Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Theranostics ; 14(2): 622-639, 2024.
Article in English | MEDLINE | ID: mdl-38169606

ABSTRACT

Rationale: Cisplatin-based chemotherapy is the first-line treatment for late-stage head and neck squamous cell carcinoma (HNSCC). However, resistance to cisplatin has become a major obstacle for effective therapy. Cancer stem cells (CSCs) are critical for tumor initiation, growth, metastasis, and chemoresistance. How to effectively eliminate CSCs and overcome chemoresistance remains a key challenge. Herein, we confirmed that MYC plays critical roles in chemoresistance, and explored targeting MYC to overcome cisplatin resistance in preclinical models. Methods: The roles of MYC in HNSCC cisplatin resistance and cancer stemness were tested in vitro and in vivo. The combined therapeutic efficiency of MYC targeting using the small molecule MYC inhibitor MYCi975 and cisplatin was assessed in a 4­nitroquinoline 1-oxide-induced model and in a patient-derived xenograft model. Results: MYC was highly-expressed in cisplatin-resistant HNSCC. Targeting MYC using MYCi975 eliminated CSCs, prevented metastasis, and overcame cisplatin resistance. MYCi975 also induced tumor cell-intrinsic immune responses, and promoted CD8+ T cell infiltration. Mechanistically, MYCi975 induced the DNA damage response and activated the cGAS-STING-IRF3 signaling pathway to increase CD8+ T cell-recruiting chemokines. Conclusions: Our findings suggested that targeting MYC might eliminate CSCs, prevent metastasis, and activate antitumor immunity to overcome cisplatin resistance in HNSCC.


Subject(s)
Antineoplastic Agents , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Cisplatin/pharmacology , Cisplatin/therapeutic use , Squamous Cell Carcinoma of Head and Neck/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/metabolism , Drug Resistance, Neoplasm/genetics , Head and Neck Neoplasms/pathology , Cell Line, Tumor , Carcinoma, Squamous Cell/pathology , Neoplastic Stem Cells/metabolism
2.
Cancers (Basel) ; 15(24)2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38136338

ABSTRACT

The altered expression of long noncoding RNAs (lncRNAs) is associated with human carcinogenesis. We performed a high-throughput analysis of lncRNA expression in strictly selected pairs of metastatic head and neck squamous cell carcinoma (HNSCC) and non-metastatic HNSCC samples. We identified a novel lncRNA, which was highly expressed in metastatic HNSCC, named Metastasis Associated Squamous Cell Carcinoma 1 (MASCC1), for further study. Using qRT-PCR, we further compared MASCC1 expression in 60 HNSCC samples. The results show that high expression of MASCC1 in patients with HNSCC was related to poor prognosis. In vitro, MASCC1 knockdown (KD) inhibited HNSCC proliferation, migration, invasion, and tumor sphere formation, while promoting apoptosis. In vivo, MASCC1 KD inhibited HNSCC growth and lymph node metastasis. Mechanistically, MASCC1 acted as a competing endogenous RNA (ceRNA) by binding to miR-195, subsequently regulating the expression of Cyclin D1, BCL-2, and YAP1. Moreover, miR-195 overexpression rescued the effects of MASCC1 on the biological behaviors of HNSCC. Taken together, our results suggest that MASCC1 is a novel oncogene that can predict the prognosis of patients with HNSCC and is a potential therapeutic target for HNSCC intervention.

3.
Cell Death Dis ; 14(3): 187, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36894542

ABSTRACT

Cancer stem cells (CSCs) cause tumor metastasis and immune evasion by as-yet-unknown molecular mechanisms. In the present study, we identify a long noncoding RNA (lncRNA), termed PVT1, which is highly expressed in CSCs and correlated closely with lymph node metastasis of head and neck squamous cell carcinoma (HNSCC). PVT1 inhibition eliminates CSCs, prevents metastasis, and stimulates anti-tumor immunity, while inhibiting HNSCC growth. Moreover, PVT1 inhibition promotes the infiltration of CD8+ T cells into the tumor microenvironment, thereby enhancing immunotherapy by PD1 blockade. Mechanistically, PVT1 inhibition stimulates the DNA damage response, which induces CD8+ T cell-recruiting chemokines, while preventing CSCs and metastasis via regulating the miR-375/YAP1 axis. In conclusion, targeting PVT1 might potentiate the elimination of CSCs via immune checkpoint blockade, prevent metastasis, and inhibit HNSCC growth.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , MicroRNAs/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Cell Line, Tumor , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/genetics , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Tumor Microenvironment
4.
Oncoimmunology ; 11(1): 2130583, 2022.
Article in English | MEDLINE | ID: mdl-36211811

