Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Transl Med ; 22(1): 407, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689292

ABSTRACT

BACKGROUND AND OBJECTIVE: Progranulin (PGRN), a multifunctional growth factor, plays indispensable roles in the regulation of cancer, inflammation, metabolic diseases, and neurodegenerative diseases. Nevertheless, its immune regulatory role in periodontitis is insufficiently understood. This study attempts to explore the regulatory effects of PGRN on macrophage polarization in periodontitis microenvironment. METHODS: Immunohistochemical (IHC) and multiplex immunohistochemical (mIHC) stainings were performed to evaluate the expression of macrophage-related markers and PGRN in gingival samples from periodontally healthy subjects and periodontitis subjects. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were polarized towards M1 or M2 macrophages by the addition of LPS or IL-4, respectively, and were treated with or without PGRN. Real-time fluorescence quantitative PCR (qRT-PCR), immunofluorescence staining (IF), enzyme-linked immunosorbent assay (ELISA), and flow cytometry were used to determine the expressions of M1 and M2 macrophage-related markers. Co-immunoprecipitation was performed to detect the interaction between PGRN and tumor necrosis factor receptor 2 (TNFR2). Neutralizing antibody was used to block TNFR2 to confirm the role of TNFR2 in PGRN-mediated macrophage polarization. RESULTS: The IHC and mIHC staining of human gingival slices showed a significant accumulation of macrophages in the microenvironment of periodontitis, with increased expressions of both M1 and M2 macrophage markers. Meanwhile, PGRN was widely expressed in the gingival tissue of periodontitis and co-expressed mainly with M2 macrophages. In vitro experiments showed that in RAW264.7 cells and BMDMs, M1 markers (CD86, TNF-α, iNOS, and IL-6) substantially decreased and M2 markers (CD206, IL-10, and Arg-1) significantly increased when PGRN was applied to LPS-stimulated macrophages relatively to LPS stimulation alone. Besides, PGRN synergistically promoted IL-4-induced M2 markers expression, such as CD206, IL-10, and Arg1. In addition, the co-immunoprecipitation result showed the direct interaction of PGRN with TNFR2. mIHC staining further revealed the co-localization of PGRN and TNFR2 on M2 macrophages (CD206+). Blocking TNFR2 inhibited the regulation role of PGRN on macrophage M2 polarization. CONCLUSIONS: In summary, PGRN promotes macrophage M2 polarization through binding to TNFR2 in both pro- and anti-inflammatory periodontal microenvironments.


Subject(s)
Cell Polarity , Macrophages , Periodontitis , Progranulins , Receptors, Tumor Necrosis Factor, Type II , Periodontitis/metabolism , Periodontitis/pathology , Macrophages/metabolism , Humans , Animals , Receptors, Tumor Necrosis Factor, Type II/metabolism , Progranulins/metabolism , Mice , RAW 264.7 Cells , Gingiva/metabolism , Gingiva/pathology , Male , Female , Adult , Macrophage Activation , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL
2.
J Clin Periodontol ; 51(6): 774-786, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38462847

ABSTRACT

AIM: To evaluate the effect of subgingival delivery of progranulin (PGRN)/gelatin methacryloyl (GelMA) complex as an adjunct to scaling and root planing (SRP) on an experimental periodontitis dog model with Class II furcation involvement (FI). MATERIALS AND METHODS: A Class II FI model was established, and the defects were divided into four treatment groups: (a) no treatment (control); (b) SRP; (c) SRP + GelMA; (d) SRP + PGRN/GelMA. Eight weeks after treatment, periodontal parameters were recorded, gingival crevicular fluid and gingival tissue were collected for ELISA and RT-qPCR, respectively, and mandibular tissue blocks were collected for micro computed tomography (micro-CT) scanning and hematoxylin and eosin (H&E) staining. RESULTS: The SRP + PGRN/GelMA group showed significant improvement in all periodontal parameters compared with those in the other groups. The expression of markers related to M1 macrophage and Th17 cell significantly decreased, and the expression of markers related to M2 macrophage and Treg cell significantly increased in the SRP + PGRN/GelMA group compared with those in the other groups. The volume, quality and area of new bone and the length of new cementum in the root furcation defects of the PGRN/GelMA group were significantly increased compared to those in the other groups. CONCLUSIONS: Subgingival delivery of the PGRN/GelMA complex could be a promising non-surgical adjunctive therapy for anti-inflammation, immunomodulation and periodontal regeneration.


Subject(s)
Dental Scaling , Furcation Defects , Hydrogels , Progranulins , Animals , Dogs , Furcation Defects/therapy , Hydrogels/therapeutic use , Dental Scaling/methods , Immunomodulation , Root Planing/methods , Disease Models, Animal , Periodontitis/therapy , Periodontitis/immunology , Gelatin , Male , X-Ray Microtomography
3.
J Periodontal Res ; 58(6): 1201-1211, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37587560

ABSTRACT

OBJECTIVES: To evaluate the effect of hyperlipidemia on the healing of bone defects. MATERIALS AND METHODS: Apolipoprotein E (ApoE)-deficient mice and wild-type (WT) C57BL/6J mice were fed with an atherogenic high-fat diet (HFD) or a standard chow diet (as control) for 6 weeks. Blood samples were collected to evaluate serum lipid levels. Closed bone defects and open tooth extraction wounds were then created in the mandibles of these animals. One or two weeks after surgery, animals were euthanized. Micro-CT analysis and histomorphometric analysis were conducted to evaluate the healing of bone defects and the alveolar ridge resorption. RESULTS: Bone regeneration of closed bone defects was considerably delayed in the hyperlipidemic Apoe-/- mice and WT mice. No obvious difference was detected in the new bone formation of the tooth extraction wounds. The HFD-fed mice showed more prominent reduction in the lingual alveolar ridge height of the tooth extraction wounds when compared with the control group. CONCLUSIONS: Hyperlipidemia results in delayed bone regeneration in large closed bone defects. Although tooth extraction wounds are small and normally regenerated in a hyperlipidemic microenvironment, the prominent reduction in the alveolar ridge height is also a challenge for future restoration of the dentition.


Subject(s)
Alveolar Bone Loss , Alveolar Ridge Augmentation , Hyperlipidemias , Animals , Mice , Tooth Socket/surgery , Hyperlipidemias/complications , Alveolar Ridge Augmentation/methods , Mice, Inbred C57BL , Mice, Knockout, ApoE , Bone Regeneration , Alveolar Bone Loss/surgery , Tooth Extraction/methods , Apolipoproteins E
4.
Oral Dis ; 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37964399

ABSTRACT

OBJECTIVES: To assess the role of TNF-α/TNFR2 axis on promoting angiogenesis in oral squamous cell carcinoma (OSCC) cells and uncover the underlying mechanisms. MATERIALS AND METHODS: The expression of TNFR2 and CD31 in OSCC tissues was examined; gene expression relationship between TNF-α/TNFR2 and angiogenic markers or signaling molecules was analyzed; the expression of angiogenic markers, signaling molecules, TNFR1, and TNFR2 in TNF-α-stimulated OSCC cells treated with or without TNFR2 neutralizing antibody (TNFR2 Nab) were assessed; the concentration of angiogenic markers in the supernatant of OSCC cells was detected; conditioned mediums of OSCC cells treated with TNF-α or TNF-α + TNFR2 Nab were applied to human umbilical vein endothelial cells (HUVECs), followed by tube formation and cell migration assays. RESULTS: Significantly elevated expression of TNFR2 and CD31 in OSCC tissues was observed. A positive gene expression correlation was identified between TNF-α/TNFR2 and angiogenic markers or signaling molecules. TNFR2 Nab inhibited the effects of TNF-α on enhancing the expression of angiogenic factors and TNFR2, the phosphorylation of the Akt/mTOR signaling pathway, HUVECs migration, and tube formation. CONCLUSIONS: TNFR2 Nab counteracts the effect of TNF-α on OSCC cells through the TNFR2/Akt/mTOR axis, indicating that blocking TNFR2 might be a promising strategy against cancer.

5.
Clin Oral Implants Res ; 32(7): 808-817, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33756026

ABSTRACT

OBJECTIVE: To investigate the effect of a bone substitute material combined with fibroblast growth factor-2 (FGF-2) loaded barrier membrane on the preservation of alveolar ridge after tooth extraction. MATERIAL AND METHODS: Four dogs were included. Six extraction sockets of each animal received the 3 treatments and were randomly divided into three groups. Group A: negative control; Group B: bovine xenografts + membrane; and Group C: bovine xenografts + FGF-2-loaded membrane. CBCT and histological analysis were performed to evaluate changes in the width and height of alveolar ridges and extraction socket bone healing 8 weeks post-extraction. RESULTS: CBCT showed that the alveolar bone in Group A was significantly thinner than that in Group B and Group C at 1 and 3 mm apically from the alveolar crest. The alveolar width at 1 mm in Group C (60.99 ± 15.36%) was significantly thicker than that in Group B (39.75 ± 30.18%). Histomorphmetrical measurements showed that the buccal alveolar width at 1 mm was significantly thicker in Groups B and C than in Group A. Additionally, buccal bone height and lingual bone width at 1 mm in Group C (87.06 ± 10.34%, 89.09 ± 10.56%) were significantly greater than in Group A (53.48 ± 23.94%, 82.72 ± 12.59%). CONCLUSION: The present findings indicate that application of bovine bone combined with barrier membrane with or without FGF-2 over tooth sockets can effectively reduce ridge absorption, especially in terms of ridge width and FGF-2 modified membrane seems to improve the outcomes obtained with membrane alone.


Subject(s)
Acellular Dermis , Alveolar Bone Loss , Alveolar Ridge Augmentation , Alveolar Bone Loss/prevention & control , Alveolar Process/diagnostic imaging , Alveolar Process/surgery , Animals , Cattle , Fibroblast Growth Factor 2/pharmacology , Heterografts , Tooth Extraction , Tooth Socket/diagnostic imaging , Tooth Socket/surgery
6.
BMC Immunol ; 21(1): 32, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32503416

ABSTRACT

BACKGROUND: Macrophage M1 polarization plays a pivotal role in inflammatory diseases. Progranulin (PGRN) has potential anti-inflammation action, however, the effect of PGRN on macrophage M1 polarization has been poorly studied. Our study aimed to investigate the effect of PGRN on lipopolysaccharide (LPS)-induced macrophage M1 polarization and clarify the underlying mechanisms. METHODS: RAW264.7 cells were polarized to M1 macrophage by LPS with or without recombinant PGRN (rPGRN) and tumor necrosis factor alpha antibody (anti-TNF-α). A cell counting kit-8 assay (CCK-8), flow cytometry, Quantitative Real-Time PCR assay (q-PCR), Western blot assay and enzyme-linked immunosorbent assay (ELISA) were used to determine the effect of different treatments on cell proliferation, expression of surface phenotype marker and expressions and secretion of inflammatory cytokines. The activation of NF-κB/mitogen-activated protein kinase (MAPK) pathways and the nuclear translocation of NF-κB p65 were detected by Western blot and immunofluorescence respectively. THP-1 and primary bone marrow-derived monocytes (BMDMs) were also used to demonstrate effect of PGRN on expressions and secretion of inflammatory cytokines induced by LPS. RESULTS: In RAW264.7 cells, rPGRN at concentrations below 80 ng/ml significantly promoted cell proliferation in dose dependent fashion. rPGRN significantly inhibited LPS-induced change of phenotype (CD86/CD206 ratio) and function (tumor necrosis factor (TNF-α) and inducible nitric oxide synthase (iNOS) expressions). LPS-stimulated secretion of TNF-α and activated phosphorylation of IKKα/ß, IкBα, p65, JNK and p38 and the nucleus translocation of NF-кB p65 were also significantly downregulated by rPGRN. In addition, recombinant TNF-α (rTNF-α) significantly boosted TNF-α and iNOS expression vs the control group. Moreover, anti-TNF-α significantly inhibited LPS-induced TNF-α and iNOS expression. In THP-1 and BMDM cells, reversing effect of rPGRN on LPS-enhanced expressions of TNF-α and iNOS and secretion of TNF-α was further demonstrated. CONCLUSIONS: PGRN down-regulates LPS-induced macrophage M1 polarization in phenotype and function via NF-κB/MAPK signaling pathways.


Subject(s)
Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Progranulins/pharmacology , Animals , Biomarkers/metabolism , Cell Line , Cell Proliferation/drug effects , Cytokines/metabolism , Humans , Inflammation/metabolism , Macrophage Activation/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , THP-1 Cells/drug effects , THP-1 Cells/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
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
8.
Oral Dis ; 26(7): 1375-1383, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32060973

ABSTRACT

Cancer-associated fibroblasts (CAFs) are "activated" fibroblasts in the tumor microenvironment (TME) and play a vital role in all steps of cancer development. Increasing evidence focusing on the function of CAFs suggests that CAFs are candidate therapeutic targets and that drugs targeting the modification of CAFs would suppress tumor progression and be beneficial to tumor treatment and prevention. In the present study, we found that curcumin reversed the phenotype of CAFs to that of peri-tumor fibroblast (PTF)-like cells by downregulating the expression of α-SMA (a special marker for CAFs) and inhibiting the secretion of pro-carcinogenic cytokines, including transforming growth factor-ß1 (TGF-ß1), matrix metalloproteinases 2 (MMP2), and stromal cell-derived factor-1 (SDF-1). We further demonstrated that the conditioned medium (CM) derived from CAFs promoted the proliferation of Cal27, and this effect was confirmed by the xenograft model. More importantly, we found that curcumin blocked the CAF-mediated enhancement of Cal27 proliferation in vitro and in vivo. In conclusion, our data suggest that curcumin reverses cell phenotype from CAF to PTF-like cells and suppresses the CAF-mediated proliferation and tumorigenicity of Cal27 by inhibiting TSCC CAFs.


Subject(s)
Cancer-Associated Fibroblasts , Curcumin , Neoplasms , Cell Proliferation , Curcumin/pharmacology , Fibroblasts , Tumor Microenvironment
9.
J Transl Med ; 17(1): 66, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30819199

ABSTRACT

BACKGROUND: As the optimal source of seed cells in periodontal tissue engineering, periodontal ligament stem cells (PDLSCs) have always been researched to improve cell expansion due to their limited resource and spontaneous differentiation in vitro cultivation. Fibroblast growth factor-2 (FGF-2) has been proven to stimulate bone marrow mesenchymal stem cells (BMMSCs) proliferation and maintain their pluripotency when being added to the culture medium. As a small molecule inhibitor of transforming growth factor-beta receptors (TGF-ßRs), A83-01 can also promote cell proliferation. Therefore, the aim of this study was to verify whether the combined application of FGF-2 and A83-01 could augment cell quantity and quality during in vitro culture. METHODS: PDLSCs were preconditioned with A83-01, FGF-2, or their combination. A cell counting kit-8 (CCK8) assay, cell apoptosis assay, ALP activity assay, Alizarin Red S staining assay, RT-PCR assay, Western blot assay and ELISA were used to determine the sustained effects of different preconditioning strategies on the proliferation, apoptosis, stemness, osteogenic differentiation and paracrine action of PDLSCs. RESULTS: The combined application of FGF-2 and A83-01 significantly augmented cell expansion, reduced cell apoptosis, magnified stemness expression, promoted later osteogenic differentiation and mineralization and increased paracrine action of PDLSCs compared with the control. Moreover, the combination presented significant advantages in enhancing proliferation, stemness expression and paracrine action over FGF-2 alone. CONCLUSIONS: The combined application of A83-01 and FGF-2 may be an improved strategy for PDLSCs biological behavior optimization in culture expansion and advantageous for reinforcing proliferation, stemness expression and cytokine secretion over FGF-2 alone.


Subject(s)
Cell Culture Techniques/methods , Fibroblast Growth Factor 2/pharmacology , Periodontal Ligament/cytology , Pyrazoles/pharmacology , Stem Cells/cytology , Thiosemicarbazones/pharmacology , Adult , Alkaline Phosphatase/metabolism , Apoptosis/drug effects , Calcification, Physiologic/drug effects , Calcium/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation/drug effects , Humans , Osteogenesis/drug effects , Paracrine Communication/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Young Adult
10.
Med Sci Monit ; 25: 4384-4389, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31188801

ABSTRACT

BACKGROUND The restoration of damaged periodontium, especially one-wall intrabony defects, is a major challenge for clinicians. Concentrated growth factors (CGF) are a 100% autologous fibrin with multiple concentrated growth factors. The rigid fibrin structure of CGF makes it possible to preserve or reconstruct the initial bone volume. The aim of this study was to evaluate the clinical healing patterns after surgical application of CGF with and without a Bio-Oss graft in one-wall infrabony defects. MATERIAL AND METHODS We randomly divided 120 one-wall intrabony defects in 54 patients into 4 groups: flap surgery alone (Group 1), flap surgery with autologous CGF (Group 2), flap surgery with Bio-Oss (Group 3), and flap surgery with CGF+Bio-Oss (Group 4). Clinical parameters such as probing depth (PD) and clinical attachment level (CAL) change were recorded at baseline and at 6 and 12 months postoperatively. RESULTS At 12 months postoperatively, Group 2 showed significant improvement in clinical parameters over Group 1 (P<0.05) and the results were significantly greater in Groups 3 and 4 compared to the other groups (P<0.05). Although no significant difference was noted between Groups 3 and 4 in clinical parameters (P>0.05) compared to Group 3, the mean change of CAL at 6-12 months in Group 4 was not significant (P>0.05). CONCLUSIONS CGF reduced periodontal intrabony defects depth and, when mixed with Bio-Oss, CGF showed better results in the early period and the effect was more stable.


Subject(s)
Bone Substitutes/pharmacology , Chronic Periodontitis/drug therapy , Intercellular Signaling Peptides and Proteins/pharmacology , Adult , Alveolar Bone Loss/drug therapy , Bone Regeneration/drug effects , China , Female , Fibrin/pharmacology , Follow-Up Studies , Humans , Male , Mandibular Diseases/drug therapy , Middle Aged , Minerals/pharmacology , Periodontal Index , Periodontal Ligament , Wound Healing/drug effects
11.
Acta Biochim Biophys Sin (Shanghai) ; 50(3): 238-245, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29385406

ABSTRACT

C-reactive protein (CRP) acts as a biomarker reflecting different degrees of inflammation. Accumulating reports have suggested that there is a close relationship between CRP and various cancers. However, the influence of CRP on the development of tongue squamous cell carcinoma (TSCC) remains unclear. The purpose of this study was to investigate the role of CRP in TSCC. The results of immunohistochemical staining and statistical analyses showed that CRP expression was associated with TSCC tumor size, lymph node metastasis and pathological differentiation. Cell Counting Kit-8 (CCK-8) assay revealed that CRP could enhance TSCC cell proliferation in a dose- and time-dependent manner. Moreover, with CRP stimulation, proliferating cell nuclear antigen (PCNA) expression patterns presented a notable time-dependent up-regulation. In addition, CRP could enhance the invasion and migration of TSCC cells, as revealed by transwell and wound-healing assays, respectively. Annexin V-FITC/PI staining showed that CRP could protect TSCC cells from starvation- and drug-induced apoptosis. With CRP stimulation, the protein expression levels of phosphorylated protein kinase B (pAkt), phosphorylated mammalian target of rapamycin (pmTOR) and phosphorylated S6 ribosomal protein (pS6) were significantly increased, as demonstrated by western blot analysis. Our data suggest that CRP may play an important role in the development of TSCC. Moreover, the biological effects of CRP on TSCC cells might be related to Akt, mTOR, and S6.


Subject(s)
C-Reactive Protein/biosynthesis , Carcinoma, Squamous Cell/metabolism , Tongue Neoplasms/metabolism , Apoptosis/drug effects , C-Reactive Protein/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Tongue Neoplasms/pathology
13.
Biol Res ; 50(1): 19, 2017 May 25.
Article in English | MEDLINE | ID: mdl-28545522

ABSTRACT

BACKGROUND: Bromodomain-containing protein 4 (BRD4) inhibition is a new therapeutic strategy for many malignancies. In this study, we aimed to explore the effect of BRD4 inhibition by JQ1 on in vitro cell growth, migration and invasion of salivary adenoid cystic carcinoma (SACC). METHODS: The human normal epithelial cells and SACC cells (ACC-LM and ACC-83) were treated with JQ1 at concentrations of 0, 0.1, 0.5 or 1 µM. Cell Counting Kit-8 (CCK-8) assay was performed to evaluate cell proliferation. Cell apoptosis and cell cycle distribution was evaluated by Flow cytometry. Immunofluorescence staining was used to examine the expression of BRD4 in SACC cells. The quantitative real-time polymerase chain reaction (qRT-PCR) assay and western blot assay were performed to examine messenger RNA (mRNA) and protein levels in SACC cells. Wound-healing assay and transwell assay were used to evaluate the activities of migration and invasion of SACC cells. RESULTS: JQ1 exhibits no adverse effects on proliferation, cell cycle and cell apoptosis of the normal human epithelial cells, while suppressed proliferation and cell cycle, and induced apoptosis of SACC cells, down-regulated the mRNA and protein levels of BRD4 in SACC cells, meanwhile reduced protein expressions of c-myc and BCL-2, two known target genes of BRD4. Moreover, JQ1 inhibited SACC cell migration and invasion by regulating key epithelial-mesenchymal transition (EMT) characteristics including E-cadherin, Vimentin and Twist. CONCLUSIONS: BRD4 is an important transcription factor in SACC and BRD4 inhibition by JQ1 may be a new strategy for SACC treatment.


Subject(s)
Azepines/pharmacology , Carcinoma, Adenoid Cystic/drug therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Neoplasm Invasiveness/pathology , Nuclear Proteins/antagonists & inhibitors , Salivary Gland Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Carcinoma, Adenoid Cystic/pathology , Cell Cycle Proteins , Cell Line, Tumor , Down-Regulation , Humans , Real-Time Polymerase Chain Reaction , Salivary Gland Neoplasms/pathology
14.
Oral Health Prev Dent ; 15(4): 337-345, 2017.
Article in English | MEDLINE | ID: mdl-28831458

ABSTRACT

PURPOSE: To present four different cases with a diagnosed cemental tear. The differences in aetiology, clinical diagnosis and treatment of cemental tears are described and discussed in order to provide guidance for clinical practice. CASES: Four patients of different ages presented with gingival swelling and other different complaints. Clinically, localised deep periodontal pockets and inflammation were noted on affected aspects in four cases. Radiographic examination revealed a prickly or flakey structure and bone loss on the affected side of the involved tooth. Different treatments, such as extraction, traditional periodontal initial therapy, periodontal flap surgery, or no clinical intervention were given based on different extents of tooth mobility and bone loss. In the first two cases, teeth with cemental tear were extracted due to poor prognosis. In case 3, deep pockets recovered to a normal condition, while cemental tears on the distal aspect of tooth 21 had no abnormal sensation or clinical symptoms. In case 4, a periapical radiograph showed distal bone loss was interrupted, and the tooth also recovered normal mobility. Histopathological evaluation of the specimens with H&E staining all resulted in a definitive diagnosis of cemental/cementodentinal tears. CONCLUSIONS: Cementodentinal or cemental tears are unique, localized, tooth-related factors associated with attachment loss. Aging, trauma and occlusal overload are the main aetiological factors. Early diagnosis and appropriate treatment will avoid unnecessary tooth extraction and result in a better prognosis. Detailed clinical and radiographic examinations as well as explorative surgery may help to make a diagnosis of cemental tears, but histopathological analysis is the only method for a definitive diagnosis.


Subject(s)
Dental Cementum/injuries , Adult , Female , Humans , Male , Middle Aged , Wounds and Injuries/diagnosis , Wounds and Injuries/etiology , Wounds and Injuries/therapy
15.
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
16.
BMC Oral Health ; 15(1): 133, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26511096

ABSTRACT

BACKGROUND: Gingival epithelial cells are the major population of the gingival tissue, acting as the front-line defense against microbial intrusion and regulating the homeostasis of the periodontal tissue in health and disease via NLR family pyrin domain-containing-3 (NLRP3) inflammasome, which recognizes pathogen- and danger-associated molecular patterns (PAMPs and DAMPs). The aim of this study was to determine whether the activation of NLRP3 inflammasome depends on infection with the periodontal pathogen Porphyromonas gingivalis (P. gingivalis), or stimulation with P. gingivalis lipopolysaccharide (LPS), and/or extracellular adenosine triphosphate (ATP). METHODS: An oral epithelial cell line was treated with P. gingivalis, P. gingivalis LPS and ATP. The gene and protein expression of NLRP3 inflammasome components were quantified by real time RT-PCR and immunoblots. Production of IL-1ß and IL-18 was measured by ELISA. RESULTS: There was no increase in NLRP3 inflammasome gene expression after P. gingivalis infection unless pre-stimulated by ATP. Obvious increases of NLRP3 inflammasome gene expression was observed after P. gingivalis LPS stimulation, even pre-stimulated by ATP at 2 h. CONCLUSIONS: The findings indicate that the activation of NLRP3 inflammasome does not rely on P. gingivalis infection, unless stimulated by P. gingivalis LPS and/or extracellular ATP, suggesting diverse signaling pathways are involved in the host immune response.


Subject(s)
Gingiva , Lipopolysaccharides , NLR Family, Pyrin Domain-Containing 3 Protein , Porphyromonas gingivalis , Adenosine Triphosphate , Cell Line , Epithelial Cells , Gingiva/metabolism , Gingiva/microbiology , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyrin Domain , Signal Transduction
17.
Cell Death Dis ; 15(4): 277, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637499

ABSTRACT

Dysbiosis of the oral microbiota is related to chronic inflammation and carcinogenesis. Fusobacterium nucleatum (Fn), a significant component of the oral microbiota, can perturb the immune system and form an inflammatory microenvironment for promoting the occurrence and progression of oral squamous cell carcinoma (OSCC). However, the underlying mechanisms remain elusive. Here, we investigated the impacts of Fn on OSCC cells and the crosstalk between OSCC cells and macrophages. 16 s rDNA sequencing and fluorescence in situ hybridization verified that Fn was notably enriched in clinical OSCC tissues compared to paracancerous tissues. The conditioned medium co-culture model validated that Fn and macrophages exhibited tumor-promoting properties by facilitating OSCC cell proliferation, migration, and invasion. Besides, Fn and OSCC cells can recruit macrophages and facilitate their M2 polarization. This crosstalk between OSCC cells and macrophages was further enhanced by Fn, thereby amplifying this positive feedback loop between them. The production of CXCL2 in response to Fn stimulation was a significant mediator. Suppression of CXCL2 in OSCC cells weakened Fn's promoting effects on OSCC cell proliferation, migration, macrophage recruitment, and M2 polarization. Conversely, knocking down CXCL2 in macrophages reversed the Fn-induced feedback effect of macrophages on the highly invasive phenotype of OSCC cells. Mechanistically, Fn activated the NF-κB pathway in both OSCC cells and macrophages, leading to the upregulation of CXCL2 expression. In addition, the SCC7 subcutaneous tumor-bearing model in C3H mice also substantiated Fn's ability to enhance tumor progression by facilitating cell proliferation, activating NF-κB signaling, up-regulating CXCL2 expression, and inducing M2 macrophage infiltration. However, these effects were reversed by the CXCL2-CXCR2 inhibitor SB225002. In summary, this study suggests that Fn contributes to OSCC progression by promoting tumor cell proliferation, macrophage recruitment, and M2 polarization. Simultaneously, the enhanced CXCL2-mediated crosstalk between OSCC cells and macrophages plays a vital role in the pro-cancer effect of Fn.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Animals , Mice , Squamous Cell Carcinoma of Head and Neck/pathology , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Fusobacterium nucleatum , NF-kappa B/metabolism , In Situ Hybridization, Fluorescence , Mice, Inbred C3H , Macrophages/metabolism , Cell Proliferation , Head and Neck Neoplasms/metabolism , Cell Line, Tumor , Tumor Microenvironment
18.
Cells Tissues Organs ; 198(6): 428-37, 2013.
Article in English | MEDLINE | ID: mdl-24777155

