Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Head Neck ; 45(9): 2161-2172, 2023 09.
Article in English | MEDLINE | ID: mdl-37466293

ABSTRACT

BACKGROUND: Cysteine-rich protein 2 (CSRP2) is discovered as oncogene. The study aims to investigate the clinical significance and potential mechanism of CSRP2 in head and neck squamous cell carcinoma (HNSCC). METHODS: CSRP2 expression was explored by immunohistochemistry tissue microarrays and Western blotting in HNSCC. The effect of CSRP2 on the cancer stemness and epithelial-to-mesenchymal transition (EMT) of HNSCC cells was investigated by sphere formation, wound healing, and transwell assays. The vitro and vivo experiments revealed that CSRP2 modulated cancer stemness and EMT phenotypes in HNSCC. RESULTS: CSRP2 was overexpressed in HNSCC patients and presented poor prognosis. CSRP2 knockdown inhibited the migration and invasion ability of the HNSCC cells. And CSRP2 expression was closely associated with CSCs markers, EMT-transcription factor, new oncoprotein, and immune checkpoint. CONCLUSION: The overexpression of CSRP2 indicates poor prognosis and plays a key role in maintaining the cancer cell stemness and EMT.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/genetics , Transcription Factors/genetics , Phenotype , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Cell Movement , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Proteins/pharmacology , Nuclear Proteins/genetics , LIM Domain Proteins/genetics
2.
Int Immunopharmacol ; 119: 110243, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37137265

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is implicated in promoting HNSCC malignant progression. However, EZH2 inhibitors, when used alone, increase the number of myeloid-derived suppressor cells (MDSCs), which are responsible for enhancing tumor stemness and promoting tumor immune escape. We aimed to determine whether combining tazemetostat (an EZH2 inhibitor) and sunitinib (a MDSC inhibitor) can improve the response rate to an immune-checkpoint-blocking (ICB) therapy. We evaluated the efficacy of the above treatment strategies by bioinformatics analysis and animal experiments. EZH2 overexpression and abundant MDSCs in patients with HNSCC are associated with tumor progression. Tazemetostat treatment alone had limited inhibitory effect on HNSCC progression in the mouse models, accompanied by a surge in the number of MDSCs in the tumor microenvironment. Conversely, the combined use of tazemetostat and sunitinib reduced the number of MDSCs and regulatory T cell populations, promoting intratumoral infiltration of T cells and inhibiting of T cell exhausting, regulating of wnt/ß-catenin signaling pathway and tumor stemness, promoting the intratumoral PD-L1 expression and improved the response rate to anti-PD-1 therapy. The combined use of EZH2 and MDSC inhibitors effectively reverses HNSCC-specific immunotherapeutic resistance and is a promising strategy for overcoming resistance to ICB therapy.


Subject(s)
Head and Neck Neoplasms , Myeloid-Derived Suppressor Cells , Mice , Animals , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Sunitinib/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Tumor Microenvironment
3.
Cancer Lett ; 563: 216193, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37088326

ABSTRACT

Immunotherapy, particularly immune checkpoint blockade (ICB), has shown great promise in the treatment of cancer and emerged as a beacon of hope for patients who have exhausted traditional therapeutic options. Despite ICB's approval for the treatment of advanced tumors, its efficacy remains limited to a small subset of patients. As a systemic disease, cancer can induce changes in the composition and function of the systemic immune system, and ICB resistance often involves a dialog between the tumor microenvironment (TME) and the systemic immune macroenvironment. While investigations into tumor progression and ICB resistance have largely focused on the TME itself, the alterations in the systemic immune system and immune macroenvironment are still poorly understood. Given the spleen's role as the largest secondary lymphoid organ, its examination and discussion may provide valuable insights into the systemic immune status and TME components. Recent studies have highlighted the importance of the spleen in tumor progression and immunotherapy, particularly in the context of erythroid progenitor cells (EPCs), a significant cell subpopulation. In this review, we discuss the mechanisms and role of splenic extramedullary hematopoiesis (EMH) as an intermediary in tumor-host interactions and explore the mechanism of EPC-TME collusions. We further summarize the progress in EPC-targeting strategies and emphasize the potential for further research into the role and mechanisms of EPCs in tumor progression and treatment, which could have far-reaching implications.


Subject(s)
Erythroid Precursor Cells , Neoplasms , Humans , Neoplasms/therapy , Immunotherapy , Spleen , Tumor Microenvironment
4.
Cancer Immunol Immunother ; 72(6): 1647-1660, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36586012

ABSTRACT

The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome plays cell- and tissue-specific roles in cancer, meaning that its activation in different tumors or cells may play different roles in tumor progression. We have previously described the tumor-promoting function of tumor-intrinsic NLRP3/IL-1ß signaling in head and neck squamous cell carcinoma (HNSCC), but its role in immune cells remains unclear. In this study, we found that NLRP3 was highly expressed in tumor-associated macrophages (TAMs) in both mouse and human HNSCC, and the expression of NLRP3 was positively correlated with the density of TAMs according to immunohistochemistry, immunofluorescence, and flow cytometry analyses. Importantly, the number of NLRP3high TAMs was related to worse overall survival in HNSCC patients. Knocking out NLRP3 inhibited M2-like macrophage differentiation in vitro. Moreover, the carcinogenic effect induced by 4-nitroquinoline-1-oxide was decreased in Nlrp3-deficient mice, which had smaller tumor sizes. Genetic depletion of NLRP3 reduced the expression of protumoral cytokines, such as IL-1ß, IL-6, IL-10, and CCL2, and suppressed the accumulation of TAMs and myeloid-derived suppressor cells (MDSCs) in mouse HNSCC. Thus, activation of NLRP3 in TAMs may contribute to tumor progression and have prognostic significance in HNSCC.


Subject(s)
Head and Neck Neoplasms , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Mice , Animals , Squamous Cell Carcinoma of Head and Neck , Tumor-Associated Macrophages/metabolism , Interleukin-1beta/metabolism , Prognosis
5.
J Cell Mol Med ; 22(2): 1337-1349, 2018 02.
Article in English | MEDLINE | ID: mdl-29193723

ABSTRACT

Epithelial-mesenchymal transition (EMT) is associated with metastasis formation, generation and maintenance of cancer stem cells (CSCs). However, the regulatory mechanisms of CSCs have not been clarified. This study aims to investigate the role of TNF receptor-associated factor 6 (TRAF6) on EMT and CSC regulation in squamous cell carcinoma of head and neck (SCCHN). We found TRAF6 was overexpressed in human SCCHN tissues, and high TRAF6 expression was associated with lymphatic metastasis and resulted in poor prognosis in patients with SCCHN. In addition, elevated TRAF6 expression was observed in several HNSCC cell lines, and wound healing and transwell assay results showed that TRAF6 knockdown inhibited the migration and invasion ability of the SCCHN cells. Moreover, the expression of Vimentin, Slug and N-cadherin was down-regulated and that of E-cadherin was elevated after TRAF6 knockdown but decreased by transforming growth factor beta 1 (TGF-ß1) and CAL27 similar to mesenchymal cells formed after TGF-ß1 induction. In addition, the expression levels of CD44, ALDH1, KLF4 and SOX2 were inhibited after TRAF6 knockdown, and the anchor-dependent colony formation number and sphere number were remarkably reduced. Flow cytometry showed TRAF6 knockdown reduced ALDH1-positive cancer stem cells. We also demonstrated that TRAF6 is closely associated with EMT process and cancer stem cells using a Tgfbr1/Pten 2cKO mice SCCHN model and human SCCHN tissue microarray. Our findings indicate that TRAF6 plays a role in EMT phenotypes, the generation and maintenance of CSCs in SCCHN, suggesting that TRAF6 is a potential therapeutic target for SCCHN.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , TNF Receptor-Associated Factor 6/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Kruppel-Like Factor 4 , Lymphatic Metastasis/pathology , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype
6.
Cell Mol Life Sci ; 75(11): 2045-2058, 2018 06.
Article in English | MEDLINE | ID: mdl-29184980

