Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
J Cancer Res Clin Oncol ; 150(5): 264, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38767747

RESUMEN

BACKGROUND: Bladder cancer (BCa) is among the most prevalent malignant tumors affecting the urinary system. Due to its highly recurrent nature, standard treatments such as surgery often fail to significantly improve patient prognosis. Our research aims to predict prognosis and identify precise therapeutic targets for novel treatment interventions. METHODS: We collected and screened genes related to the TGF-ß signaling pathway and performed unsupervised clustering analysis on TCGA-BLCA samples based on these genes. Our analysis revealed two novel subtypes of bladder cancer with completely different biological characteristics, including immune microenvironment, drug sensitivity, and more. Using machine learning classifiers, we identified SMAD6 as a hub gene contributing to these differences and further investigated the role of SMAD6 in bladder cancer in the single-cell transcriptome data. Additionally, we analyzed the relationship between SMAD6 and immune checkpoint genes. Finally, we performed a series of in vitro assays to verify the function of SMAD6 in bladder cancer cell lines. RESULTS: We have revealed two novel subtypes of bladder cancer, among which C1 exhibits a worse prognosis, lower drug sensitivity, a more complex tumor microenvironment, and a 'colder' immune microenvironment compared to C2. We identified SMAD6 as a key gene responsible for the differences and further explored its impact on the molecular characteristics of bladder cancer. Through in vitro experiments, we found that SMAD6 promoted the prognosis of BCa patients by inhibiting the proliferation and migration of BCa cells. CONCLUSION: Our study reveals two novel subtypes of BCa and identifies SMAD6 as a highly promising therapeutic target.


Asunto(s)
Aprendizaje Automático , Proteína smad6 , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Pronóstico , Proteína smad6/genética , Proteína smad6/metabolismo , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Proliferación Celular , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica
2.
J Med Genet ; 61(4): 363-368, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38290823

RESUMEN

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.


Asunto(s)
Craneosinostosis , Radio (Anatomía)/anomalías , Sinostosis , Cúbito/anomalías , Humanos , Masculino , Craneosinostosis/diagnóstico , Craneosinostosis/genética , Radio (Anatomía)/metabolismo , Cúbito/metabolismo , Mutación Missense/genética , Proteína smad6/genética , Proteína smad6/metabolismo
3.
Gene ; 896: 147994, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-37977316

RESUMEN

Craniosynostosis is one of the most common congenital craniofacial birth defects. The genetic etiology is complex, involving syndromic developmental diseases, chromosomal abnormalities, and monogenic non-syndromic diseases. Herein, we presented a proband of craniosynostosis, who firstly displayed structural abnormalities. This research conducted dynamic ultrasound monitoring a fetus with gradually developing intrauterine growth retardation (IUGR). A novel de novo variant c.41G > A: p.W14* in SMAD6 was identified by pedigree analysis and genetic examination approaches. Recombinant plasmid carrying wild-type sequence and mutant that carries c.41G > A in SMAD6 were constructed and transfected into HEK293T cells. mRNA and protein expression of SMAD6 were reduced in SMAD6 mutants compared to the wild type. Cycloheximide (CHX) treatment and si-UPF1 transfection rescued the SMAD6 mRNA expression in the mutant construct, indicating that c.41G > A: p.W14* in SMAD6 triggered nonsense-mediated mRNA degradation (NMD) process and thus led to haploinsufficiency of the protein product. Our study demonstrated that whole-exome sequencing (WES) was a powerful tool for further diagnosis and etiological identification once fetal malformation was detected by ultrasound. Novel de novo c.41G > A: p.W14* in SMAD6 is pathogenic and potentially leads to craniosynostosis via NMD process.


Asunto(s)
Craneosinostosis , Embarazo , Femenino , Humanos , Células HEK293 , Craneosinostosis/diagnóstico por imagen , Craneosinostosis/genética , Feto , ARN Mensajero/genética , China , Proteína smad6/genética , Transactivadores , ARN Helicasas
4.
Oral Dis ; 29(8): 3447-3459, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35957556

RESUMEN

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.


