Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Biochem Pharmacol ; 216: 115796, 2023 10.
Article in English | MEDLINE | ID: mdl-37690572

ABSTRACT

Oral lichen planus (OLP) is a chronic T cell-mediated mucocutaneous disease characterized by T cell infiltration at the connective tissue-epithelium interface. Traditionally, topical corticosteroids are used as the first-line drugs to treat OLP. However, long-term use of corticosteroids may lead to drug tolerance, secondary candidiasis, and autoimmune adrenal insufficiency. Although topical tacrolimus has often been recommended for short-term use in corticosteroid-refractory OLP, the precise role of tacrolimus in epithelial cells remains elusive. This study showed that tacrolimus could directly upregulate the expression of IL-37 in human gingival epithelial cells by promoting the TGF-ßRI/Smad3 pathway independently of calcineurin inhibition and MAPKs. In contrast, dexamethasone, one of the corticosteroids, did not have the same effect. Moreover, IL-37 could inhibit the proliferation of activated T cells and the secretion of effector cytokines and alleviate epithelial cell apoptosis and death caused by activated T cells ina co-culturesystem. Furthermore, compared with healthy controls, IL-37 and p-Smad3 levels significantly increased in the oral mucosa affected by OLP, especially in the epithelium. IL-37 might have mediated a negative feedback mechanism to curb excessive inflammation in OLP. However, the expression of IL-37 was not associated with the infiltration of CD8+ T cells and Tregs in OLP, implying that IL-37 might mostly affect T cell activation rather than T cell differentiation and migration. Overall, this study discovered a potential novel mechanism by which tacrolimus might indirectly inhibit T cell-mediated immune damage by upregulating IL-37 in human gingival epithelial cells.


Subject(s)
Lichen Planus, Oral , Tacrolimus , Humans , Adrenal Cortex Hormones/therapeutic use , CD8-Positive T-Lymphocytes , Epithelial Cells/metabolism , Lichen Planus, Oral/drug therapy , Lichen Planus, Oral/metabolism , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Up-Regulation
2.
Biochem Pharmacol ; 216: 115767, 2023 10.
Article in English | MEDLINE | ID: mdl-37634599

ABSTRACT

Oral lichen planus (OLP) is a T cell-mediated autoimmune disease of oral mucosa concerning with the redox imbalance. Although glutamine uptake mediated by alanine-serine-cysteine transporter 2 (ASCT2) is critical to T cell differentiation, the exact mechanism remains ambiguous. Here, we elucidate a novel regulatory mechanism of ASCT2-mediated uptake in the differentiation and proliferation of T cells through maintaining redox balance in OLP. The results of immunohistochemistry (IHC) showed that both ASCT2 and glutaminase (GLS) were obviously upregulated compared to controls in OLP. Moreover, correlation analyses indicated that ASCT2 expression was significantly related to GLS level. Interestingly, the upregulation of glutamine metabolism in epithelial layer was consistent with that in lamina propria. Functional assays in vitro revealed the positive association between glutamine metabolism and lymphocytes infiltration. Additionally, multiplex immunohistochemistry (mIHC) uncovered a stronger colocalization among ASCT2 and CD4 and IFN-γ, which was further demonstrated by human Th1 differentiation assay in vitro. Mechanistically, targeting glutamine uptake through interference with ASCT2 using L-γ-Glutamyl-p-nitroanilide (GPNA) decreased the glutamine uptake of T cells and leaded to the accumulation of intracellular reactive oxygen species (ROS), which promoted dual specificity phosphatase 2 (DUSP2/PAC1) expression through activation of early growth response 1 (EGR1) to induce dephosphorylation of signal transducer and activator of transcription 3 (STAT3) and inhibit Th1 differentiation in turn. These results demonstrated that glutamine uptake mediated by ASCT2 induced Th1 differentiation by ROS-EGR1-PAC1 pathway, and restoring the redox dynamic balance through targeting ASCT2 may be a potential treatment for T cell-mediated autoimmune diseases.


Subject(s)
Amino Acid Transport System ASC , Glutamine , Lichen Planus, Oral , Humans , Alanine , Cell Differentiation , Cysteine , Early Growth Response Protein 1 , Glutamine/metabolism , Reactive Oxygen Species/metabolism , Amino Acid Transport System ASC/metabolism
3.
J Exp Clin Cancer Res ; 42(1): 162, 2023 Jul 08.
Article in English | MEDLINE | ID: mdl-37420300