ABSTRACT

MYC plays critical roles in tumorigenesis and is considered an attractive cancer therapeutic target. Small molecules that directly target MYC and are well tolerated in vivo represent invaluable anti-cancer therapeutic agents. Here, we aimed to investigate the therapeutic effect of MYC inhibitors in head and neck squamous cell carcinoma (HNSCC). The results showed that pharmacological and genetic inhibition of MYC inhibited HNSCC proliferation and migration. MYC inhibitor 975 (MYCi975), inhibited HNSCC growth in both cell line-derived xenograft and syngeneic murine models. MYC inhibition also induced tumor cell-intrinsic immune responses, and promoted CD8+ T cell infiltration. Mechanistically, MYC inhibition increased CD8+ T cell-recruiting chemokines by inducing the DNA damage related cGAS-STING pathway. High expression of MYC combined with a low level of infiltrated CD8+ T cell in HNSCC correlated with poor prognosis. These results suggested the potential of small-molecule MYC inhibitors as anti-cancer therapeutic agents in HNSCC.


Subject(s)
Antineoplastic Agents , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Mice , Nucleotidyltransferases , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics
5.
FASEB J ; 36(12): e22627, 2022 12.
Article in English | MEDLINE | ID: mdl-36314562

ABSTRACT

Mechanical stress regulates various cellular functions like cell inflammation, immune responses, proliferation, and differentiation to maintain tissue homeostasis. However, the impact of mechanical signals on macrophages and the underlying mechanisms by which mechanical force regulates bone remodeling during orthodontic tooth movement remain unclear. NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome has been reported to promote osteoclastic differentiation to regulate alveolar bone resorption. But the relationship between the compressive force and NLRP3 inflammasome in macrophages remains unknown. In this study, immunohistochemical staining results showed elevated expression of NLRP3 and interleukin-1ß, as well as an increased number of macrophages expressing NLRP3, on the compression side of the periodontal tissues, after force application for 7 days. Furthermore, the number of tartrate-resistant acid phosphatase-positive osteoclasts, and the mRNA and protein expression levels of osteoclast-related genes in the periodontal tissue decreased in the Nlrp3-/- mice compared to the WT mice group after orthodontic movement. In vitro mechanical force activates the NLRP3 inflammasome and inhibits autophagy. Intraperitoneal injection of the autophagy inhibitor 3-methyladenine in Nlrp3-/- mice promoted orthodontic tooth movement. This result indicates that the absence of NLRP3 inflammasome activation can be partially compensated for by autophagy inhibitors. Mechanistically, force-induced activation of the NLRP3 inflammasome in macrophages via the cGAS/P2X7R axis. In conclusion, compressive force regulates orthodontic tooth movement via activating the NLRP3 inflammasome.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tooth Movement Techniques , Interleukin-1beta/metabolism , Macrophages/metabolism , Osteoclasts/metabolism
6.
Stem Cells Int ; 2022: 4185972, 2022.
Article in English | MEDLINE | ID: mdl-36159746

ABSTRACT

Root resorption is a common dental challenge that can lead to tooth loosening or even tooth loss. Among the cells involved in root resorption, cementoblasts are responsible for laying down the cementum, while macrophages with different phenotypes have also been shown to have bidirectional effects on root resorption. However, the relationship between macrophages and cementoblasts remains largely unknown. In this study, we examined the effect of macrophages with different polarization phenotypes on the mineralization of cementoblasts. Using the transwell coculture system and a conditioned medium-based coculture system, we found that compared with M0 (unpolarized macrophages), M1-polarized macrophages attenuated cementoblast mineralization, while M2-polarized macrophages enhanced cementoblast mineralization. Furthermore, by extracting M0/M1/M2 macrophage exosomes and examining their effects on the mineralization of cementoblasts, we found that the effects of macrophages on cementoblast mineralization were, at least partially, exerted by exosomes. Moreover, in vivo studies also indicated that an increased M1/M2 ratio could suppress cementoblast mineralization and bring about root resorption. During mechanical force-induced orthodontic tooth movement (OTM), root resorption was evident on the compression side of periodontal tissue, and a higher M1/M2 ratio and weaker cementoblast mineralization were observed on the compression side than on the tension side. We also used localized lipopolysaccharide (LPS) injection to increase the M1/M2 ratio around the roots of maxillary molars, where root resorption and decreased cementoblast mineralization were also observed. Furthermore, when we injected the exosomes from M0 and M1- and M2-polarized macrophages into mice, it was observed that the cementoblast mineralization was attenuated in the group injected with M1-polarized macrophage exosomes, while it was augmented in the group injected with M2-polarized macrophage exosomes.

7.
J Clin Periodontol ; 49(10): 1038-1051, 2022 10.
Article in English | MEDLINE | ID: mdl-35713268

ABSTRACT

AIM: We investigated the role of long non-coding RNAs and small nucleolar RNA host gene 5 (SNHG5) in the pathogenesis of periodontitis. MATERIALS AND METHODS: A ligature-induced periodontitis mouse model was established, and gingival tissues were collected from patients with periodontitis and healthy controls. Inflammatory cytokines were detected using quantitative reverse transcription-polymerase chain reaction and western blotting analyses. Direct interactions between SNHG5 and p65 were detected by RNA pull-down and RNA immunoprecipitation assays. Micro-computed tomography, haematoxylin and eosin staining, and immunohistochemical staining were used to measure periodontal bone loss. RESULTS: SNHG5 expression was down-regulated in human and mouse periodontal tissues compared to that in the healthy controls. In vitro experiments demonstrated that SNHG5 significantly ameliorated tumour necrosis factor α-induced inflammation. Mechanistically, SNHG5 directly binds to the nuclear factor-kappa B (NF-κB) p65 subunit and inhibits its translocation, thereby suppressing the NF-κB signalling pathway activation and reducing the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing three inflammasome expression. Locally injecting si-SNHG5 aggravated the periodontal destruction. CONCLUSION: This study revealed that SNHG5 mediates periodontal inflammation through the NF-κB signalling pathway, providing a potential therapeutic target for periodontitis treatment.


Subject(s)
Periodontitis , RNA, Long Noncoding , Animals , Cytokines/metabolism , Eosine Yellowish-(YS)/therapeutic use , Humans , Inflammasomes/metabolism , Inflammasomes/therapeutic use , Inflammation/metabolism , Interleukin-1beta/metabolism , Mice , NF-kappa B/metabolism , Nucleotides/therapeutic use , Periodontitis/drug therapy , RNA, Long Noncoding/genetics , RNA, Long Noncoding/therapeutic use , RNA, Small Nucleolar/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , X-Ray Microtomography
8.
Stem Cell Res Ther ; 13(1): 130, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35346361

ABSTRACT

BACKGROUND: The treatment of bone loss has posed a challenge to clinicians for decades. Thus, it is of great significance to identify more effective methods for bone regeneration. However, the role and mechanisms of long non-coding RNA small nucleolar RNA host gene 5 (SNHG5) during osteogenic differentiation remain unclear. METHODS: We investigated the function of SNHG5, Yin Yang 1 (YY1), miR-212-3p and growth differentiation factor 5 (GDF5) in osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) in vitro and in vivo. Molecular mechanisms were clarified by chromatin immunoprecipitation assay and dual luciferase reporter assay. RESULTS: We found SNHG5 expression was upregulated during osteogenesis of hBMSCs. Knockdown of SNHG5 in hBMSCs inhibited osteogenic differentiation while overexpression of SNHG5 promoted osteogenesis. Moreover, YY1 transcription factor directly bound to the promoter region of SNHG5 and regulated SNHG5 expression to promote osteogenesis. Dual luciferase reporter assay confirmed that SNHG5 acted as a miR-212-3p sponge and miR-212-3p directly targeted GDF5 and further activated Smad1/5/8 phosphorylation. miR-212-3p inhibited osteogenic differentiation, while GDF5 promoted osteogenic differentiation of hBMSCs. In addition, calvarial defect experiments showed knockdown of SNHG5 and GDF5 inhibited new bone formation in vivo. CONCLUSION: Our results demonstrated that the novel pathway YY1/SNHG5/miR-212-3p/GDF5/Smad regulates osteogenic differentiation of hBMSCs and may serve as a potential target for the treatment of bone loss.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , RNA, Long Noncoding , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Humans , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , RNA, Long Noncoding/genetics
9.
FASEB J ; 36(1): e22120, 2022 01.
Article in English | MEDLINE | ID: mdl-34958157

ABSTRACT

The mineralization capability of cementoblasts is the foundation for repairing orthodontic treatment-induced root resorption. It is essential to investigate the regulatory mechanism of mineralization in cementoblasts under mechanical compression to improve orthodontic therapy. Autophagy has a protective role in maintaining cell homeostasis under environmental stress and was reported to be involved in the mineralization process. Long noncoding RNAs are important regulators of biological processes, but their functions in compressed cementoblasts during orthodontic tooth movement remain unclear. In this study, we showed that compressive force downregulated the expression of mineralization-related markers. LincRNA-p21 was strongly enhanced by compressive force. Overexpression of lincRNA-p21 downregulated the expression of mineralization-related markers, while knockdown of lincRNA-p21 reversed the compressive force-induced decrease in mineralization. Furthermore, we found that autophagy was impeded in compressed cementoblasts. Then, overexpression of lincRNA-p21 decreased autophagic activity, while knockdown of lincRNA-p21 reversed the autophagic process decreased by mechanical compression. However, the autophagy inhibitor 3-methyladenine abolished the lincRNA-p21 knockdown-promoted mineralization, and the autophagy activator rapamycin rescued the mineralization inhibited by lincRNA-p21 overexpression. Mechanistically, the direct binding between lincRNA-p21 and FoxO3 blocked the expression of autophagy-related genes. In a mouse orthodontic tooth movement model, knockdown of lincRNA-p21 rescued the impeded autophagic process in cementoblasts, enhanced cementogenesis, and alleviated orthodontic force-induced root resorption. Overall, compressive force-induced lincRNA-p21 inhibits the mineralization capability of cementoblasts by impeding the autophagic process.


Subject(s)
Antigens, Differentiation/biosynthesis , Autophagy , Calcification, Physiologic , Compressive Strength , Dental Cementum/metabolism , Down-Regulation , RNA, Long Noncoding/biosynthesis , Animals , Male , Mice
10.
Stem Cells Int ; 2021: 6647651, 2021.
Article in English | MEDLINE | ID: mdl-34422062

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) are intraosseous stem cells, and the effects of tensile strain on BMSC differentiation mediate several bone-related treatments. To study the response of BMSCs under tension, we designed and developed a small cellular tension instrument, iStrain. When iStrain applied tension on BMSCs, these cells exhibited convergence in the alignment direction and lengthening of the cell processes and cell body. Real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting demonstrated that iStrain-mediated cyclic tension promotes the differentiation of BMSCs toward osteogenesis and fibrogenesis. And the mRNA and protein expression of differentiation-related genes changes with the extension of tension time.

11.
J Periodontol ; 92(11): 128-138, 2021 11.
Article in English | MEDLINE | ID: mdl-34231875

ABSTRACT

BACKGROUND: Migration of cementoblasts to resorption lacunae is the foundation for repairing root resorption during orthodontic tooth movement. Previous studies reported that autophagy was activated by compression in periodontal ligament cells. The aim of this study was to investigate the migration of cementoblasts and determine whether autophagy is involved in the regulation of cementoblast migration under compressive force. METHODS: Flow cytometry was employed to examine the apoptosis of murine cementoblasts (OCCM-30) at different compression times (0, 6, 12, and 24 hours) and magnitudes (0, 1.0, 1.5, and 2.0 g/cm2 ). Cell proliferation was examined using the CCK-8 method. Wound healing migration assays and transwell migration assays were performed to compare the migration of cementoblasts. Chloroquine (CQ) and rapamycin were used to inhibit and activate autophagy, respectively. The level of autophagy was determined using western blotting and immunofluorescence staining. The expression of matrix metalloproteinases (MMPs) was assessed using quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blot analysis, and enzyme-linked immunosorbent assay (ELISA). RESULTS: Cell apoptosis and proliferation did not significantly change in OCCM-30 cells under mechanical compression at magnitude of 1.5 g/cm2 for 12 hours. However, the migration of cementoblasts was significantly inhibited after the application of compressive force. MMP2, MMP9, and MMP13 mRNA expression was decreased, and MMP9 and MMP13 protein expression and secretion level were also decreased. Further, autophagic activity was inhibited in cementoblasts under compressive force. Treatment with chloroquine reduced the cellular migration, and rapamycin partially relieved the inhibition of cementoblast migration induced by the compressive force. MMP9 and MMP13 mRNA expression, protein expression, and secretion levels showed a similar trend. CONCLUSION: Migration of OCCM-30 cells was inhibited under compressive force partially dependent on the inhibition of MMPs, which was mediated by downregulation of autophagy. The findings provide new insights into the role of autophagy in biological behaviors of cementoblasts under compressive force and a potential therapeutic strategy for reducing external root resorption.


Subject(s)
Dental Cementum , Root Resorption , Animals , Autophagy , Down-Regulation , Mice , Periodontal Ligament
12.
Adv Sci (Weinh) ; 8(13): 2003376, 2021 07.
Article in English | MEDLINE | ID: mdl-34258151

ABSTRACT

Cancer stemness and immune evasion are closely associated, and play critical roles in tumor development and resistance to immunotherapy. However, little is known about the underlying molecular mechanisms that coordinate this association. Here, it is reported that elevated circular RNA FAT1 (circFAT1) in squamous cell carcinoma (SCC) unifies and regulates the positive association between cancer stemness and immune evasion by promoting STAT3 activation. circFAT1 knockdown (KD) reduces tumorsphere formation of SCC cells in vitro and tumor growth in vivo. Bioinformatic analysis reveals that circFAT1 KD impairs the cancer stemness signature and activates tumor cell-intrinsic immunity. Mechanistically, circFAT1 binding to STAT3 in the cytoplasm prevents STAT3 dephosphorylation by SHP1 and promotes STAT3 activation, resulting in inhibition of STAT1-mediated transcription. Moreover, circFAT1 KD significantly enhances PD1 blockade immunotherapy by promoting CD8+ cell infiltration into tumor microenvironment. Taken together, the results demonstrate that circFAT1 is an important regulator of cancer stemness and antitumor immunity.


Subject(s)
Cadherins/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , STAT3 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Animals , Cadherins/immunology , Cadherins/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , RNA, Circular/genetics , RNA, Circular/immunology , RNA, Circular/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
13.
STAR Protoc ; 2(2): 100484, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33982017

ABSTRACT

BMI1-expressing cancer stem cells (CSCs) play a key role in the development, progression, therapy resistance, recurrence, and metastasis of head and neck squamous cell carcinoma (HNSCC). Here, we present a chemically-induced HNSCC mouse model, genetically and pathologically similar to human HNSCC. This protocol describes how to use genetic lineage tracing based on the Cre-loxP recombination strategy, which allows us to study the regulation and targeting of BMI1+ CSCs in primary tumors and lymph node metastases. For complete details on the use and execution of this protocol, please refer to Chen et al. (2017) and Jia et al. (2020).


Subject(s)
Head and Neck Neoplasms , Neoplasms, Experimental , Neoplastic Stem Cells , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins , Squamous Cell Carcinoma of Head and Neck , Animals , Cell Lineage/genetics , Cells, Cultured , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Immunohistochemistry , Male , Mice , Neoplastic Stem Cells/classification , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
14.
Cell Stem Cell ; 28(9): 1597-1613.e7, 2021 09 02.
Article in English | MEDLINE | ID: mdl-33945793

ABSTRACT

Immunosurveillance is a critical mechanism guarding against tumor development and progression. Checkpoint inhibitors have shown significant success in cancer treatment, but expression of key factors such as PD-L1 in putative cancer stem cell (CSC) populations in squamous cell carcinoma has been inconclusive, suggesting that CSCs may have developed other mechanisms to escape immune surveillance. Here we show that CSCs upregulate the immune checkpoint molecule CD276 (B7-H3) to evade host immune responses. CD276 is highly expressed by CSCs in mouse and human head and neck squamous cell carcinoma (HNSCC) and can be used to prospectively isolate tumorigenic CSCs. Anti-CD276 antibodies eliminate CSCs in a CD8+ T cell-dependent manner, inhibiting tumor growth and lymph node metastases in a mouse HNSCC model. Single-cell RNA sequencing (RNA-seq) showed that CD276 blockade remodels SCC heterogeneity and reduces epithelial-mesenchymal transition. These results show that CSCs utilize CD276 for immune escape and suggest that targeting CD276 may reduce CSCs in HNSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Animals , Cell Line, Tumor , Mice , Neoplastic Stem Cells , Squamous Cell Carcinoma of Head and Neck
15.
Mol Cell ; 81(10): 2148-2165.e9, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33743195