ABSTRACT

Gingiva-derived mesenchymal stromal cells (GMSCs) have been considered as a promising alternative strategy for periodontal regeneration based on their potential for multilineage differentiation in vitro and the ability to form new bone in vivo. In order to investigate the capacity of GMSCs for periodontal regeneration and the fate of GMSCs during periodontal tissue repair, enhanced green fluorescent protein-labeled GMSCs were transplanted into class III furcation defects created in beagle dogs. The results showed that the transplanted GMSCs significantly enhanced the regeneration of the damaged periodontal tissue, including the alveolar bone, cementum and functional periodontal ligament (PDL). Moreover, GMSCs were able to differentiate into osteoblasts, cementoblasts and PDL fibroblasts in vivo. These findings indicate that GMSCs represent a novel cell source for periodontal tissue reconstruction.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Gingiva/cytology , Guided Tissue Regeneration, Periodontal , Mesenchymal Stem Cell Transplantation , Periodontal Ligament/cytology , Adolescent , Adult , Animals , Bone Regeneration , Cell Differentiation , Dental Cementum/cytology , Dogs , Green Fluorescent Proteins , Humans , Male , Mesenchymal Stem Cells/cytology , Osteogenesis , Periodontitis/therapy , Transplantation, Heterologous , Wound Healing , Young Adult
19.
Arch Pharm (Weinheim) ; 345(11): 870-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22836682

ABSTRACT

A series of novel N-aryl-3-aryl-1-arylmethyl-1H-pyrazole-5-carboxamide derivatives 4a-l was synthesized by the reaction of 3-aryl-1-arylmethyl-1H-pyrazole-5-carbonyl chloride with substituted aniline in good to excellent yields. Structures of the compounds were determined by IR, (1) H NMR, and HR-MS spectroscopy. The molecular structure was confirmed by the X-ray crystal analysis of one compound (4j) that was prone to crystallization. These compounds were used to induce mouse osteoblast precursors MC3T3-E1 into osteoblasts and the induction was assessed by alkaline phosphatase (ALP) activity and the gene expression of bone sialoprotein (BSP). The results showed that the compounds 4a-d, 4g, 4h, and 4k could increase the ALP activity in comparison with the negative control group and compound 4h was the most effective one which could induce osteogenesis. Furthermore, mRNA expression of BSP which is a marker of osteogenesis was up-regulated by the compound 4h.


Subject(s)
Integrin-Binding Sialoprotein/genetics , Osteoblasts/drug effects , Osteogenesis/drug effects , Pyrazoles/pharmacology , 3T3 Cells , Alkaline Phosphatase/metabolism , Animals , Cell Line , Crystallization , Crystallography, X-Ray , Magnetic Resonance Spectroscopy/methods , Mice , Osteoblasts/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , RNA, Messenger/metabolism , Spectrophotometry, Infrared , Up-Regulation/drug effects
20.
ACS Omega ; 7(13): 11405-11414, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35415372

ABSTRACT

M2 macrophages are generally recognized to have a protumor role, while the effect of M1 macrophages in cancer is controversial. Here, the in vitro and in vivo effects of conditioned medium from M1 macrophages (M1-CM) on oral squamous cell carcinoma (OSCC) cells and a potential mechanism were studied. CCK-8, colony formation, EdU labeling, xenograft growth, and Transwell assays were utilized to observe cell survival/proliferation and migration/invasion, respectively, in OSCC cell lines treated with basic medium (BM) and M1-CM. The ErbB2 phosphorylation inhibitor (CI-1033) and GDF15 knockout cell lines were used to appraise the role of ErbB2 and GDF15 in mediating the effects of M1-CM. Compared with BM, M1-CM significantly enhanced the survival/proliferation of SCC25 cells. The migration/invasion of SCC25 and CAL27 cells also increased. Mechanically, M1-CM promoted GDF15 expression and increased the phosphorylation of ErbB2, AKT, and ErK. CI-1033 significantly declined the M1-CM-induced activation of p-AKT and p-ErK and its protumor effects. M1-CM stimulated enhancement of p-ErbB2 expression was significantly decreased in cells with GDF15 gene knockout vs without. In xenograft, M1-CM pretreatment significantly promoted the carcinogenic potential of OSCC cells. Our results demonstrate that M1 macrophages induce the proliferation, migration, invasion, and xenograft development of OSCC cells. Mechanistically, this protumor effect of M1 macrophages is partly associated with inducing GDF15-mediated ErbB2 phosphorylation.

SELECTION OF CITATIONS
SEARCH DETAIL