos/enzimologia , Neovascularização Fisiológica/efeitos dos fármacos , Saliva/enzimologia , Proteína Smad6/metabolismo , Enzimas de Conjugação de Ubiquitina , Animais , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia , Saliva/citologia , Pele/lesões , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/farmacologia , Cicatrização/efeitos dos fármacos
20.
Genet Med ; 21(11): 2577-2585, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31138930

RESUMO

PURPOSE: Radioulnar synostosis (RUS) can be syndromic or nonsyndromic. The genetic basis for several RUS syndromes have been reported. However, the genetic cause of nonsyndromic RUS (nsRUS) remains unknown. METHODS: We performed Giemsa (GTG) banding, Sanger sequencing, and exome sequencing on patients (n = 140) and families (n = 11) who suffered from RUS. RESULTS: GTG banding identified 10% RUS sporadic cases affected by sex chromosome aneuploidy. Sanger sequencing on candidate genes revealed noggin (NOG) rarely mutated in nsRUS. Exome sequencing identified 16 loss-of-function (LOF) and 6 missense variants (minor allele frequency [MAF] < 0.0001) in 22/117 nsRUS sporadic patients. Genetic association analysis found a significant association between SMAD6-LOF variants and nsRUS risk (odds ratio [OR] = 430, 95% confidence interval [CI]: 238-780, P < 0.000001). SMAD6 mutated in nsRUS was further confirmed by direct Sanger sequencing of SMAD6-coding regions on other unrelated cohorts of nsRUS cases or families. In summary, we detected 27 SMAD6 rare variants in nsRUS, most of which were LOF variants, 4 were de novo, and 3 were transmitted in families with autosomal dominant inheritance. CONCLUSION: As an intracellular bone morphogenetic protein (BMP) antagonist gene, SMAD6 is frequently mutated in nsRUS. NOG, which encodes an extracellular BMP antagonist, is rarely mutated in nsRUS. This work is the first genetic study on nsRUS.


Assuntos
Rádio (Anatomia)/anormalidades , Proteína Smad6/genética , Sinostose/genética , Ulna/anormalidades , Adolescente , Adulto , Alelos , Criança , Pré-Escolar , Família , Feminino , Frequência do Gene/genética , Humanos , Lactente , Recém-Nascido , Masculino , Mutação de Sentido Incorreto/genética , Linhagem , Penetrância , Análise de Sequência de DNA/métodos , Proteína Smad6/metabolismo , Sequenciamento do Exoma/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA