Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Phytomedicine ; 129: 155688, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38728920

ABSTRACT

BACKGROUND: Malignant breast cancer cells trigger the over-activation of osteoclast precursor cells, leading to bone loss and severe pain. Targeted inhibition of osteoclast differentiation has emerged as an important strategy for treating bone syndromes induced by breast cancer. PURPOSE: The objective is to discover natural osteoclast inhibitor to treat osteoclastogenesis and bone destruction induced by breast cancer, and clarify the specific mechanisms. METHODS: Recepteur d'origine Nantais (RON) protein was employed to search the natural osteoclast inhibitor for breast cancer-induced osteoclastogenesis by molecular docking, molecular dynamics simulation and cellular thermal shift assay (CETSA). In the in vitro experiment, breast cancer MDA-MB-231 cell-conditioned medium (MDA-MB-231 CM) was used to induce osteoclastogenesis in murine bone marrow-derived macrophages (BMMs), aiming to elucidate the effects and mechanisms of the natural osteoclast inhibitor. In the in vivo model, MDA-MB-231 cells was injected into the mouse tibia to evaluate the therapeutic effect of drug on breast cancer-induced bone destruction. RESULTS: We discovered a significant increase in the expression of RON during MDA-MB-231 CM-induced osteoclast differentiation in vitro. Molecular docking analysis found that oroxylin A (OA), a flavonoid derived from the Chinese medicine Scutellaria baicalensis Georgi, showed binding ability with RON, while its impact and mechanism on breast cancer-induced osteoclastogenesis and osteolysis remains unclear. Molecular dynamics simulation and CETSA further revealed that OA bound directly to the RON protein, and it also decreased RON expression in breast cancer CM-induced osteoclastogenesis. Correspondingly, OA suppressed the MDA-MB-231 CM-induced osteoclastogenesis and bone resorption in vitro. The downstream signals of RON including Src and NFATc1, as well as the osteoclast-specific genes, were downregulated by OA. Of interesting, the suppressive effect of OA on osteoclastogenesis induced by MDA-MB-231 CM was abolished after RON was knocked down by the specific RON-siRNA, this further confirmed that OA showed inhibitory effects on osteoclasts through targeting RON. In addition, we found that OA attenuated MDA-MB-231 cell-induced osteolysis and reduced the number of osteoclasts in vivo. CONCLUSION: Our results indicate that OA acts as a natural RON inhibitor to suppress breast cancer-induced osteoclastogenesis and osteolysis. This provides new strategy for treating breast cancer-induced bone destruction and related syndromes.


Subject(s)
Breast Neoplasms , Flavonoids , Molecular Docking Simulation , Osteoclasts , Osteogenesis , Osteolysis , Animals , Female , Humans , Mice , Breast Neoplasms/drug therapy , Cell Differentiation/drug effects , Cell Line, Tumor , Flavonoids/pharmacology , Macrophages/drug effects , Mice, Inbred BALB C , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteolysis/drug therapy , Receptor Protein-Tyrosine Kinases , Mice, Nude
2.
J Adv Res ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38556044

ABSTRACT

INTRODUCTION: Breast cancer-related bone metastasis can lead to skeletal-related events (SREs), which decrease patient quality of life. Inhibition of osteoclastogenesis is a key treatment for SREs; however, the availability of clinical drugs remains limited, and all existing ones disrupt physiological bone formation, while exhibiting no effect on patient survival time. OBJECTIVES: This study aimed to identify a novel osteoclast inhibitor for the treatment of breast cancer-induced SREs. METHODS: The MDA-MB-231 breast cancer cell-induced bone loss model was used to investigate the therapeutic effects of erianin in vivo. Then, we evaluated the inhibitory effects of erianin on osteoclastogenesis and signalling in bone marrow-derived macrophages (BMMs) induced by conditioned medium from MDA-MB-231 breast cancer cells (231 CM) and receptor activator of nuclear factor-κB ligand (RANKL) in vitro. Next, a Cellular Thermal Shift Assay and siRNA-mediate knockdown were performed, to investigate the target of erianin during osteoclast formation. The effects of erianin on human osteoclastogenesis were evaluated using CD14+ monocytes obtained from patients with breast cancer. RESULTS: Erianin effectively improved breast cancer cells-induced bone destruction at doses of 2 and 20 mg/kg/day in vivo, while suppressing osteoclastogenesis and the upregulation of SRC-NFATc1, INTEGRIN ß3-MMP9 signals induced by 231 CM and RANKL in vitro. Furthermore, erianin interacted with NFATc1 but not SRC, and Nfatc1 knockdown eliminated the inhibitory effects of erianin on osteoclastogenesis. Notably, lower expression of NFATc1 positively correlated with longer survival in patients with cancer and a high risk of bone metastasis. We further revealed that 62.5-250 nM erianin suppresses NFATc1 and excessive osteoclastogenesis in CD14+ monocytes from patients with breast cancer. CONCLUSION: Erianin acts as an NFATc1 inhibitor that attenuates breast cancer-induced osteoclastogenesis and bone destruction.

3.
Viruses ; 15(11)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-38005826

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic. The interplay between innate and adaptive immune responses plays a crucial role in managing COVID-19. Cell therapy has recently emerged as a promising strategy to modulate the immune system, offering immense potential for the treatment of COVID-19 due to its customizability and regenerative capabilities. This review provides an overview of the various subsets of immune cell subsets implicated in the pathogenesis of COVID-19 and a comprehensive summary of the current status of immune cell therapy in COVID-19 treatment.


Subject(s)
COVID-19 , Humans , COVID-19/therapy , SARS-CoV-2 , COVID-19 Drug Treatment , Immunotherapy , Cell- and Tissue-Based Therapy , Immunity, Innate
4.
Immunology ; 170(4): 567-578, 2023 12.
Article in English | MEDLINE | ID: mdl-37688314

ABSTRACT

Sepsis is a life-threatening disease characterized by multiple organ dysfunction. B cells play a pivotal role in sepsis. Here, we first observed the significantly reduced Flot2 gene expression in B cells from patients with bacterial sepsis and endotoxin-induced septic mice. However, the effects of Flot2 on sepsis and B-cell immunity remain unknown. Thus, we sorted B cells from Flot2 knockout (Flot2-/- ) mice, RNA-seq revealed significantly upregulated effector B cell (Beff) cytokines such as Il6, Il1b and Cxcl10 after Flot2 deficiency, while it showed no effect on the expression of regulatory B cell (Breg) cytokines such as Il10, Tgfb. Consistently, elevated Beff cytokine IL-6 and unchanged Breg cytokine IL-10 were shown in B cells from Flot2-/- mice. Similar results were subsequently observed in B cell-specific Flot2 knockout chimeric mice. Notably, Flot2 deficiency aggravated sepsis with increased lung injury and shortened survival time in vivo by facilitating Beffs but not Bregs. Taken together, our data identify Flot2 as a novel controller of B cells, Flot2 deficiency amplifies inflammation by affecting Beffs to participate in the pathogenesis and progression of sepsis.


Subject(s)
B-Lymphocytes, Regulatory , Sepsis , Animals , Mice , Cytokines/metabolism , Inflammation/genetics
5.
Int Immunopharmacol ; 120: 110316, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37253315

ABSTRACT

Bone is a preferred metastatic site of advanced breast cancer and the 5-year overall survival rate of breast cancer patients with bone metastasis is only 22.8%. Targeted inhibition of osteoclasts can treat skeletal-related events (SREs) in breast cancer patients. Polyphyllin VII (PP7), a pennogenyl saponin isolated from traditional Chinese herb Paris polyphylla, exhibits strong anti-inflammatory and anti-cancer activities. In this study, we evaluated the effect of PP7 on metastatic breast cancer-induced bone destruction in vivo and the underlying mechanisms. We found that intraperitoneal injection of 1 mg/kg PP7 significantly ameliorated the breast cancer MDA-MB-231 cell-induced osteolysis in mice. Mechanistically, PP7 (0.125-0.5 µM) inhibited the conditioned medium of MDA-MB-231 cells (MDA-MB-231 CM)-induced osteoclast formation in bone marrow-derived macrophages (BMMs). Furthermore, PP7 markedly reduced MDA-MB-231 CM-induced osteoclastic bone resorption and F-actin rings formation in vitro. During MDA-MB-231 CM-induced osteoclastogenesis, the activation of c-Fos and NFATc1 signaling was significantly downregulated by PP7, and finally osteoclast-related genes such as Oscar, Atp6v0d2, Mmp9 and ß3 integrin were decreased. In addition, the formation of osteoblast was promoted by PP7 treatment. Our current findings revealed PP7 as a potential safe agent for preventing and treating bone destruction in breast cancer patients with bone metastases.


Subject(s)
Bone Resorption , Neoplasms , Osteolysis , Saponins , Animals , Mice , Osteogenesis , Osteolysis/drug therapy , Osteoclasts , Saponins/pharmacology , Proto-Oncogene Proteins c-fos , RANK Ligand/pharmacology , Cell Differentiation
6.
Immunotherapy ; 15(10): 737-750, 2023 07.
Article in English | MEDLINE | ID: mdl-37139963

ABSTRACT

Aim: To evaluate the efficacy and safety of first-line immunochemotherapy in the treatment of advanced esophageal squamous cell carcinoma (CRD42021287033). Methods: PubMed, Embase, Cochrane Library and Web of Science were systematically searched to obtain randomized controlled trials, and the outcome indicators of the reports were compared and analyzed. Results: A total of 3163 patients from five reported randomized controlled trials were included in the meta-analysis. The results showed the comprehensive benefits of toripalimab combined with chemotherapy, in terms of overall survival (hazard ratio: 0.59; 95% CI: 0.43-0.81) and progression-free survival (hazard ratio: 0.58; 95% CI: 0.46-0.73). Conclusion: Toripalimab combined with chemotherapy may be a better choice for first-line immunochemotherapy, although this needs to be verified by clinical studies.


The treatment of cancer is an issue of importance to the general public. A number of drugs are available to treat cancer, including those that enhance the body's natural defense. Such drugs are also the first choice for cancer of the esophagus, which cannot be removed surgically. In this study, we analyzed five different first-choice drugs in different ways, including how long the patient survives and how long the patient lives without their disease getting worse. We found that toripalimab, which strengthens the body's natural defense, may be the best combination choice for use in combination with chemotherapy. Although further studies are needed to increase the reliability of the conclusions, we preliminarily consider that this drug is particularly promising.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Network Meta-Analysis , Immunotherapy/methods
7.
Biomedicines ; 11(3)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36979675

ABSTRACT

For hepatocellular carcinoma (HCC) patients, we attempted to establish a new oxidative stress (OS)-related prognostic model for predicting prognosis, exploring immune microenvironment, and predicting the immunotherapy response. Significantly differently expressed oxidative stress-related genes (DEOSGs) between normal and HCC samples from the Cancer Genome Atlas (TCGA) were screened, and then based on weighted gene coexpression network analysis (WGCNA), HCC-related hub genes were discovered. Based on the least absolute shrinkage and selection operator (LASSO) and cox regression analysis, a prognostic model was developed. We validated the prognostic model's predictive power using an external validation cohort: the International Cancer Genome Consortium (ICGC).Then a nomogram was determined. Furthermore, we also examined the relationship of the risk model and clinical characteristics as well as immune microenvironment. 434 DEOSGs, comprising 62 downregulated and 372 upregulated genes (p < 0.05 and |log2FC| ≥ 1), and 257 HCC-related hub genes were recognized in HCC. Afterward, we built a five-DEOSG (LOX, CYP2C9, EIF2B4, EZH2, and SRXN1) prognostic risk model. Using the nomogram, the risk model was shown to have good prognostic value. Compared to the low risk group, HCC patients with high risk had poorer outcomes, worse pathological grades, and advanced tumor stages (p < 0.05). There were significant increases in LOX, EIF2B4, EZH2, and SRXN1 expression in HCC samples, while CYP2C9 expression was decreased. Finally, Real-time PCR (RT-qPCR) confirmed the mRNA expressions of five genes (CYP2C9, EIF2B4, EZH2, SRXN1, LOX) in HCC cell lines. Our study constructed a prognostic OS-related model with strong predictive power and potential as an immunosuppressive biomarker for HCC leading to improving prediction and providing new insights for HCC immunotherapy.

8.
BMC Cancer ; 22(1): 613, 2022 Jun 04.
Article in English | MEDLINE | ID: mdl-35659630

ABSTRACT

BACKGROUND: A malignancy of the liver, hepatocellular carcinoma (HCC) is among the most common and second-leading causes of cancer-related deaths worldwide. A reliable prognosis model for guidance in choosing HCC therapies has yet to be established. METHODS: A consensus clustering approach was used to determine the number of immune clusters in the Cancer Genome Atlas and Liver Cancer-RIKEN, JP (LIRI_JP) datasets. The differentially expressed genes (DEGs) among these groups were identified based on RNA sequencing data. Then, to identify hub genes among signature genes, a co-expression network was constructed. The prognostic value and clinical characteristics of the immune clusters were also explored. Finally, the potential key genes for the immune clusters were determined. RESULTS: After conducting survival and correlation analyses of the DEGs, three immune clusters (C1, C2, and C3) were identified. Patients in C2 showed the longest survival time with the greatest abundance of tumor microenvironment (TME) cell populations. MGene mutations in Ffibroblast growth factor-19 (FGF19) and catenin (cadherin-associated protein),ß1(CTNNB1) were mostly observed in C2 and C3, respectively. The signature genes of C1, C2, and C3 were primarily enriched in 5, 23, and 26 pathways, respectively. CONCLUSIONS: This study sought to construct an immune-stratification model for the prognosis of HCC by dividing the expression profiles of patients from public datasets into three clusters and discovering the unique molecular characteristics of each. This stratification model provides insights into the immune and clinical characteristics of HCC subtypes, which is beneficial for the prognosis of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology , Prognosis , Tumor Microenvironment/genetics
9.
Cancer Biol Med ; 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34591414

ABSTRACT

OBJECTIVE: The aims of this study were to examine the prognostic value of SHP-1 in breast cancer, its roles in the regulation of breast cancer cell growth and metastasis, and the underlying mechanisms. METHODS: Tumor specimens from 160 patients with breast cancer and 160 noncancerous tissues were used to examine the expression of SHP-1 and to analyze its association with overall survival through Kaplan-Meier and multivariate Cox regression analyses. RNA sequencing data and the expression and clinical importance of SHP-1 in breast cancer were evaluated with data from The Cancer Genome Atlas. In vitro and in vivo assays were performed to elucidate the effects of SHP-1 on breast cancer cell proliferation and invasion. Confocal immunofluorescence and GST pulldown assays were used to demonstrate the interaction between SHP-1 and epidermal growth factor receptor, as well as its downstream pathways. Immunohistochemistry and The Cancer Genome Atlas database were used to investigate the clinical association between SHP-1 and EGFR in human breast cancer. RESULTS: SHP-1 expression was associated with better survival in patients with breast cancer, whereas SHP-1 expression was negatively correlated with EGFR in human breast cancer. Ectopic SHP-1 expression significantly suppressed breast cancer cell proliferation, migration, and invasion. SHP-1 knockdown induced a more invasive phenotype and accelerated cell growth. Mechanistically, EGFR, a protein directly interacting with SHP-1, mediates the SHP-1-induced inactivation of Ras/Erk/GSK3ß signaling and its downstream effectors. CONCLUSIONS: SHP-1 is an important prognostic biomarker in patients with breast cancer, and the SHP-1-EGFR axis is a promising target for treatment.

10.
Can J Gastroenterol Hepatol ; 2021: 9990338, 2021.
Article in English | MEDLINE | ID: mdl-34557456

ABSTRACT

Long noncoding RNAs (lncRNAs) have been substantially reported to have critical roles in regulating tumorigenesis in recent years. However, the expression pattern and biological function of SNHG17 in hepatocellular carcinoma (HCC) remain unclear. Bioinformatics analysis and qRT-PCR were performed to detect the expression pattern of SNHG17 in HCC tissues, adjacent nontumorous tissues, and cell lines. The effect of SNHG17 on proliferation, migration, and apoptosis of HCC was investigated by knockdown and overexpressing SNHG17 in HCC cell lines. RNA sequencing was utilized to explore the underlying mechanism. Utilizing publicly available TCGA-LIHC, GSE102079 HCC datasets, and qRT-PCR, we found SNHG17 was significantly upregulated in HCC tissues and cell lines and was notably associated with larger tumor size, poorly differentiation, presence of vascular invasion, and advanced TNM stage. Furthermore, gain- and loss-of-function studies demonstrated that SNHG17 promoted cell proliferation and migration and inhibited apoptosis of HCC. By employing RNA sequencing, we found knockdown of SNHG17 caused 1037 differentially expressed genes, highly enriched in several pathways, including metabolic, PI3K-Akt, cell adhesion, regulation of cell proliferation, and apoptotic pathway; among them, 92 were overlapped with SNHG17-related genes in the TCGA-LIHC dataset. Furthermore, ERH, TBCA, TDO2, and PDK4 were successfully validated and found significantly dysregulated in HCC tissues. Moreover, HCC patients with higher SNHG17 expression had a relatively poor overall survival and disease-free survival, and ERH and PDK4 also played a marked role in the prognosis of HCC. Broadly, our findings illustrate that SNHG17 acts as a noncoding oncogene in HCC progression, suggesting its potential value as a novel target for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , RNA, Long Noncoding , Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , RNA, Long Noncoding/genetics
11.
Mol Ther Nucleic Acids ; 25: 328-341, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34484860