ABSTRACT

BACKGROUND: A growing body of research has revealed the connection of metabolism reprogramming and tumor progression, yet how metabolism reprogramming affects inter-patient heterogeneity and prognosis in head and neck squamous cell carcinoma (HNSCC) still requires further explorations. METHODS: A cellular hierarchy framework based on metabolic properties discrepancy, METArisk, was introduced to re-analyze the cellular composition from bulk transcriptomes of 486 patients through deconvolution utilizing single-cell reference profiles from 25 primary and 8 metastatic HNSCC sample integration of previous studies. Machine learning methods were used to identify the correlations between metabolism-related biomarkers and prognosis. The functions of the genes screened out in tumor progression, metastasis and chemotherapy resistance were validated in vitro by cellular functional experiments and in vivo by xenograft tumor mouse model. RESULTS: Incorporating the cellular hierarchy composition and clinical properties, the METArisk phenotype divided multi-patient cohort into two classes, wherein poor prognosis of METArisk-high subgroup was associated with a particular cluster of malignant cells with significant activity of metabolism reprogramming enriched in metastatic single-cell samples. Subsequent analysis targeted for phenotype differences between the METArisk subgroups identified PYGL as a key metabolism-related biomarker that enhances malignancy and chemotherapy resistance by GSH/ROS/p53 pathway, leading to poor prognosis of HNSCC. CONCLUSION: PYGL was identified as a metabolism-related oncogenic biomarker that promotes HNSCC progression, metastasis and chemotherapy resistance though GSH/ROS/p53 pathway. Our study revealed the cellular hierarchy composition of HNSCC from the cell metabolism reprogramming perspective and may provide new inspirations and therapeutic targets for HNSCC in the future.


Subject(s)
Head and Neck Neoplasms , Humans , Animals , Mice , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Reactive Oxygen Species , Tumor Suppressor Protein p53/genetics , Prognosis , Gene Expression Regulation, Neoplastic
4.
Int Immunopharmacol ; 119: 110216, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37116342

ABSTRACT

BACKGROUND: Oral lichen planus (OLP) is a chronic inflammatory disease characterized by T cell infiltration at lesion sites. T cell migration is greatly facilitated by chemokines produced by epithelial cells. Studies have noted the potential role of glutamine uptake in OLP and other inflammatory diseases. Here, we investigated the effect of altered glutamine uptake of epithelial cells on T cell infiltration and its underlying mechanisms in OLP. METHODS: Immunohistochemistry was used to identify the expressions of glutamine transporter alanine-serine-cysteine transporter 2 (ASCT2) and C-C motif chemokine ligand 5 (CCL5) in oral tissues of OLP and healthy controls. Human gingival epithelial cells (HGECs) were treated with glutamine deprivation and ASCT2 inhibiter GPNA respectively to detect the expressions of CCL5 and its related signaling molecules. Additionally, we had determined the impact of epithelial cell-derived CCL5 on T-cell migration using a co-culture system in vitro. RESULTS: ASCT2 and CCL5 expressions in OLP were significantly higher than healthy controls and positively correlated with the density of inflammatory infiltrations. Glutamine supplement significantly increased CCL5 production in HGECs, which was effectively inhibited by GPNA. Besides, glutamine could inhibit reactive oxygen species (ROS) production to activate the signal transducer and activator of transcription 3 (STAT3) causing higher expression level of CCL5 in HGECs. Simultaneously, T cell migration could be blocked by anti-CCL5 neutralizing antibody and STAT3 inhibitor stattic in the co-culture system. CONCLUSION: The upregulated ASCT2-mediated glutamine uptake in epithelial cells promotes CCL5 production via ROS-STAT3 signaling, which boosts the T-cell infiltration in OLP lesion.


Subject(s)
Amino Acid Transport System ASC , Lichen Planus, Oral , T-Lymphocytes , Humans , Epithelial Cells/metabolism , Glutamine/metabolism , Ligands , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Amino Acid Transport System ASC/metabolism , Chemokine CCL5/metabolism
5.
PeerJ ; 11: e14824, 2023.
Article in English | MEDLINE | ID: mdl-36811004

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the 11th most prevalent tumor worldwide. Despite advantages of therapeutic approaches, the 5-year survival rate of patients with OSCC is less than 50%. It is urgent to elucidate mechanisms underlying OSCC progression for developing novel treatment strategies. Our recent study has revealed that Keratin 4 (KRT4) suppresses OSCC development, which is downregulated in OSCC. Nevertheless, the mechanism downregulating KRT4 in OSCC remains unknown. In this study, touchdown PCR was utilized to detect KRT4 pre-mRNA splicing, while m6A RNA methylation was identified by methylated RNA immunoprecipitation (MeRIP). Besides, RNA immunoprecipitation (RIP) was used to determine RNA-protein interaction. Herein, this study indicated that intron splicing of KRT4 pre-mRNA was suppressed in OSCC. Mechanistically, m6A methylation of exon-intron boundaries prevented intron splicing of KRT4 pre-mRNA in OSCC. Besides, m6A methylation suppressed the binding of splice factor DGCR8 microprocessor complex subunit (DGCR8) to exon-intron boundaries in KRT4 pre-mRNA to prohibit intron splicing of KRT4 pre-mRNA in OSCC. These findings revealed the mechanism downregulating KRT4 in OSCC and provided potential therapeutic targets for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , MicroRNAs , Mouth Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck , Carcinoma, Squamous Cell/genetics , Methylation , Mouth Neoplasms/genetics , RNA Precursors/metabolism , Keratin-4/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/genetics
6.
J Oral Pathol Med ; 52(2): 150-160, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36459062

ABSTRACT

BACKGROUND: Although abnormal cell proliferation and apoptosis are associated with the pathogenesis of oral lichen planus (OLP), the exactly mechanism of which is not yet known. It has been reported that glutamine (Gln) can promote cell proliferation and inhibit apoptosis of various tumor cells. This study aims to evaluate the effect of Gln metabolism on the balance of proliferation and apoptosis in epithelial cells of OLP. METHODS: Thirty human OLP specimens and 11 normal controls were stained by immunohistochemistry to detect the levels of proliferation and Gln metabolism related proteins. Then, the critical role of Gln in cell proliferation and apoptosis was determined by Gln deprivation or treatment with glutaminase inhibitor (CB-839) to intervene Gln metabolism in human gingival epithelial cells. Cell proliferation was detected using CCK8, p-mTOR and p-S6 proteins were detected using Western Blot, cell apoptosis and cell cycle were detected using flow cytometry, and cell stress was detected using immunofluorescence. RESULTS: Compared with normal controls, OLP specimens showed higher levels of Ki-67 and Gln metabolism-related proteins, including Gln transporter (ASCT2), glutaminase (GLS), and pathway proteins (p-mTOR and p-S6). In vitro, Gln promoted cell proliferation and simultaneously upregulated the activity of mTOR/S6 pathway. Moreover, rapamycin, an mTOR pathway inhibitor, could effectively block the Gln-induced cell proliferation. MHY1485, an mTOR pathway agonist, could effectively reverse the decline of cell proliferation under Gln deprivation. In addition, inhibiting Gln metabolism caused the accumulation of intracellular radical oxygen species (ROS) and induced cell apoptosis. However, N-acetylcysteine reversed this state and then decreased cell apoptosis by eliminating intracellular ROS. CONCLUSION: Gln metabolism is essential to maintain the balance of proliferation and apoptosis in oral epithelial cells, and inhibition of Gln metabolism may have a beneficial effect on OLP treatment.


Subject(s)
Glutamine , Lichen Planus, Oral , Humans , Glutamine/pharmacology , Glutaminase/pharmacology , Lichen Planus, Oral/pathology , Reactive Oxygen Species , TOR Serine-Threonine Kinases/metabolism , Epithelial Cells/pathology , Cell Proliferation , Apoptosis
7.
Front Genet ; 13: 1065320, 2022.
Article in English | MEDLINE | ID: mdl-36437918

ABSTRACT

Retinoids, natural and synthetic derivatives of vitamin A, have many regulatory functions in human body, including regulating cellular proliferation, differentiation, apoptosis. Moreover, retinoids have been used successfully for the treatment of certain malignancies, especially acute promyelocytic leukemia (APL) in adults and neuroblastoma in children. However, retinoids have not yet been translated into effective systemic treatments for most solid cancers. Some recent studies have shown that retinoids promote tumorigenesis. Therefore, we performed this meta-analysis to systematically evaluate the efficacy of retinoids in the chemoprevention and treatment of cancers. We performed literature search of several electronic databases, including PubMed, Embase and Cochrane Library from 2000 January to 2021 November. Various outcomes were applied to investigate the potential of retinoids for prevention and treatment of cancers. The primary outcomes in this study were disease recurrence and clinical response. The secondary outcomes included overall survival (OS), cancer development, disease progression and event-free survival. We identified 39 randomized controlled trials with 15,627 patients in this study. Our results showed that lower recurrence rate and better clinical response were obtained in retinoids treated patients with cancer or premalignancy as compared with control. The differences were statistically significant (RR = 0.85, 95% CI = 0.74-0.96, p = 0.01; RR = 1.24, 95% CI = 1.03-1.49, p = 0.02, respectively). Retinoids treatment was not associated with improvement in overall survival, cancer development, disease progression or event-free survival. Subgroup analysis conducted based on cancer type showed that patients benefited from retinoids treatment in APL, renal cell carcinoma, hepatocellular carcinoma, lung cancer, Kaposi sarcoma, and complete hydatidiform mole. No significant therapeutic effect was noted in head and neck cancer, acute myeloid leukemia (AML), melanoma, breast cancer, bladder cancer, cervical intraepithelial neoplasia (CIN) or cervical carcinoma. Subgroup analysis based on tumor classification demonstrated that retinoids group obtained a lower recurrence rate and better clinical response than control group in solid cancers. In conclusion, clinical application of retinoids was associated with reduction in disease recurrence and improvement in clinical response, illustrating that retinoids play a key role in cancer prevention and therapy. Further research is needed to broaden the utility of retinoids in other types of cancers. Systematic Review Registration: PROSPERO, identifier CRD42022296706.

8.
World J Stem Cells ; 14(2): 146-162, 2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35432735

ABSTRACT

Cancer stem cells (CSCs) comprise a subpopulation of cancer cells with stem cell properties, which exhibit the characteristics of high tumorigenicity, self-renewal, and tumor initiation and are associated with the occurrence, metastasis, therapy resistance, and relapse of cancer. Compared with differentiated cells, CSCs have unique metabolic characteristics, and metabolic reprogramming contributes to the self-renewal and maintenance of stem cells. It has been reported that CSCs are highly dependent on lipid metabolism to maintain stemness and satisfy the requirements of biosynthesis and energy metabolism. In this review, we demonstrate that lipid anabolism alterations promote the survival of CSCs, including de novo lipogenesis, lipid desaturation, and cholesterol synthesis. In addition, we also emphasize the molecular mechanism underlying the relationship between lipid synthesis and stem cell survival, the signal trans-duction pathways involved, and the application prospect of lipid synthesis reprogramming in CSC therapy. It is demonstrated that the dependence on lipid synthesis makes targeting of lipid synthesis metabolism a promising therapeutic strategy for eliminating CSCs. Targeting key molecules in lipid synthesis will play an important role in anti-CSC therapy.

9.
J Dent Educ ; 86(10): 1399-1404, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35442534

ABSTRACT

BACKGROUND: During the past decades, the rapid development of modern teaching tools and methods has been observed, and the new teaching module constructed through digitization or networking has been widely used in the field of medical education. This study aimed to investigate the effectiveness and acceptability of the online learning combined with case-based discussion (CBD) approach in oral medicine education. METHODS: Sixty senior students majoring in stomatology were randomly divided into two groups. One group (new module-based teaching group [NG], n = 30) watched an online teaching video and discussed clinical cases in groups. The control group (traditional lecture-based group [TG], n = 30) was assigned to the traditional lecture-based teaching. Subsequently, a theory assessment was conducted on the topics taught, and the scores of both groups were compared. Feedback about this teaching model was obtained from the NG. RESULTS: The results showed that students in the NG had significantly better performance in terms of mastering professional knowledge than did students in the TG. Moreover, most students in the NG had a high degree of satisfaction with this new teaching method, as they agreed that it can mobilize their learning enthusiasm and promote their engagement, interaction, and cooperation in the learning of oral medicine. CONCLUSIONS: Our findings indicate the effectiveness and high satisfaction of CBD combined with online learning in the teaching of oral medicine. This study is expected to provide new ideas for improving the dental teaching quality.


Subject(s)
Education, Distance , Education, Medical , Oral Medicine , Educational Measurement , Humans , Learning , Teaching
10.
Cell Death Dis ; 13(2): 158, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177591

ABSTRACT

For head and neck squamous cell carcinoma (HNSCC), the local invasion and distant metastasis represent the predominant causes of mortality. Targeted inhibition of chemokines and their receptors is an ongoing antitumor strategy established on the crucial roles of chemokines in cancer invasion and metastasis. Herein, we showed that C-C motif chemokine ligand 2 (CCL2)- C-C motif chemokine receptor 4 (CCR4) signaling, but not the CCL2- C-C motif chemokine receptor 2 (CCR2) axis, induces the formation of the vav guanine nucleotide exchange factor 2 (Vav2)- Rac family small GTPase 1 (Rac1) complex to activate the phosphorylation of myosin light chain (MLC), which is involved in the regulation of cell motility and cancer metastasis. We identified that targeting CCR4 could effectively interrupt the activation of HNSCC invasion and metastasis induced by CCL2 without the promoting cancer relapse observed during the subsequent withdrawal period. All current findings suggested that CCL2-CCR4-Vav2-Rac1-p-MLC signaling plays an essential role in cell migration and cancer metastasis of HNSCC, and CCR4 may serve as a new potential molecular target for HNSCC therapy.


Subject(s)
Chemokine CCL2 , Head and Neck Neoplasms , Neoplasm Recurrence, Local , Receptors, CCR4 , Squamous Cell Carcinoma of Head and Neck , Cell Line, Tumor , Cell Movement , Chemokine CCL2/genetics , Guanine Nucleotide Exchange Factors , Head and Neck Neoplasms/genetics , Humans , Receptors, CCR4/genetics , Squamous Cell Carcinoma of Head and Neck/genetics
11.
J Oral Pathol Med ; 51(1): 63-73, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34133803

ABSTRACT

BACKGROUND: Symptomatic oral lichen planus is a common chronic T-cell-mediated disorder characterized by pain and inflammation. The meta-analysis aimed to compare and evaluate the effects and safety of tacrolimus for treating patients with symptomatic oral lichen planus. METHODS: A comprehensive literature review was performed, including PubMed, the Cochrane Library, Embase, and Web of Science published up to and including December 2020. ClinicalTrials.gov was searched for ongoing trials. There were no restrictions on language or date of publication. Using the Cochrane Collaboration tool, we assessed the risk of bias for randomized controlled trials and estimated the proportion of between-trial heterogeneity. RESULTS: A total of 9 RCTs evaluating the effects of tacrolimus were included in this study. The results revealed no significant difference in clinical resolution and relapse between tacrolimus and corticosteroids. However, tacrolimus may be more likely to cause mild adverse effects. In particular, clinical resolution was not significantly different between tacrolimus and clobetasol propionate, and between tacrolimus and triamcinolone acetonide, while tacrolimus was more likely to cause adverse effects than triamcinolone acetonide and clobetasol propionate. Moreover, there was no significant difference in pain resolution between tacrolimus and clobetasol. Furthermore, adverse effects were not significantly different between tacrolimus and pimecrolimus. CONCLUSIONS: This systematic review and meta-analysis of 9 clinical trials supported the short-term application of tacrolimus as an effective regimen in OLP patients resistant to other topical and systemic therapies. Furthermore, the adverse effects of tacrolimus were minor and transient and did not affect tacrolimus' continued application.


Subject(s)
Lichen Planus, Oral , Tacrolimus , Administration, Topical , Calcineurin Inhibitors/therapeutic use , Humans , Lichen Planus, Oral/drug therapy , Randomized Controlled Trials as Topic , Tacrolimus/adverse effects , Treatment Outcome
12.
Cell Death Dis ; 12(10): 946, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650054

ABSTRACT

Although obesity has been associated with an increased risk and aggressiveness of many types of carcinoma, whether it promotes squamous cell carcinoma remains unclear. To reveal the role of obesity in oral squamous cell carcinoma (OSCC) initiation and development, we used 4NQO-induced OSCC model mice to examine the impact of dietary obesity on carcinogenesis. The results showed that high-fat diet (HFD)-induced obesity significantly promoted the incidence of OSCC and altered the local immune microenvironment with the expansion of CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs). The underlying mechanism that induced an immunosuppressive local microenvironment in obesity was the recruitment of MDSCs through the CCL9/CCR1 axis and enhancement of MDSC immunosuppressive function via intracellular fatty acid uptake. Furthermore, clinical samples verified the increase in infiltrated CD33+ (a marker of human MDSCs) cells in obese OSCC patients, and data from the TCGA dataset confirmed that CD33 expression was positively correlated with local adipocytes in OSCC. Survival analysis showed that enrichment of adipocytes and high expression of CD33 were associated with poor prognosis in OSCC patients. Strikingly, depletion of MDSCs significantly ameliorated HFD-promoted carcinogenesis in 4NQO-induced model mice. These findings indicate that obesity is also an important risk factor for OSCC, and cancer immunotherapy, especially targeting MDSCs, may exhibit greater antitumor efficacy in obese patients.


Subject(s)
Carcinogenesis/pathology , Mouth Neoplasms/etiology , Mouth Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Obesity/complications , 4-Nitroquinoline-1-oxide , Adipocytes/metabolism , Animals , Antigens, Ly , CD11b Antigen/metabolism , Chemokines, CC , Diet, High-Fat , Disease Models, Animal , Humans , Immunosuppression Therapy , Macrophage Inflammatory Proteins , Male , Mice, Inbred C57BL , Models, Biological , Quinolones , Receptors, CCR1/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Signal Transduction , Survival Analysis , Tongue/metabolism , Tongue/pathology , Tumor Microenvironment/drug effects
13.
Front Immunol ; 12: 696605, 2021.
Article in English | MEDLINE | ID: mdl-34248996

