Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Commun ; 15(1): 3884, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719909

ABSTRACT

Only a minority of cancer patients benefit from immune checkpoint blockade therapy. Sophisticated cross-talk among different immune checkpoint pathways as well as interaction pattern of immune checkpoint molecules carried on circulating small extracellular vesicles (sEV) might contribute to the low response rate. Here we demonstrate that PD-1 and CD80 carried on immunocyte-derived sEVs (I-sEV) induce an adaptive redistribution of PD-L1 in tumour cells. The resulting decreased cell membrane PD-L1 expression and increased sEV PD-L1 secretion into the circulation contribute to systemic immunosuppression. PD-1/CD80+ I-sEVs also induce downregulation of adhesion- and antigen presentation-related molecules on tumour cells and impaired immune cell infiltration, thereby converting tumours to an immunologically cold phenotype. Moreover, synchronous analysis of multiple checkpoint molecules, including PD-1, CD80 and PD-L1, on circulating sEVs distinguishes clinical responders from those patients who poorly respond to anti-PD-1 treatment. Altogether, our study shows that sEVs carry multiple inhibitory immune checkpoints proteins, which form a potentially targetable adaptive loop to suppress antitumour immunity.


Subject(s)
B7-1 Antigen , B7-H1 Antigen , Extracellular Vesicles , Programmed Cell Death 1 Receptor , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Programmed Cell Death 1 Receptor/metabolism , Humans , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Animals , Mice , Cell Line, Tumor , Female , Neoplasms/immunology , Neoplasms/pathology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immune Tolerance , Mice, Inbred C57BL , Male , Tumor Microenvironment/immunology
2.
Biomolecules ; 14(2)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38397460

ABSTRACT

MicroRNAs (miRNAs) in oral squamous cell carcinoma (OSCC)-derived small extracellular vesicles (sEVs) play a pivotal role in modulating intercellular communications between tumor cells and other cells in the microenvironment, thereby influencing tumor progression and the efficacy of therapeutic interventions. However, a comprehensive inventory of these secretory miRNAs in sEVs and their biological and clinical implications remains elusive. This study aims to profile the miRNA content of OSCC cell line sEVs and computationally elucidate their biological and clinical relevance. We conducted miRNA sequencing to compare the miRNA profiles of OSCC cells and their corresponding sEVs. Our motif enrichment analysis identified specific sorting motifs that are implicated in either cellular retention or preferential sEV secretion. Target cell analysis suggested that the sEV miRNAs potentially interact with various immune cell types, including natural killer cells and dendritic cells. Additionally, we explored the clinical relevance of these miRNAs by correlating their expression levels with TNM stages and patient survival outcomes. Intriguingly, our findings revealed that a distinct sEV miRNA signature is associated with lymph node metastasis and poorer survival in patients in TCGA-HNSC dataset. Collectively, this research furthers our understanding of the miRNA sorting mechanisms in OSCC and underscores their clinical implications.


Subject(s)
Carcinoma, Squamous Cell , Extracellular Vesicles , Head and Neck Neoplasms , MicroRNAs , Mouth Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Squamous Cell Carcinoma of Head and Neck , Carcinoma, Squamous Cell/pathology , Lymphatic Metastasis , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Cell Line, Tumor , Extracellular Vesicles/metabolism , Tumor Microenvironment
3.
Front Plant Sci ; 14: 1261518, 2023.
Article in English | MEDLINE | ID: mdl-37900740

ABSTRACT

Although Brassica juncea has demonstrated potential as a hyperaccumulator crop, it was not entirely clear how cadmium (Cd) accumulates in plants. Here, we found that BjNRAMP1 (Natural Resistance-Associated Macrophage Protein 1) plays a crucial role in the accumulation of Cd and manganese (Mn) through its expression in yeast and Arabidopsis thaliana. The high concentration of Cd exposure could induce the expression of BjNRAMP1. The ectopic expression of BjNRAMP1 in yeast led to higher accumulation of Cd and Mn compared to the vector control. BjNARAMP1 was localized to the plasma membrane and expressed in the vascular system of roots, leaves, and flowers. The overexpression of BjNRAMP1 in A. thaliana resulted in an increased accumulation of Cd in both roots and shoots, which inhibited the normal growth of transgenic lines. Moreover, Mn uptake in roots was activated by the increase in Cd stress. Together, our results indicated that BjNRAMP1 significantly contributes to the uptake of Mn and Cd in B. juncea.

4.
Cell Death Dis ; 14(2): 107, 2023 02 11.
Article in English | MEDLINE | ID: mdl-36774351

ABSTRACT

In oral squamous cell carcinoma (OSCC), a highly aggressive and frequently lethal malignancy, the role and action mechanism of the microtubule regulatory protein CDK5RAP2 have not been fully understood. Here, we show that CDK5RAP2 is highly expressed in OSCC and its expression correlates with clinical stage and lymph node metastasis of the disease. The expression of CDK5RAP2 is regulated by the Wnt signaling pathway. Depletion of CDK5RAP2 inhibits the tumorigenesis and migration of OSCC cells and alters the OSCC cancer stem (-like) cell (CSC) signature. Notably, suppression of CDK5RAP2 expression disrupts spindle orientation during mitosis. Collectively, these results identify CDK5RAP2 as a potential CSC marker and reveal a mechanism that controls the CSC population in OSCC.


Subject(s)
Cell Cycle Proteins , Mouth Neoplasms , Nerve Tissue Proteins , Squamous Cell Carcinoma of Head and Neck , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics
5.
Biomater Transl ; 4(3): 142-150, 2023.
Article in English | MEDLINE | ID: mdl-38283090

ABSTRACT

Periodontitis is a prevalent oral disease. It can cause tooth loss and has a significant impact on patients' quality of life. While existing treatments can only slow the progression of periodontitis, they are unable to achieve complete regeneration and functional reconstruction of periodontal tissues. As a result, regenerative therapies based on biomaterials have become a focal point of research in the field of periodontology. Despite numerous studies reporting the superiority of new materials in periodontal regeneration, limited progress has been made in translating these findings into clinical practice. This may be due to the lack of appropriate animal models to simulate the tissue defects caused by human periodontitis. This review aims to provide an overview of established animal models for periodontal regeneration, examine their advantages and limitations, and outline the steps for model construction. The objective is to determine the most relevant animal models for periodontal regeneration based on the hypothesis and expected outcomes.

6.
Biochim Biophys Acta Mol Cell Res ; 1869(12): 119358, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36084732

ABSTRACT

N6-methyladenosine (m6A) is the most prevalent mRNA modification which plays crucial roles in various biological processes, but its role in cementogenesis remains largely unknown. Here, using time-series transcriptomic analysis, we reveal that mRNA m6A demethylase Fat mass and obesity-associated protein (FTO) is involved in cementogenesis. Knocking down FTO decreases cementoblast differentiation and mineralization in both OCCM-30 cellular model and murine ectopic bone formation model. Mechanistically, we find that FTO directly binds Runt-related transcription factor 2 (Runx2) mRNA, an important cementogenesis factor, thus protecting it from YTH domain-containing family protein 2 (YTHDF2) mediated degradation, when cementoblasts are differentiating. Knocking down YTHDF2 restores the expression of Runx2 in FTO-knockdown cells. Moreover, under inflammatory conditions, TNF-α inhibits cementoblast differentiation and mineralization partly through FTO/RUNX2 axis. Collectively, our study reveals an important regulatory role of FTO/RUNX2 axis in normal and pathological cementogenesis.


