Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Laryngoscope ; 133(9): 2174-2182, 2023 09.
Article in English | MEDLINE | ID: mdl-36286082

ABSTRACT

OBJECTIVES: We sought to evaluate the impact of the time interval from surgical resection to local recurrence (TTLR) on clinical outcomes in head and neck soft tissue sarcoma (HNSTS). METHODS: A total of 401 patients who underwent R0 resection for primary HNSTS were included in this study. Patients with local recurrence as the first event after their initial resection were divided into early local recurrence (ELR) or late local recurrence (LLR) groups according to TTLR. Multiple survival analyses were performed to identify the independent prognostic predictors of overall survival (OS) and survival after local recurrence (SAR). RESULTS: Two hundred and nine of the 401 patients (52.1%) developed local recurrence during a median follow-up period of 134.6 months. Patients in the ELR group had a shorter median OS time (35.0 vs. 120.6, p < 0.001) and lower 5-year OS rate (47.7% vs. 80.9%, p < 0.001) than those in the LLR group. Moreover, the ELR group exhibited worse SAR (p = 0.001) than the LLR group, and multivariate analyses demonstrated TTLR as an independent prognostic factor for SAR (p = 0.048) and OS (p = 0.004). Additionally, re-resection significantly prolonged SAR than other salvage interventions or no treatment (p < 0.001). CONCLUSION: In patients with HNSTS, ELR after R0 resection presents adverse effects on OS and SAR than those with LLR, and TTLR could serve as a promising predictor for survival. Salvage therapies, especially the re-resection could improve SAR and should be recommended when there are surgical indications after recurrence. LEVEL OF EVIDENCE: 3 Laryngoscope, 133:2174-2182, 2023.


Subject(s)
Sarcoma , Humans , Adult , Retrospective Studies , Prognosis , Survival Analysis , Time Factors , Neoplasm Recurrence, Local , Survival Rate
2.
Oncotarget ; 9(18): 14124-14137, 2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29581832

ABSTRACT

The underlying cause of treatment failure in many cancer patients is intrinsic and acquired resistance to chemotherapy. Recently, histone deacetylase (HDAC) inhibitors have developed into a promising cancer treatment. However, resistance mechanism induced by HDAC inhibitors remains largely unknown. Here we report that a HDAC inhibitor, JNJ-2648158 induced transcription of XIAP by activating AP-1 expression, which conferring resistance to chemotherapeutics. Our results showed that high expression of c-Fos caused by HDAC inhibitor promoted AP-1 formation during acquired resistance towards chemo-drugs, indicating an extremely poor clinical outcome in breast cancers and liver cancers. Our study reveals a novel regulatory mechanism towards chemo-drug resistance, and suggests that XIAP may serve as a potential therapeutic target in those chemo-resistant cancer cells.

3.
J Cell Mol Med ; 21(5): 860-870, 2017 05.
Article in English | MEDLINE | ID: mdl-28165192

ABSTRACT

Natural killer (NK) cells play an important role in preventing cancer development. NK group 2 member D (NKG2D) is an activating receptor expressed in the membrane of NK cells. Tumour cells expressing NKG2DL become susceptible to an immune-dependent rejection mainly mediated by NK cells. The paradoxical roles of transforming growth factor beta (TGF-ß) in regulation of NKG2DL are presented in many studies, but the mechanism is unclear. In this study, we showed that TGF-ß up-regulated the expression of NKG2DLs in both PC3 and HepG2 cells. The up-regulation of NKG2DLs was characterized by increasing the expression of UL16-binding proteins (ULBPs) 1 and 2. TGF-ß treatment also increased the expression of transcription factor SP1. Knockdown of SP1 significantly attenuated TGF-ß-induced up-regulation of NKG2DLs in PC3 and HepG2 cells, suggesting that SP1 plays a key role in TGF-ß-induced up-regulation of NKG2DLs. TGF-ß treatment rapidly increased SP1 protein expression while not mRNA level. It might be due to that TGF-ß can elevate SP1 stability by activating PI3K/AKT signalling pathway, subsequently inhibiting GSK-3ß activity and decreasing the association between SP1 and GSK-3ß. Knockdown of GSK-3ß further verified our findings. Taken together, these results revealed that AKT/GSK-3ß-mediated stabilization of SP1 is required for TGF-ß induced up-regulation of NKG2DLs. Our study provided valuable evidence for exploring the tumour immune modulation function of TGF-ß.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, NK Cell Lectin-Like/metabolism , Sp1 Transcription Factor/metabolism , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , Hep G2 Cells , Humans , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Protein Stability/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects
4.
Cancer Immunol Immunother ; 66(3): 355-366, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27915371

ABSTRACT

Myeloid-derived suppressor cells (MDSC) have been identified as a population of immature myeloid cells that suppress anti-tumor immunity. MDSC are increased in tumor-bearing hosts; thus, depletion of MDSC may enhance anti-tumor immunity. Histone deacetylase inhibitors (HDACi) are chemical agents that are primarily used against hematologic malignancies. The ability of these agents to modulate anticancer immunity has recently been extensively studied. However, the effect of HDACi on MDSC has remained largely unexplored. In the present study, we provide the first demonstration that HDACi treatment decreases MDSC accumulation in the spleen, blood and tumor bed but increases the proportion of T cells (particularly the frequency of IFN-γ- or perforin-producing CD8+ T cells) in BALB/C mice with 4T1 mammary tumors. In addition, HDACi exposure of bone marrow (BM) cells significantly eliminated the MDSC population induced by GM-CSF or the tumor burden in vitro, which was further demonstrated as functionally important to relieve the inhibitory effect of MDSC-enriched BM cells on T cell proliferation. Mechanistically, HDACi increased the apoptosis of Gr-1+ cells (almost MDSC) compared with that of Gr-1- cells, which was abrogated by the ROS scavenger N-acetylcysteine, suggesting that the HDACi-induced increase in MDSC apoptosis due to increased intracellular ROS might partially account for the observed depletion of MDSC. These findings suggest that the elimination of MDSC using an HDACi may contribute to the overall anti-tumor properties of these agents, highlighting a novel property of HDACi as potent MDSC-targeting agents, which may be used to enhance the efficacy of immunotherapeutic regimens.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/pathology
5.
Arch Biochem Biophys ; 608: 34-41, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27592306

ABSTRACT

Nodal is a member of transforming growth factor beta (TGF-ß) superfamily. Nodal promotes the self-renewal of human cancer stem cells (CSCs) and triggers carcinogenesis of human cancers via an autocrine manner through Smad2/3 pathway. In our study, generation of Nodal-overexpressed cancer cells was constructed, and the effect of Nodal on the stem cell marker Oct-4 was evaluated by overexpression or blocked Nodal/ALKs signaling pathway in non-small cell lung cancer cells A549 and prostate cancer cells PC3. Functionally, Nodal also increased the proliferation via the ß-catenin nuclear translocation. This increase was attributed to GSK-3ß dephosphorylating, and activin receptor-like kinase 4/7 (ALK4/7) played a major role in human cancer cells. Our study provides a positive understanding of Nodal function in cancer cells and suggests a potential novel target for clinical therapeutic research.


Subject(s)
Active Transport, Cell Nucleus , Gene Expression Regulation, Neoplastic , Nodal Protein/metabolism , Octamer Transcription Factor-3/metabolism , Prostatic Neoplasms/metabolism , beta Catenin/metabolism , A549 Cells , Activin Receptors, Type I/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , Humans , Male , Signal Transduction , Transfection
6.
Biochem Biophys Res Commun ; 478(2): 710-5, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27498029

ABSTRACT

Calotropin (M11), an active compound isolated from Asclepias curasavica L., was found to exert strong inhibitory and pro-apoptotic activity specifically against cisplatin-induced resistant non-small cell lung cancer (NSCLC) cells (A549/CDDP). Molecular mechanism study revealed that M11 induced cell cycle arrest at the G2/M phase through down-regulating cyclins, CDK1, CDK2 and up-regulating p53 and p21. Furthermore, M11 accelerated apoptosis through the mitochondrial apoptotic pathway which was accompanied by increase Bax/Bcl-2 ratio, decrease in mitochondrial membrane potential, increase in reactive oxygen species production, activations of caspases 3 and 9 as well as cleavage of poly ADP-ribose polymerase (PARP). The activation and phosphorylation of JNK was also found to be involved in M11-induced apoptosis, and SP610025 (specific JNK inhibitor) partially prevented apoptosis induced by M11. In contrast, all of the effects that M11 induce cell cycle arrest and apoptosis in A549/CDDP cells were not significant in A549 cells. Drugs with higher sensitivity against resistant tumor cells than the parent cells are rather rare. Results of this study supported the potential application of M11 on the non-small lung cancer (NSCLC) with cisplatin resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Asclepias/chemistry , Cardenolides/pharmacology , Drug Resistance, Neoplasm/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , A549 Cells , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/genetics , CDC2 Protein Kinase , Cardenolides/isolation & purification , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cisplatin/pharmacology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/agonists , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Drug Resistance, Neoplasm/genetics , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Plant Extracts/chemistry , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proteolysis , Proto-Oncogene Proteins c-bcl-2/agonists , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
7.
Oxid Med Cell Longev ; 2016: 5874127, 2016.
Article in English | MEDLINE | ID: mdl-27057280

ABSTRACT

Some evidence indicated that chemoresistance associates with the acquisition of cancer stem-like properties. Recent studies suggested that chemokines can promote the chemoresistance and stem cell properties in various cancer cells, while the underling mechanism is still not completely illustrated. In our study, we found that CCL21 can upregulate the expression of P-glycoprotein (P-gp) and stem cell property markers such as Bmi-1, Nanog, and OCT-4 in colorectal cancer (CRC) HCT116 cells and then improve the cell survival rate and mammosphere formation. Our results suggested that Snail was crucial for CCL21-mediated chemoresistance and cancer stem cell property in CRC cells. Further, we observed that CCL21 treatment increased the protein but not mRNA levels of Snail, which suggested that CCL21 upregulates Snail via posttranscriptional ways. The downstream signals AKT/GSK-3ß mediated CCL21 induced the upregulation of Snail due to the fact that CCL21 treatment can obviously phosphorylate both AKT and GSK-3ß. The inhibitor of PI3K/Akt, LY294002 significantly abolished CCL21 induced chemoresistance and mammosphere formation of HCT116 cells. Collectively, our results in the present study revealed that CCL21 can facilitate chemoresistance and stem cell property of CRC cells via the upregulation of P-gp, Bmi-1, Nanog, and OCT-4 through AKT/GSK-3ß/Snail signals, which suggested a potential therapeutic approach to CRC patients.


Subject(s)
Chemokine CCL21/pharmacology , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Up-Regulation/drug effects
8.
Oncotarget ; 7(21): 31466-83, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27129170

ABSTRACT

Multidrug resistance (MDR) mediated by P-glycoprotein (P-gp) is a major cause of cancer therapy failure. In this study, we identified a novel C21 steroidal glycoside, asclepiasterol, capable of reversing P-gp-mediated MDR. Asclepiasterol (2.5 and 5.0µM) enhanced the cytotoxity of P-gp substrate anticancer drugs in MCF-7/ADR and HepG-2/ADM cells. MDR cells were more responsive to paclitaxel in the presence of asclepiasterol, and colony formation of MDR cells was only reduced upon treatment with a combination of asclepiasterol and doxorubicin. Consistent with these findings, asclepiasterol treatment increased the intracellular accumulation of doxorubicin and rhodamine 123 (Rh123) in MDR cells. Asclepiasterol decreased expression of P-gp protein without stimulating or suppressing MDR1 mRNA levels. Asclepiasterol-mediated P-gp suppression caused inhibition of ERK1/2 phosphorylation in two MDR cell types, and EGF, an activator of the MAPK/ERK pathway, reversed the P-gp down-regulation, implicating the MAPK/ERK pathway in asclepiasterol-mediated P-gp down-regulation. These results suggest that asclepiasterol could be developed as a modulator for reversing P-gp-mediated MDR in P-gp-overexpressing cancer variants.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Asclepias/chemistry , Drug Resistance, Multiple/drug effects , Glycosides/pharmacology , Phytosterols/pharmacology , Saponins/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Resistance, Multiple/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glycosides/chemistry , Hep G2 Cells , Humans , MCF-7 Cells , Molecular Structure , Phosphorylation/drug effects , Phytosterols/chemistry , Saponins/chemistry
9.
Leuk Res ; 46: 37-44, 2016 07.
Article in English | MEDLINE | ID: mdl-27123832