ABSTRACT

Developing strategies to activate tumor-cell-intrinsic immune response is critical for improving tumor immunotherapy by exploiting tumor vulnerability. KDM4A, as a histone H3 lysine 9 trimethylation (H3K9me3) demethylase, has been found to play a critical role in squamous cell carcinoma (SCC) growth and metastasis. Here we report that KDM4A inhibition promoted heterochromatin compaction and induced DNA replication stress, which elicited antitumor immunity in SCC. Mechanistically, KDM4A inhibition promoted the formation of liquid-like HP1γ puncta on heterochromatin and stall DNA replication, which activated tumor-cell-intrinsic cGAS-STING signaling through replication-stress-induced cytosolic DNA accumulation. Moreover, KDM4A inhibition collaborated with PD1 blockade to inhibit SCC growth and metastasis by recruiting and activating CD8+ T cells. In vivo lineage tracing demonstrated that KDM4A inhibition plus PD1 blockade efficiently eliminated cancer stem cells. Altogether, our results demonstrate that targeting KDM4A can activate anti-tumor immunity and enable PD1 blockade immunotherapy by aggravating replication stress in SCC cells.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , DNA Replication/genetics , Epigenesis, Genetic , Histone Demethylases/metabolism , Immunity/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Stress, Physiological/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Chemokines/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage/genetics , Epithelial Cells/metabolism , Gene Deletion , Humans , Lymphatic Metastasis , Mice, Transgenic , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Programmed Cell Death 1 Receptor/metabolism , Receptors, CXCR3/metabolism , Th1 Cells/immunology
16.
Front Cell Dev Biol ; 9: 631181, 2021.
Article in English | MEDLINE | ID: mdl-33604341

ABSTRACT

Orthodontic tooth movement is achieved by periodontal tissue remodeling triggered by mechanical force. It is essential to investigate the reaction of periodontal ligament stem cells (PDLSCs) for improving orthodontic therapeutic approaches. Autophagy is an endogenous defense mechanism to prevent mechanical damage of environmental change. Long non-coding RNAs (lncRNAs) are key regulators in gene regulation, but their roles are still largely uncharacterized in the reaction of PDLSCs during orthodontic tooth movement. In this study, we showed that autophagy was significantly induced in PDLSCs under compressive force, as revealed by the markers of autophagy, microtubule-associated protein light chain 3 (LC3) II/I and Beclin1, and the formation of autophagosomes. After the application of compressive force, lncRNA FER1L4 was strongly upregulated. Overexpression of FER1L4 increased the formation of autophagosome and autolysosomes in PDLSCs, while knockdown of FER1L4 reversed the autophagic activity induced by mechanical force. In mechanism, FER1L4 inhibited the phosphorylation of protein kinase B (AKT) and subsequently increased the nuclear translocation of forkhead box O3 (FOXO3) and thus mediated autophagic cascades under compressive strain. In mouse model, the expression of Lc3 as well as Fer1l4 was increased in the pressure side of periodontal ligament during tooth movement. These findings suggest a novel mechanism of autophagy regulation by lncRNA during periodontal tissue remodeling of orthodontic treatment.

17.
Oral Dis ; 27(5): 1268-1282, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32890413

ABSTRACT

OBJECTIVE: This study aimed to investigate how mechanical force affects the proliferation of human periodontal ligament stem cells (hPDLSCs). METHODS: CCK-8 assays and staining of ki67 were performed to evaluate hPDLSCs proliferation. qRT-PCR, ELISA, or Western blot analysis were used to measure the expression levels of interleukin (IL)-6, miR-31 host gene (MIR31HG), DNA methyltransferase 1 (DNMT1), and DNA methyltransferase 3B (DNMT3B). Dual-luciferase reporter assays and chromatin immunoprecipitation (ChIP) assays were conducted to determine whether MIR31HG was targeted by DNMT1 and DNMT3B. MassARRAY mass spectrometry was used to quantify DNA methylation levels of the MIR31HG promoter. RESULTS: Mechanical force inhibited hPDLSCs proliferation with the downregulation of MIR31HG and upregulation of IL-6, DNMT1 and DNMT3B. Knockdown of MIR31HG suppressed hPDLSCs proliferation, and knockdown of DNMT1 or DNMT3B reversed mechanical force-induced downregulation of MIR31HG. Dual-luciferase and ChIP assays revealed DNMT1 and DNMT3B bound MIR31HG promoter in the region 1,015 bp upstream of the transcriptional start site. Treatment with 5'-aca-2'-deoxycytidine downregulated DNA methylation level in MIR31HG gene promoter, while mechanical force promoted the methylation of MIR31HG gene promoter. CONCLUSIONS: These findings elucidated how mechanical force affects proliferation via MIR31HG in hPDLSCs, providing clues for possible MIR31HG-based orthodontic therapeutic approaches.