Subject(s)
Biological Phenomena , Core Binding Factor Alpha 1 Subunit , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Dental Cementum/metabolism , Mice , RNA, Messenger/metabolism , Transcription Factors , Tumor Necrosis Factor-alpha
7.
Int J Oral Sci ; 14(1): 47, 2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36167732

ABSTRACT

Interferon-γ (IFN-γ), a key effector molecule in anti-tumor immune response, has been well documented to correlate with the intratumoral infiltration of immune cells. Of interest, however, a high level of IFN-γ has been reported in salivary adenoid cystic carcinoma (SACC), which is actually a type of immunologically cold cancer with few infiltrated immune cells. Investigating the functional significance of IFN-γ in SACC would help to explain such a paradoxical phenomenon. In the present study, we revealed that, compared to oral squamous cell carcinoma cells (a type of immunologically hot cancer), SACC cells were less sensitive to the growth-inhibition effect of IFN-γ. Moreover, the migration and invasion abilities of SACC cells were obviously enhanced upon IFN-γ treatment. In addition, our results revealed that exposure to IFN-γ significantly up-regulated the level of programmed death ligand 1 (PD-L1) on SACC cell-derived small extracellular vesicles (sEVs), which subsequently induced the apoptosis of CD8+ T cells through antagonizing PD-1. Importantly, it was also found that SACC patients with higher levels of plasma IFN-γ also had higher levels of circulating sEVs that carried PD-L1 on their surface. Our study unveils a mechanism that IFN-γ induces immunosuppression in SACC via sEV PD-L1, which would account for the scarce immune cell infiltration and insensitivity to immunotherapy.


Subject(s)
Carcinoma, Adenoid Cystic , Carcinoma, Squamous Cell , Mouth Neoplasms , Salivary Gland Neoplasms , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Adenoid Cystic/metabolism , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Humans , Immunosuppression Therapy , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Mouth Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Salivary Gland Neoplasms/pathology
8.
Blood ; 138(1): 71-85, 2021 07 08.
Article in English | MEDLINE | ID: mdl-33763698

ABSTRACT

RNA-binding proteins (RBPs) are critical regulators of transcription and translation that are often dysregulated in cancer. Although RBPs are increasingly recognized as being important for normal hematopoiesis and for hematologic malignancies as oncogenes or tumor suppressors, RBPs that are essential for the maintenance and survival of leukemia remain elusive. Here we show that YBX1 is specifically required for maintaining myeloid leukemia cell survival in an N6-methyladenosine (m6A)-dependent manner. We found that expression of YBX1 is significantly upregulated in myeloid leukemia cells, and deletion of YBX1 dramatically induces apoptosis and promotes differentiation coupled with reduced proliferation and impaired leukemic capacity of primary human and mouse acute myeloid leukemia cells in vitro and in vivo. Loss of YBX1 has no obvious effect on normal hematopoiesis. Mechanistically, YBX1 interacts with insulin-like growth factor 2 messenger RNA (mRNA)-binding proteins (IGF2BPs) and stabilizes m6A-tagged RNA. Moreover, YBX1 deficiency dysregulates the expression of apoptosis-related genes and promotes mRNA decay of MYC and BCL2 in an m6A-dependent manner, which contributes to the defective survival that results from deletion of YBX1. Thus, our findings have uncovered a selective and critical role of YBX1 in maintaining myeloid leukemia survival, which might provide a rationale for the therapeutic targeting of YBX1 in myeloid leukemia.


Subject(s)
Adenosine/analogs & derivatives , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Y-Box-Binding Protein 1/metabolism , Adenosine/metabolism , Animals , Apoptosis/genetics , Cell Survival/genetics , Gene Deletion , Gene Expression Regulation, Leukemic , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice, Inbred C57BL , Protein Stability , Proto-Oncogene Proteins c-myc/metabolism , RNA, Neoplasm/metabolism , RNA-Binding Proteins/metabolism , Y-Box-Binding Protein 1/genetics
9.
Cell Stem Cell ; 27(1): 81-97.e8, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32402251

ABSTRACT

N6-methyladenosine (m6A) is a commonly present modification of mammalian mRNAs and plays key roles in various cellular processes. m6A modifiers catalyze this reversible modification. However, the underlying mechanisms by which these m6A modifiers are regulated remain elusive. Here we show that expression of m6A demethylase ALKBH5 is regulated by chromatin state alteration during leukemogenesis of human acute myeloid leukemia (AML), and ALKBH5 is required for maintaining leukemia stem cell (LSC) function but is dispensable for normal hematopoiesis. Mechanistically, KDM4C regulates ALKBH5 expression via increasing chromatin accessibility of ALKBH5 locus, by reducing H3K9me3 levels and promoting recruitment of MYB and Pol II. Moreover, ALKBH5 affects mRNA stability of receptor tyrosine kinase AXL in an m6A-dependent way. Thus, our findings link chromatin state dynamics with expression regulation of m6A modifiers and uncover a selective and critical role of ALKBH5 in AML that might act as a therapeutic target of specific targeting LSCs.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Leukemia, Myeloid, Acute , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Animals , Chromatin , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, Myeloid, Acute/genetics , RNA Stability , Stem Cells/metabolism
10.
Cancer Immunol Res ; 8(2): 179-191, 2020 02.
Article in English | MEDLINE | ID: mdl-31771985

ABSTRACT

CMTM6, a regulator of PD-L1 expression, also modulates tumor immunity. Little is known about the function of CMTM6 and its mechanism of action in head and neck squamous cell carcinoma (HNSCC). In this study, we found by IHC analysis that CMTM6 overexpression predicted a poor prognosis for patients with HNSCC. We discovered that CMTM6 expression was correlated with increased activity through the Wnt/ß-catenin signaling pathway, which is essential for tumorigenesis, maintenance of cancer stem cells (CSC), and the epithelial-to-mesenchymal transition (EMT) characteristic of multiple cancers. We used short hairpin RNA to eliminate expression of CMTM6, which led, in HNSCC cells, to reduced expression of nuclear ß-catenin as well as inhibition of stem cell-like properties, TGFß-induced EMT, and cell proliferation. Consistent with these results, we identified a significant positive correlation between expression of CMTM6 and EMT- and CSC-related genes in The Cancer Genome Atlas (TCGA). We found positive correlations for both RNA and protein between expression of CMTM6 and immune checkpoint components. CMTM6 silencing-induced PD-L1 downregulation delayed SCC7 tumor growth and increased CD8+ and CD4+ T-cell infiltration. The proportions of PD-1+, TIM-3+, VISTA+, LAG-3+, and B7-H3+ exhausted T cells were decreased significantly in the CMTM6 knockdown group. CMTM6 thus regulates stemness, EMT, and T-cell dysfunction and may be a promising therapeutic target in the treatment of HNSCC.


Subject(s)
Head and Neck Neoplasms/immunology , Membrane Proteins/antagonists & inhibitors , Neoplastic Stem Cells/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Animals , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Gene Knockout Techniques , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunity, Cellular/immunology , MARVEL Domain-Containing Proteins , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Myelin Proteins , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Survival Rate , Wnt Signaling Pathway
11.
Adv Exp Med Biol ; 1143: 41-57, 2019.
Article in English | MEDLINE | ID: mdl-31338814

ABSTRACT

Leukemia stem cells (LSCs) are leukemia-initiating population with the capacity to self-renew, differentiate, and stay quiescent. Human hematopoietic malignancies such as chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) are derived from this cell population. LSCs are also responsible for disease relapse due to its resistance to drug treatment. This rare cell population is phenotypically and functionally heterogeneous. Increasing evidence indicates that this heterogeneous cellular state of LSCs might determine the different drug sensitivity and is the major reason for disease relapse. In here, focusing on myeloid leukemia stem cells, we describe the biological features including cellular and molecular state, heterogeneity of LSCs, and the dynamic cross talk between LSCs and bone marrow microenvironment. These specific features of LSCs highlight the dynamic cellular state of LSCs, and further exploring on it might provide potential therapeutic targets that are important for eliminating LSCs.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid, Acute , Neoplastic Stem Cells , Bone Marrow/physiology , Drug Resistance, Neoplasm , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Leukemia, Myeloid, Acute/physiopathology , Tumor Microenvironment/physiology
12.
Int J Cancer ; 143(6): 1494-1504, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29663369

ABSTRACT

The adenosine-induced immunosuppression hampers the immune response toward tumor cells and facilitates the tumor cells to evade immunosurveillance. CD73, an ecto-5-nucleotidase, is the ectoenzyme dephosphorylating extracellular AMP to adenosine. Here, using immunocompetent transgenic head and neck squamous cell carcinoma (HNSCC) mouse model, immune profiling showed high expression of CD73 on CD4+ and CD8+ T cells was associated with an "exhausted" phenotype. Further, treatment with anti-CD73 monoclonal antibody (mAb) significantly blunted the tumor growth in the mouse model, and the blockade of CD73 reversed the "exhausted" phenotype of CD4+ and CD8+ T cells through downregulation of total expression of PD-1 and CTLA-4 on T cells. Whereas the population of CD4+ CD73hi /CD8+ CD73hi T cells expressed higher CTLA-4 and PD-1 as compared to untreated controls. In addition, the human tissue microarrays showed the expression of CD73 is upregulated on tumor infiltrating immune cells in patients with primary HNSCC. Moreover, CD73 expression is an independent prognostic factor for poor outcome in our cohort of HNSCC patients. Altogether, these findings highlight the immunoregulatory role of CD73 in the development of HNSCC and we propose that CD73 may prove to be a promising immunotherapeutic target for the treatment of HNSCC.


Subject(s)
5'-Nucleotidase/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Immune Tolerance/immunology , Lymphocytes, Tumor-Infiltrating/immunology , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , Biomarkers, Tumor , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Follow-Up Studies , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Mice, Transgenic , PTEN Phosphohydrolase/physiology , Phenotype , Prognosis , Receptor, Transforming Growth Factor-beta Type I/physiology , Survival Rate , Tumor Cells, Cultured
13.
Int J Cancer ; 142(5): 999-1009, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29047105

ABSTRACT

Immune evasion is a hallmark feature of cancer, and it plays an important role in tumour initiation and progression. In addition, tumour immune evasion severely hampers the desired antitumour effect in multiple cancers. In this study, we aimed to investigate the role of the Notch pathway in immune evasion in the head and neck squamous cell carcinoma (HNSCC) microenvironment. We first demonstrated that Notch1 signaling was activated in a Tgfbr1/Pten-knockout HNSCC mouse model. Notch signaling inhibition using a γ-secretase inhibitor (GSI-IX, DAPT) decreased tumour burden in the mouse model after prophylactic treatment. In addition, flow cytometry analysis indicated that Notch signaling inhibition reduced the sub-population of myeloid-derived suppressor cells (MDSCs), tumour-associated macrophages (TAMs) and regulatory T cells (Tregs), as well as immune checkpoint molecules (PD1, CTLA4, TIM3 and LAG3), in the circulation and in the tumour. Immunohistochemistry (IHC) of human HNSCC tissues demonstrated that elevation of the Notch1 downstream target HES1 was correlated with MDSC, TAM and Treg markers and with immune checkpoint molecules. These results suggest that modulating the Notch signaling pathway may decrease MDSCs, TAMs, Tregs and immune checkpoint molecules in HNSCC.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Carcinoma, Squamous Cell/immunology , Diamines/pharmacology , Disease Models, Animal , Head and Neck Neoplasms/immunology , Myeloid Cells/immunology , T-Lymphocytes, Regulatory/immunology , Thiazoles/pharmacology , Animals , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunosuppression Therapy , Mice , Mice, Knockout , Myeloid Cells/drug effects , PTEN Phosphohydrolase/physiology , Protein Serine-Threonine Kinases/physiology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/physiology , T-Lymphocytes, Regulatory/drug effects , Tumor Cells, Cultured , Tumor Escape/drug effects
14.
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
15.
Mol Carcinog ; 57(3): 429-439, 2018 03.
Article in English | MEDLINE | ID: mdl-29215754

ABSTRACT

Angiogenesis is an essential event in tumor growth and metastasis, and immune system also contributes to the tumor evasion. Emerging evidences have suggested the bidirectional link between angiogenesis and immunosuppression. Myeloid-derived suppressor cell (MDSC) is a kind of immunosuppressive cells and plays an important role in this process. However, the actual regulatory mechanisms of angiogenesis and MDSCs in head and neck squamous cell carcinoma (HNSCC) were unclear. In this study, through analyzing the immunohistochemistry staining of human HNSCC tissue microarray, we found that the microvascular density (MVD) was significantly increased in HNSCC patients. We also characterized angiogenic factors p-STAT3, VEGFA, CK2, and MDSCs marker CD11b in HNSCC tissue array, and found the close expression correlation among these markers. To determine the role of JAK2/STAT3 pathway in tumor microenvironment of HNSCC, we utilized AG490 (an inhibitor of JAK2/STAT3) for further research. Results showed that inhibition of JAK2/STAT3 suppressed angiogenesis by decreasing VEGFA and HIF1-α both in vitro and vivo. Moreover, in HNSCC transgenic mouse model, inhibiting JAK2/STAT3 not only suppressed angiogenesis but also reduced MDSCs in the tumor microenvironment through suppressing VEGFA and CK2. Our findings demonstrated the close relationship between angiogenesis and MDSCs in HNSCC, and inhibition of JAK2/STAT3 could reduce tumor-induced angiogenesis and decrease MDSCs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Janus Kinase 2/antagonists & inhibitors , Myeloid-Derived Suppressor Cells/drug effects , Neovascularization, Pathologic/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Human Umbilical Vein Endothelial Cells , Humans , Janus Kinase 2/metabolism , Mice, Transgenic , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tyrphostins/pharmacology
16.
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
17.
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
18.
Sci Rep ; 7(1): 6629, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747781

ABSTRACT

Although conventional chemoradiotherapy is effective for most anal squamous cell carcinoma (ASCC) patients, HPV-negative ASCC patients respond poorly to this treatment and new therapeutic approach is required. Our group has previously established an HPV-negative ASCC mouse model and demonstrated that signal transducer and activation of transcription 3 (STAT3) is hyper-activated in the model. Here, we show that in vivo inhibition of STAT3 by S3I-201 effectively delays tumor growth in ASCC mouse model indicated by significantly smaller tumor size and burden in the treatment group compared with control group at the same point. Further analysis shows that survivin and Ki67, important biomarkers for tumor cell survival and proliferation, are significantly reduced after S3I-201 treatment. Additionally, flow cytometry and immunohistofluorescent assays reveal decreased Myeloid-derived suppressor cell (MDSC) and tumor-associated macrophage (TAM) populations in the S3I-201 treatment group, which indicates a reversion of the immunosuppressive environment, unraveling the potential role for S3I-201 in immunosuppression in ASCC. Together these results for the first time demonstrated the anti-tumor effects of STAT3 inhibitor S3I-201 in HPV-negative ASCC mouse model and its multiple effects on cancer cells and immune system. Thus we conclude that S3I-201 may be a novel therapeutic approach for HPV-negative ASCC patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Anus Neoplasms/drug therapy , Benzenesulfonates/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Immunotherapy/methods , STAT3 Transcription Factor/antagonists & inhibitors , Aminosalicylic Acids/administration & dosage , Animals , Anus Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Disease Models, Animal , Histocytochemistry , Immunohistochemistry , Ki-67 Antigen/analysis , Mice , Phosphorylation , Protein Processing, Post-Translational , Survivin/analysis , Treatment Outcome
19.
Tumour Biol ; 39(4): 1010428317695930, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28381191

ABSTRACT

Tumor necrosis factor receptor-associated factor 1, an adaptor protein of tumor necrosis factor 2, is involved in classical nuclear factor (NF)-κB activation and lymphocyte recruitment. However, less is known about the expression and association of tumor necrosis factor receptor-associated factor 1 with cancer stem cell markers in oral squamous cell carcinoma. This study aimed to investigate the expression of tumor necrosis factor receptor-associated factor 1 and stem cell characteristic markers (lin28 homolog B, B cell-specific Moloney murine leukemia virus integration site 1, and aldehyde dehydrogenase 1) in oral squamous cell carcinoma and analyze their relations. Paraffin-embedded tissues of 78 oral squamous cell carcinomas, 39 normal oral mucosa, and 12 oral dysplasia tissues were employed in tissue microarrays, and the expression of tumor necrosis factor receptor-associated factor 1, B cell-specific Moloney murine leukemia virus integration site 1, aldehyde dehydrogenase 1, and lin28 homolog B was measured by immunohistostaining and digital pathological analysis. The expression of tumor necrosis factor receptor-associated factor 1 was higher in the oral squamous cell carcinoma group as compared with the expression in the oral mucosa (p < 0.01) and oral dysplasia (p < 0.001) groups. In addition, the expression of tumor necrosis factor receptor-associated factor 1 was associated with those of B cell-specific Moloney murine leukemia virus integration site 1, aldehyde dehydrogenase 1, and lin28 homolog B (p = 0.032, r2 = 0.109; p < 0.0001, r2 = 0.64; and p < 0.001, r2 = 0.16) in oral squamous cell carcinoma. The patient survival rate was lower in the highly expressed tumor necrosis factor receptor-associated factor 1 group, although the difference was not significant. The clustering analysis showed that tumor necrosis factor receptor-associated factor 1 was most related to aldehyde dehydrogenase 1. These findings suggest that tumor necrosis factor receptor-associated factor 1 has potential direct/indirect regulations with the cancer stem cell markers in oral squamous cell carcinoma, which may help in further analysis of the cancer stem cell characteristics.


Subject(s)
Carcinoma, Squamous Cell/chemistry , Isoenzymes/analysis , Mouth Neoplasms/chemistry , Neoplastic Stem Cells/chemistry , Polycomb Repressive Complex 1/analysis , RNA-Binding Proteins/analysis , Retinal Dehydrogenase/analysis , TNF Receptor-Associated Factor 1/analysis , Aldehyde Dehydrogenase 1 Family , Biomarkers , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Humans , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology
20.
Am J Transl Res ; 9(2): 507-519, 2017.
Article in English | MEDLINE | ID: mdl-28337279

ABSTRACT

Salivary adenoid cystic carcinoma (AdCC) is a common head and neck cancer with the propensity for local spread and distant metastasis. In our previous study, elevated expression of Anterior gradient 2 (AGR2) was detected in head and neck squamous cell carcinoma (HNSCC), associated with epithelial-mesenchymal transition (EMT) and cancer stemness. However, to date, the expression and function of AGR2 in AdCC has yet to be elucidated. In the present study, human AdCC tissue microarrays including 18 cases of normal salivary gland (NSG), 12 cases of pleomorphic adenoma (PMA) and 72 cases of AdCC were employed for immunohistochemical staining analysis. Results indicated that AGR2, which was remarkably correlated with Ki-67, transforming growth factor beta-1 (TGF-ß1) and CD147, was significantly elevated in human salivary AdCC tissues. Knockdown of AGR2 significantly repressed the proliferation and migration of human SACC-83 and SACC-LM cell lines. Additionally, AGR2 silencing obviously reversed the EMT phenomena induced by TGF-ß1. Taken together, our present study revealed the potential pro-metastasis role of AGR2 in AdCC, indicating that AGR2 might be a novel therapeutic target of AdCC with distant metastasis.

SELECTION OF CITATIONS
SEARCH DETAIL
...