ABSTRACT

Persistent activation of NF-κB is a prerequisite for development of adult T cell leukemia-lymphoma (ATL) caused by human T cell leukemia virus type 1 (HTLV-1). HTLV-1 genome encodes a viral transforming protein named Tax, which constitutively activates the canonical IκB kinases (IKK), the central regulator of NF-κB signaling. However, the role of the non-canonical IκB kinases, TBK1 and IKKε, in the pathogenesis of HTLV-1-associated leukemia has not been evaluated. We here show that TBK1/IKKε are crucial pro-survival molecules by maintaining persistent activity of Stat3. Consistent with this finding, silencing Stat3 by the specific shRNA or by the chemical inhibitor ruxolitinib results in drastic impediment of leukemia cell growth. We further find that in HTLV-1-transformed T cells expressing Tax, TBK1 co-localizes with the canonical IκB kinases and Tax in the lipid raft microdomains. The wild type Tax, but not the Tax mutant defective in activating the canonical IKK, promotes the lipid raft translocation of TBK1. This phenomenon correlates with Tax activation of both NF-κB and Stat3. Tax does not interact directly with TBK1/IKKε, and it rather engages a molecular crosstalk between the canonical IKKs and TBK1/IKKε. Our data, therefore, demonstrate a key role of TBK1/IKKε in the survival and proliferation of HTLV-1-transformed T cells and implicate a potential therapy targeting TBK1/IKKε and Stat3 in controlling HTLV-1-mediated oncogenesis.


Subject(s)
Cell Transformation, Viral , Human T-lymphotropic virus 1 , I-kappa B Kinase/physiology , Leukemia-Lymphoma, Adult T-Cell/pathology , Protein Serine-Threonine Kinases/physiology , T-Lymphocytes/virology , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Gene Products, tax , Humans , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , NF-kappa B/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
10.
Free Radic Biol Med ; 95: 230-42, 2016 06.
Article in English | MEDLINE | ID: mdl-27021965

ABSTRACT

Resveratrol gains a great interest for its strong antioxidant properties, while the molecular mechanisms underlie the beneficial effects on psychosocial stress remain controversial. In this study, we demonstrated that resveratrol protected peritoneal macrophages and RAW 264.7 cells from stress-induced decrease in the total cell count, phagocytic capability, reactive oxygen species generation, monodansylcadaverine and mitochondrial membrane potential in stressed mice. Resveratrol promoted stress-induced autophagy in both models. Modulation of autophagy by rapamycin or 3-methyladenine regulated the protective effect of resveratrol, suggesting a role of autophagy in the protective mechanisms of resveratrol. The comparison studies revealed that distinct mechanisms were implicated in the protective effect of resveratrol and other antioxidants (vitamin C and edaravone). Resveratrol promoted autophagy via upregulating SIRT3 expression and phosphorylation of AMP-activated protein kinase (AMPK). Knockdown of SIRT3 resulted in decreased autophagy and abolished protective effect of resveratrol. SIRT1 was also involved in the protective mechanism of resveratrol, although its effect on autophagy was unnoticeable. Pharmacological manipulation of autophagy modulated the effects of resveratrol on SIRT3 and AMPK, revealing the engagement of a positive feedback loop. In sharp contrast, vitamin C and edaravone effectively protected macrophages from stress-induced cytotoxicity, accompanied by downregulated SIRT3 expression and AMPK phosphorylation, and decreased level of autophagy response. Taken together, we conclude that a SIRT3/AMPK/autophagy network orchestrates in the protective effect of resveratrol in macrophages.


