Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Periodontal Res ; 55(3): 363-373, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31854464

ABSTRACT

OBJECTIVE: To investigate the molecular mechanism of Progranulin (PGRN) in promoting osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) in inflammatory environment. BACKGROUND: Progranulin is an antagonist of tumor necrosis factor (TNF) receptors (TNFRs) and is known to promote inflammatory periodontal bone defect regeneration. METHODS: TNFR1- and TNFR2-silenced hPDLSCs designed as hPDLSCs-sh-TNFR1 and hPDLSCs-sh-TNFR2 were cultured with osteoinductive medium containing TNF-α and (or) PGRN. Immunofluorescence, quantitative real-time PCR, and western blot were used to, respectively, detect expressions of TNFR1\TNFR2 and osteogenic differentiation markers as well as phosphorylation level in NF-κB\MAPK-related pathways. RESULTS: Immunofluorescence and real-time PCR showed that TNFR1 and TNFR2 positively expressed in hPDLSCs. TNF-α stimulation could significantly decrease the expressions of ALP and RUNX2 in hPDLSCs, whereas PGRN treatment could significantly enhance their expressions, and reverse TNF-α-mediated expression suppression of ALP and RUNX2 in hPDLSCs. In hPDLSCs-sh-TNFR1, TNF-α mediated osteogenic inhibition decreased, but both TNF-α + PGRN and alone PGRN significantly promoted expression of ALP and RUNX2. PGRN significantly enhanced expression of P-ERK1/2 and P-JNK, while corresponding inhibitors eliminated PGRN-stimulated osteogenic differentiation. In hPDLSCs-sh-TNFR2, no significant difference existed in osteogenic markers and P-JNK expression between the PGRN group and the control group. However, PGRN still activated P-ERK1/2 expression. Besides, PGRN antagonized TNF-α-enhanced NF-κB P65 expression. CONCLUSION: Progranulin promotes osteogenic differentiation of hPDLSCs via TNFR1 to inhibit TNF-α-sensitized NF-κB and via TNFR2 to activate JNK signaling. The mechanism by which PGRN activates ERK signaling remains to be explored.


Subject(s)
Osteogenesis , Periodontal Ligament/cytology , Progranulins/pharmacology , Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Chemokine CCL27/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , MAP Kinase Signaling System , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology
2.
Transl Cancer Res ; 8(5): 1709-1717, 2019 Sep.
Article in English | MEDLINE | ID: mdl-35116920

ABSTRACT

BACKGROUND: This study was performed to investigate the effect of insulin-like growth factor binding protein 3 (IGFBP3) on the biological behavior of tumor cells and tumorigenesis of oral squamous cell carcinoma (OSCC). METHODS: OSCC HB96, CAL27, and Tca8113 cells were transfected with the following plasmids: siRNA-IGFBP3, pcDNA-0-IGFBP3, or siRNA-NC (negative control). The effect of aberrant IGFBP3 on cell viability, apoptosis, and colony formation was assessed. Quantitative real-time PCR (qRT-PCR) and western blot analysis were used to measure IGFBP3 mRNA and protein levels, respectively. HB96 and CAL27 cells were transfected with IGFBP3-expressing lentiviral plasmids and then transplanted into nude mice to monitor xenograft tumor formation. RESULTS: An optimal transfection efficiency was obtained with 50 pmol siRNA-IGFBP3. Transient silencing of IGFBP3 significantly reduced cell viability, and increased apoptosis in comparison with the non-targeting negative control (NC). Overexpressing IGFBP3 promoted cell viability. Additionally, in comparison with the NC group, both cell growth and colony formation were reduced, while apoptosis was elevated in stably transfected cells. Moreover, silencing IGFBP3 inhibited cell viability and tumor formation in nude mice after 3 weeks, and colony formation, diminished tumorigenesis in nude mice, but promoted cell apoptosis in OSCC cells. CONCLUSIONS: Collectively, our study revealed a protumorigenic role for IGFBP3 in OSCC cancer cells, and demonstrated a potential mechanism for the dysregulation of IGFBP3 in cell growth. Therefore, IGFBP3 may be a potential therapeutic target for the treatment of OSCC.

3.
Mediators Inflamm ; 2016: 6430407, 2016.
Article in English | MEDLINE | ID: mdl-28077917

ABSTRACT

The important role of ephrinB2-EphB4 signaling pathway in bone remodeling has been well established. However, it is still unclear whether this bidirectional signaling also has effects on the regenerative processes of bone defects created in an inflammatory microenvironment. In this study, an experimental animal model of bone defects treated with lentiviruses was prepared and an inflammatory microenvironment was established. Expression levels of bone marker genes were monitored in the newly formed bone tissue using quantitative reverse transcriptase polymerase chain reaction and western blot. Immunohistochemical (IHC) staining and histomorphometric analysis were also performed to evaluate bone healing processes. Compared with the pLenti6.3-ctrl group, the pLenti6.3-ephb4siRNA group exhibited lower expression levels of bone formation marker genes and a higher level of NFATc1 in the new bone tissue. In addition, the newly formed bone was thinner and the number of giant osteoclasts was higher in the pLenti6.3-ephb4siRNA group than that in the pLenti6.3-ctrl group. In contrast, there was no significant difference between the pLenti6.3-efnb2siRNA group and the pLenti6.3-ctrl group. In conclusion, EphB4 plays an irreplaceable role in bone regeneration in an inflammatory microenvironment, whereas the functional loss of ephrinB2 can be effectively compensated, most possibly by other ephrins with similar chemical structures.


Subject(s)
Bone Regeneration , Inflammation , NFATC Transcription Factors/metabolism , Receptor, EphB2/metabolism , Receptor, EphB4/metabolism , Animals , Bone Remodeling , Bone and Bones/metabolism , Cell Differentiation , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Immunohistochemistry , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , RNA, Small Interfering/metabolism , Signal Transduction
4.
Int J Clin Exp Pathol ; 8(9): 10261-71, 2015.
Article in English | MEDLINE | ID: mdl-26617734

ABSTRACT

Pulp regeneration caused by endogenous cells homing has become the new research spot in endodontics. However, the source of functional cells that are involved in and contributed to the reconstituting process has not been identified. In this study, the possible role of systemical BMSC in pulp regeneration and the effect of stromal cell-derived factor-1 (SDF-1) on stem cell recruitment and angiogenesis were evaluated. 54 mice were divided into three groups: SDF-1 group (subcutaneous pockets containing roots with SDF-1 absorbed neutralized collagen gel and the green fluorescent protein (GFP) positive BMSCs transplantation via the tail vein), SDF-1-free group (pockets containing roots with gel alone and GFP + BMSCs transplantation) and Control group (pockets containing roots with gel alone). The animals were sacrificed after the roots were implanted into subcutaneous pockets for 3 weeks. Histomorphometric analysis was performed to evaluate the regenerated tissue in the canal by hematoxylin and eosin (HE) staining. The homing of the transplanted BMSCs was monitored with a fluorescence microscope and immunohistochemical analysis. The expression of ALP in new formed tissue was detected immunohistochemically. Dental-pulp-like tissue and new vessels were regenerated and GFP-positive BMSCs and expression of ALP could be observed in both SDF-1 group and SDF-1-free group. Furthermore, more GFP+ cells, stronger expression of ALP and stronger angiogenesis were found in the SDF-1 group than in the SDF-1-free group. To conclude, systemic BMSC can home to the root canal and participate in dental-pulp-like tissue regeneration. Intracanal application of SDF-1 may enhance BMSC homing efficiency and angiogenesis.