ABSTRACT

The NLRP3 inflammasome is a critical innate immune pathway responsible for producing active interleukin (IL)-1ß, which is associated with tumor development and immunity. However, the mechanisms regulating the inflammatory microenvironment, tumorigenesis and tumor immunity are unclear. Herein, we show that the NLRP3 inflammasome was over-expressed in human HNSCC tissues and that the IL-1ß concentration was increased in the peripheral blood of HNSCC patients. Additionally, elevated NLRP3 inflammasome levels were detected in tumor tissues of Tgfbr1/Pten 2cKO HNSCC mice, and elevated IL-1ß levels were detected in the peripheral blood serum, spleen, draining lymph nodes and tumor tissues. Blocking NLRP3 inflammasome activation using MCC950 remarkably reduced IL-1ß production in an HNSCC mouse model and reduced the numbers of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and tumor-associated macrophages (TAMs). Moreover, inhibiting NLRP3 inflammasome activation increased the numbers of CD4+ and CD8+ T cells in HNSCC mice. Notably, the numbers of exhausted PD-1+ and Tim3+ T cells were significantly reduced. A human HNSCC tissue microarray showed that NLRP3 inflammasome expression was correlated with the expression of CD8 and CD4, the Treg marker Foxp3, the MDSC markers CD11b and CD33, and the TAM markers CD68 and CD163, PD-1 and Tim3. Overall, our results demonstrate that the NLRP3 inflammasome/IL-1ß pathway promotes tumorigenesis in HNSCC and inactivation of this pathway delays tumor growth, accompanied by decreased immunosuppressive cell accumulation and an increased number of effector T cells. Thus, inhibition of the tumor microenvironment through the NLRP3 inflammasome/IL-1ß pathway may provide a novel approach for HNSCC therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Humans , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , T-Lymphocytes, Regulatory/immunology
7.
J Exp Clin Cancer Res ; 36(1): 116, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28865486

ABSTRACT

BACKGROUND: NLRP3 inflammasome acts as a danger signal sensor that triggers and coordinates the inflammatory response. However, the roles of NLRP3 inflammasome in the tumorigenesis and development of cancer stem cells (CSCs) of squamous cell carcinoma of the head and neck (SCCHN) remain ambiguous. METHODS: In our study, tissue microarrays, ELISA, sphere-forming assay, colony formation assay and Western blot analysis were performed to evaluate the effect of NLRP3 inflammasome on the development of CSCs in human SCCHN tissue specimen, cell lines, and transgenic mouse SCCHN model. RESULTS: The components of NLRP3 inflammasome, namely, NLRP3, ASC, Caspase-1, and IL-18 were correlated with CSCs markers BMI1, ALDH1 and CD44 in human SCCHN specimens. Moreover, NLRP3, Caspase-1, IL-1ß, and IL-18 were highly expressed in SCCHN cell lines. NLRP3 inflammasome activated by LPS and ATP promoted sphere-forming and colony formation capacities along with an upregulation of BMI1, ALDH1 and CD44. In addition, NLRP3 inflammasome blockade by NLRP3 inhibitor MCC950 reduced sphere and colony number, also decreased the expression of BMI1, ALDH1 and CD44 in SCCHN cell lines. Expression of NLRP3, ASC, Caspase-1, IL-1ß, IL-18, BMI1, ALDH1 and CD44 was upregulated in Tgfbr1/Pten 2cKO mouse SCCHN model, and NLRP3 inflammasome expression was closely related to those CSCs makers in mice SCCHN. However, MCC950 treatment reduced the expression of NLRP3 inflammasome, CSCs markers BMI1, ALDH1 and CD44 in Tgfbr1/Pten 2cKO mice SCCHN. In addition, blockade of NLRP3 inflammasome can also delayed the tumor-burdened speed in SCCHN mice. CONCLUSIONS: Our study demonstrates that NLRP3 inflammasome was upregulated and associated with the carcinogenesis and CSCs self-renewal activation in SCCHN. NLRP3 inflammasome can be a potential target in the development of novel approaches for head and neck squamous cell carcinoma therapy.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Inflammasomes/genetics , Inflammation/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neoplasm Proteins/genetics , Animals , Carcinogenesis/genetics , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic/genetics , Head and Neck Neoplasms/pathology , Humans , Inflammasomes/antagonists & inhibitors , Inflammation/drug therapy , Inflammation/pathology , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis
8.
Cancer Immunol Immunother ; 66(5): 627-636, 2017 May.
Article in English | MEDLINE | ID: mdl-28236118

ABSTRACT

V-domain Ig suppressor of T cell activation (VISTA), a novel immune checkpoint regulatory molecule, suppresses T cell mediated immune responses. The aim of the present study was to profile the immunological expression, clinical significance and correlation of VISTA in human oral squamous cell carcinoma (OSCC). Human tissue microarrays, containing 165 primary OSCCs, 48 oral epithelial dysplasias and 43 normal oral mucosae, were applied to investigate the expression levels of VISTA, CD8, cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), programmed death ligand 1 (PD-L1), PI3Kα p110, IL13Rα2, phospho-STAT3 at tyrosine 705 (p-STAT3) and myeloid-derived suppressor cell (MDSC) markers (CD11b and CD33) by immunohistochemistry and digital pathology analysis. The results demonstrated that the protein level of VISTA was significantly higher in human OSCC specimens, and that VISTA expression in primary OSCCs was correlated with lymph node status. VISTA expression did not serve as an independent predictor for poor prognosis, while patient subgroup with VISTA high and CD8 low expression (22/165) had significantly poorer overall survival compared with other subgroups based on the multivariate and Cox hazard analyses among the primary OSCC patients in the present cohort. Additionally, the expression of VISTA was significantly correlated with PD-L1, CTLA-4, IL13Rα2, PI3K, p-STAT3, CD11b and CD33 according to Pearson's correlation coefficient test. Taken together, the results indicated that the VISTA high and CD8 low group, as an immunosuppressive subgroup, might be associated with a poor prognosis in primary OSCC. These findings indicated that VISTA might be a potential immunotherapeutic target in OSCC treatment.


Subject(s)
B7 Antigens/biosynthesis , CD8 Antigens/immunology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Mouth Neoplasms/immunology , B7-H1 Antigen/biosynthesis , CTLA-4 Antigen/biosynthesis , Carcinoma, Squamous Cell/pathology , Humans , Immunosuppression Therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Prognosis , STAT3 Transcription Factor/biosynthesis , Survival Analysis
9.
Mol Oncol ; 11(2): 235-247, 2017 02.
Article in English | MEDLINE | ID: mdl-28102051

ABSTRACT

T-cell immunoglobulin mucin 3 (TIM3) contributes to immune suppression during progression of many cancers, but the precise role of TIM3 in head and neck squamous cell carcinoma (HNSCC) is not clearly understood. In this study, we report that TIM3 expression was significantly up-regulated in patients with HNSCC and associated with lymph node metastasis. Additionally, TIM3 expression was increased in patients with recurrent HNSCC and patients with preradiotherapy or prechemotherapy. We also characterized CD8+ T cells and CD11b+ CD33+ myeloid-derived suppressor cells (MDSCs) in human HNSCC, and found that their expression was positively correlated with TIM3 expression. To determine the underlying mechanism of TIM3 in immune response during HNSCC progression, we utilized the Tgfbr1/Pten 2cKO HNSCC mouse model with TIM3 overexpression. Treatment with anti-TIM3 monoclonal antibody effectively suppressed tumor growth through restoring effector T-cell function by targeting CD4+ TIM3+ cells and CD8+ TIM3+ cells and decreasing MDSCs. Our findings demonstrate TIM3 expression in patients with HNSCC and suggest anti-TIM3 immunotherapy as a novel therapeutic approach for effective treatment of HNSCC.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Head and Neck Neoplasms/immunology , Immunity, Cellular , Mucin-3/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Female , Head and Neck Neoplasms/pathology , Humans , Male , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Neoplasms, Experimental/pathology
10.
Sci Rep ; 6: 24704, 2016 04 25.
Article in English | MEDLINE | ID: mdl-27108536

ABSTRACT

Cancer stem cells (CSCs) are considered responsible for tumor initiation and chemoresistance. This study was aimed to investigate the possibility of targeting head neck squamous cell carcinoma (HNSCC) by NOTCH1 pathway inhibition and explore the synergistic effect of combining NOTCH inhibition with conventional chemotherapy. NOTCH1/HES1 elevation was found in human HNSCC, especially in tissue post chemotherapy and lymph node metastasis, which is correlated with CSCs markers. NOTCH1 inhibitor DAPT (GSI-IX) significantly reduces CSCs population and tumor self-renewal ability in vitro and in vivo. Flow cytometry analysis showed that NOTCH1 inhibition reduces CSCs frequency either alone or in combination with chemotherapeutic agents, namely, cisplatin, docetaxel, and 5-fluorouracil. The combined strategy of NOTCH1 blockade and chemotherapy synergistically attenuated chemotherapy-enriched CSC population, promising a potential therapeutic exploitation in future clinical trial.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Diamines/therapeutic use , Head and Neck Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Receptor, Notch1/antagonists & inhibitors , Thiazoles/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Self Renewal/drug effects , Cisplatin/therapeutic use , Diamines/pharmacology , Docetaxel , Drug Synergism , Drug Therapy, Combination , Female , Fluorouracil/therapeutic use , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/physiology , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Taxoids/therapeutic use , Thiazoles/pharmacology , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(10): 11651-63, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26872381

ABSTRACT

To assess the role of transcription factor µE3 (TFE3) in the tumorigenesis of head and neck squamous cell carcinoma (HNSCC), human HNSCC tissue arrays were investigated for TFE3 expression. Human HNSCC tissues with neoadjuvant inductive chemotherapey (docetaxel, cisplatin and fluorouracil, TPF) and mice HNSCC tissues from transgenic mice model were evaluated for TFE3 expression and the hypoxia pathway. The roles of EGF/EGFR mediated hypoxia in TFE3 nuclear expression were analyzed in vitro and in vivo. TFE3 expression was higher in human HNSCC tissues compared with that in normal oral mucosa. Moreover, high TFE3 expression was related to HIF-1α, PAI-1, and EGFR, which demonstrated the activation of the hypoxia pathway in HNSCC tissues. Furthermore, elevated TFE3 expression was observed in HNSCC after cisplatin-based chemotherapy, and high TFE3 expression may indicate poor response to TPF inductive chemotherapy. Furthermore, similar changes with increased TFE3 were observed in HNSCC of the transgenic mouse HNSCC model. Hypoxic culture in the human HNSCC cell line increased TFE3 expression, which promoted cell survival under hypoxia. EGFR inhibiton by cetuximab could attenuate hypoxia-induced TFE3 in the HNSCC cell line and transgenic mouse HNSCC model. These findings indicated that TFE3 was an important hypoxia-induced transcriptional factor in HNSCC. TFE3 could be regarded as a durgable therapeutic oncotarget by EGFR inhibition.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Carcinoma, Squamous Cell/metabolism , Cell Hypoxia/physiology , Head and Neck Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Hypoxia/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , ErbB Receptors/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Plasminogen Activator Inhibitor 1/metabolism , Serpin E2/metabolism , Squamous Cell Carcinoma of Head and Neck , Transfection
12.
Pathol Oncol Res ; 22(1): 109-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26350055

ABSTRACT

Salivary Adenoid Cystic Carcinoma (SACC) is a tumor characterized by inevitable local progression and terminal hematogenous metastasis. This study aimed to investigate the expression of LC3, LAMP2, KEAP1 and NRF2 in SACC. Human salivary gland tissue microarray which contains 74 SACC, 12 pleomorphic adenoma and 18 normal salivary gland specimens. High expression of LC3, LAMP2, KEAP1 and NRF2 were found in SACC patients, and LC3, LAMP2, KEAP1 and NRF2 expression were significantly higher in SACC than as compared with pleomorphic adenoma and (or) normal salivary gland. The expression of NRF2 was correlated with pathological type of human SACC (P < 0.05). Moreover, the high-expression of KEAP1 had significant correlations with LC3 (P < 0.001, R = 0.3195), and LAMP2 (P < 0.001, R = 0.3346) and NRF2 (P < 0.05, R = 0.2246) by using the Pearson correlation coefficient test. Our findings demonstrated that up-regulation of LC3, LAMP2, KEAP1 and NRF2 were associated with carcinogenesis and progression of SACC patients, suggesting that they may be useful molecular targets in salivary adenoid cystic carcinoma.


Subject(s)
Adenoma, Pleomorphic/metabolism , Carcinoma, Adenoid Cystic/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Microtubule-Associated Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Salivary Gland Neoplasms/metabolism , Adenoma, Pleomorphic/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Adenoid Cystic/pathology , Case-Control Studies , Cohort Studies , Humans , Immunoenzyme Techniques , Kelch-Like ECH-Associated Protein 1 , Neoplasm Grading , Neoplasm Staging , Prognosis , Salivary Gland Neoplasms/pathology , Salivary Glands/metabolism
13.
Oncotarget ; 6(39): 41944-58, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26556875

ABSTRACT

Signaling transducer and activator 3 (STAT3) and cancer stem cells (CSCs) have garnered huge attention as a therapeutic focus, based on evidence that they may represent an etiologic root of tumor initiation and radio-chemoresistance. Here, we investigated the high phosphorylation status of STAT3 (p-STAT3) and its correlation with self-renewal markers in head neck squamous cell carcinoma (HNSCC). Over-expression of p-STAT3 was found to have increased in post chemotherapy HNSCC tissue. We showed that blockade of p-STAT3 eliminated both bulk tumor and side population (SP) cells with characteristics of CSCs in vitro. Inhibition of p-STAT3 using small molecule S3I-201 significantly delayed tumorigenesis of spontaneous HNSCC in mice. Combining blockade of p-STAT3 with cytotoxic drugs cisplatin, docetaxel, 5-fluorouracil (TPF) enhanced the antitumor effect in vitro and in vivo with decreased tumor sphere formation and SP cells. Taken together, our results advocate blockade of p-STAT3 in combination with conventional chemotherapeutic drugs enhance efficacy by improving CSCs eradication in HNSCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Side-Population Cells/drug effects , Aminosalicylic Acids/pharmacology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Self Renewal/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Docetaxel , Dose-Response Relationship, Drug , Fluorouracil/pharmacology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice, Knockout , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Phosphorylation , STAT3 Transcription Factor/metabolism , Side-Population Cells/metabolism , Side-Population Cells/pathology , Squamous Cell Carcinoma of Head and Neck , Taxoids/pharmacology , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Oncotarget ; 6(41): 43581-93, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26561201

ABSTRACT

Autophagy is considered as a double-edged sword. It can prolong the survival of cancer cells and enhance its resistance to apoptosis, and paradoxically, defective autophagy has been linked to increased tumorigenesis, but the mechanism behind this phenomenon is unclear. In this study, we demonstrated that decreased phosphorylation of signal transducer and activator of transcription 3 (p-STAT3) was correlated with increased autophagy through the Akt/mTOR and Erk signaling pathways in human head and neck squamous cell carcinoma (HNSCC). We also showed that blockage of STAT3 by NSC74859 could markedly induce apoptotic cell death and autophagy. Meanwhile, increased autophagy inhibited apoptosis. The pharmacological or genetic inhibition of autophagy and STAT3 further sensitized HNSCC cells to apoptosis. Furthermore, evidence from xenograft model proved that suppressed STAT3 activity combined with inhibition of autophagy promoted tumor regression better than either treatment alone. Taken together, this present study demonstrated that autophagy alleviates apoptotic cell death in HNSCC, and combination of inhibition of STAT3 by NSC74859 and autophagy might be a promising new therapeutic strategy for HNSCC.


Subject(s)
Autophagy/physiology , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/physiology , Aminosalicylic Acids/pharmacology , Animals , Autophagy/drug effects , Benzenesulfonates/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , Female , Head and Neck Neoplasms/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Phosphorylation , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis , Xenograft Model Antitumor Assays
15.
Oncotarget ; 6(39): 42067-80, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26573233

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) and tumor associated macrophages (TAMs) play key roles in the tumor immune suppressive network and tumor progression. However, precise roles of programmed death-1 (PD-1) in immunological functions of MDSCs and TAMs in head and neck squamous cell carcinoma (HNSCC) have not been clearly elucidated. In the present study, we show that PD-1 and PD-L1 levels were significantly higher in human HNSCC specimen than in normal oral mucosa. MDSCs and TAMs were characterized in mice and human HNSCC specimen, correlated well with PD-1 and PD-L1 expression. αPD-1 treatment was well tolerated and significantly reduced tumor growth in the HNSCC mouse model along with significant reduction in MDSCs and TAMs in immune organs and tumors. Molecular analysis suggests a reduction in the CD47/SIRPα pathway by PD-1 blockade, which regulates MDSCs, TAMs, dendritic cell as well as effector T cells. Hence, these data identify that PD-1/PD-L1 axis is significantly increased in human and mouse HNSCC. Adoptive αPD-1 immunotherapy may provide a novel therapeutic approach to modulate the micro- and macro-environment in HNSCC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , Head and Neck Neoplasms/drug therapy , Myeloid Cells/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/immunology , Antigens, Differentiation/genetics , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Blotting, Western , CD47 Antigen/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Immunohistochemistry , Macrophages/drug effects , Macrophages/metabolism , Mice, Knockout , Myeloid Cells/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Papillomaviridae/physiology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Immunologic/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Am J Cancer Res ; 5(5): 1751-61, 2015.
Article in English | MEDLINE | ID: mdl-26175943

ABSTRACT

In this study, we accessed the expression and correlation of p-STAT3 with Survivin, Cyclin D1, CD147, Slug and Ki67 by immunohistochemical staining of human tissue microarray which contains 72 adenoid cystic carcinoma (AdCC), 12 pleomorphic adenoma (PMA) and 18 normal salivary gland (NSG) using digital pathological scanner and scoring system. We found that the expression of p-STAT3, Survivin, Slug, Cyclin D1 and CD147 was significantly increased in AdCC as compared with PMA and (or) NSG (p<0.05). While, the level of p-STAT3 and expression of Cyclin D1 and CD147 was not associated with pathological type of human AdCC (p>0.05). Correlation analysis of these proteins revealed that p-STAT3 up-regulates the expression of Survivin, Slug, Cyclin D1 and CD147 (p<0.05). Moreover, the activation of STAT3 was associated with proliferation marker Ki-67 (p<0.05). Selective inhibition of STAT3 by a small molecule S3I-201 significantly reduced human SACC-83 and SACC-LM cells proliferation, migration and invasion with the corresponding decrease in expression of Survivin, Slug, Cyclin D1 and CD147. These findings indicate that high phosphorylation level of STAT3 in AdCC is related to Survivin, Slug, Cyclin D1 and CD147. We suggest that the inhibition of STAT3 may be a novel strategy for neoadjuvant chemotherapeutic treatment of AdCC.

17.
Oncotarget ; 6(11): 8807-21, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25871396

ABSTRACT

Anterior gradient protein 2 (AGR2) is a novel biomarker with potential oncogenic role. We sought to investigate the diagnostic and prognostic role of AGR2 on head and neck squamous cell carcinoma (HNSCC) with an emphasis on its correlation of cancer stemloid cells (CSC) and epithelial mesenchymal transition (EMT). We found that AGR2 protein levels were higher in HNSCC than in normal oral mucosa. High levels of AGR2 were associated with the T category, pathological grade and lymph node metastasis of HNSCC. Expression of AGR2 increased in recurring HNSCC after radiotherapy and in post cisplatin-based chemotherapeutic tissues. In HNSCC cell lines, knock-down of AGR2 induced apoptosis, reduced sphere formation, and down-regulated Survivin, Cyclin D1, Bcl2, Bcl2l1, Slug, Snail, Nanog and Oct4. In addition, over-expressed AGR2 in transgenic mice with spontaneous HNSCC was associated with lost function of Tgfbr1 and/ or lost function of Pten. In vitro knockdown TGFBR1 in HNSCC cell lines increased AGR2 expression. These results suggest that AGR2 is involved in EMT and self-renewal of CSC and may present a potential therapeutic target (oncotarget) for HNSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition/genetics , Head and Neck Neoplasms/pathology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/metabolism , Proteins/physiology , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , Cell Self Renewal/genetics , Cisplatin/administration & dosage , Cisplatin/pharmacology , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Mice, Knockout , Mouth Mucosa/chemistry , Mucoproteins , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oncogene Proteins , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , RNA, Small Interfering , Radiotherapy, Adjuvant , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Spheroids, Cellular , Taxoids/administration & dosage , Taxoids/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics
18.
Am J Transl Res ; 7(1): 162-74, 2015.
Article in English | MEDLINE | ID: mdl-25755838

ABSTRACT

Epithelial-mesenchymal transition (EMT) is considered to have pivotal roles in the invasive and metastatic of Adenoid cystic carcinoma (AdCC) which is marked by local infiltration and distant metastasis. Notch signaling abnormity has been implicated as important molecular events in recent next generation sequencing studies of AdCC, but the detail is still unclear. This study was designed to investigate the expression of Notch signaling pathway and its relation with EMT program in AdCC. We constructed custom-made Tissue microarray (TMA) to evaluate the immunoreactivity of Notch signaling and EMT program and found that Notch signaling increase consecutively from NSG, PMA to AdCC, suggesting Notch signaling pathway may be associated with human AdCC progression. Then, we carried out Pearson correlation analysis and showed a close correlation of Notch signaling and EMT progression. When blocking Notch signaling pathway with γ-secretase inhibitor DAPT, EMT progression was decreased and migration and invasion ability were declined. Collectively, these findings suggest the vital roles of Notch signaling pathway in AdCC progression through their relationship with EMT progress. Targeting Notch signaling may provide further understanding of the mechanism of invasion and metastasis of AdCC as well as potential clinical therapeutics.

19.
Am J Cancer Res ; 5(12): 3505-15, 2015.
Article in English | MEDLINE | ID: mdl-26885441

ABSTRACT

C4.4A, a member of the Ly6/uPAR family of membrane proteins, has been identified as a metastasis-associated molecule, but little is known about its actual expression and possible function in head and neck squamous cell carcinoma (HNSCC). To explore diagnostic and prognostic roles of C4.4A in HNSCC, we investigated the expression of C4.4A in human HNSCC tissue array which contains 43 HNSCC, 6 epithelial dysplasia and 16 normal oral mucosa. Expression of C4.4A was significantly increased in epithelial dysplasia and HNSCC when compared with normal oral mucosa. Moreover, high C4.4A expression indicated a rather poor prognosis of HNSCC patients. To better understand the function of C4.4A in HNSCC progression, we investigated epithelial to mesenchymal transition (EMT) associated proteins including transforming growth factor (TGF-ß1), Slug and CD147 in HNSCC. The expression of TGF-ß1, Slug, and CD147 was significantly increased in HNSCC when compared with normal oral mucosa. Meanwhile, the expression of C4.4A was significantly correlated with TGF-ß1, Slug, and CD147 in HNSCC tissue array. Furthermore, knockdown of C4.4A decreased the cell invasion and migration in CAL27 cell line and suppressed the EMT with increased E-cadherin and decreased N-cadherin and Slug. Our study demonstrated that C4.4A was a potential marker for prognosis of HNSCC, and C4.4A participated in EMT program in HNSCC progression.

20.
PLoS One ; 9(12): e114051, 2014.
Article in English | MEDLINE | ID: mdl-25485635

ABSTRACT

In the present study, we explored the expression and correlation of survivin with HIF-1α, TGF-ß1 and TFE3 in adenoid cystic carcinoma (AdCC). The expression of survivin, HIF-1α, TGF-ß1 and TFE3 was assessed by immunohistochemical staining of a tissue microarray containing tissue samples of normal salivary gland (NSG), pleomorphic adenoma (PA) and AdCC. Correlation analysis of these proteins revealed that increased survivin expression was associated with the overexpression of HIF-1α (P<0.001, r = 0.5599), TGF-ß1 (P<0.001, r = 0.6616) and TFE3 (P<0.001, r = 0.7747). The expression of survivin, HIF-1α, TGF-ß1 and TFE3 was not correlated with the pathological type of human AdCC (P>0.05). Selective inhibition of survivin by YM155 and siRNA significantly reduced human SACC-83 cell proliferation, with the corresponding decrease in expression of HIF-1α, TGF-ß1 and TFE3. The data indicate that the overexpression of survivin in AdCC is related to HIF-1α, TGF-ß1 and TFE3. We hypothesize from these findings that the inhibition of survivin may be a novel strategy for neoadjuvant chemotherapeutic and radiosensitive treatment of AdCC.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Adenoid Cystic/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Salivary Gland Neoplasms/metabolism , Transforming Growth Factor beta1/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/pathology , Case-Control Studies , Cell Line, Tumor , Cluster Analysis , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Imidazoles/pharmacology , Immunohistochemistry , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Naphthoquinones/pharmacology , Neoplasm Grading , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Survivin , Transforming Growth Factor beta1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...