Subject(s)
Oxidative Stress/drug effects , Protein Kinases/genetics , Sirtuin 3/genetics , Stilbenes/administration & dosage , Stress, Psychological/drug therapy , AMP-Activated Protein Kinase Kinases , Animals , Antioxidants/administration & dosage , Antipyrine/administration & dosage , Antipyrine/analogs & derivatives , Apoptosis/drug effects , Ascorbic Acid/administration & dosage , Autophagy/drug effects , Edaravone , Gene Expression Regulation/drug effects , Macrophages/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Resveratrol , Sirolimus/administration & dosage , Stress, Psychological/genetics
11.
Oncotarget ; 7(4): 4122-41, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26716641

ABSTRACT

Transforming growth factor-ß1 (TGF-ß1) present in tumor microenvironment acts in a coordinated fashion to either suppress or promote tumor development. However, the molecular mechanisms underlying the effects of TGF-ß1 on tumor microenvironment are not well understood. Our clinical data showed a positive association between TGF-ß1 expression and cancer-associated fibroblasts (CAFs) in tumor microenvironment of breast cancer patients. Thus we employed starved NIH3T3 fibroblasts in vitro and 4T1 cells mixed with NIH3T3 fibroblasts xenograft model in vivo to simulate nutritional deprivation of tumor microenvironment to explore the effects of TGF-ß1. We demonstrated that TGF-ß1 protected NIH3T3 fibroblasts from Star-induced growth inhibition, mitochondrial damage and cell apoptosis. Interestingly, TGF-ß1 induced the formation of CAFs phenotype in starvation (Star)-treated NIH3T3 fibroblasts and xenografted Balb/c mice, which promoted breast cancer tumor growth. In both models, autophagy agonist rapamycin increased TGF-ß1-induced protective effects and formation of CAFs phenotypes, while autophagy inhibitor 3-methyladenine, Atg5 knockdown or TGF-ß type I receptor kinase inhibitor LY-2157299 blocked TGF-ß1 induced these effects. Taken together, our results indicated that TGF-ß/Smad autophagy was involved in TGF-ß1-induced protective effects and formation of CAFs phenotype in tumor microenvironment, which may be used as therapy targets in breast cancer.


Subject(s)
Autophagy , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Protective Agents/pharmacology , Transforming Growth Factor beta1/pharmacology , Tumor Microenvironment/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Am J Cancer Res ; 5(6): 2098-112, 2015.
Article in English | MEDLINE | ID: mdl-26269769

ABSTRACT

Nodal, an important embryonic morphogen, has been reported to modulate tumorigenesis. Epithelial-mesenchymal transition (EMT) plays an important role in cancer metastasis. We have previously reported that recombinant Nodal treatment can promote melanoma undergoing EMT, but the effects of endogenous Nodal on EMT are still unknown. Here we generated both Nodal-overexpression and -knockdown stable cell lines to investigate the in vitro and in vivo characteristics of Nodal-induced EMT in murine melanoma cells. Nodal-overexpression cells displayed increased migration ability, accompanied by typical phenotype changes of EMT. In contrast, Nodal-knockdown stable cells repressed the EMT phenotype as well as reduced cell motility. Results of animal experiments confirmed that overexpression of Nodal can promote the metastasis of melanoma tumor in vivo. Mechanistically, we found that Nodal-induced expression of Snail and Slug involves its activation of ALK/Smads and PI3k/AKT pathways, which is an important process in the Nodal-induced EMT. However, we also found that the EMT phenotype was not completely inhibited by blocking the paracrine activity of Nodal in Nodal overexpression cell line suggesting the presence of additional mechanism(s) in the Nodal-induced EMT. This study provides a better understanding of Nodal function in melanoma, and suggests targeting Nodal as a potential strategy for melanoma therapey.

13.
Mol Immunol ; 65(1): 34-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25618241

ABSTRACT

Human leukocyte antigen class I antigens (HLA-I) is essential in immune response by presenting antigenic peptides to cytotoxic T lymphocytes. Downregulation of HLA-I is observed in primary and metastatic prostate cancers, which facilitates them escape from immune surveillance, thereby promotes prostate cancer progression. In addition, elevated level of growth factors like TGF-ß or EGF in microenvironment is related to the prostate cancer deterioration. Thus, we wondered whether TGF-ß or EGF was involved in the regulation of HLA-I during the development of prostate cancer cells. In this study, we demonstrated that TGF-ß and EGF both downregulated the expression of HLA-I, thereby attenuated the cytotoxic T cell mediated lysis of prostate cancer cells. Next, we revealed that TGF-ß and EGF induced downregulation of HLA-I is associated with classical epithelial-mesenchymal transition (EMT) morphological changes and expression profiles. We further illustrated that overexpression of Snail is crucial for HLA-I downregulation and its association with EMT. At last, we discussed that NF-κB/p65 is the plausible target for Snail to induce HLA-I downregulation. Taken together, this is the first evidence to reveal that both TGF-ß and EGF can induce HLA-I downregulation which is then proven to be associated with EMT in prostate cancer cells. These discoveries provide a deeper understanding of growth factors induced immune escape and introduce potential therapeutic targets for prostate cancers.


Subject(s)
Epidermal Growth Factor/immunology , Epithelial-Mesenchymal Transition/immunology , Histocompatibility Antigens Class I/immunology , Prostatic Neoplasms/immunology , Transforming Growth Factor beta/immunology , Cell Line, Tumor , Down-Regulation , Epidermal Growth Factor/biosynthesis , Humans , Male , RNA Interference , RNA, Small Interfering , Snail Family Transcription Factors , T-Lymphocytes, Cytotoxic/immunology , Transcription Factor RelA/genetics , Transcription Factors/biosynthesis , Transcription, Genetic/genetics , Transforming Growth Factor beta/biosynthesis , Tumor Escape/immunology , Up-Regulation
14.
Biochim Biophys Acta ; 1840(10): 3096-105, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25088797

ABSTRACT

BACKGROUND: Epithelial-mesenchymal transition (EMT) plays a pivotal role in the development of metastatic cancers. Basic fibroblast growth factor (bFGF) is significantly elevated in metastatic prostate cancers, which has been mentioned mainly to induce EMT in normal cells. However, there is no description about bFGF induced EMT and its underlying mechanism in prostate cancer cells. METHODS: Western blotting, immunofluorescence and qRT-PCR assays were used to study protein or mRNA expression profiles of the EMT. Wound healing scratch, migration and invasion assays were used to test the motility of cells undergoing EMT. More methods were used to explore the underlying mechanisms. RESULTS: We demonstrated that bFGF promoted EMT and motility of human prostate cancer PC-3 cells. Both protein and mRNA expression of Snail were rapidly increased after bFGF treatment. Ectopic expression of Snail triggered EMT and enhanced cell motility in PC-3 cells, and knockdown of Snail almost abolished bFGF induced EMT, suggesting the critical role of Snail. Mechanistic study demonstrated that bFGF promoted the stability, nuclear localization and transcription of Snail by inhibiting the activity of glycogen synthase kinase 3 beta (GSK-3ß) through phosphatidylinositide 3 kinases (PI3K)/protein kinase B (AKT) signaling pathway. CONCLUSIONS: It is concluded that bFGF can promote EMT and motility of PC-3 cells, and AKT/GSK-3ß signaling pathway controls the stability, localization and transcription of Snail which is crucial for this bFGF induced EMT. GENERAL SIGNIFICANCE: To our knowledge, this is the first study to demonstrate that bFGF can induce EMT via AKT/GSK-3ß/Snail signaling pathway in prostate cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition , Fibroblast Growth Factor 2/metabolism , Glycogen Synthase Kinase 3/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , Fibroblast Growth Factor 2/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
15.
Cell Tissue Res ; 358(2): 491-502, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25124796

ABSTRACT

Cancer metastasis is considered a major challenge in cancer therapy. Recently, epidermal growth factor (EGF)/epidermal growth factor receptor (EGFR) signaling has been shown to induce epithelial-mesenchymal transition (EMT) and thereby to promote cancer metastasis. However, the underlying mechanism has not been fully elucidated. We demonstrate that EGF can induce EMT in human prostate and lung cancer cells and thus promote invasion and migration. EGF-induced EMT has been characterized by the cells acquiring mesenchymal spindle-like morphology and increasing their expression of N-cadherin and fibronectin, with a concomitant decrease of E-cadherin. Both protein and mRNA expression of transcription factor Snail rapidly increases after EGF treatment. The knockdown of Snail significantly attenuates EGF-induced EMT, suggesting that Snail is crucial for this process. To determine the way that Snail is accumulated, we demonstrate (1) that EGF promotes the stability of Snail via inhibiting the activity of glycogen synthase kinase 3 beta (GSK-3ß), (2) that protein kinase C (PKC) rather than the phosphatidylinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway is responsible for GSK-3ß inhibition and (3) that GSK-3ß inhibition promotes the transcription of Snail. Taken together, these results reveal that the PKC/GSK-3ß signaling pathway controls both the stability and transcription of Snail, which is crucial for EMT induced by EGF in PC-3 and A549 cells. Our study suggests a novel signaling pathway for Snail regulation and provides a better understanding of growth-factor-induced tumor EMT and metastasis.


