Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Pharmacol Res ; 195: 106863, 2023 09.
Article in English | MEDLINE | ID: mdl-37480971

ABSTRACT

Human papillomavirus (HPV) infection is a causative agent of cervical cancer (CC). N6-methyladenosine (m6A) modification is implicated in carcinogenesis and tumor progression. However, the involvement of m6A modification in HPV-involved CC remains unclear. Here we showed that HPV E6/7 oncoproteins affected the global m6A modification and E7 specifically promoted the expression of ALKBH5. We found that ALKBH5 was significantly upregulated in CC and might serve as a valuable prognostic marker. Forced expression of ALKBH5 enhanced the malignant phenotypes of CC cells. Mechanistically, we discovered that E7 increased ALKBH5 expression through E2F1-mediated activation of the H3K27Ac and H3K4Me3 histone modifications, as well as post-translational modification mediated by DDX3. ALKBH5-mediated m6A demethylation enhanced the expression of PAK5. The m6A reader YTHDF2 bound to PAK5 mRNA and regulated its stability in an m6A-dependent manner. Moreover, ALKBH5 promoted tumorigenesis and metastasis of CC by regulating PAK5. Overall, our findings herein demonstrate a significant role of ALKBH5 in CC progression in HPV-positive cells. Thus, we propose that ALKBH5 may serve as a prognostic biomarker and therapeutic target for CC patients.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Papillomavirus Infections/genetics , Carcinogenesis/genetics , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism
2.
Cell Signal ; 110: 110803, 2023 10.
Article in English | MEDLINE | ID: mdl-37437827

ABSTRACT

Renal cell carcinoma (RCC) is an aggravating cancer with a poor prognosis and a high rate of metastasis. PAK5, a p21-activated kinases, has shown to be overexpressed in a variety of cancers, including RCC. In previous studies, we discovered that PAK5 regulates cell migration and invasion in RCC cell lines. However, the underlying mechanisms remain obscure. In this study, we consolidated that PAK5 confers a pro-metastatic phenotype RCC cells in vitro and exacerbates metastasis in vivo. High PAK5 expression was associated with an advanced TNM stage and a lower overall survival. Furthermore, PAK5 increases the expression level of N-cadherin. In terms of mechanism, PAK5 bound to Slug and phosphorylated it at serine 87. As a result, phosphorylated Slug transactivated N-cadherin, accelerating the epithelial-mesenchymal transition. Collectively, Slug is a novel PAK5 substrate, and PAK5-mediated phosphorylation of Slug-S87 increases N-cadherin and the pro-metastatic phenotype of RCC, implying that phosphorylated Slug-S87 could be a therapeutic target in progressive RCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Snail Family Transcription Factors , Humans , Cadherins/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Phosphorylation , Snail Family Transcription Factors/metabolism , Transcriptional Activation
3.
Clin Transl Med ; 12(11): e1113, 2022 11.
Article in English | MEDLINE | ID: mdl-36394206

ABSTRACT

BACKGROUND: Dysregulation of alternative splicing (AS) induced by serine/arginine-rich proteins has recently been linked to cancer metastasis. Nonetheless, as a member of the serine/arginine-rich protein family, the involvement of SRSF11 in colorectal cancer (CRC) is unknown. METHODS: The TCGA dataset and clinical samples were used to assess SRSF11 expression levels in CRC. For SRSF11, functional experiments were conducted both in vitro and in vivo. RNA-seq technology was used to analyze and screen SRSF11-triggered AS events, which were then confirmed by in vivo UV crosslinking and immunoprecipitation (CLIP) and mini-gene reporter assays. Jalview software was used to determine the preferential binding motif with relation to exon skipping (ES) events. Furthermore, coimmunoprecipitation (Co-IP) and Phospho-tag SDS-PAGE experiments were used to investigate PAK5-mediated phosphorylation regulation on SRSF11, and in vitro kinase experiments validated the interaction. RESULTS: In CRC, SRSF11 was discovered to be overexpressed and associated with a poor prognosis. And SRSF11 played a pro-metastatic role in vitro and in vivo. By screening SRSF11-regulated AS events, we identified the binding motif of SRSF11-triggered splicing-switching of HSPA12A AS, which specifically regulated HSPA12A AS by directly binding to a motif in exon 2. Mechanistically, the HSPA12A transcript with exon 2 retention increased N-cadherin expression by promoting RNA stability. Furthermore, the oncogenic kinase PAK5 phosphorylated SRSF11 at serine 287, protecting it from ubiquitination degradation. CONCLUSIONS: SRSF11 exerts pro-metastatic effects in CRC by inhibiting the AS of HSPA12A pre-RNA. Our findings point to SRSF11-regulated HSPA12A splicing as a novel relationship between SRSF11-regulated splicing and CRC metastasis and suggest a PAK5/SRSF11/HSPA12A axis as a potential therapeutic target and prognostic biomarker in CRC.


Subject(s)
Alternative Splicing , Colorectal Neoplasms , Humans , Alternative Splicing/genetics , Arginine/genetics , Arginine/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , RNA/metabolism , Serine/genetics , Serine/metabolism
4.
DNA Cell Biol ; 41(9): 790-809, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35947859

ABSTRACT

[Formula: see text] Alternative splicing (AS) can generate distinct transcripts and subsequent isoforms that play differential functions from the same pre-mRNA. Recently, increasing numbers of studies have emerged, unmasking the association between AS and cancer. In this review, we arranged AS events that are closely related to cancer progression and presented promising treatments based on AS for cancer therapy. Obtaining proliferative capacity, acquiring invasive properties, gaining angiogenic features, shifting metabolic ability, and getting immune escape inclination are all splicing events involved in biological processes. Spliceosome-targeted and antisense oligonucleotide technologies are two novel strategies that are hopeful in tumor therapy. In addition, bioinformatics applications based on AS were summarized for better prediction and elucidation of regulatory routines mingled in. Together, we aimed to provide a better understanding of complicated AS events associated with cancer biology and reveal AS a promising target of cancer treatment in the future.


Subject(s)
Alternative Splicing , Neoplasms , Alternative Splicing/genetics , Computational Biology , Humans , Neoplasms/drug therapy , Neoplasms/therapy , RNA Precursors/genetics , RNA Precursors/therapeutic use , Spliceosomes/genetics
5.
J Biomed Sci ; 28(1): 56, 2021 Aug 02.
Article in English | MEDLINE | ID: mdl-34340705

ABSTRACT

BACKGROUND: Sorafenib is a kinase inhibitor that is used as a first-line therapy in advanced hepatocellular carcinoma (HCC) patients. However, the existence of sorafenib resistance has limited its therapeutic effect. Through RNA sequencing, we demonstrated that miR-138-1-3p was downregulated in sorafenib resistant HCC cell lines. This study aimed to investigate the role of miR-138-1-3p in sorafenib resistance of HCC. METHODS: In this study, quantitative real-time PCR (qPCR) and Western Blot were utilized to detect the levels of PAK5 in sorafenib-resistant HCC cells and parental cells. The biological functions of miR-138-1-3p and PAK5 in sorafenib-resistant cells and their parental cells were explored by cell viability assays and flow cytometric analyses. The mechanisms for the involvement of PAK5 were examined via co-immunoprecipitation (co-IP), immunofluorescence, dual luciferase reporter assay and chromatin immunoprecipitation (ChIP). The effects of miR-138-1-3p and PAK5 on HCC sorafenib resistant characteristics were investigated by a xenotransplantation model. RESULTS: We detected significant down-regulation of miR-138-1-3p and up-regulation of PAK5 in sorafenib-resistance HCC cell lines. Mechanistic studies revealed that miR-138-1-3p reduced the protein expression of PAK5 by directly targeting the 3'-UTR of PAK5 mRNA. In addition, we verified that PAK5 enhanced the phosphorylation and nuclear translocation of ß-catenin that increased the transcriptional activity of a multidrug resistance protein ABCB1. CONCLUSIONS: PAK5 contributed to the sorafenib resistant characteristics of HCC via ß-catenin/ABCB1 signaling pathway. Our findings identified the correlation between miR-138-1-3p and PAK5 and the molecular mechanisms of PAK5-mediated sorafenib resistance in HCC, which provided a potential therapeutic target in advanced HCC patients.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm , Liver Neoplasms/drug therapy , MicroRNAs/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sorafenib/pharmacology , Animals , Antineoplastic Agents/pharmacology , Female , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred BALB C , Mice, Nude
7.
Cancer Manag Res ; 11: 10411-10418, 2019.
Article in English | MEDLINE | ID: mdl-31849530

ABSTRACT

MicroRNAs (miRNAs) are small non-coding and highly conserved RNAs that act in biological processes including cell proliferation, invasion, apoptosis, metabolism, signal transduction, and tumorigenesis. The previously identified miRNA-326 (miR-326) has been reported to participate in cellular apoptosis, tumor growth, cell invasion, embryonic development, immunomodulation, chemotherapy resistance, and oncogenesis. This review presents a detailed overview of what is known about the effects of miR-326 on cell invasion, metastasis, drug resistance, proliferation, apoptosis, and its involvement in signaling pathways.

8.
Cell Death Differ ; 26(6): 994-1006, 2019 06.
Article in English | MEDLINE | ID: mdl-30082769

ABSTRACT

p21-activated kinase 5 (PAK5) is involved in several oncogenic signaling pathways and its amplification or overexpression has been found in various types of cancer; however, the pathophysiologic role of PAK5 in cervical cancer (CC) remains elusive. This study aims to elucidate the effects of PAK5 on CC metastasis and its specific regulation mechanism. We performed western blotting and immunohistochemistry (IHC) analysis and found that the expression levels of PAK5 were significantly upregulated in CC cells and tissues. In addition, statistical analysis via IHC showed that increased PAK5 significantly correlated with CC progression. Mn2+-Phos-tag SDS-PAGE, western blotting, immunofluorescence and dual luciferase reporter assays were utilized to determine the involvement of SATB1 in PAK5-mediated epithelial-mesenchymal transition (EMT). We found that PAK5-mediated special AT-rich binding protein-1 (SATB1) phosphorylation on Ser47 initiated EMT cascade and promoted migration and invasion of CC cells. Furthermore, overexpression of PAK5 induced lung metastasis of CC cells in xenograft modes. Taken together, we conclude that PAK5 is a novel prognostic indicator and plays an important role in the CC metastasis.


Subject(s)
Matrix Attachment Region Binding Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , p21-Activated Kinases/metabolism , Cell Movement , Cell Proliferation , Female , HeLa Cells , Humans , Phosphorylation , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology , Wound Healing
9.
Oncol Lett ; 15(4): 5137-5142, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29552148

ABSTRACT

c-Jun activation domain-binding protein-1 (Jab1) is a multifunctional protein involved in cell proliferation and apoptosis, DNA damage and repair and genome stability. In a number of types of human carcinoma, the abnormal expression of Jab1 is associated with poor prognosis, suggesting that Jab1 serves a vital function in tumorigenesis. However, the functional effects and the underlying molecular mechanisms of Jab1 in laryngeal squamous cell carcinoma (LSCC) progression remain poorly understood. The results of the present study demonstrate that downregulating Jab1 expression promotes LSCC apoptosis while inhibiting the proliferation of LSCC cells. Furthermore, Jab1 inhibition results in decreased protein kinase B phosphorylation accompanied by increased caspase-3 cleavage and p53 expression. It has been identified that the increased expression of Jab1 is markedly associated with LSCC progression, therefore Jab1 may be used as a novel target for the treatment of laryngeal cancer.

10.
Cell Death Dis ; 8(10): e3155, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29072688

ABSTRACT

MicroRNA-106a-5p (MiR-106a-5p), a small non-coding RNA, has been reported to be downregulated in astrocytoma, osteosarcoma and colorectal cancer. However, the expression levels and biological function in renal cell carcinoma (RCC) have not been studied yet. In this study, we found that the miR-106a-5p was significantly downregulated in RCC tissues and cell lines, and that overexpression of miR-106a-5p led to decreased cell metastasis ability in a xenograft model. Inhibition of miR-106a-5p in RCC cell lines altered the cell migration, invasion and wound healing abilities. Mechanistic studies demonstrated that miR-106a-5p directly bound to the 3'-UTR of the PAK5 mRNA and mediated a decrease in the protein expression of PAK5. We further proved that PAK5 protein levels were negatively correlated with the miR-106a-5p expression in both patient samples and xenograft model. In epigenetics, methylation specific PCR experiments indicated that the upstream gene promoter of miR-106a-5p was hypermethylated in RCC, which might be responsible for its downregulation. Our findings suggested that miR-106a-5p might be a potential gene therapy target for the treatment of RCC metastasis.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , MicroRNAs/genetics , p21-Activated Kinases/genetics , Animals , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement , Down-Regulation , Female , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neoplasm Invasiveness , Transfection , p21-Activated Kinases/metabolism
11.
J Exp Clin Cancer Res ; 36(1): 146, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29041983

ABSTRACT

BACKGROUND: Abnormal proliferation is significantly associated with the promotion of malignant tumor. Growing evidence suggest that the signal pathways of p21cdc42/rac1-activated kinase 5 (PAK5) have been found in various tumor progression, however, the role of PAK5 in breast cancer remains largely unclear. METHODS: We evaluated PAK5 and p65 staining in breast cancer tissues (BCTs) and paired non-cancerous tissues (NTs) using tissue microarray (TMA) technology. The functions of PAK5 were studied in vitro and in vivo. Cell Counting Kit-8 (CCK-8) and flow cytometry were performed to determine proliferation of breast cancer cells. Phosphorylation assay and co-immunoprecipitation (co-IP) were employed to identify the regulation mechanism of p65 by PAK5. The activation of Cyclin D1 promoter was measured with luciferase reporter assay. Xenograft models in nude mice were established to explore the roles of PAK5 in breast cancer growth. RESULTS: In this study, we show that PAK5 is highly expressed in breast cancer tissues and the increased PAK5 is significantly associated with breast cancer progression. Overexpression of PAK5 promotes the proliferation and cell-cycle progression by increasing the expression of Cyclin D1 in vitro and in vivo. Mechanistic studies demonstrated that PAK5 can promote the phosphorylation and the nuclear translocation of p65 subunit of nuclear factor-kappaB (NF-κB). Furthermore, p65 can directly bind to the promoter of Cyclin D1 and mediate an increase in its protein expression. CONCLUSIONS: Taken together, our findings suggest that PAK5 may serve as a potential prognosis marker and therapeutic target for human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Transcription Factor RelA/metabolism , p21-Activated Kinases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin D1/metabolism , Disease Models, Animal , Female , Follow-Up Studies , Gene Expression , Genes, Reporter , Heterografts , Humans , Immunohistochemistry , Mice , Neoplasm Staging , Phosphorylation , Prognosis , Promoter Regions, Genetic , Protein Transport , Signal Transduction , Transcriptional Activation , Tumor Burden , p21-Activated Kinases/genetics
12.
Sci Rep ; 7(1): 6623, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747626

ABSTRACT

Rap2a, a member of the small GTPase superfamily, belongs to Ras superfamily, and its function in cancer progression is still poorly understood. Our previous study indicated that the ectopic expression of Rap2a enhanced the migration and invasion ability of lung cancer cells. However, its expression and molecular mechanism on renal cell carcinoma (RCC) have not been characterized. This study explored the clinical significance and biological function of Rap2a in human RCC. The clinical relevance of Rap2a in RCC was evaluated by immunohistochemical staining using tissue microarray. Our data showed that Rap2a expression was dramatically increased in RCC tissues compared with normal renal tissues. The ectopic expression of Rap2a enhanced the migration and invasive ability of cancer cells. In contrast, downregulation of Rap2a inhibited cell invasion. Rap2a had no effect on the proliferation of RCC cell lines. Meanwhile, Rap2a can regulate the phosphorylation level of Akt in vitro. In vivo studies also showed that Rap2a positively regulated metastasis of renal cancer cells and the expression of p-Akt. These findings indicate that Rap2a promotes RCC metastasis and may serve as a candidate RCC prognostic marker and a potential therapeutic target.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/pathology , rap GTP-Binding Proteins/analysis , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Tissue Array Analysis
13.
Anticancer Agents Med Chem ; 17(7): 896-901, 2017.
Article in English | MEDLINE | ID: mdl-27604574

ABSTRACT

BACKGROUND: Deleted in liver cancer 1 (DLC-1) In human was originally isolated from rats brain and was often found to be deleted in hepatocellular carcinoma (HCC). METHODS: We undertook a structured search of bibliographic databases for peer-reviewed research literature using a focused review question and inclusion/exclusion criteria. RESULTS: Subsequent studies have demonstrated that DLC-1 is generally expressed in normal human tissues as well as in rats, while it always exists inactivated or even lost in many human cancers, which characterizes DLC-1 as a potential tumor suppressor. Additionally, the RhoGAP (Rho-GTPase activating proteins) activity was found to play a pivotal role in regulating DLC-1. CONCLUSION: Although emerging studies in a variety of cancers have identified DLC-1 and its downstream signaling molecules as potential therapeutic targets for treatments of DLC-1-related cancers, the mechanisms linked to DLC-1 remain undefined.


Subject(s)
GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Female , GTPase-Activating Proteins/analysis , GTPase-Activating Proteins/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/pathology , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/metabolism
14.
Bioorg Med Chem Lett ; 26(22): 5373-5377, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27780637

ABSTRACT

MicroRNAs (miRNAs), which are characterized by highly conserved and small non-coding RNAs, have been a hot spot regarding biological processes such as cellular proliferation, apoptosis and metabolism as well as cellular differentiation, signal transduction and carcinogenesis. MiRNA-106a (miR-106a), a member of the miR-17 family, has been validated to be aberrantly regulated in the diversity of tumors. The purpose of this review is supposed to deliver an intricate overview of miR-106a, including its role in cell proliferation, apoptosis, cell cycle, invasion and metastasis, involvement in drug resistance as well as its interactions with the target proteins and signaling pathways involved.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Animals , Cell Proliferation , Drug Resistance, Neoplasm , Humans , MicroRNAs/analysis , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...