Asunto(s)
MicroARNs , Osteogénesis , Humanos , Osteogénesis/genética , MicroARNs/genética , MicroARNs/metabolismo , Regulación hacia Abajo , Hibridación Fluorescente in Situ , Microtomografía por Rayos X , Diferenciación Celular/genética , Proliferación Celular/genética , Proteína smad6/genética , Proteína smad6/metabolismo
5.
Ann Anat ; 244: 151977, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35787440

RESUMEN

BACKGROUND: Multiple signaling molecules have been shown to play crucial roles in dental root development. Therefore, we aimed to investigate the prevalence of S-shaped roots and also to investigate, if single nucleotide polymorphisms (SNPs) in BMP2, BMP4 and SMAD6 are associated with this phenotype in humans. METHODS: This is a cross-sectional phenotype-genotype association study that used radiographs to determine the phenotypes and DNA to investigate SNPs in candidate genes. During the radiographic exam, teeth presenting root canal(s) doubly curved were considered S-shaped roots. SNPs in BMP2 (rs1005464 and rs235768), BMP4 (rs17563) and SMAD6 (rs2119261 and rs3934908) were blindly genotyped by real-time PCR using TaqMan assay. The relative and absolute frequency of S-shaped roots were calculated. Chi-square test was used to compare the genotype distributions between control and S-shaped groups. RESULTS: Among the 578 subjects, 61 (10.6 %) presented at least one tooth with an S-shaped root. The most commonly affected type of tooth was the premolar. rs1005464 in BMP2 was statistically associated with an S-shaped root (p = 0.036). rs235768 in BMP2 was associated with an S-shaped root also in mandibular teeth (p = 0.017). A statistical significance was observed for the rs3934908 in SMAD6 (p = 0.049) for S-shaped root in the mandible. In the analysis stratified according to the type of tooth, rs235768 in BMP2 was associated with S-shaped roots in premolars (p = 0.029). CONCLUSION: The prevalence of S-shaped roots is 10.6 % in permanent teeth. SNPs in BMP2 and SMAD6 could be involved in a higher chance to present S-shaped roots.


Asunto(s)
Proteína Morfogenética Ósea 2 , Tomografía Computarizada de Haz Cónico , Proteína smad6 , Raíz del Diente , Humanos , Estudios Transversales , Cavidad Pulpar , Mandíbula , Prevalencia , Raíz del Diente/diagnóstico por imagen , Proteína Morfogenética Ósea 2/genética , Proteína smad6/genética
6.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(5): 588-599, 2022 May 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-35753729

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Esfingosina N-Aciltransferasa/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , ADN Complementario , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/patología , Invasividad Neoplásica/genética , Oxidorreductasas , ARN Mensajero/genética , ARN Interferente Pequeño , Proteína smad6/genética , Proteína smad6/metabolismo
7.
Stem Cell Rev Rep ; 18(3): 1078-1096, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34449013

RESUMEN

Mesenchymal stem cells (MSCs) constitute a promising therapy for spinal cord injury (SCI) because they can provide a favorable environment for the regrowth of neurons by inhibiting receptor-regulated Smads (R-Smads) expression in endogenous neural stem cells (NSCs). However, their mechanism of action and effect on the expression of inhibitory Smads (I-Smads) remain unclear. Herein, we demonstrated that extracellular vesicles (EVs) from MSCs were able to upregulate the Smad 6 expression by carrying TGF-ß, and the Smad 6 knockdown in NSCs partially weakened the bone marrow MSC (BMSC)-EV-induced effect on neural differentiation. We found that the expression of Smad 6 did not reduced owing to the TGF-ß type I receptor kinase inhibitor, SB 431,542, treatment in the acute phase of injury in rats with SCI, thereby indicating that the Smad 6 expression was not only mediated by TGF-ß, but also by the inflammatory factors and bone morphogenetic proteins (BMPs) as well. However, in the later phase of SCI, the Smad 6 expression decreased by the addition of SB 431,542, suggesting that TGF-ß plays a key role in the mediation of Smad 6 expression in this phase. In addition, immunohistochemistry staining; hematoxylin-eosin staining; and the Basso, Beattie, and Bresnahan (BBB) scores revealed that the early inhibition of TGF-ß did not increase neuron regrowth. However, this inhibition increased the cavity and the caspase-3 expression at 24 h post-injury, leading to a worse functional outcome. Conversely, the later treatment with the TGF-ß inhibitor promoted the regrowth of neurons around the cavity, resulting in a better neurological outcome. Together, these results indicate that Smad 6 acts as a feedback regulator to prevent the over-differentiation of NSCs to astrocytes and that BMSC-EVs can upregulate Smad 6 expression by carrying TGF-ß.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Proteína smad6 , Traumatismos de la Médula Espinal , Animales , Vesículas Extracelulares/metabolismo , Neuronas , Ratas , Proteína smad6/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/terapia , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
8.
Mol Genet Genomic Med ; 10(1): e1850, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34953066

RESUMEN

BACKGROUND: SMAD6 variants have been reported in patients with radioulnar synostosis (RUS). This study aimed to investigate the genotypes and phenotypes for a large cohort of patients with RUS having mutant SMAD6. METHODS: Genomic DNA samples were isolated from 251 RUS sporadic patients (with their parents) and 27 RUS pedigrees. Sanger sequencing was performed for the SMAD6 coding regions. For positive probands, co-segregation and parental-origin analysis of SMAD6 variants and phenotypic re-evaluation were performed for their family members. RESULTS: We identified 50 RUS probands with SMAD6 variants (13 co-segregated with RUS in pedigrees and 37 in RUS-sporadic patients). Based on the new and previous data, we identified SMAD6 mutated in 16/38 RUS pedigrees and 61/393 RUS sporadic patients, respectively. Overall, 93 SMAD6 mutant patients with RUS were identified, among which 29 patients had unilateral RUS, where the left side was more involved than the right side (left:right = 20:9). Female protective effects and non-full penetrance were observed, in which only 6.90% mothers (vs. ~50% fathers) of SMAD6 mutant RUS probands had RUS. Pleiotropy was observed as a re-evaluation of SMAD6 mutant families identified: (a) three families had axial skeletal malformations; (b) two families had polydactyly; and (c) eight families had other known malformations. CONCLUSION: SMAD6 was mutated in 42.11% RUS pedigrees and 15.52% RUS sporadic patients. The RUS patients with SMAD6 variants exhibit both non-full-penetrance, variable expressivity, pleiotropy, female protective effects, and the left side is more susceptible than the right side.


Asunto(s)
Proteína smad6 , Sinostosis , Femenino , Genotipo , Humanos , Masculino , Fenotipo , Radio (Anatomía)/anomalías , Proteína smad6/genética , Sinostosis/genética , Cúbito/anomalías
9.
Genes (Basel) ; 12(7)2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34208845

RESUMEN

Objective, the application of genomic sequencing in clinical practice has allowed us to appreciate the contribution of co-occurring pathogenic variants to complex and unclassified clinical phenotypes. Besides the clinical relevance, these findings have provided evidence of previously unrecognized functional links between genes in the context of developmental processes and physiology. Patients and Methods, a 5-year-old patient showing an unclassified phenotype characterized by developmental delay, speech delay, peculiar behavioral features, facial dysmorphism and severe cardiopathy was analyzed by trio-based whole exome sequencing (WES) analysis to identify the genomic events underlying the condition. Results, two co-occurring heterozygous truncating variants in CNOT3 and SMAD6 were identified. Heterozygous loss-of-function variants in CNOT3, encoding a subunit of the CCR4-NOT protein complex, have recently been reported to cause a syndromic condition known as intellectual developmental disorder with speech delay, autism and dysmorphic facies (IDDSADF). Enrichment of rare/private variants in the SMAD6 gene, encoding a protein negatively controlling transforming growth factor ß/bone morphogenetic protein (TGFB/BMP) signaling, has been described in association with a wide spectrum of congenital heart defects. We dissected the contribution of individual variants to the complex clinical manifestations and profiled a previously unappreciated set of facial features and signs characterizing IDDSADF. Conclusions, two concomitant truncating variants in CNOT3 and SMAD6 are the cause of the combination of features documented in the patient resulting in the unique multisystem neurodevelopmental condition. These findings provide evidence for a functional link between the CCR4-NOT complex and TGFB/BMP signaling in processes controlling cardiac development. Finally, the present revision provides evidence that IDDSADF is characterized by a distinctive facial gestalt.


Asunto(s)
Trastorno Autístico/patología , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/patología , Trastornos del Desarrollo del Lenguaje/patología , Proteína smad6/genética , Factores de Transcripción/genética , Trastorno Autístico/genética , Preescolar , Heterocigoto , Humanos , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Masculino , Secuenciación del Exoma
10.
Sci Rep ; 11(1): 12715, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135450

RESUMEN

This study investigated, if genetic variants in BMP2, BMP4 and SMAD6 are associated with variations in the palatal rugae pattern in humans. Dental casts and genomic DNA from 75 patients were evaluated. Each patient was classified as follows: total amount of rugae; bilateral symmetry in the amount, length and shape of the palatal rugae; presence of secondary or fragmentary palatal rugae; presence of unifications; predominant shape; and predominant direction of the palatal rugae. The genetic variants in BMP2 (rs1005464 and rs235768), BMP4 (rs17563) and SMAD6 (rs2119261 and rs3934908) were genotyped. Genotype distribution was compared between palatal rugae patterns using the chi-square test (alpha = 0.05). The allele A was associated with the presence of secondary or fragmentary rugae for rs1005464 (OR = 2.5, 95%CI 1.1-6.3; p = 0.014). Secondary or fragmentary rugae were associated with the G allele in rs17563 (OR = 2.1, 95%CI 1.1-3.9; p = 0.017). rs17563 was also associated with rugae unification (p = 0.017 in the additive model). The predominant shape (wavy) was associated with rs2119261 (p = 0.023 in the additive model). The left-right symmetry of the length of primary rugae was associated with rs3934908 in the recessive model (OR = 3.6, 95%CI 1.2-11.7; p = 0.025). In conclusion, genetic variants in the BMP pathway impacted on palatal rugae pattern.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 4/genética , Paladar Duro/anatomía & histología , Polimorfismo de Nucleótido Simple , Proteína smad6/genética , Adolescente , Adulto , Alelos , Variación Anatómica , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Niño , Femenino , Genotipo , Humanos , Masculino , Mucosa Bucal/anatomía & histología , Fenotipo , Transducción de Señal , Proteína smad6/metabolismo , Adulto Joven
11.
Mol Immunol ; 136: 128-137, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34139553

RESUMEN

Transcription factor small mothers against decapentaplegic (Smad) family SMAD proteins are the essential intracellular signal mediators and transcription factors for transforming growth factor ß (TGF-ß) signal transduction pathway, which usually exert pleiotropic actions on cell physiology, including immune response, cell migration and differentiation. In this study, the Smad family was identified in the most primitive vertebrates through the investigation of the transcriptome data of lampreys. The topology of phylogenetic tree showed that the four Smads (Smad1, Smad3, Smad4 and Smad6) in lampreys were subdivided into four different groups. Meanwhile, homology analysis indicated that most Smads were conserved with typical Mad Homology (MH) 1 and MH2 domains. In addition, Lethenteron reissneri Smads (Lr-Smads) adopted general Smads folding structure and had high tertiary structural similarity with human Smads (H-Smads). Genomic synteny analysis revealed that the large-scale duplication blocks were not found in lamprey genome and neighbor genes of lamprey Smads presented dramatic differences compared with jawed vertebrates. Importantly, quantitative real-time PCR analysis demonstrated that Smads were widely expressed in lamprey, and the expression level of Lr-Smads mRNA was up-regulated with different pathogenic stimulations. Moreover, depending on the weighted gene co-expression network analysis (WGCNA), four Lr-Smads were identified as two meaningful modules (green and gray). The functional analysis of these two modules showed that they might have a correlation with ployI:C. And these genes presented strong positive correlation during the immune response from the results of Pearson's correlation analysis. In conclusion, our results would not only enrich the information of Smad family in jawless vertebrates, but also lay the foundation for immunity in further study.


Asunto(s)
Lampreas/genética , Lampreas/inmunología , Proteínas Smad/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Secuencia de Aminoácidos , Animales , Evolución Molecular , Regulación de la Expresión Génica/genética , Genoma/genética , Filogenia , Poli I-C/inmunología , Conformación Proteica , Transducción de Señal/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína smad6/genética , Proteína smad6/metabolismo
12.
Biol Chem ; 402(4): 469-480, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33938174

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/metabolismo , MicroARNs/metabolismo , Proteína smad6/genética , Proteína smad7/genética , Factor de Crecimiento Transformador beta/metabolismo , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Neoplasias Colorrectales/patología , Biología Computacional , Humanos , MicroARNs/genética , Transducción de Señal , Proteína smad6/metabolismo , Proteína smad7/metabolismo , Células Tumorales Cultivadas
13.
FEBS Lett ; 595(3): 389-403, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33264418