Subject(s)
DNA Methylation , Periodontal Ligament , Cell Proliferation , Down-Regulation , Humans , Up-Regulation
18.
Mol Cell Biochem ; 476(1): 231-246, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32918186

ABSTRACT

Circular RNAs(circRNAs) are a large family of RNAs shaping covalently closed ring-like molecules and have become a hotspot with thousands of newly published studies. Stem cells are undifferentiated cells and have great potential in medical treatment due to their self-renewal ability and differentiation capacity. Abundant researches have unveiled that circRNAs have unique expression profile during the differentiation of stem cells and could serve as promising biomarkers of these cells. There are key circRNAs relevant to the differentiation, proliferation, and apoptosis of stem cells with certain mechanisms such as sponging miRNAs, interacting with proteins, and interfering mRNA translation. Moreover, several circRNAs have joined in the interplay between stem cells and lymphocytes. Our review will shed lights on the emerging roles of circRNAs in regulating the fate of diverse stem cells.


Subject(s)
RNA, Circular , Stem Cells/metabolism , Animals , Apoptosis , Autoimmune Diseases/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Humans , Mice , MicroRNAs/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/cytology , Pluripotent Stem Cells/cytology
19.
J Inflamm Res ; 13: 635-646, 2020.
Article in English | MEDLINE | ID: mdl-33061528

ABSTRACT

PURPOSE: Periodontitis is the leading cause of tooth loss. The role of long non-coding RNA (lncRNA) in periodontal inflammation remains unclear. The aim of this study was to investigate the role of lncRNA H19 in periodontitis and its possible regulation of autophagy in periodontitis. MATERIAL AND METHODS: Inflammation level was determined by quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) in periodontal ligament cells (PDLCs). Western blotting, flow cytometric analysis, and immunofluorescence staining were used to detect the autophagy flux. Overexpression or knockdown of H19 was used to confirm its function. Ligature-induced periodontitis model in mice and periodontitis-affected human gingival tissue were used in vivo. RNA sequencing was performed to determine the differentially expressed genes. RESULTS: Autophagy was significantly increased in PDLCs after inflammatory stimulation as well as in a ligature-induced periodontitis model in mice and periodontitis-affected human gingival tissue. During the inflammatory process, H19 expression was also significantly upregulated. Further, the levels of autophagic markers were significantly upregulated after overexpressing H19 in PDLCs, and the increased autophagic activity induced by inflammatory stimulation was reversed by H19 knockdown. RNA sequencing showed that the expression profiles of mRNAs were significantly altered after H19 overexpression, and the differentially expressed genes were enriched in the PI3K/AKT signaling pathway, which was confirmed by the decreased p-AKT protein expression in the H19 overexpression group. CONCLUSION: Periodontal inflammation activates autophagy flux, and H19 mediates the activation of autophagy via AKT pathway in periodontitis. This study expands our understanding of molecular regulation in periodontitis.

20.
Stem Cells Int ; 2020: 4813140, 2020.
Article in English | MEDLINE | ID: mdl-32908541

ABSTRACT

As the most important bone-resorbing cells, osteoclasts play fundamental roles in bone remodeling and skeletal health. Much effort has been focused on identifying the regulators of osteoclast metabolism. Noncoding RNAs (ncRNAs) reportedly regulate osteoclast formation, differentiation, survival, and bone-resorbing activity to participate in bone physiology and pathology. The present review intends to provide a general framework for how ncRNAs and their targets regulate osteoclast differentiation and the important events of osteoclastogenesis they are involved in, including osteoclast precursor generation, early differentiation, mononuclear osteoclast fusion, and multinucleated osteoclast function and survival. This framework is beneficial for understanding bone biology and for identifying the potential biomarkers or therapeutic targets of bone diseases. The review also summarizes the results of in vivo experiments and classic experiment methods for osteoclast-related researches.

SELECTION OF CITATIONS
SEARCH DETAIL
...