ABSTRACT

IL-37 is a recently discovered cytokine in the IL-1 family exerting broad protective effects on inflammatory diseases, autoimmune diseases, and cancer. Immune and non-immune cells produce the IL-37 precursor upon pro-inflammatory stimuli. Intracellularly, caspase-1 cleaves and activates IL-37, and its mature form binds to Smad3; this complex translocates into the nucleus where it suppresses cytokine production, consequently reducing inflammation. Extracellularly, IL-37 forms a complex with IL-18Rα and IL-1R8 (formerly TIR8 or SIGIRR) that transduces anti-inflammatory signals by the suppression of NF-κB and MAPK and the activation of Mer-PTEN-DOK pathways. During inflammation, IL-37 suppresses the expression of several pro-inflammatory cytokine in favor to the expression of the anti-inflammatory ones by the regulation of macrophage polarization, lipid metabolism, inflammasome function, TSLP synthesis and miRNAs function. Moreover, IL-37 not only regulates the innate and acquired immunity, but also improves aging-associated immunosenescence. Furthermore, IL-37 exerts an inhibitory effect on tumor angiogenesis and metastasis, and progression. Finally, IL-37 may have a potential ability to reduce excessive inflammation since it is aberrantly expressed in patients with inflammatory diseases, autoimmune diseases, and cancer, thus, it may be used as a marker for different types of diseases. Therefore, this review provides an updated view of the role of IL-37 in human health and disease, and discusses the potential of IL-37 as a therapeutic target and biomarker in inflammatory diseases, autoimmune diseases, and cancer.


Subject(s)
Autoimmune Diseases/metabolism , Inflammation/metabolism , Interleukin-1/metabolism , Neoplasms/metabolism , Adaptive Immunity , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Interleukin-1/genetics , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction , Tumor Microenvironment/immunology
14.
J Periodontal Res ; 56(4): 782-788, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33729573

ABSTRACT

OBJECTIVE: The aim of this study was to determine the potential role of TLR-4 in the osteoimmunological imbalance of periodontitis. BACKGROUND: Although current evidence supports that TLR-4 plays an important role in the inflammatory response of periodontal tissues triggered by microorganisms, little information is available regarding the function of TLR-4 in the osteoimmune regulation of homeostasis in periodontitis. METHODS: Human gingival epithelial cells (HGEC) were isolated from the gingival tissues of 3 healthy volunteers and the expression of osteoclastogenic cytokines was evaluated by ELISA and real time RT-PCR. In addition, 30 C57BL/6 mice were used and randomly divided into three groups: control group, periodontitis group (CP) and periodontitis+TAK-242 (a specific inhibitor of TLR-4) group (TAK-242) and the expression of osteoclastogenic cytokines and the osteoclast density in the periodontal tissue were evaluated by immunohistochemical staining and tartrate resistant acid phosphatase staining. Moreover, micro-computed tomography (Micro-CT) was used to assess bone resorption. RESULTS: The in vitro results showed that TAK-242 blocked the overproduction of IL-1, IL-6, TNF-α and RANKL in HGEC treated with LPS. The in vivo results revealed that TAK-242 also effectively decreased these osteoclastogenic cytokines in periodontal tissue of mice with periodontitis. More importantly, Micro-CT analysis showed a significant reduction of the alveolar bone loss in the TAK-242 group compared with the CP group. Furthermore, the TRAP staining showed a significant lower density of osteoclasts in the alveolar bone area of the TAK-242 group. CONCLUSION: TLR-4 inhibition decreased the differentiation of osteoclast through the inhibition of the overproduction of osteoclastogenic cytokines and the prevention of the alveolar bone absorption in mouse periodontitis models. Therefore, the use of TAK-242 might contribute to the recovery of the osteoimmunological homeostasis and might provide a potential strategy to treat periodontal diseases.


Subject(s)
Alveolar Bone Loss , Periodontitis , Animals , Mice , Alveolar Bone Loss/diagnostic imaging , Mice, Inbred C57BL , Osteoclasts , Periodontitis/diagnostic imaging , Periodontitis/drug therapy , Toll-Like Receptor 4 , X-Ray Microtomography
15.
Int J Cancer ; 148(7): 1548-1561, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33091960

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most common malignancy representing 90% of all forms of oral cancer worldwide. Although great efforts have been made in the past decades, the 5-year survival rate of OSCC patients is no more than 60% due to tumor metastasis and subsequent recurrence. The metastasis from the primary site is due to a complex process known as epithelial-to-mesenchymal transition (EMT). During the EMT, epithelial cells gradually acquire the structural and functional characteristics of mesenchymal cells, leading to the upregulation of cell migration and the promotion of tumor cell dissemination. Therefore, EMT attracted broad attention due to its close relationship with cancer invasion and metastasis. Therefore, in the present review, an extensive description of the current research on OSCC and the role of EMT in this cancer type is provided, including diverse EMT markers, regulatory networks and crucial EMT-inducing transcription factors in OSCC. Moreover, a brief summary was made regarding the current application of EMT-correlated indexes in the prognostic analysis of OSCC patients, and the potential therapeutic approaches against OSCC and difficulties in the development of an effective anti-EMT treatment are discussed. Our aim is to provide novel insights to develop new strategies to combat OSCC by targeting EMT.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Tumor Microenvironment/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Movement , Epithelial-Mesenchymal Transition , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Prognosis , Transcription Factors/genetics
16.
Neoplasia ; 22(11): 617-629, 2020 11.
Article in English | MEDLINE | ID: mdl-33045527

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) is an important type II arginine methyltransferase that can play roles in cancers in a highly tissue-specific manner, but its role in the carcinogenesis and metastasis of head and neck squamous cell carcinoma (HNSCC) remains unclear. Here, we detected PRMT5 expression in HNSCC tissues and performed series of in vivo and in vitro assays to investigate the function and mechanism of PRMT5 in HNSCC. We found that PRMT5 was overexpressed in dysplastic and cancer tissues, and associated with lymph node metastasis and worse patient survival. PRMT5 knockdown repressed the malignant phenotype of HNSCC cells in vitro and in vivo. PRMT5 specific inhibitor blocked the formation of precancerous lesion and HNSCC in 4NQO-induced tongue carcinogenesis model, prevented lymph node metastasis in tongue orthotopic xenograft model and inhibited cancer development in subcutaneous xenograft model and Patient-Derived tumor Xenograft (PDX) model. Mechanistically, PRMT5-catalyzed H3R2me2s promotes the enrichment of H3K4me3 in the Twist1 promoter region by recruiting WDR5, and subsequently activates the transcription of Twist1. The rescue experiments indicated that overexpressed Twist1 abrogated the inhibition of cell invasion induced by PRMT5 inhibitor. In summary, this study elucidates that PRMT5 inhibition could reduce H3K4me3-mediated Twist1 transcription and retard the carcinogenesis and metastasis of HNSCC.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , tRNA Methyltransferases/antagonists & inhibitors , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Models, Biological , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Transcription, Genetic , Xenograft Model Antitumor Assays
17.
Cancer Med ; 9(10): 3489-3499, 2020 05.
Article in English | MEDLINE | ID: mdl-32162845

ABSTRACT

BACKGROUND: Alanine-serine-cysteine transporter 2 (ASCT2), a major glutamine transporter, is essential for cell growth and tumor development in a variety of cancers. However, the clinicopathological significance and pathological role of ASCT2 in OSCC (oral squamous cell carcinoma) lesions remain unclear. METHODS: Sections from 89 OSCC patients and 10 paracancerous tissue controls were stained by immunohistochemistry (IHC) to detect the expression of ASCT2, glutaminase, and Ki-67. Survival analysis was carried out to determine the predictive value of ASCT2 expression using the log-rank test. Moreover, the critical role of ASCT2 in tumor growth was determined by a series of in vitro and in vivo assays. Cell Counting Kit-8 (CCK8), Western Blotting (WB), Reactive Oxygen Species (ROS), and Glutathione (GSH) detection were applied to explore the molecular mechanism of ASCT2 involvement in tumor development. RESULTS: In OSCC lesions, ASCT2 expression was significantly increased and associated with cell proliferation index (Ki-67) and GLS expression. Moreover, survival analysis showed that OSCC patients with high ASCT2 expression had lower overall survival (P = 0.0365). In OSCC cell lines, the high level of ASCT2 was inherent and related to the glutamine addiction of tumor cells. In vitro and in vivo functional experiments revealed that targeted silencing of ASCT2 can effectively inhibit OSCC cell proliferation and tumor growth. Mechanistically, targeting ASCT2 knockdown reduced glutamine uptake and intracellular GSH levels, which contribute to the accumulation of ROS and induce apoptosis in OSCC cells. CONCLUSION: ASCT2 is a significant factor for predicting overall survival in patients with OSCC, and targeting ASCT2 to inhibit glutamine metabolism may be a promising strategy for OSCC treatment.


Subject(s)
Amino Acid Transport System ASC/genetics , Cell Proliferation/genetics , Glutamine/metabolism , Minor Histocompatibility Antigens/genetics , Mouth Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Aged , Amino Acid Transport System ASC/metabolism , Animals , Cell Line, Tumor , Female , Gene Knockdown Techniques , Glutaminase/metabolism , Glutathione/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Middle Aged , Minor Histocompatibility Antigens/metabolism , Mouth Neoplasms/metabolism , Neoplasm Transplantation , Reactive Oxygen Species/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism
18.
J Oral Pathol Med ; 48(6): 477-482, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31077446

ABSTRACT

BACKGROUND: Although a few studies suggested that the chemokine CCL2 might be involved in the development of oral squamous cell carcinoma (OSCC), the exact mechanism remains unclear. In this study, we aimed to determine the resource of CCL2 in lesions and explored a potential mechanism that CCL2 promotes tumor progression. The study was an effort to provide new insights into the pathological role of CCL2 in OSCC. METHODS: Specimens of OSCC and normal oral mucosa were stained using immunohistochemistry (IHC) to assess the CCL2 expression. Enzyme-linked immunosorbent assay (ELISA) was used to detect the difference of CCL2 between OSCC and normal oral mucosa cell lines. In addition, we treated OSCC cells with exogenous rCCL2 combined with or without CCL2 neutralizing antibody and then determined the changes of in epithelial-mesenchymal transition (EMT) markers and cell migration capacity using immunofluorescence, Western blotting, transwell migration, and wound healing assays. RESULTS: We have found that CCL2 expression was upregulated significantly in both lesions and cell culture supernatant of OSCC compared with controls. IHC staining demonstrated that CCL2 expression was primarily located in the cytoplasm and cell membrane of cells. We have also found that rCCL2 could effectively induce EMT through upregulating Snail in OSCC cells, which was demonstrated by the decrease of E-cadherin and the increase of vimentin. In addition, we have found that CCL2 neutralizing antibody could block EMT induced by CCL2 in OSCC. CONCLUSIONS: CCL2 secreted by cancer cells can promote cell migration by inducing EMT via paracrine or autocrine in OSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Chemokine CCL2/metabolism , Epithelial-Mesenchymal Transition , Mouth Neoplasms/pathology , Antigens, CD , Cadherins , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Humans , Mouth Neoplasms/metabolism
19.
Cancer Lett ; 454: 108-119, 2019 07 10.
Article in English | MEDLINE | ID: mdl-30981761

ABSTRACT

Treatment of oral squamous cell carcinoma (OSCC) remains a challenge because of the lack of effective early treatment strategies and high incidence of relapse. Here, we showed that combined 4SC-202 (a novel selective class I HDAC inhibitor) and INK128 (a selective mTORC1/C2 inhibitor) treatment exhibited synergistic effects on inhibiting cell growth, sphere-forming ability, subcutaneous tumor formation and ALDH1+ cancer stem cells (CSCs) in OSCC. The initiation of OSCC was significantly inhibited by combined treatment in 4NQO-induced rat model. In addition, upregulated SOX2 was associated with advanced and metastatic tumors in OSCC patients and was responsible for the drug-resistance property of OSCC cells. The inhibitory effect of combined treatment on cell viability and ALDH1+ CSCs were attenuated by SOX2 verexpression. Furthermore, combined treatment can effectively overcome chemoresistance and inhibit the growth of recurrent OSCC in vitro and in vivo. Mechanistically, 4SC-202 and INK128 repressed SOX2 expression through miR-429/miR-1181-mediated mRNA degradation and preventing cap-dependent mRNA translation, respectively. These results suggest that combined class I histone deacetylase and mTORC1/C2 inhibition suppresses the carcinogenesis and recurrence of OSCC by repressing SOX2.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mouth Neoplasms/drug therapy , SOXB1 Transcription Factors/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Benzamides/administration & dosage , Benzamides/pharmacology , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacology , Carcinogenesis , Drug Synergism , Female , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred BALB C , Mouth Neoplasms/metabolism , Neoplasm Recurrence, Local , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Random Allocation , Rats , Squamous Cell Carcinoma of Head and Neck/metabolism , Xenograft Model Antitumor Assays
20.
Cancer Lett ; 434: 172-183, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30026052

ABSTRACT

The dysregulation of glycolysis has been suggested to lead to alteration of cell drug resistance signals, proliferation and metastasis. Emerging evidence indicates that lncRNAs play a key role in the cellular processes of tumor cells, including glycolysis, growth, and movement. However, the role and potential mechanism of lncRNAs in glycolysis-mediated metastasis has not been explored. In this study, we identified a novel lncRNA lnc-p23154 which is associated with OSCC patient metastasis and the promotion of OSCC cell migration and invasion in vitro and in vivo. Furthermore, we found that lnc-p23154 also participates in OSCC glycolysis by facilitating Glut1 expression. Rescue of lnc-p23154 reversed the suppression of OSCC cell migration and invasion induced by Glut1 knockdown. In addition, lnc-p23154 is mainly located in the nucleus and binds to the promoter region of miR-378a-3p, which represses Glut1 expression by targeting to its 3'UTR directly. Therefore, we concluded that lnc-p23154 may play an important role in Glut1-mediated glycolysis by inhibiting miR-378a-3p transcription and accelerate OSCC metastasis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/genetics , Glycolysis/genetics , Mouth Neoplasms/genetics , RNA, Long Noncoding/genetics , 3' Untranslated Regions/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Female , Glucose Transporter Type 1/metabolism , HEK293 Cells , Humans , Male , MicroRNAs/genetics , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL