Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Cell Sci ; 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39037211

RESUMO

Muscle stem cells (MuSCs) play an indispensable role in postnatal muscle growth and hypertrophy in adult. MuSCs also retain a highly regenerative capacity and are therefore considered a promising stem cell source for regenerative therapy of muscle diseases. In this study, we identify tumor-suppressor protein Tob1 as a Pax7-target gene that negatively controls the population expansion of MuSCs. Tob1 protein is undetectable in the quiescent state but is upregulated during activation in MuSCs. Tob1 ablation in mice accelerates MuSC population expansion and boosts muscle regeneration. Moreover, inactivation of Tob1 in MuSCs ameliorates the efficiency of MuSC transplantation in a murine muscular dystrophy model. Collectively, selective targeting of Tob1 may be a therapeutic option for the treatment of muscular diseases, including muscular dystrophy and age-related sarcopenia.

2.
Dig Dis Sci ; 68(4): 1316-1331, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36002674

RESUMO

BACKGROUND: TOB1, a member of the transducer of erbB-2 /B-cell translocation gene family, was detected to be down-regulated in ESCC by RNA sequencing. TOB1-AS1, a head-to-head antisense lncRNA with TOB1, was down-regulated in several cancers. However, the roles of them in esophageal squamous cell carcinoma (ESCC) remained unclarified. AIMS: To investigate the roles and functions of TOB1-AS1 and TOB1 in ESCC tumorigenesis. MATERIALS AND METHODS: The expression levels, methylation status, biological function and mechanisms of TOB1-AS1 and TOB1 in ESCC were, respectively, detected. RESULTS: Frequent down-regulation of TOB1-AS1 and TOB1 was verified in esophageal cancer cells and ESCC tissues, and there was a positive correlation between them in ESCC tissues. The CpG sites hypermethylation within proximal promoter of TOB1-AS1 and TOB1 could lead to transcriptional inhibition of both genes. Furthermore, expression and proximal promoter methylation status of TOB1-AS1 or TOB1 may be associated with ESCC patients' prognosis. TOB1-AS1 and TOB1 may function as tumor suppressors by inhibiting growth, migration, and invasion of esophageal cancer cells. Up-regulation of TOB1-AS1 increased expression level of TOB1, and TOB1-AS1 could work as a ceRNA to modulate ATF3 expression via competitively binding with miR-103a-2-5p. Meanwhile, ATF3, as a transcription factor, could regulate transcription of TOB1; down-regulation of TOB1-AS1 in ESCC led to decreased expression of ATF3 through ceRNA mechanism, and further influenced the transcription of TOB1. CONCLUSION: TOB1-AS1 and TOB1 may act as tumor suppressors and may serve as potential targets for antitumor therapy in ESCC.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma de Células Escamosas do Esôfago/patologia , Regulação para Baixo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Progressão da Doença , Linhagem Celular Tumoral , Prognóstico , Metilação de DNA , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
3.
Pharmacology ; 108(4): 321-330, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37232038

RESUMO

INTRODUCTION: Tumor exosome-derived miRNAs play important roles in the human laryngocarcinoma. However, it is still unknown if exosome miR-552 is involved in the laryngocarcinoma. The aim of the current study was to explore exosome miR-552's role in laryngocarcinoma and its underlying mechanisms. METHODS: Hep-2 exosome was characterized by transmission electron microscopy and nanoparticle tracking technology. CCK-8 was used to determine cell viability, and a xenograft animal model was used to determine the tumorigenicity. qPCR and Western blotting were used to measure the changes in target biomarkers. Luciferase reporter assay was used to evaluate the interactions between miR-552 and PTEN. miRNA sequencing was used to check the changes in miRNA profiles. RESULTS: miR-552 was upregulated in the laryngocarcinoma patients and was positively correlated to the cell proliferation and tumor growth. PTEN was identified as a direct target of miR-552. Hep-2 exosome is featured by high expression of miR-552 and treatment of Hep-2 exosome enhanced cell proliferation and tumorigenicity. The underlying mechanisms revealed that treatment of exosomes enhanced the malignant transformation of recipient cells in part by regulating epithelial-mesenchymal transition. CONCLUSION: Exosome miR-552 promotes laryngocarcinoma cells' malignant progression in part by the regulation of the PTEN/TOB1 axis.


Assuntos
Exossomos , MicroRNAs , Animais , Humanos , Exossomos/genética , Exossomos/metabolismo , Transdução de Sinais , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Proteínas Supressoras de Tumor/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo
4.
Biochem Biophys Res Commun ; 526(4): 1005-1012, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32307083

RESUMO

Triple negative breast cancer (TNBC) is a malignant breast cancer subtype with poor prognosis. Recent studies have revealed the critical roles of dysregulated long non-coding RNAs (lncRNAs) in many cancer types, including TNBC. LncRNA WEE2 antisense RNA 1 (WEE2-AS1) has been reported to be able to promote the progression of hepatocellular carcinoma, but the function of WEE2-AS1 in TNBC is still unknown. Therefore, in this study, we specifically researched the role of WEE2-AS1 and probed its molecular mechanism in TNBC cells. Our results showed that WEE2-AS1 was up-regulated in TNBC cell lines, and WEE2-AS1 knockdown could inhibit TNBC cell proliferation, promote apoptosis, and suppress migration and invasion. Further, we found that miR-32-5p was down-regulated in TNBC cells and could be sponged by WEE2-AS1. Moreover, miR-32-5p could target its downstream gene transducer of ERBB2, 1 (TOB1), which was highly expressed and could play the oncogenic role in TNBC cells. Through rescue assays, we proved that WEE2-AS1/miR-32-5p/TOB1 axis could modulate cancer progression in TNBC cells. In conclusion, our results demonstrated the oncogenic function of lncRNA WEE2-AS1 in TNBC cells, providing a novel insight into TNBC therapy.


Assuntos
Apoptose/genética , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteínas Supressoras de Tumor/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Técnicas de Silenciamento de Genes , Humanos , MicroRNAs/genética , RNA Longo não Codificante/genética , Transdução de Sinais/genética
5.
Mol Cancer ; 17(1): 159, 2018 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-30447686

RESUMO

Tob1, a Tob/BTG anti-proliferative protein family member, functions as a tumour suppressor in many cancers. Here, we reveal a unique oncogenic role of Tob1 in colon cancer. Tob1 expression was upregulated during colon cancer progression, was significantly correlated with tumour size and tumour differentiation, and was a prognostic indicator of colon cancer. Unlike in other cancers, where nuclear Tob1 performs anticancer activity, Tob1 is predominantly localized in the cytosol of colon cancer cells, where this protein binds and stabilizes ß-catenin to activate Wnt/ß-catenin signalling, which in turn enhances Tob1 expression, thus forming a positive feedback loop to promote cell proliferation. Moreover, Tob1 deficiency led to reduced tumourigenesis in AOM/DSS-treated and ApcMin/+ mice. Our findings provide important insights into a previously unrecognized oncogenic role of Tob1 in colon cancer and suggest that Tob1 is an adverse prognostic factor and therapeutic target for colon cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Expressão Gênica , Via de Sinalização Wnt , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Biomarcadores Tumorais , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/mortalidade , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Prognóstico , Curva ROC
6.
Biochem Biophys Res Commun ; 493(2): 901-908, 2017 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-28947212

RESUMO

Although the exact pathogenesis of multiple sclerosis (MS) remains largely unclear, Th17 cells have been suggested as an essential regulator in the disease induction. Emerging evidence have demonstrated that noncoding RNAs, especially microRNAs (miRs), play a crucial role in modulation of Th17 cell differentiation and autoimmune disease development. Here, we revealed that miR-590 expression was markedly increased in periphery blood mononuclear cells (PBMC) and cerebrospinal fluid (CSF) of patients with MS, and positively correlated with the disease severity. Th17 cells were found to express high level of miR-590. We further demonstrated that miR-590 was able to facilitate Th17 differentiation and pathogenicity. Notably, we identified that miR-590 directly targeted Tob1, a known suppressor of Th17 differentiation. The expression level of Tob1 was observed to be significantly decreased in PBMC of patients with MS. Our finding suggest that miR-590 could enhance pathogenic Th17 differentiation in MS and augment inflammation in central nervous system (CNS) through inhibiting Tob1.


Assuntos
Regulação para Baixo , Peptídeos e Proteínas de Sinalização Intracelular/genética , MicroRNAs/genética , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Células Th17/patologia , Proteínas Supressoras de Tumor/genética , Regulação para Cima , Diferenciação Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/análise , MicroRNAs/análise , Células Th17/metabolismo , Proteínas Supressoras de Tumor/análise
7.
Semin Immunol ; 25(4): 299-304, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24211040

RESUMO

T helper 17 (Th17) cells have been reported to be responsible for several chronic inflammatory diseases. However, a peculiar feature of human Th17 cells is that they are very rare in the inflammatory sites in comparison with Th1 cells. The first reason for this rarity is the existence of some self-regulatory mechanisms that limit their expansion. The limited expansion of human Th17 cells is related to the retinoic acid orphan (ROR)C-dependent up-regulation of the interleukin (IL)-4 induced gene 1 (IL4I1), which encodes for a l-phenylalanine oxidase, that has been shown to down-regulate CD3ζ expression in T cells. This results in abnormalities of the molecular pathway which is responsible for the impairment of IL-2 production and therefore for the lack of cell proliferation in response to T-cell receptor (TCR) signalling. IL4I1 up-regulation also associates with the increased expression of Tob1, a member of the Tob/BTG anti-proliferative protein family, which is involved in cell cycle arrest. A second reason for the rarity of human Th17 cells in the inflammatory sites is their rapid shifting into the Th1 phenotype, which is mainly related to the activity of IL-12 and TNF-α. We have named these Th17-derived Th1 cells as non-classic because they differ from classic Th1 cells for the expression of molecules specific for Th17 cells, such as RORC, CD161, CCR6, IL4I1, and IL-17 receptor E. This distinction may be important for defining the respective pathogenic role of Th17, non-classic Th1 and classic Th1 cells in many human inflammatory disorders.


Assuntos
Inflamação/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th17/imunologia , Animais , Humanos , Inflamação/patologia , Subpopulações de Linfócitos T/patologia , Células Th17/patologia
8.
Cell Tissue Res ; 364(2): 443-51, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26662055

RESUMO

Tob1 is a member of the BTG/TOB family of proteins with established antiproliferative function. In Danio rerio and Xenopus laevis, the Tob1 gene is expressed from the one-cell stage through to early gastrula stages, followed in later development by discrete expression in many tissues including the notochord and somites. In both mouse and human, Tob1 is expressed in many adult tissues including the testis and ovary; however, the specific cell types are unknown. We examine Tob1 gene expression in mouse in developing germ cells and in sorted male germ cells (gonocytes, spermatogonia, pachytene spermatocytes and round spermatids) by reverse transcription and droplet digital polymerase chain reaction (RT-ddPCR) and in adult ovary and testis by immunofluorescence with anti-Tob1 protein staining. By RT-ddPCR, Tob1 expression was low in developing male germ cells but was highly expressed in round spermatids. In developing female germ cells undergoing entry into meiosis, it increased 10-fold. Tob1 was also highly expressed in round spermatids and in oocytes in all stages of folliculogenesis. Notably, a marker for P-bodies, Dcp-2, was also highly expressed in round spermatids and all oocyte stages examined. The cytoplasmic presence of Tob1 protein in round spermatids and oocytes and the association of Tob1 protein with Dcp2 in both cell types suggest that Tob1 protein plays a role in post-transcriptional mechanisms.


Assuntos
Proteínas de Transporte/biossíntese , Células Germinativas Embrionárias/metabolismo , Endorribonucleases/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Oócitos/metabolismo , Espermátides/metabolismo , Espermatócitos/metabolismo , Espermatogônias/metabolismo , Animais , Biomarcadores/metabolismo , Feminino , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oogênese/fisiologia , Ovário/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/fisiologia , Testículo/metabolismo
9.
Eur J Immunol ; 44(3): 646-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24497109

RESUMO

Th17 cells are key players in antibacterial and antifungal immunity, but have also been implicated in autoimmunity. Interestingly, Th17 cells poorly proliferate upon stimulation, a phenotype that was attributed to a decreased sensitivity to T-cell receptor (TCR) stimulation, and to low IL-2 production by Th17 cells. In this issue of the European Journal of Immunology, Santarlasci et al. [Eur. J. Immunol. 2014. 44: 654-661] shed further light on the molecular mechanism that keeps Th17 cells at bay. They identify the transcriptional regulator TOB1, which not only impairs IL-2 production in Th17 cells, but also blocks the expression of cell cycle genes. Strikingly, TOB1 suppresses Th17-cell proliferation through several pathways, including impaired signal transduction, transcription, and possibly also post-transcriptional regulation.


Assuntos
Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , L-Aminoácido Oxidase/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Humanos
10.
Eur J Immunol ; 44(3): 654-61, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24307243

RESUMO

Human Th17 cells have a limited proliferative capacity compared to other T-cell subsets. We have shown that human Th17 cells display impaired IL-2 production due to IL-4-induced gene 1 (IL4I1) upregulation. Here, we show that in human Th17 cells, IL4I1 also maintains high levels of Tob1, a member of the Tob/BTG (B-cell traslocation gene) antiproliferative protein family, which prevents cell-cycle progression mediated by TCR stimulation. Indeed, Th17 cells exhibited higher levels of Tob1 than Th1 cells in both resting and TCR-activated conditions. Accordingly, the expression of positive regulators of the cell cycle (cyclin A, B, C, and E and Cdk2), as well as of Skp2, which promotes Tob1 degradation, was lower in Th17 cells than in Th1 cells. Tob1 expression in human Th17 cells correlated with both RAR (retinoic acid receptor)-related orphan receptor C (RORC) and IL4I1 levels. However, RORC was not directly involved in the regulation of Tob1 expression, whereas IL4I1 silencing in Th17 cells induced a substantial decrease of Tob1 expression. These data suggest that IL4I1 upregulation in human Th17 cells limits their TCR-mediated expansion not only by blocking the molecular pathway involved in the activation of the IL-2 promoter, but also by maintaining high levels of Tob1, which impairs entry into the cell cycle.


Assuntos
Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , L-Aminoácido Oxidase/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Humanos , Modelos Biológicos , Proteínas Quinases Associadas a Fase S/metabolismo , Células Th1/imunologia , Células Th1/metabolismo
11.
Cell Biol Int ; 39(12): 1425-30, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26377349

RESUMO

The aim of the present study was to evaluate the effect of Tob1 on the radiosensitivity of breast cancer cells. The results showed that overexpression of Tob1 reduced the clonogenic growth of 231 cells and induced the rate of apoptosis. Tob1 caused an accumulation of cells in the G0 /G1 phase and decreased the percentage of cells in S phase. We also found that overexpression of Tob1 significantly reduced the phosphorylation of JNK and p38. The activator of JNK and p38, anisomycin, attenuated the blockage of Tob1 on the cell cycle and reversed the effect of Tob1 on apoptosis. Taken together, Tob1 enhanced radiosensitivity of breast cancer cells through regulation of the JNK and p38 pathways. The results indicated that Tob1 might be a promising molecular in gene therapy for the treatment of breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Sistema de Sinalização das MAP Quinases/fisiologia , Tolerância a Radiação/fisiologia , Proteínas Supressoras de Tumor/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos da radiação , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Proteínas Supressoras de Tumor/efeitos da radiação , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos da radiação
12.
Front Immunol ; 15: 1369087, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38617839

RESUMO

Introduction: The ErbB-2.1(TOB1) signaling transducer protein is a tumor-suppressive protein that actively suppresses the malignant phenotype of gastric cancer cells. Yet, TOB1 negatively regulates the activation and growth of different immune cells. Understanding the expression and role of TOB1 in the gastric cancer immune environment is crucial to maximize its potential in targeted immunotherapy. Methods: This study employed multiplex immunofluorescence analysis to precisely delineate and quantify the expression of TOB1 in immune cells within gastric cancer tissue microarrays. Univariate and multivariate Cox analyses were performed to assess the influence of clinical-pathological parameters, immune cells, TOB1, and double-positive cells on the prognosis of gastric cancer patients. Subsequent experiments included co-culture assays of si-TOB1-transfected neutrophils with AGS or HGC-27 cells, along with EdU, invasion, migration assays, and bioinformatics analyses, aimed at elucidating the mechanisms through which TOB1 in neutrophils impacts the prognosis of gastric cancer patients. Results: We remarkably revealed that TOB1 exhibits varying expression levels in both the nucleus (nTOB1) and cytoplasm (cTOB1) of diverse immune cell populations, including CD8+ T cells, CD66b+ neutrophils, FOXP3+ Tregs, CD20+ B cells, CD4+ T cells, and CD68+ macrophages within gastric cancer and paracancerous tissues. Significantly, TOB1 was notably concentrated in CD66b+ neutrophils. Survival analysis showed that a higher density of cTOB1/nTOB1+CD66b+ neutrophils was linked to a better prognosis. Subsequent experiments revealed that, following stimulation with the supernatant of tumor tissue culture, the levels of TOB1 protein and mRNA in neutrophils decreased, accompanied by enhanced apoptosis. HL-60 cells were successfully induced to neutrophil-like cells by DMSO. Neutrophils-like cells with attenuated TOB1 gene expression by si-TOB1 demonstrated heightened apoptosis, consequently fostering a malignant phenotype in AGS and HCG-27 cells upon co-cultivation. The subsequent analysis of the datasets from TCGA and TIMER2 revealed that patients with high levels of TOB1 combined neutrophils showed better immunotherapy response. Discussion: This study significantly advances our comprehension of TOB1's role within the immune microenvironment of gastric cancer, offering promising therapeutic targets for immunotherapy in this context.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Neutrófilos , Linfócitos T CD8-Positivos , Imunoterapia , Microambiente Tumoral , Proteínas Supressoras de Tumor , Peptídeos e Proteínas de Sinalização Intracelular/genética
13.
J Orthop Surg Res ; 18(1): 402, 2023 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-37268992

RESUMO

BACKGROUND: Ankylosing spondylitis (AS) is a chronic inflammatory arthritis. Upregulation of microRNA (miR)-92b-3p is associated with enhanced osteoblastic differentiation. The current study sought to investigate the functional mechanism of miR-92b-3p in osteogenic differentiation of AS fibroblasts. METHODS: First, fibroblasts were isolated from AS and non-AS patients and cultured. Next, cell morphology was observed, cell proliferation was assessed and the vimentin expression pattern was determined. Alkaline phosphatase (ALP) activity and levels of osteogenic markers RUNX2, OPN, OSX, and COL I were additionally measured, followed by determination of miR-92b-3p and TOB1 levels. The binding site of miR-92b-3p and TOB1 was predicted, and their target relationship was validated. Lastly, miR-92b-3p inhibitor, si-TOB1, and the BMP/Smad signaling pathway inhibitor LDN193189 were delivered into AS fibroblasts to evaluate the osteogenic differentiation of AS fibroblasts and the activation of the BMP/Smad pathway. RESULTS: miR-92b-3p was highly expressed in AS fibroblasts. AS fibroblasts showed enhanced osteogenic differentiation and proliferation, while inhibition of miR-92b-3p suppressed osteogenic differentiation and proliferation of AS fibroblasts. miR-92b-3p targeted TOB1, and TOB1 was poorly expressed in AS fibroblasts. The concurrent downregulation of TOB1 and inhibition of miR-92b-3p elevated the levels of RUNX2, OPN, OSX, and COL I and ALP activity and further enhanced the proliferation of AS fibroblasts. The BMP/Smad pathway was activated in AS fibroblasts. Silencing miR-92b-3p could inhibit the activation of the BMP/Smad pathway by upregulating TOB1. Inhibition of the BMP/Smad pathway reduced the number of calcified nodules and hindered the osteogenic differentiation and proliferation of AS fibroblasts. CONCLUSION: Our findings highlighted that silencing miR-92b-3p inhibited the osteogenic differentiation and proliferation of AS fibroblasts by upregulation of TOB1 and inhibition of the BMP/Smad pathway.


Assuntos
MicroRNAs , Espondilite Anquilosante , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Osteogênese/genética , Espondilite Anquilosante/genética , Diferenciação Celular/genética , Fibroblastos/metabolismo , Células Cultivadas , Proteínas Supressoras de Tumor , Peptídeos e Proteínas de Sinalização Intracelular/genética
14.
PeerJ ; 10: e12904, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35186488

RESUMO

BACKGROUND: We previously identified the tumor suppressor gene TOB1 as related to gastric cancer. The purpose of this study was to explore whether TOB1 induces autophagy through the AKT/mTOR signaling pathway in gastric cancer. METHODS: Western blotting was used to detect the protein levels of TOB1, LC3, AKT, mTOR, phosphorylated (p) AKT, and p-mTOR. A double fluorescent GFP-RFP-LC3 fusion protein was used to trace autophagy by laser confocal microscopy. Autophagosomes were observed by transmission electron microscopy. RESULTS: The conversion of LC3-I to LC3-II and the LC3-II/LC3-I ratio were significantly increased in AGS cells overexpressing TOB1 compared with control cells. Fluorescence imaging showed LC3 puncta at 48 h, and these puncta increased significantly at 72 h after TOB1 transfection compared with control tumor cells. The presence of autophagosomes in AGS cells was observed at 72 h after TOB1 transfection by transmission electron microscopy, and no autophagosomes were found in the control cells. Moreover, the levels of p-AKT and p -mTOR were lower in AGS cells than in control cancer cells. CONCLUSION: Our results provide novel insight that TOB1 might suppress gastric cancer by inducing autophagy, possibly through decreasing phosphorylation and the subsequent activation of the AKT/mTOR signaling pathway.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Neoplasias Gástricas , Humanos , Proteínas Proto-Oncogênicas c-akt/genética , Neoplasias Gástricas/genética , Apoptose , Linhagem Celular Tumoral , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Autofagia/genética , Proteínas Supressoras de Tumor/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética
15.
J Cancer ; 13(3): 890-905, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154457

RESUMO

Purpose Gastric cancer (GC) is associated with rapid disease progression and poor patient prognosis, highlighting the pressing need for new biomarkers to facilitate disease management. Exosomes are released by all cells and are ubiquitous in body fluids, thus giving them great potential as diagnostic biomarkers and therapeutic targets. MicroRNAs (miRNAs) can be transported by exosomes, and are a common target for regulation in cancer. Methods Our screen of miRNAs in the Gene Expression Omnibus and The Cancer Genome Atlas databases identified miR-552-5p as the most overexpressed miRNA in GC, and we investigated its function and mechanism of action. Results We detected high expression of miR-552-5p in GC tissues, plasma samples and cell lines. We found that miR-552-5p binds directly to the 3'-untranslated region of PTEN, and the resulting downregulation of PTEN in turn downregulates the tumor suppressor TOB1. Furthermore, experiments in cell culture and mice showed that miR-552-5p in exosomes is internalized by recipient cells, where it enhances proliferation, migration and the epithelial-mesenchymal transition, while suppressing the caspase-3 apoptotic pathway. These effects were reversed by inhibiting miR-552-5p. Conclusion GC-derived exosomal miR-552-5p facilitates tumorigenesis by interfering with the PTEN/TOB1 axis, providing new potential therapeutic targets.

16.
Cell Mol Gastroenterol Hepatol ; 13(4): 1201-1221, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34920145

RESUMO

BACKGROUND & AIMS: TOB1 is an anti-proliferative protein of Tob/BTG family and typically involved in the tumorigenesis and T cell activation. Although TOB1 is associated with T helper 17 cell-related autoimmunity, its role in modulating T cell-mediated immune responses in IBD remains poorly understood. Here, we explored its expression and the underlying mechanisms involved in the pathogenesis of inflammatory bowel disease (IBD). METHODS: TOB1 and ID2 expression in IBD patients was examined by quantitative real time polymerase chain reaction and immunohistochemistry. IBD CD4+ T cells were transfected with lentivirus expressing TOB1, ID2, TOB1 short hairpin RNA and ID2 short hairpin RNA, respectively, and Tob1-/-CD4+ T cells were transfected with lentivirus expressing Id2. Experimental colitis was established in Tob1-/- mice by trinitrobenzene sulfonic acid enema and in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells to further explore the role of Tob1 in intestinal mucosal inflammation. Splenic CD4+ T cells of Tob1-/- mice were sorted to determine transcriptome differences by RNA sequencing. RESULTS: TOB1 expression was decreased in inflamed mucosa and peripheral blood CD4+ T cells of IBD patients compared with healthy subjects. Overexpression of TOB1 downregulated IBD CD4+ T cells to differentiate into Th1/Th17 cells compared with control subjects. Severe colitis was observed in Tob1-/- mice through trinitrobenzene sulfonic acid enema or in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells, compared with control animals. RNA sequencing analysis revealed ID2 as functional target of TOB1 to inhibit IBD CD4+ T cell differentiation into Th1/Th17 cells. Mechanistically, TOB1 was associated with Smad4/5 to induce ID2 expression and restrain Th1/Th17 cell differentiation. CONCLUSIONS: TOB1 restrains intestinal mucosal inflammation through suppressing Th1/Th17 cell-mediated immune responses via the Smad4/5-ID2 pathway. It may serve as a novel therapeutic target for treatment of human IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Proteínas de Homeodomínio/metabolismo , Humanos , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Mucosa Intestinal/metabolismo , Ativação Linfocitária , Camundongos , RNA Interferente Pequeno/metabolismo , Ácidos Sulfônicos/metabolismo , Ácidos Sulfônicos/uso terapêutico , Células Th1 , Células Th17/metabolismo , Células Th17/patologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
17.
Int J Mol Sci ; 12(7): 4315-26, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21845080

RESUMO

Cell proliferation is an important biological process during myogenesis. Tob1 encoded a member of the Tob/BTG family of anti-proliferative proteins. Our previous LongSAGE (Long Serial Analysis of Gene Expression) analysis suggested that Tob1 was differentially expressed during prenatal skeletal muscle development. In this study, we isolated and characterized the swine Tob1 gene. Subsequently, we examined Tob1 chromosome assignment, subcellular localization and dynamic expression profile in prenatal skeletal muscle (33, 65 and 90 days post-conception, dpc) from Landrace (lean-type) and Tongcheng pigs (obese-type). The Tob1 gene was mapped to pig chromosome 12 (SSC12). The Tob1 protein was distributed throughout the nucleus and cytoplasm of PK15 cells. During prenatal skeletal muscle development, Tob1 was up-regulated and highly expressed in skeletal muscle at 90 dpc in Tongcheng pigs but peaked at 65 dpc in Landrace pigs. This result suggested that there were different proliferation patterns during myogenesis between Tongcheng and Landrace pigs. During postnatal skeletal muscle development, the expression of Tob1 increased with aging, indicating that the proliferation potential of myoblasts decreased in postnatal muscle development. In tissues of adult wuzhishan miniature pigs, the Tob1 gene was highly expressed in skeletal muscle. The expression of Tob1 was significantly increased at day 6 during C2C12 differentiation time, suggesting a possible role in skeletal muscle development. Therefore, this study indicated that Tob1 perhaps played an important role in skeletal muscle development.


Assuntos
Proteínas de Transporte/metabolismo , Músculo Esquelético/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Diferenciação Celular , Linhagem Celular , Mapeamento Cromossômico , Peptídeos e Proteínas de Sinalização Intracelular , Dados de Sequência Molecular , Desenvolvimento Muscular , Mioblastos/citologia , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Suínos , Fatores de Tempo , Transcriptoma
18.
Cancer Med ; 9(4): 1503-1514, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31891232

RESUMO

TOB1 participates in various kinds of cancers. However, its role in pancreatic cancer has rarely been reported. In this study, we explored the expression and mechanisms of TOB1 in regulating the malignant phenotype of pancreatic cancer cells. TOB1 expression was determined by data mining and immunohistochemistry (IHC), and its localization was observed by immunofluorescence. CCK-8 cell proliferation, colony formation, flow cytometric, transwell migration, and Western blot (WB) assays were used to examine how it impacts the malignant phenotype of pancreatic cancer. Furthermore, Foxa2 binding to TOB1 was tested by dual-luciferase reporter assays, and RNA-Seq was performed to identify signaling pathways. We found TOB1 was downregulated in pancreatic cancer tissues and was mainly located in the cytoplasm. TOB1 overexpression reduced the proliferation of K-Ras wild-type pancreatic cancer cells but made no difference to cell migration and invasion. Foxa2 overexpression significantly enhanced TOB1 promoter activity. Moreover, overexpressing TOB1 substantially enriched the calcium pathway in K-Ras wild-type pancreatic cancer cells. In conclusion, TOB1 may suppress the proliferation of K-Ras wild-type pancreatic cancer cells by regulating calcium pathway genes.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Pancreáticas/genética , Proteínas Supressoras de Tumor/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Conjuntos de Dados como Assunto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , RNA-Seq , Análise Serial de Tecidos
19.
Cancer Biother Radiopharm ; 35(10): 760-764, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32407174

RESUMO

A recent study reported the oncogenic function of lncRNA NR2F1-AS1 in liver cancer. Interestingly, by analyzing TCGA data set, downregulation of NR2F1-AS1 in colorectal cancer (CRC) was observed. This observation triggered interest to analyze the functions of NR2F1-AS1 in CRC. It was observed that NR2F1-AS1 was downregulated in CRC and predicted poor survival. NR2F1-AS1 can directly interact with miR-371a-3p but their overexpression failed to affect the expression of each other. However, NR2F1-AS1 overexpression led to the upregulation of TOB1, a target of miR-371a-3p. Cell proliferation analysis revealed reduced proliferation rate of CRC cells after NR2F1-AS1 and TOB1 overexpression. MiR-371a-3p overexpression played an opposite role and reduced the effects of NR2F1-AS1 and TOB1 overexpression. In conclusion, NR2F1-AS1 regulates miR-371a-3p/TOB1 axis to suppress proliferation of CRC cells.


Assuntos
Neoplasias Colorretais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adulto , Idoso , Fator I de Transcrição COUP/genética , Proliferação de Células/fisiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Antissenso/genética , RNA Longo não Codificante/genética , Proteínas Supressoras de Tumor/genética
20.
Neuromolecular Med ; 22(1): 100-110, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31482275

RESUMO

The hallmark of multiple sclerosis (MS) pathogenesis is the breakdown of peripheral tolerance in the immune system. However, its molecular mechanism is not completely understood. Since long non-coding RNAs (lncRNAs) has played important roles in regulation of immunological pathways, here, we evaluated the expression of a novel lncRNA, TOB1-AS1, and its putative associated coding genes in the mechanism of maintaining immune tolerance in peripheral blood of MS patients to assess their possible roles in MS pathogenesis. In this study, 39 MS patients and 32 healthy matched controls were recruited. Real-time PCR standard curve method was used to quantify transcript levels of TOB1-AS1, TOB1, SKP2, and TSG. In addition, the potential sex hormone receptor binding sites on target genes promoter were analyzed using JASPR software. This work demonstrates a negative correlation between TOB1-AS1 expression and EDSS of patients. Also, a robust dysregulation of co-expression of TOB1-AS1 lncRNA and the coding genes in MS patients compared to controls was observed. Such dysregulation in this pathway may be related to MS pathogenesis and response to interferon treatment.


Assuntos
Tolerância Imunológica/genética , Esclerose Múltipla Crônica Progressiva/genética , Esclerose Múltipla Recidivante-Remitente/genética , RNA Longo não Codificante/imunologia , Adulto , Simulação por Computador , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Interferon beta/uso terapêutico , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Esclerose Múltipla Crônica Progressiva/imunologia , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/imunologia , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/química , RNA Longo não Codificante/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Curva ROC , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Quinases Associadas a Fase S/biossíntese , Proteínas Quinases Associadas a Fase S/química , Proteínas Quinases Associadas a Fase S/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa