Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Cell Int ; 22(1): 162, 2022 Apr 23.
Article in English | MEDLINE | ID: mdl-35461277

ABSTRACT

BACKGROUND: Piezo1 has been revealed to play a regulatory role in vascular development and progression of variety tumors. However, whether and how the progression of hepatocellular carcinoma (HCC) regulated by Piezo1 remains elusive. This study aimed to elucidate the effect and mechanisms of Piezo1 in HCC. METHODS: The mRNA and protein expression level of Piezo1 in HCC samples and cell lines was determined by qRT-PCR, western blot and immunohistochemistry analyses. Two independent study cohorts containing 280 patients were analyzed to reveal the association between Piezo1 expression and clinicopathological characteristics. Series of in vitro and in vivo experiments were used to validate the function of Piezo1 in HCC. Gene set enrichment analysis (GSEA) was performed to explore the signaling pathway of Piezo1. Immunoprecipitation, immunofluorescence and in vitro and in vivo experiments were used to explore the molecular mechanism of Piezo1 in HCC progression. RESULTS: Our results demonstrated the Piezo1 expression was significantly upregulated in HCC tissues and cell lines, and upregulation of Piezo1 closely correlated with aggressive clinicopathological features and poor prognosis. Knockdown of Piezo1 in HCCLM3 and Hep3B cells significantly restrained proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of HCC cells in vitro, and tumor growth, metastasis, EMT in vivo. TGF-ß signaling pathway was most significant enriched pathway in GSEA. Finally, tumor promotion effect of Piezo1 was found to exerted through recruiting and combining Rab5c to activating TGF-ß signaling pathway. CONCLUSIONS: Piezo1 significantly related to poor prognosis and promotes progression of hepatocellular carcinoma via activating TGF-ß signaling, which suggesting that Piezo1 may serve as a novel prognostic predictor and the potential therapeutic target for HCC patients.

2.
Carcinogenesis ; 42(11): 1357-1369, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34657150

ABSTRACT

Gαq subfamily proteins play critical roles in many biological functions including cardiovascular development, angiogenesis, and tumorigenesis of melanoma. However, the understanding of G Protein Subunit Alpha 14 (GNA14) in diseases, especially in cancers is limited. Here, we revealed that GNA14 was significantly low expression in Human hepatocellular carcinoma (HCC) samples. Low GNA14 expression was correlated with aggressive clinicopathological features. Moreover, the overall survival (OS) and disease-free survival (DFS) of high GNA14 expression HCC patients were much better than low GNA14 expression group. Lentivirus-mediated GNA14 knockdown significantly promoted the growth of liver cancer in vitro and in vivo. However, opposing results were observed when GNA14 is upregulated. Mechanistically, We identified receptor for activated C kinase 1 (RACK1) as a binding partner of GNA14 by co-immunoprecipitation and mass spectrometry (MS). Glutathione-S-transferase (GST) pull-down assay further verified the direct interaction between GNA14 and RACK1. RNA-Seq and loss- and gain-of-function assays also confirmed that GNA14 reduced the activity of both MAPK/JNK and PI3K/AKT signaling pathways through RACK1. GNA14 synergized with U73122 (PLC inhibitor) to enhance this effect. Further studies suggested that GNA14 potentially competed with protein kinase C (PKC) to bind with RACK1, consequently reducing the stability of PKC. Moreover, we also showed that GNA14'supression of p-AKT protein level depended on sufficient RACK1 expression. In conclusion, we indicated a different role of GNA14, which acted as a suppressor inhibiting liver cancer progression through MAPK/JNK and PI3K/AKT signaling pathways. Due to this, GNA14 served as a potentially valuable prognostic biomarker for liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Liver Neoplasms/metabolism , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors for Activated C Kinase/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Humans , Liver Neoplasms/pathology , Prognosis , Protein Kinase C/metabolism , Signal Transduction
3.
Int J Gen Med ; 14: 4897-4911, 2021.
Article in English | MEDLINE | ID: mdl-34475781

ABSTRACT

PURPOSE: Patients with gastric cancer (GC) often die from metastasis. However, the exact molecular mechanism underlying GC metastasis is complicated and still remains elusive. Epidermal growth factor, latrophilin and seven-transmembrane domain-containing 1 (ELTD1), has been reported to be involved in cancer metastasis, but its role in GC is still missing. PATIENTS AND METHODS: We first analyzed the expression of ELTD1 in GC using public databases (TCGA, Oncomine, and GEO) and our clinical samples. The functions of ELTD1 in GC proliferation, invasion and metastasis were determined by in vitro and in vivo experiments. The functional mechanism of ETLD1 in GC was also investigated. Finally, the association between ELTD1 expression and the overall survival of GC patients was analyzed using public databases. RESULTS: ELTD1 is significantly upregulated in GC tissues. Knockdown of ELTD1 inhibits GC cell proliferation, migration and invasion in vitro as well as tumor growth and metastasis in vivo, while ELTD1 overexpression obtains opposite results. Moreover, ELTD1 could promote epithelial to mesenchymal transition (EMT) in GC. Mechanistically, ELTD1 exerts its tumor-promoting effect by activating MAPK/ERK signaling. Subsequent studies demonstrated that ELTD1 could interact with C-terminal Src kinase (CSK) and inhibit its expression, which finally lead to MAPK/ERK activation. Data from TGCA and GEO both revealed that GC patients with high ELTD1 expression had poorer prognosis and the combination of ELTD1 with CSK showed better predictive performance. CONCLUSION: ELTD1 plays an oncogene role in GC through MAPK/ERK signaling via inhibiting CSK, which may be a useful prognostic predictor and potential therapeutic target for GC.

4.
Cell Death Dis ; 12(9): 849, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521817

ABSTRACT

Tumour metastasis is the main cause of postoperative tumour recurrence and mortality in patients with hepatocellular carcinoma (HCC), but the underlying mechanism remains unclear. Accumulating evidence has demonstrated that programmed cell death 10 (PDCD10) plays an important role in many biological processes. However, the role of PDCD10 in HCC progression is still elusive. In this study, we aimed to explore the clinical significance and molecular function of PDCD10 in HCC. PDCD10 is significantly upregulated in HCC, which also correlates with aggressive clinicopathological characteristics and predicts poor prognosis of HCC patients after liver resection. High PDCD10 expression promotes HCC cell proliferation, migration, and invasion in vitro and tumour growth, metastasis in vivo. In addition, PDCD10 could facilitate epithelial-to-mesenchymal transition (EMT) of HCC cells. In terms of the mechanism, PDCD10 directly binds to the catalytic subunit of protein phosphatase 2A (PP2Ac) and increases its enzymatic activity, leading to the interaction of YAP and dephosphorylation of the YAP protein. This interaction contributes to YAP nuclear translocation and transcriptional activation. PP2Ac is necessary for PDCD10-mediated HCC progression. Knocking down PP2Ac abolished the tumour-promoting role of PDCD10 in the migration, invasion and EMT of HCC. Moreover, a PP2Ac inhibitor (LB100) could restrict tumour growth and metastasis of HCC with high PDCD10 expression. Collectively, PDCD10 promotes EMT and the progression of HCC by interacting with PP2Ac to promote YAP activation, which provides new insight into the mechanism of cancer metastasis. PDCD10 may be a potential prognostic biomarker and therapeutic target for HCC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition , Liver Neoplasms/pathology , Membrane Proteins/metabolism , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins/metabolism , YAP-Signaling Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/pathology , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Male , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Biological , Neoplasm Invasiveness , Prognosis , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics , Wound Healing
5.
Mol Cancer Res ; 19(9): 1441-1453, 2021 09.
Article in English | MEDLINE | ID: mdl-34158395

ABSTRACT

Arginyltransferase (ATE1) plays critical roles in many biological functions including cardiovascular development, angiogenesis, adipogenesis, muscle contraction, and metastasis of cancer. However, the role of ATE1 in hepatocellular carcinoma (HCC) remains unknown. In this study, we find that ATE1 plays an essential role in growth and malignancy of liver cancer. ATE1 expression is significantly reduced in human HCC samples compared with normal liver tissue. In addition, low ATE1 expression is correlated with aggressive clinicopathologic features and is an independent poor prognostic factor for overall survival and disease-free survival of patients with HCC. Lentivirus-mediated ATE1 knockdown significantly promoted liver cancer growth, migration, and disease progression in vitro and in vivo. Opposing results were observed when ATE1 was upregulated. Mechanistically, ATE1 accelerated the degradation of ß-catenin and inhibited Wnt signaling by regulating turnover of Regulator of G Protein Signaling 5 (RGS5). Loss- and gain-of-function assays confirmed that RGS5 was a key effector of ATE1-mediated regulation of Wnt signaling. Further studies indicated that RGS5 might be involved in regulating the activity of GSK3-ß, a crucial component of the cytoplasmic destruction complex. Treatment with a GSK inhibitor (CHIR99021) cooperated with ablation of ATE1 or RGS5 overexpression to promote Wnt/ß-catenin signaling, but overexpression of ATE1 or RGS5 knockdown did not reverse the effect of GSK inhibitor. IMPLICATIONS: ATE1 inhibits liver cancer progression by suppressing Wnt/ß-catenin signaling and can serve as a potentially valuable prognostic biomarker for HCC.


Subject(s)
Aminoacyltransferases/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , RGS Proteins/metabolism , Wnt1 Protein/metabolism , beta Catenin/metabolism , Aminoacyltransferases/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , RGS Proteins/genetics , Survival Rate , Tumor Cells, Cultured , Wnt1 Protein/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
6.
Cancer Lett ; 506: 128-141, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33705863

ABSTRACT

Metastasis remains the major obstacle of improving the survival of patients with hepatocellular carcinoma (HCC). Epithelial-mesenchymal transition (EMT) is critical to cancer metastasis. Successful induction of EMT requires dramatic cytoskeleton rearrangement. However, the significance of microtubule (MT), one of the core components of cell cytoskeleton, in this process remains largely unknown. Here we revealed that STMN2, an important MT dynamics regulator, is barely expressed in normal live tissues but markedly up-regulated in HCCs, especially in those with early recurrence. High STMN2 expression correlates with aggressive clinicopathological features and predicts poor prognosis of HCC patients. STMN2 overexpression in HCC cells promotes EMT, invasion and metastasis in vitro and in vivo, whereas STMN2 knockdown has opposite results. Mechanistically, STMN2 modulates MTs disassembly, disrupts MT-Smad complex, and facilitates release from MT network, phosphorylation and nuclear translocation of Smad2/3 even independent of TGFß stimulation, thereby enhancing TGFß signaling. Collectively, STMN2 orchestrates MT disassembly to facilitate EMT via TGF-ß signaling, providing a novel insight into the mechanisms underlying cancer metastasis. STMN2 is a promising prognostic biomarker and potential therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Smad2 Protein/genetics , Stathmin/genetics , Transforming Growth Factor beta/genetics , Active Transport, Cell Nucleus/genetics , Aged , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Signal Transduction/genetics , Smad3 Protein/genetics
7.
Clin Sci (Lond) ; 135(7): 847-864, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33755094

ABSTRACT

Metastasis is the main cause of poor postoperative survival of hepatocellular carcinoma (HCC) patients. Cytoskeleton rearrangement is a key event in cancer metastasis. However, the significance of microtubule (MT), one of the core components of cytoskeleton, in this process is only beginning to be revealed. Here, we find that the MT dynamics regulator end-binding protein 2 (EB2) is highly expressed in HCC and predicts poor prognosis of HCC patients. Functional studies show that EB2 overexpression promotes HCC proliferation, invasion and metastasis in vitro and in vivo, while EB2 knockdown has opposite results. Mechanistically, EB2 mediates MTs destabilization, increases Src (Src proto-oncogene non-receptor tyrosine kinase) activity, and thus facilitates extracellular signal-regulated kinase (ERK) signaling activation, which could in turn promote EB2 expression in HCC, eventually resulting in enhanced HCC proliferation, invasion and metastasis. Furthermore, U0126, a specific ERK inhibitor, could effectively inhibit EB2-mediated HCC proliferation and metastasis in vitro and in vivo. In conclusion, EB2 coordinates MT cytoskeleton and intracellular signal transduction, forming an EB2-MT-ERK positive feedback loop, to facilitate HCC proliferation, invasion and metastasis. EB2 could serve as a promising prognostic biomarker and potential therapeutic target for HCC; HCC patients with high EB2 expression may benefit from treatment with ERK inhibitors.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System , Microtubules/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/metabolism , Middle Aged , Neoplasm Metastasis , Proto-Oncogene Mas , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...