Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 12(3): 275, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33723215

ABSTRACT

Ovarian cancer (OC) causes more deaths than any other gynecological cancer. Many cellular pathways have been elucidated to be associated with OC development and progression. Specifically, the insulin-like growth factor 1 receptor/insulin receptor substrate 1 (IGF1R/IRS1) pathway participates in OC development. Moreover, accumulating evidence has shown that microRNA deregulation contributes to tumor initiation and progression. Here, our study aimed to investigate the molecular functions and regulatory mechanisms of miR-150, specifically, in OC. We found that the expression of miR-150-5p/3p and their precursor, mir-150, was downregulated in OC tissues; lower mir-150 levels were associated with poor OC patient outcomes. Ectopic mir-150 expression inhibited OC cell growth and metastasis in vitro and in vivo. Furthermore, both IRS1 and IGF1R were confirmed as direct targets of miR-150-5p/3p, and the miR-150-IGF1R/IRS1 axis exerted antitumor effects via the PI3K/AKT/mTOR pathway. Forkhead box protein 3 (FoxP3) positively regulated the expression of miR-150-5p/3p by binding to the mir-150 promoter. In turn, the PI3K/AKT/mTOR pathway downregulated FoxP3 and miR-150-5p/3p. Taken together, these findings indicate that a complex FoxP3-miR-150-IGF1R/IRS1-PI3K/AKT/mTOR feedback loop regulates OC pathogenesis, providing a novel mechanism for miR-150 as a tumor suppressor miRNA in OC.


Subject(s)
Cell Movement , Cell Proliferation , Forkhead Transcription Factors/metabolism , Insulin Receptor Substrate Proteins/metabolism , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , Receptor, IGF Type 1/metabolism , Animals , Apoptosis , Cell Line, Tumor , Feedback, Physiological , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Insulin Receptor Substrate Proteins/genetics , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Burden
2.
Int J Biol Sci ; 17(1): 236-246, 2021.
Article in English | MEDLINE | ID: mdl-33390846

ABSTRACT

MicroRNAs (miRNAs) are a class of short non-coding RNAs of approximately 22 nucleotides in length, which function by binding to the 3' UTR sequences of their target mRNAs. It has been reported that dysregulated miRNAs play pivotal roles in numerous diseases, including cancers, such as gastric, breast, colorectal, ovarian, and other cancers. Recent research efforts have been devoted to translating these basic discoveries into clinical applications that could improve the therapeutic outcome in patients with cancer. Early studies have shown that miR-340 may act either as an oncogene or a tumor suppressor by targeting genes related to proliferation, apoptosis, and metastasis, as well as those associated with diagnosis, treatment, chemoresistance, and prognosis. miR-340 has been shown to have a role in other diseases, such as autoimmune diseases, acute stroke, and alcoholic steatohepatitis. Nevertheless, the roles of miR-340 in human malignancies are still unclear, and the associated mechanisms are complex, involving a variety of signaling pathways, such as Wnt/ß-catenin and the JAK-STAT pathways. Herein, we review the crucial roles of miR-340 in human cancers through the analysis of the latest research studies, with the aim of clarifying miR-340 function in malignant disease diagnosis, treatment, and prognosis, and to propose further investigations.


Subject(s)
MicroRNAs/metabolism , Neoplasms/metabolism , Humans , Neoplasm Metastasis
3.
Rheumatology (Oxford) ; 60(6): 2979-2989, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33247940

ABSTRACT

OBJECTIVE: To determine the upregulation of IL-21-inducible genes in minor salivary glands (MSGs) in 28 primary SS (pSS) patients and 12 non-pSS subjects and correlate it with disease characteristics. METHODS: RNA sequencing was utilized to compare IL-21-inducible genes expression in the MSGs between pSS and non-pSS subjects. The subgroups were characterized according to the IL-21 score calculated by seven IL-21-inducible genes. Furthermore, the disease characteristics and transcripts implicated in hypoxia and interferon signalling were assessed in two pSS subgroups. RESULTS: We observed that the expression of the IL-21-inducible genes (IL-21, IL-21R, JAK3, STAT1, HLA-B, CCR7 and CXCL10), the so-called IL-21 signature genes, was significantly increased in pSS patients. The upregulation of JAK3 expression may be induced by hypomethylation of the JAK3 promoter in pSS patients and putatively associated with POU2F2. The patients with increased IL-21 signature gene expression showed an increased EULAR Sjögren's Syndrome Disease Activity Index score and increased enrichment of B cells, memory B cells, CD4+ T cells and CD8+ T cells. Furthermore, the IL-21 scores in the anti-SSA+, SSB+, ANA+ and high IgG samples were higher than those in the respective antibody-negative samples and normal IgG. In addition, we found both hypoxia and IFN-relevant genes showed strong correlation with IL-21 signature gene expression, indicating their interaction in pSS. CONCLUSION: IL-21 signature gene was associated with typical disease characteristics in pSS, which provides insight into the contribution of the IL-21 signalling pathway to the pathogenesis of the disease and might provide a novel treatment strategy for this subtype of pSS.


Subject(s)
Interleukins/genetics , Salivary Glands, Minor/pathology , Sjogren's Syndrome/genetics , Adult , Female , Gene Expression , Humans , Male , Middle Aged , Protein Interaction Maps , Up-Regulation
4.
J Exp Clin Cancer Res ; 38(1): 249, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31186036

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the third most frequent cancer and the second leading cause of cancer-related death worldwide. Increasing evidence indicates that the deregulation of long noncoding RNAs (lncRNAs) contributes to tumor initiation and progression; however, little is known about the biological role of cancer susceptibility candidate 9 (CASC9) in CRC. METHODS: Novel lncRNAs potentially involved in CRC tumorigenesis were identified from datasets downloaded from The Cancer LncRNome Atlas and The Atlas of Noncoding RNAs in Cancer. The CRC cell lines HCT-116, HCT-116 p53-/-, SW620, SW480, HT-29, LoVo, LS-174T, and RKO were used. Colony-formation, MTS, cell-cycle, apoptosis, and in-vivo tumorigenesis assays were used to determine the role of CASC9 in CRC cell growth in vitro and in vivo. Potential interaction between CASC9 and cleavage and polyadenylation specificity factor subunit 3 (CPSF3) was evaluated using RNA immunoprecipitation and RNA-protein pull-down assays. RNA-sequencing was performed to analyze gene expression following CASC9 knockdown. RT-qPCR, western blotting, and mRNA decay assays were performed to study the mechanisms involved. RESULTS: CASC9 was frequently upregulated in CRC, which was correlated with advanced TNM stage, and higher CASC9 levels were associated with poor patient outcomes. Knockdown of CASC9 inhibited growth and promoted apoptosis in CRC cells, whereas ectopic CASC9 expression promoted cell growth in vitro and in vivo. We demonstrated that CPSF3 is a CASC9-interacting protein, and knockdown of CPSF3 mimicked the effects of CASC9 knockdown in CRC cells. Furthermore, we found that CASC9 exerts its oncogenic activity by modulating TGFß2 mRNA stability and upregulating the levels of TGFß2 and TERT, resulting in an increase in phosphorylated SMAD3 and activation of TGF-ß signaling, and enhanced TERT complex function in CRC cells. Finally, CPSF3 was significantly upregulated in CRC tissues as compared with adjacent or non-adjacent normal colon tissues, and CASC9, CPSF3, and TGFß2 levels in human CRC tissues were positively correlated. CONCLUSIONS: CASC9 is a promising prognostic predictor for patients with CRC and the CASC9-CPSF3-TGFß2 axis is a potential therapeutic target for CRC treatment.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , RNA, Long Noncoding/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Aged , Aged, 80 and over , Apoptosis/genetics , Biomarkers, Tumor , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , RNA Interference , Tumor Burden
5.
Cell Death Dis ; 10(5): 372, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31068580

ABSTRACT

Although increasing evidence indicated that deregulation of microRNAs (miRNAs) contributed to tumor initiation and progression, but little is known about the biological role of miR-340 in ovarian cancer (OC). In this study, we found that miR-340 expression was downregulated in OC tissues compared with its expression in normal ovarian epithelium and endometrium, and treatment with 5-aza-2'-deoxycytidine (5-Aza-dC) or trichostatin A (TSA) increased miR-340 expression in OC cells. In addition, ectopic miR-340 expression inhibited OC cell growth and metastasis in vitro and in vivo. Four and a half LIM domains protein 2 (FHL2) was confirmed as a direct target of miR-340 and silencing FHL2 mimicked the effects of miR-340 in OC cells. Further mechanistic study showed that miR-340 inhibited the Wnt/ß-catenin pathway by targeting FHL2, as well as downstream cell cycle and epithelial-to-mesenchymal transition (EMT) signals in OC cells. Moreover, the greatest association between miR-340 and FHL2 was found in 481 ovarian serous cystadenocarcinoma tissues via pan-cancer analysis. Finally, we revealed that lower miR-340 or higher FHL2 was associated with poor OC patient outcomes. Our findings indicate that the miR-340-FHL2 axis regulates Wnt/ß-catenin signaling and is involved in tumorigenesis in OC. Therefore, manipulating the expression of miR-340 or its target genes is a potential strategy in OC therapy.


Subject(s)
Cell Proliferation , LIM-Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Muscle Proteins/metabolism , Ovarian Neoplasms/pathology , Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Movement , Cystadenocarcinoma/metabolism , Cystadenocarcinoma/pathology , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Humans , LIM-Homeodomain Proteins/antagonists & inhibitors , LIM-Homeodomain Proteins/genetics , Mice , Mice, Nude , MicroRNAs/chemistry , MicroRNAs/genetics , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Ovarian Neoplasms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...