RESUMEN

Bone morphogenetic protein (BMP) 9 is one of the most osteogenic BMPs, but its mechanism of action has not been fully elucidated. Hes1, a transcriptional regulator with a basic helix-loop-helix domain, is a well-known effector of Notch signaling. Here, we find that BMP9 induces periodic increases of Hes1 mRNA and protein expression in osteoblasts, presumably through an autocrine negative feedback mechanism. BMP9-mediated Hes1 induction is significantly inhibited by an ALK inhibitor and overexpression of Smad7, an inhibitory Smad. Luciferase and ChIP assays revealed that two Smad-binding sites in the 5' upstream region of the mouse Hes1 gene are essential for transcriptional activation by BMP9. Thus, our data indicate that BMP9 induces Hes1 expression in osteoblasts via the Smad signaling pathway.


Asunto(s)
Factor 2 de Diferenciación de Crecimiento/genética , Osteoblastos/metabolismo , Transducción de Señal/genética , Proteína smad7/genética , Factor de Transcripción HES-1/genética , Animales , Animales Recién Nacidos , Comunicación Autocrina , Secuencia de Bases , Diferenciación Celular , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis/genética , Cultivo Primario de Células , Regiones Promotoras Genéticas , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Cráneo/citología , Cráneo/metabolismo , Proteína smad6/genética , Proteína smad6/metabolismo , Proteína smad7/metabolismo , Factor de Transcripción HES-1/metabolismo
14.
J Endod ; 47(2): 278-285, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33245975

RESUMEN

INTRODUCTION: This study aimed to evaluate the interplay among single-nucleotide polymorphisms (SNPs) in the encoding genes BMP2, BMP4, SMAD6, and RUNX2 in persistent apical periodontitis (PAP). METHODS: In this multicentric study, 272 patients diagnosed with pulp necrosis with apical periodontitis before root canal therapy who attended regular follow-up visits for at least 1 year were screened. Periapical radiographs and clinical aspects were evaluated, and the participants were classified as PAP (n = 110) or repaired (n = 162). Genomic DNA was used for the genotyping of the following SNPs: rs1005464 and rs235768 in bone morphogenetic protein 2 (BMP2), rs17563 in bone morphogenetic protein 4 (BMP4), rs2119261 and rs3934908 in SMAD family member 6 (SMAD6), and rs59983488 and rs1200425 in runt-related transcription factor 2 (RUNX2). The chi-square test was used to compare genotype distributions between groups. The multifactor dimensionality reduction method was applied to identify SNP-SNP interactions. The alpha for all the analysis was 5%. RESULTS: The multifactor dimensionality reduction suggested the rs235768 in BMP2 and rs59983488 in RUNX2 as the best SNP-SNP interaction model (cross-validation = 10/10, testing balanced accuracy = 0.584, P = .026) followed by rs17563 in BMP4 and rs2119261 in SMAD6 (cross validation = 10/10, testing balanced accuracy = 0.580, P = .031). In the rs235768 in BMP2 and rs59983488 in RUNX2 model, the high-risk genotype was TT + TT (odds ratio = 4.36; 95% confidence interval, 0.44-42.1). In model rs17563 in BMP4 and rs2119261 in SMAD6, GG + TT (odds ratio = 2.63; 95% confidence interval, 0.71-11.9) was the high-risk genotype. CONCLUSIONS: The interactions between rs235768 in BMP2 and rs59983488 in RUNX2 and between rs17563 in BMP4 and rs2119261 in SMAD6 are associated with PAP, suggesting that an interplay of these SNPs is involved in the higher risk of developing PAP.


Asunto(s)
Proteína Morfogenética Ósea 2 , Periodontitis Periapical , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4 , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Predisposición Genética a la Enfermedad , Humanos , Periodontitis Periapical/diagnóstico por imagen , Periodontitis Periapical/genética , Polimorfismo de Nucleótido Simple/genética , Proteína smad6/genética
15.
J Cell Mol Med ; 24(24): 14539-14548, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33174391

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteína smad6/genética , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , Inmunohistoquímica , Masculino , Neoplasias de la Próstata/patología , Unión Proteica , Proteína smad6/metabolismo
16.
Eur Rev Med Pharmacol Sci ; 24(14): 7681-7689, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32744694

RESUMEN

OBJECTIVE: MiRNA family gene is an evolutionarily conserved non-coding small RNA that directly participates in a variety of physiological processes and cancer development via regulating gene expression in the biological level of transcription. To research the specific mechanism by which miR-186 regulates apoptosis within gliomas. PATIENTS AND METHODS: RT-qPCR was performed to verify the transcriptional level of miR-186 within glioma tissues and glioma cells. miRanda and Dual-Luciferase assay were performed to predict and confirm that Smad6 gene is an effective target of miR-186 within glioma. The expression of Smad6 protein was tested by Western blot following cell effective transfection. Apoptosis of gliomas was analyzed by inverted fluorescence microscopy and flow cytometry. RESULTS: The mRNA level of miR-186 was suppressed within glioma tissues and glioma U87 cells. MiR-186 is associated with apoptosis in glioma. Overexpression of miR-186 promoted U87 cell apoptosis, whereas suppression of miR-186 had the opposite effect. Besides, miR-186 directly targeted Smad6 and suppress its expression in glioma. The expression of Smad6 affected the regulation of miR-186 on glioma cell apoptosis, restoration of Smad6 rescued apoptosis of glioma U87 cells induced by miR-186 mimics, whereas inhibition of Smad6 promoted apoptosis. CONCLUSIONS: As noted above, miR-186 exerts a tumor-suppressing effect by targeting Smad6. We propose that miR-186 can be used as a novel biomarker for glioma diagnosis in the future, or as a new pharmacy target in the cure of gliomas.


Asunto(s)
Apoptosis , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , MicroARNs/metabolismo , Proteína smad6/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Humanos , MicroARNs/genética , Transducción de Señal , Proteína smad6/genética
17.
Mol Genet Genomic Med ; 8(10): e1406, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32748548

RESUMEN

BACKGROUND: Bicuspid aortic valve (BAV) is the most common cardiovascular malformation in adults, with a prevalence of 0.5%-2%. The prevalence of BAV in cohorts who were ascertained due to thoracic aortic aneurysms or acute aortic dissections (TAD) is as high as 20%. However, the contribution of causal BAV genes to TAD is not known. Therefore, we evaluated rare deleterious variants of GATA4, NOTCH1, SMAD6, or ROBO4 in patients with BAV who presented with TAD. METHODS: Our cohort consisted of 487 probands with Heritable Thoracic Aortic Aneurysms or Dissections (HTAD, 12% BAV, 29% female) and 63 probands with Early onset complications of Bicuspid Aortic Valve disease (EBAV, 63% TAD, 34% female). After whole exome sequencing, we functionally annotated GATA4, NOTCH1, SMAD6, and ROBO4 variants and compared the prevalence of rare variants in these genes to controls without HTAD. RESULTS: We identified 11 rare deleterious variants of GATA4, SMAD6, or ROBO4 in 12 (18%) EBAV cases. The burden of rare SMAD6 and GATA4 variants was significantly enriched in EBAV but not in HTAD cases, even among HTAD cases with BAV (p < .003). CONCLUSION: Rare variants of NOTCH1, ROBO4, SMAD6, or GATA4 do not significantly contribute to BAV in cohorts with HTAD. We conclude that BAV patients who present with HTAD are a genetically distinct subgroup with implications for genetic testing and prognosis.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Enfermedad de la Válvula Aórtica Bicúspide/genética , Factor de Transcripción GATA4/genética , Receptor Notch1/genética , Receptores de Superficie Celular/genética , Proteína smad6/genética , Adulto , Aneurisma de la Aorta Torácica/complicaciones , Aneurisma de la Aorta Torácica/patología , Enfermedad de la Válvula Aórtica Bicúspide/complicaciones , Enfermedad de la Válvula Aórtica Bicúspide/patología , Femenino , Frecuencia de los Genes , Humanos , Masculino , Persona de Mediana Edad , Mutación
18.
Genet Med ; 22(9): 1498-1506, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32499606

RESUMEN

PURPOSE: Enrichment of heterozygous missense and truncating SMAD6 variants was previously reported in nonsyndromic sagittal and metopic synostosis, and interaction of SMAD6 variants with a common polymorphism nearBMP2 (rs1884302) was proposed to contribute to inconsistent penetrance. We determined the occurrence of SMAD6 variants in all types of craniosynostosis, evaluated the impact of different missense variants on SMAD6 function, and tested independently whether rs1884302 genotype significantly modifies the phenotype. METHODS: We performed resequencing of SMAD6 in 795 unsolved patients with any type of craniosynostosis and genotyped rs1884302 in SMAD6-positive individuals and relatives. We examined the inhibitory activity and stability of SMAD6 missense variants. RESULTS: We found 18 (2.3%) different rare damaging SMAD6 variants, with the highest prevalence in metopic synostosis (5.8%) and an 18.3-fold enrichment of loss-of-function variants comparedwith gnomAD data (P < 10-7). Combined with eight additional variants, ≥20/26 were transmitted from an unaffected parent but rs1884302 genotype did not predict phenotype. CONCLUSION: Pathogenic SMAD6 variants substantially increase the risk of both nonsyndromic and syndromic presentations of craniosynostosis, especially metopic synostosis. Functional analysis is important to evaluate missense variants. Genotyping of rs1884302 is not clinically useful. Mechanisms to explain the remarkable diversity of phenotypes associated with SMAD6 variants remain obscure.


Asunto(s)
Craneosinostosis , Craneosinostosis/genética , Genotipo , Humanos , Mutación Missense/genética , Penetrancia , Fenotipo , Proteína smad6/genética
19.
Biol Reprod ; 103(3): 572-582, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32432313

RESUMEN

In this study, we aimed to determine the origin of the difference, in terms of anti-Müllerian hormone production, existing between the bovine and porcine ovaries. We first confirmed by quantitative real-time-Polymerase-Chain Reaction, ELISA assay and immunohistochemistry that anti-Müllerian hormone mRNA and protein production are very low in porcine ovarian growing follicles compared to bovine ones. We then have transfected porcine and bovine granulosa cells with vectors containing the luciferase gene driven by the porcine or the bovine anti-Müllerian hormone promoter. These transfection experiments showed that the porcine anti-Müllerian hormone promoter is less active and less responsive to bone morphogenetic protein stimulations than the bovine promoter in both porcine and bovine cells. Moreover, bovine but not porcine granulosa cells were responsive to bone morphogenetic protein stimulation after transfection of a plasmidic construction including a strong response element to the bone morphogenetic proteins (12 repetitions of the GCCG sequence) upstream of the luciferase reporter gene. We also showed that SMAD6, an inhibitor of the SMAD1-5-8 pathway, is strongly expressed in porcine compared to the bovine granulosa cells. Overall, these results suggest that the low expression of anti-Müllerian hormone in porcine growing follicles is due to both a lack of activity/sensitivity of the porcine anti-Müllerian hormone promoter, and to the lack of responsiveness of porcine granulosa cells to bone morphogenetic protein signaling, potentially due to an overexpression of SMAD6 compared to bovine granulosa cells. We propose that the low levels of anti-Müllerian hormone in the pig would explain the poly-ovulatory phenotype in this species.


Asunto(s)
Hormona Antimülleriana/biosíntesis , Células de la Granulosa/metabolismo , Ovario/metabolismo , Animales , Hormona Antimülleriana/genética , Proteínas Morfogenéticas Óseas/biosíntesis , Bovinos , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ovario/citología , Regiones Promotoras Genéticas , Transducción de Señal/efectos de los fármacos , Proteína smad6/biosíntesis , Proteína smad6/genética , Especificidad de la Especie , Porcinos
20.
J Cell Mol Med ; 24(5): 2802-2818, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32022446

RESUMEN

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.


Asunto(s)
Colágeno Tipo V/genética , Factores de Transcripción Forkhead/metabolismo , Proteína smad6/metabolismo , Adherencias Tisulares/genética , Enfermedades Uterinas/genética , Animales , Ciclo Celular/genética , Línea Celular , Proliferación Celular/genética , Colágeno Tipo V/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Endometrio/metabolismo , Endometrio/patología , Femenino , Fibrosis , Factores de Transcripción Forkhead/genética , Humanos , Ratas Sprague-Dawley , Proteína smad6/genética , Células del Estroma/metabolismo , Adherencias Tisulares/patología , Transcripción Genética , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/genética , Enfermedades Uterinas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...