Subject(s)
Epidermal Growth Factor/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Glycogen Synthase Kinase 3/metabolism , Neoplasms/pathology , Protein Kinase C/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/genetics , Protein Stability/drug effects , Protein Transport/drug effects , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription, Genetic/drug effects , Up-Regulation/drug effects
16.
Cell Immunol ; 289(1-2): 42-8, 2014.
Article in English | MEDLINE | ID: mdl-24721110

ABSTRACT

Macrophages can be divided into two groups as M1 and M2 phenotype. Our results and other groups revealed that IFN-γ can up-regulate the IDO expression and differentiate THP-1 cells to M1 phenotype. Therefore we hypothesized that IDO may play potential roles in macrophage differentiation. Interesting, our results indicated that the ectopic IDO increases the expression of M2 markers such as IL-10 and CXCR4 while decreases the M1 markers such as CCR7 and IL-12p35. In contrast, the knockdown of IDO expression in THP-1 cells resulted in increased M1 markers and lower M2 markers. Our results suggested that the expression intensity of IDO modulates macrophages differentiation. These finding support the counter-regulatory role for IDO with regarding to the polarization of macrophages to restrain excessive or inappropriate immune activation in inflammatory or tumor microenvironment. It throws new light on the mechanisms about the immunosuppressive effect of IDO in tumor or inflammatory diseases.


Subject(s)
Cell Differentiation/immunology , Cell Polarity/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Macrophages/immunology , Cell Line, Tumor , Humans , Immune Tolerance/immunology , Immunologic Factors/genetics , Immunologic Factors/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interleukin-10/biosynthesis , Interleukin-12 Subunit p35/biosynthesis , Leukemia/immunology , Macrophages/classification , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Receptors, CCR7/biosynthesis , Receptors, CXCR4/biosynthesis
17.
Mol Biosyst ; 10(6): 1320-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24651282

ABSTRACT

Curcumin (diferuloylmethane) is a polyphenol natural product of the plant Curcuma longa, and has a diversity of antitumor activities. However, the clinical application of curcumin remains limited due to its poor pharmacokinetic characteristics. It is therefore critical to develop structural analogues of curcumin with increasing anticancer activity. T63, a new 4-arylidene curcumin analogue, was synthesized in our previous studies and exhibited higher in vitro and in vivo anti-tumor activities compared to curcumin. However, the precise molecular mechanism of its anti-tumor effects has not been well elucidated. Using a two-dimensional gel electrophoresis (2-DE)-based proteomic approach, we identified 66 differentially expressed proteins. Similarly to curcumin, T63 showed a diverse range of molecular targets. We proposed that induction of ROS generation and mitochondrial dysfunction, inhibition of proteasome, HSP90, and 14-3-3 proteins play important roles in T63-induced cell cycle arrest and apoptosis. These data indicate that the novel curcumin analogue T63 is a potent anti-tumor agent, which can induce cell cycle arrest and apoptosis, and also provided valuable resources for further study of the anti-tumor effects and molecular mechanisms of T63.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Mitochondria/drug effects , Oxidation-Reduction , Proteomics
18.
Eur J Immunol ; 44(1): 173-83, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24114072

ABSTRACT

Nodal, a member of the TGF-ß superfamily, is an embryonic morphogen that is upregulated in different types of tumors. Nodal increases the tumorigenesis by inducing angiogenesis and promoting metastasis. Importantly, Nodal inhibition suppresses the growth and invasion of tumor. Since tumor-associated macrophages (TAMs) are the major infiltrating leukocytes in most cancers, we investigated whether Nodal is involved in the differentiation of TAMs. Our results revealed that Nodal inhibition in tumor microenvironment upregulated the production of IL-12 in macrophages and reversed TAMs to classically activated macrophage phenotype. In contrast, treatment with recombinant Nodal (rNodal) decreased the expression of IL-12 in murine macrophages. Furthermore, rNodal promoted macrophage polarization to an alternatively activated macrophage-like/TAM phenotype and modulated its function. These results suggest that Nodal may play an important role in macrophage polarization and downregulation of IL-12. The rescued antitumor function of TAMs via the inhibition of Nodal expression could be a new therapeutic strategy for cancer treatment.


Subject(s)
Bone Marrow Cells/immunology , Interleukin-12/metabolism , Macrophages/immunology , Neoplasms/immunology , Nodal Protein/metabolism , Recombinant Proteins/metabolism , Animals , Carcinogenesis , Cell Differentiation , Cell Line, Tumor , Down-Regulation , Humans , Lymphocyte Culture Test, Mixed , Macrophage Activation , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Nodal Protein/genetics , Nodal Protein/immunology , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Th2 Cells/immunology
19.
Eur J Pharmacol ; 723: 156-66, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24333218

ABSTRACT

Cisplatin is a first-line chemotherapeutic agent in the treatment of non-small cell lung cancer (NSCLC), but the therapeutic effect is disappointing, partly due to drug resistance. Emerging evidence showed that chemoresistance associates with acquisition of epithelial-mesenchymal transition (EMT) phenotype and cancer stem cell-like properties. However, the underlying mechanism is not entirely clear. In this study, we showed that cisplatin-resistant A549 cells (A549/CDDP) acquire EMT phenotype associated with migratory and invasive capability. A549/CDDP cells also displayed enhanced cancer stem cell-like properties. Increased expression of transcription factor Snail, but not ZEB1, Slug and Twist, was observed in A549/CDDP cells. Knockdown of Snail reversed EMT and significantly attenuated migration, invasion and cancer stem cell-like properties of A549/CDDP cells. Conversely, overexpressed Snail in A549 cells induced EMT and cancer stem cell-like properties. Finally, we demonstrated that activated AKT signal leads to increased ß-catenin expression and subsequently up-regulates Snail in A549/CDDP cells. Taken together, these results revealed that AKT/ß-catenin/Snail signaling pathway is mechanistically associated with cancer stem cell-like properties and EMT features of A549/CDDP cells, and thus, this pathway could be a novel target for the treatment of NSCLC.


Subject(s)
Drug Resistance, Neoplasm/physiology , Epithelial-Mesenchymal Transition/physiology , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Antineoplastic Agents , Cell Line, Tumor , Cisplatin , Humans , Neoplastic Stem Cells , Phenotype , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/genetics
20.
Eur J Pharmacol ; 714(1-3): 48-55, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23769744

ABSTRACT

Metastasis induced by chronic inflammation has been considered as a major challenge during cancer therapy. Epithelial-mesenchymal transition (EMT) is associated with cancer invasion and metastasis promoted by pro-inflammatory cytokine TNFα. However, the mechanisms underlying TNFα-induced EMT in prostate cancer cells is not entirely clear. Here we showed that EMT induced by longstanding stimulation with TNFα in prostate cancer PC3 cells is mediated by up-regulation of the transcriptional repressor Snail. TNFα-mediated EMT was characterized by acquiring mesenchymal fusiform morphology, increasing the expression of Vimentin and decreasing the expression of E-cadherin. Exposure to TNFα increased the expression of transcription factor Snail via post-transcriptional regulation process and induced Snail nuclear localization in PC3 cells. Moreover, overexpressed Snail in PC3 cells induced EMT. Conversely, suppressing Snail expression abrogated TNFα-induced EMT, suggesting that Snail plays a crucial role in TNFα-induced EMT in prostate cancer cells. Finally, we showed that TNFα time-dependently activated NF-κB, AKT, ERK, p38 MAPK signaling pathways, and elevated Snail stability by activating AKT pathway that subsequently inhibited GSK-3ß activity. Taken together, these results reveal that stabilization of Snail via AKT/GSK-3ß signaling pathway is required for TNFα-induced EMT in prostate cancer cells. This study offers a better understanding of TNFα-induced metastasis and provides an effective therapeutic strategy for prostate cancer treatment.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Glycogen Synthase Kinase 3/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Protein Stability/drug effects , Protein Transport/drug effects , Snail Family Transcription Factors , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...