Subject(s)
Chemokine CXCL12/metabolism , Dental Pulp/metabolism , Mesenchymal Stem Cells/metabolism , Regeneration/physiology , Animals , Cell Movement/physiology , Mesenchymal Stem Cell Transplantation , Mice
5.
J Oral Pathol Med ; 43(1): 28-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23710769

ABSTRACT

BACKGROUND: Although molecular mechanism of growth differentiation factor 15 (GDF15) in tumorigenesis of oral squamous cell carcinoma (OSCC) is not clear, the diagnostic and prognostic value of serum GDF15 detection has been noticed. However, serum GDF15 levels in patients with oral leukoplakia and GDF15 as a potential predictive biomarker for response to induction chemotherapy in patients with OSCC have not been reported. METHODS: Pretreatment serum GDF15 concentration was detected using an enzyme-linked immunosorbent assay in 30 healthy persons, 24 patients with oral leukoplakia, and 60 patients with OSCC. RESULTS: Serum GDF15 concentration was significantly higher in patients with oral leukoplakia and OSCC, compared with healthy controls (F = 13.701, df = 2, P < 0.001). From a diagnostic standpoint, a cutoff value of 346.9 ng/l of serum GDF15 concentration was calculated using receiver operating characteristic curve, with a sensitivity of 0.750, specificity of 0.867, Youden's Index of 0.617, and area under curve of 0.863. From a prognostic standpoint, patients with serum GDF15 concentration <346.9 ng/l had an improved 3-year disease-free survival rate (64.3% vs 56.5%) compared with those above 346.9 ng/l, but the difference was not statistically significant. A decreased concentration of GDF15 (<346.9 ng/l) showed a predictive trend toward an improved response to induction chemotherapy compared with elevated concentration with clinical response rates of 100% and 71.4%, respectively, but the difference was not significant. CONCLUSION: Elevated GDF15 level may be not only a diagnostic biomarker for oral leukoplakia, but also a prognostic/predictive biomarker associated with decreased survival and diminished response to induction chemotherapy for patients with OSCC.


Subject(s)
Carcinoma, Squamous Cell/blood , Growth Differentiation Factor 15/blood , Leukoplakia, Oral/blood , Mouth Neoplasms/blood , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/secondary , Carcinoma, Squamous Cell/surgery , Case-Control Studies , Disease-Free Survival , Female , Humans , Induction Chemotherapy , Leukoplakia, Oral/surgery , Male , Middle Aged , Mouth Neoplasms/surgery , Neoadjuvant Therapy , Neoplasm Grading , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , ROC Curve , Radiotherapy, Adjuvant , Remission Induction , Retrospective Studies , Sensitivity and Specificity , Treatment Outcome
6.
BMC Cancer ; 13: 301, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23786757

ABSTRACT

BACKGROUND: The benefit of induction chemotherapy in locally advanced oral squamous cell carcinoma (OSCC) remains to be clearly defined. Induction chemotherapy is likely to be effective for biologically distinct subgroups of patients and biomarker development might lead to identification of the patients whose tumors are to respond to a particular treatment. Annexin A1 may serve as a biomarker for responsiveness to induction chemotherapy. The aim of this study was to investigate Annexin A1 expression in pre-treatment biopsies from a cohort of OSCC patients treated with surgery and post-operative radiotherapy or docetaxel, cisplatin and 5-fluorouracil (TPF) induction chemotherapy followed by surgery and post-operative radiotherapy. Furthermore we sought to assess the utility of Annexin A1 as a prognostic or predictive biomarker. METHODS: Immunohistochemical staining for Annexin A1 was performed in pre-treatment biopsies from 232 of 256 clinical stage III/IVA OSCC patients. Annexin A1 index was estimated as the proportion of tumor cells (low and high, <50% and ≥50% of stained cells, respectively) to Annexin A1 cellular membrane and cytoplasm staining. RESULTS: There was a significant correlation between Annexin A1 expression and pathologic differentiation grade (P=0.015) in OSCC patients. The proportion of patients with low Annexin A1 expression was significantly higher amongst those with moderate/poorly differentiated tumor (78/167) compared to those with well differentiated tumor (18/65). Multivariate Cox model analysis showed clinical stage (P=0.001) and Annexin A1 expression (P=0.038) as independent prognostic risk factors. Furthermore, a low Annexin A1 expression level was predictive of longer disease-free survival (P=0.036, HR=0.620) and locoregional recurrence-free survival (P=0.031, HR=0.607) compared to high Annexin A1 expression. Patients with moderate/poorly differentiated tumor and low Annexin A1 expression benefited from TPF induction chemotherapy as measured by distant metastasis-free survival (P=0.048, HR=0.373) as well as overall survival (P=0.078, HR=0.410). CONCLUSIONS: Annexin A1 can be used as a prognostic biomarker for OSCC. Patients with moderate/poorly differentiated OSCC and low Annexin A1 expression can benefit from the addition of TPF induction chemotherapy to surgery and post-operative radiotherapy. Annexin A1 expression can potentially be used as a predictive biomarker to select OSCC patients with moderate/poorly differentiated tumor who may benefit from TPF induction chemotherapy.


Subject(s)
Annexin A1/biosynthesis , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/metabolism , Induction Chemotherapy/methods , Mouth Neoplasms/metabolism , Annexin A1/analysis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Combined Modality Therapy , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Neoplasm Grading , Oral Surgical Procedures , Prognosis , Proportional Hazards Models , Radiotherapy
7.
Mol Cancer Ther ; 12(6): 1112-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23515614

ABSTRACT

Induction chemotherapy is likely to be effective for biologically distinct subgroups of patients with cancer with biomarker detection. To investigate the prognostic and predictive values of cyclin D1 expression in patients with oral squamous cell carcinoma (OSCC) who were treated in a prospective, randomized, phase III trial evaluating standard treatment with surgery and postoperative radiotherapy preceded or not by induction docetaxel, cisplatin, and 5-fluorouracil (TPF), immunohistochemical staining for cyclin D1 was conducted in pretreatment biopsy specimens of 232 out of 256 clinical stage III/IVA OSCC patients randomized to the clinical trial. Cyclin D1 index was estimated as the proportion of tumor cells with cyclin D1 nuclear staining. A low cyclin D1 expression predicted significantly better overall survival (OS; P = 0.001), disease-free survival (P = 0.005), locoregional recurrence-free survival (P = 0.003), and distant metastasis-free survival (DMFS; P = 0.002) compared with high cyclin D1 expression. Cyclin D1 expression levels were not predictive of benefit from induction TPF in the population overall. However, patients with nodal stage cN2 whose tumors had high cyclin D1 expression treated with TPF had significantly greater OS (P = 0.025) and DMFS (P = 0.025) when compared with high cyclin D1 cN2 patients treated with surgery upfront. Patients with low cyclin D1 level or patients with cN0 or cN1 disease did not benefit from induction chemotherapy. This study indicates that cN2 OSCC patients with high cyclin D1 expression can benefit from the addition of TPF induction chemotherapy to standard treatment. Cyclin D1 expression could be used as a biomarker in further validation studies to select cN2 patients that could benefit from induction therapy.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Combined Modality Therapy , Cyclin D1/biosynthesis , Mouth Neoplasms/drug therapy , Aged , Apoptosis/drug effects , Biomarkers, Tumor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Clinical Trials, Phase III as Topic , Cyclin D1/genetics , Disease-Free Survival , Docetaxel , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Staging , Prognosis , Taxoids/administration & dosage
8.
Oral Oncol ; 49(6): 542-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23403091

ABSTRACT

OBJECTIVES: Functional role of Annexin A1 in tumorigenesis is poorly understood. The aim of this study was to investigate the relationship between Annexin A1 protein expression and pathological differentiation grade in biopsy samples from a large cohort of patients with oral squamous cell carcinoma (OSCC); and to evaluate the potential role of Annexin A1 on cell proliferation and tumorigenesis of OSCC. MATERIALS AND METHODS: We investigated the relationship between Annexin A1 expression by immunohistochemical staining and pathological differentiation grade of biopsy samples from 232 OSCC patients, and the relationship between Annexin A1 expression and cell proliferation as well as tumor formation using both in vitro and in vivo OSCC models. RESULTS: Annexin A1 expression correlated significantly with pathological differentiation grade in OSCC patients, a lower Annexin A1 expression correlating with a poorer differentiation grade. Forced Annexin A1 overexpression in OSCC cell lines, CAL27 and Tca8113, significantly reduced the cell proliferation whereas down-regulation of Annexin A1 expression in OSCC cell line, HB96, significantly increased proliferation of HB96 cells. Tumors formed from CAL27 cells overexpressing Annexin A1 grown significantly slower compared to the parental CAL27 cells in nude mice and showed a significantly reduced nuclear Ki-67 labeling index. Interestingly, these tumors also showed a well differentiated histology pattern whereas the tumors formed from the parental cells were consistently moderately differentiated. CONCLUSIONS: These data support a significant correlation between Annexin A1 expression and pathological differentiation grade, and a functional role of Annexin A1 in inhibiting cell proliferation and cell differentiation in OSCC.


Subject(s)
Annexin A1/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Down-Regulation , Mouth Neoplasms/metabolism , Animals , Annexin A1/genetics , Base Sequence , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , DNA Primers , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Mouth Neoplasms/pathology , Prospective Studies , Real-Time Polymerase Chain Reaction
9.
PLoS One ; 7(3): e31601, 2012.
Article in English | MEDLINE | ID: mdl-22403616

ABSTRACT

BACKGROUND: Approximately 60-80% of patients with advanced head and neck squamous cell carcinoma (HNSCC) die within five years after diagnosis. Cisplatin-based chemotherapy is the most commonly used palliative treatment for these patients. To evaluate the prognostic value of X-linked inhibitor of apoptosis (XIAP) level as a potential biomarker in these patients, we investigated the relationship between XIAP expression and cisplatin response of these patients and their prognosis. METHODOLOGY/PRINCIPAL FINDINGS: Sixty patients with advanced HNSCC were recruited in this study. Expression of XIAP was examined both before and after chemotherapy and was correlated with chemotherapy response, clinicopathology parameters and clinical outcomes of the patients. We found that XIAP was expressed in 17 (20.83%) of the 60 advanced HNSCC samples and the expression was significantly associated with cisplatin resistance (P = 0.036) and poor clinical outcome (P = 0.025). Cisplatin-based chemotherapy induced XIAP expression in those post-chemotherapy samples (P = 0.011), was further associated with poorer clinical outcome (P = 0.029). Multivariate analysis demonstrated that only alcohol consumption, lymph node metastasis and XIAP level were independently associated with the prognosis of advanced HNSCC patients. Inhibiting XIAP expression with siRNA in XIAP overexpressed HNSCC cells remarkably increased their sensitivity to cisplatin treatment to nearly a 3 fold difference. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that XIAP overexpression plays an important role in the disease course and cisplatin-resistance of advanced HNSCC. XIAP is a valuable predictor of cisplatin-response and prognosis for patients with advanced head and neck cancer. Down-regulation of XIAP might be a promising adjuvant therapy for those patients of advanced HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Antineoplastic Agents/therapeutic use , Base Sequence , Cell Line, Tumor , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/drug therapy , Humans , Male , Neoplasm Staging , Prognosis , Treatment Outcome , X-Linked Inhibitor of Apoptosis Protein/genetics
10.
Shanghai Kou Qiang Yi Xue ; 17(5): 492-5, 2008 Oct.
Article in Chinese | MEDLINE | ID: mdl-18989590

ABSTRACT

PURPOSE: Candida albicans is one of the main opportunistic pathogen for human , the aim of this study is to investigate the phenomena of apoptosis in oral Candida albicans induced by acetic acid. METHODS: The Candida albicans of clinical strains were induced to apoptosis by using a weak acid acetic acid.The apoptosis was detected by flow cytometry and TEM. The data were processed for Chi-square test using SPSS11.5 software package. RESULTS: Oral Candida albicans had classic apoptosis when induced by proper concentration of acetic acid, and different concentrations of acetic acid had variable ability of inducing apoptosis of Candida albicans. CONCLUSIONS: Apoptosis can be detected in clinical strains of Candida albicans, the mechanism of apoptosis needs further research for the purpose of developing new antifungal drugs. Supported by National Natural Science Foundation of China(Grant No.30400498) and 2007 National College Student Innovative Planning Project.


Subject(s)
Apoptosis , Candida albicans/pathogenicity , Acetic Acid , Antifungal Agents , Candidiasis , Humans , In Vitro Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...