ABSTRACT

Rapid tumor progression, metastasis, and diagnosis in advanced stages of disease are the main reasons for the short survival time and high mortality rate of patients with hepatocellular carcinoma (HCC). Ephrin A4 (EFNA4), the ligand of the EPH family, participates in the development of blood vessels and epithelium by regulating cell migration and rejection. In our study, based on bioinformatics analyses, we found that EFNA4 was highly expressed and led to poor prognosis in patients with HCC. We demonstrated that overexpression of EFNA4 significantly promoted HCC cell proliferation and migration in vivo or in vitro. In addition, knockdown of EFNA4 inhibited the proliferation and migration of HCC cells. Furthermore, EFNA4 was found to directly interact with EPHA2 and promote its phosphorylation at Ser897, followed by recruitment of phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2) and activation of the glycogen synthase kinase-3beta (GSK3ß)/ß-catenin signaling pathway. Moreover, overexpression of ß-catenin further promoted the expression of PIK3R2, which formed a positive feedback loop. The results revealed that abnormal expression of EFNA4 is the main switch of the PIK3R2/GSK3ß/ß-catenin loop that influenced the proliferation and migration of HCC cells and suggest that EFNA4 is a potential prognostic marker and a prospective therapeutic target in patients with HCC.

12.
Cancer Cell Int ; 20: 196, 2020.
Article in English | MEDLINE | ID: mdl-32508532

ABSTRACT

BACKGROUND: Forkhead box C2 (FOXC2) is a crucial factor involving in various cancers. However, its functions in hepatocellular carcinoma (HCC) is unknown. Here, we explored the role of FOXC2 in the progression of HCC and its potential mechanisms. METHODS: FOXC2 expression in HCC tissue and cells were detected by immunohistochemistry or western blot and real-time PCR. CCK8, wound healing and transwell assay were used to measure cell growth and invasion. Tumor formation experiment was carried out to assess the tumorigenicity of HCC cells. Regulation of FOXC2 on Ang-2 was validated by luciferase assay and complementary experiments. RESULTS: Increased FOXC2 expression was found to be associated positively with more aggressive clinicopathologic features. HCC patients with higher FOXC2 expression had significantly shorter overall survival. FOXC2 expression was indentified as an independent risk factor for resectable HCC. Increased FOXC2 expression accelerated the migration and invasion of HCC cells, accompanied by enhanced Ang-2 expression. Likewise, FOXC2 knockdown yielded opposite results. Moreover, FOXC2 stimulated the activation of the Ang-2 promoter. Suppression of Ang-2 expression hindered the FOXC2-mediated EMT processs, cell migration and invasion of HCC. CONCLUSIONS: FOXC2 is a novel prognostic predictor for HCC and may facilitate the growth and invasion through Ang-2.

13.
Biochem Biophys Res Commun ; 500(2): 177-183, 2018 06 02.
Article in English | MEDLINE | ID: mdl-29626478

ABSTRACT

Nuclear receptor coactivator 5 (NCOA5) plays important roles in the development of a variety of malignancies. However, the underlying mechanisms remain obscure. In this study, we successfully generated the NCOA5 knockout hepatocellular carcinoma (HCC) cells by CRISPR/Cas9 - mediated genome editing and found that knockout of NCOA5 inhibited the proliferation and tumor microsphere formation of HCC cells significantly. Moreover, the migration ability of NCOA5 knockout HCC cells declined. Mechanistic analyses indicated that knockout of NCOA5 can suppress the epithelial - mesenchymal transition (EMT) in HCC cells. In conclusion, our findings provide a mechanistic insight into the role of NCOA5 in HCC progression.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement , Epithelial-Mesenchymal Transition , Gene Knockout Techniques , Liver Neoplasms/pathology , Nuclear Receptor Coactivators/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
14.
J Exp Clin Cancer Res ; 36(1): 175, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29208003

ABSTRACT

BACKGROUND: Forkhead box A1 (FOXA1) expression is associated with various types of tumors; however, the function and underlying mechanism of FOXA1 in the development of hepatocellular carcinoma (HCC) remains obscure. METHODS: Here, we investigated the role of FOXA1 in the development of HCC by applying gene function gain and loss analysis to HepG2 and Hep3B cell lines, and comparing outcomes with those of clinical HCC samples. RESULTS: Phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), which encodes protein PI3Kp85 (p85), was identified as a FOXA1 target gene. Analyses of the mechanism and function revealed that FOXA1 suppresses hepatocellular carcinoma cell viability and motility by inhibiting PI3K/Akt signaling through direct inhibition of PIK3R1 transcription. Moreover, in clinical samples from male HCC patients, FOXA1 expression was much lower, whereas PI3Kp85 levels were much higher in tumor than in non-tumor tissues. Elevated PI3Kp85 is an unfavorable factor in HCC. CONCLUSIONS: As a tumor suppressor, FOXA1 targets PIK3R1 directly to inhibit PI3K/Akt signaling pathway, thus exerting a negative regulatory effect on proliferation, migration, and invasion of HCC in male patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Liver Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase , Disease Progression , Hep G2 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Transfection
15.
Cancer Lett ; 391: 152-161, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28137631

ABSTRACT

Type 2 Diabetes (T2D) is a risk factor for hepatocellular carcinoma (HCC). We have previously described that haploinsufficiency of nuclear receptor coactivator 5 (NCOA5) is a genetic defect linking glucose intolerance to HCC. Here we report identification and characterization of a single nucleotide variation (T445A) in NCOA5, causing an amino acid Thr to Ala substitution, in adjacent non-tumorous liver tissues derived from patients with concurrent HCC and T2D. By using Tet-On inducible expression cells, we show that ectopic expression of NCOA5wt suppressed proliferation of HCC cells via induction of G2/M arrest, while ectopic expression of NCOA5T445A had a significantly lesser effect compared to ectopic expression of NCOA5wt. Furthermore, ectopic expression of NCOA5wt increased the occurrence of DNA damage and cell senescence, whereas expression of NCOA5T445A partly lost this activity. Xenograft tumor model analysis demonstrated that ectopic NCOA5wt expression reduced HCC tumor growth and the T445A variation impairs its tumor growth inhibitory function. Collectively, our data show that the T445A variation impairs the ability of NCOA5 to inhibit growth of HCC, suggesting that this variation may have potential to increase susceptibility to HCC comorbid with T2D.


Subject(s)
Carcinoma, Hepatocellular/genetics , Diabetes Mellitus, Type 2/complications , Liver Neoplasms/genetics , Nuclear Receptor Coactivators/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Mice , Mice, Nude , Nuclear Receptor Coactivators/metabolism , Transfection
16.
Mol Med Rep ; 13(1): 605-12, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26718891

ABSTRACT

Glioma is an aggressive malignancy with limited effective treatment and poor prognosis. Therefore, the identification of novel prognostic markers and effective therapeutic targets is important for the treatment of human glioma. TIP30 is a tumor suppressor involved in the regulation of numerous cellular processes, including tumor cell growth, metastasis, and angiogenesis in various human cancers. The present study investigated whether Tat­interacting protein (TIP)30 was able to regulate tumorigenesis and predict the clinical outcome of patients with glioma. A total of 92 human glioma tissue samples and 10 normal brain tissue samples were examined by immunostaining. The results indicated that the expression levels of TIP30 significantly decreased in glioma tissue samples. as compared with normal brain tissue samples. Furthermore, TIP30 expression was inversely correlated with tumor histological classification, pathological grade, tumor size, and epidermal growth factor receptor (EGFR) expression; however, no association was detected between TIP30 expression and patient age and gender. In addition, patients with positive TIP30 expression exhibited significantly longer median overall survival rates, as compared with those with negative TIP30 expression. In vitro experiments revealed that upregulation of TIP30 expression by lentiviral vector transfection inhibited cell growth and induced cell apoptosis, as determined by MTT assay and Annexin V­fluorescein isothiocyanate staining, respectively. In addition, TIP30 expression markedly attenuated cell migration and invasion, as determined by wound healing and transwell assays. Upregulation of TIP30 expression in glioma cells decreased the expression levels of EGFR and its associated downstream molecules phosphorylated extracellular signal­regulated kinases (ERK) and phosphorylated AKT, as determined by western blot analysis. The results of the present study indicated that TIP30 may suppress oncogenesis and glioma progression, thereby improving the prognosis of patients with glioma. Therefore, TIP30 may prove useful as a prognostic biomarker, and as a potential target for glioma therapy.


Subject(s)
Acetyltransferases/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Transcription Factors/metabolism , Adult , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lentivirus/metabolism , Male , Multivariate Analysis , Neoplasm Invasiveness , Phosphorylation , Proportional Hazards Models , Proto-Oncogene Proteins c-akt/metabolism
17.
Oncotarget ; 7(5): 5327-41, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26683365

ABSTRACT

Although protein kinase D (PKD) has been shown to contribute to invasion and metastasis in several types of cancer, the role of PKD in the epithelial mesenchymal transition (EMT) of hepatocellular carcinoma (HCC) has remained unclear. We found that PKD2 is up-regulated in HCC and is correlated with the metastasis of HCC. PKD2 positively regulated TNF-α-induced EMT and metastasis of HCC. Mechanistic studies revealed TNF-α-induced PKD2 activation is mediated by the formation of a TNFR1/TRAF2 complex. PKD2 bound directly to the p110α and p85 subunits of PI3K and promoted the PI3K/Akt/GSK-3ß signaling cascade to stimulate EMT. In conclusion, our results have uncovered a novel role for the regulation of EMT and suggest inhibition of PKD2 as a potential therapeutic strategy for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Glycogen Synthase Kinase 3/metabolism , Liver Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , beta Catenin/metabolism , Carcinoma, Hepatocellular/metabolism , Class I Phosphatidylinositol 3-Kinases , Epithelial-Mesenchymal Transition/drug effects , Humans , Liver Neoplasms/metabolism , Protein Kinase D2 , Protein Kinases , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
18.
Int J Clin Exp Pathol ; 8(2): 1938-45, 2015.
Article in English | MEDLINE | ID: mdl-25973086

ABSTRACT

OBJECTIVE: Sirt7, as one of the seven Sirtuin family members, which plays distinct roles in cancer progression, is bringing emerging attention due to its oncogenic characteristic. The expression of Sirt7 in breast cancer remained unclear, and the aim of this study was to elucidate its role in breast cancer. METHODS: A total of 188 cases included in this study were immunohistochemically evaluated for Sirt7, and western blot assay was used to assess its expression in breast cell lines as well as 36 breast cancer tissues and 36 paired non-cancerous tissues. RESULTS: Upregulation of Sirt7 was found in breast cancer cell lines and breast cancer tissues (P < 0.001) by western blot analysis. Sirt7 was highly expressed in breast cancer tissue samples (67.8%) compared to adjacent normal breast tissues (31.8%) by immunohistochemical assay. It was also observed that the high expression level of Sirt7 was significantly correlated with high histological grade (P = 0.039) and negatively related to overall survival (P = 0.006). Sirt7 proved to be an independent prognostic factor (P = 0.007) in breast cancer. CONCLUSIONS: Sirt7 expression was implicated with high histological grade and independently predicted poor clinical outcome in patients with breast cancer, suggesting that Sirt7 might play a role in the malignant progression of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Sirtuins/metabolism , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Humans , Middle Aged , Neoplasm Grading , Prognosis , Survival Rate
19.
J BUON ; 19(4): 1132, 2014.
Article in English | MEDLINE | ID: mdl-25536634
20.
Cancer Lett ; 354(1): 200-9, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25135222

ABSTRACT

Aberrant epidermal growth factor (EGF)-dependent signaling plays a key role in the progression of human carcinomas. We found that TIP30, a tumor suppressor protein, translocated into the nucleus of human lung adenocarcinoma cells following EGF treatment, and the selective inhibitors of EGFR signaling pathways blocked this effect. Chromatin immunoprecipitation assays revealed that TIP30 negatively regulated EGF-dependent transcriptional activation of CCND1 through a HDAC1-dependent mechanism. In lung adenocarcinoma patients, the level of nuclear TIP30 was inversely correlated with that of EGFR and cyclin D1. These findings suggest that nuclear TIP30-induced downregulation of cyclin D1 transcription antagonizes EGFR signaling and suppresses tumorigenesis.


Subject(s)
Acetyltransferases/metabolism , Active Transport, Cell Nucleus , Adenocarcinoma/metabolism , Cyclin D1/metabolism , Epidermal Growth Factor/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Protein Binding , Signal Transduction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...