Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(11): 759, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37993428

ABSTRACT

Cancer-associated fibroblasts (CAFs) are abundant and heterogeneous in tumor microenvironment (TME). Cross-talk between cancer cells and CAFs results in cancer progression. Here, we demonstrated that a distinct cancer-associated fibroblasts subset with podoplanin (PDPN) positive expression (PDPN+ CAFs) was correlated with poor survival in oral squamous cell carcinoma (OSCC). PDPN+ CAFs promoted the progression of OSCC by transferring exosomal lncRNA FTX to OSCC cells. Mechanically, FTX bound to flap endonuclease-1 (FEN1), forming an RNA‒protein complex. FTX enhanced promoter demethylation of FEN1 by recruiting ten-eleven translocation-2 (TET2). In addition, FTX/FEN1 axis promoted OSCC cells motility by inhibiting ferroptosis. In xenograft experiments, RSL-3, a ferroptosis-inducing agent, suppressed the tumorigenesis potential of FEN1-overexpressed OSCC cells. Furthermore, Acyl-CoA synthetase long-chain family member 4 (ACSL4) was confirmed to participate in the motility promotion induced by FEN1 overexpression. FEN1 could bind to promoter region of ACSL4 and then inhibit ferroptosis in OSCC cells. Our study reveals that PDPN+ CAFs promote the invasiveness of OSCC cells by inhibiting ferroptosis through FTX/FEN1/ACSL4 signaling cascade. PDPN+ CAFs may serve as a novel potential therapeutic target for OSCC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Ferroptosis , Head and Neck Neoplasms , Mouth Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Squamous Cell/pathology , Cancer-Associated Fibroblasts/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mouth Neoplasms/pathology , Ferroptosis/genetics , Fibroblasts/metabolism , Head and Neck Neoplasms/metabolism , Tumor Microenvironment , Membrane Glycoproteins/metabolism
2.
Eur J Oral Sci ; 131(5-6): e12956, 2023.
Article in English | MEDLINE | ID: mdl-37849216

ABSTRACT

Pulpotomy is an effective treatment for retaining vital pulp after pulp exposure caused by caries removal and/or trauma. The expression of alpha smooth muscle actin (α-SMA) is increased during the wound-healing process, and α-SMA-positive fibroblasts accelerate tissue repair. However, it remains largely unknown whether α-SMA-positive fibroblasts influence pulpal repair. In this study, we established an experimental rat pulpotomy model and found that the expression of α-SMA was increased in dental pulp after pulpotomy relative to that in normal dental pulp. In vitro results showed that the expression of α-SMA was increased during the induction of odontogenic differentiation in dental pulp stem cells (DPSCs) compared with untreated DPSCs. Moreover, α-SMA overexpression promoted the odontogenic differentiation of DPSCs via increasing mitochondrial function. Mechanistically, α-SMA overexpression activated the mammalian target of rapamycin (mTOR) signaling pathway. Inhibition of the mTOR signaling pathway by rapamycin decreased the mitochondrial function in α-SMA-overexpressing DPSCs and suppressed the odontogenic differentiation of DPSCs. Furthermore, we found that α-SMA overexpression increased the secretion of transforming growth factor beta-1 (TGF-ß1). In sum, our present study demonstrates a novel mechanism by which α-SMA promotes odontogenic differentiation of DPSCs by increasing mitochondrial respiratory activity via the mTOR signaling pathway.


Subject(s)
Actins , Dental Pulp , Odontogenesis , Animals , Rats , Actins/metabolism , Actins/pharmacology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/growth & development , Stem Cells , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Pulpotomy
3.
J Oral Pathol Med ; 52(8): 727-737, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37433101

ABSTRACT

BACKGROUND: Long non-coding RNA BRAF-activated non-protein coding RNA plays bidirectional roles in human cancers. However, function and molecular mechanism of BRAF-activated non-protein coding RNA in oral squamous cell carcinoma still need to clarify further. METHODS: Long non-coding RNA microarray assay, in situ hybridization staining, clinicopathological data analysis were performed to investigate expression pattern of BRAF-activated non-protein coding RNA in oral squamous cell carcinoma tissue samples. Constructing ectopically expressed BRAF-activated non-protein coding RNA in oral squamous cell carcinoma cells via plasmids or siRNAs, then changeable abilities of proliferation and motility of these cells were observed in vitro and in vivo. RNA-protein pulldown, RNA immunoprecipitation, and bioinformatics analyses were performed to explore potential pathways involved in BRAF-activated non-protein coding RNA-based regulation of malignant progression in oral squamous cell carcinoma. RESULTS: BRAF-activated non-protein coding RNA was identified upregulated in oral squamous cell carcinoma tissue and correlated with nodal metastasis and clinical severity of patients. Overexpressed BRAF-activated non-protein coding RNA increased percentage of 5-ethynyl-2'-deoxyuridine-positive cells, viability, migration, and invasion rates of oral squamous cell carcinoma cells, while silenced BRAF-activated non-protein coding RNA could observe weakened effects in vitro. Xenograft tumor formed by BRAF-activated non-protein coding RNA-overexpressed cells had bigger volume, faster growth rates, higher weight, and more Ki67+ cells. Pulmonary metastasis induced by BRAF-activated non-protein coding RNA-silenced cells had fewer colony nodes, Ki67+ cells, and CD31+ blood vessels. Furthermore, BRAF-activated non-protein coding RNA was mainly localized in nucleus of oral squamous cell carcinoma cells and bound Ras-associated binding 1A. Silencing Ras-associated binding 1A could damage mobile ability and phosphorylation levels of nuclear factor-κB in oral squamous cell carcinoma cells induced by overexpressing BRAF-activated non-protein coding RNA. Opposite trend was also observed. CONCLUSION: Acting as a promoter in oral squamous cell carcinoma metastasis, BRAF-activated non-protein coding RNA promotes oral squamous cell carcinoma cells proliferation and motility by regulating the BRAF-activated non-protein coding RNA/Ras-associated binding 1A complex, which activates nuclear factor-κB signaling pathway.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Proto-Oncogene Proteins B-raf/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , NF-kappa B/metabolism , Ki-67 Antigen/metabolism , Mouth Neoplasms/genetics , Signal Transduction/genetics , Head and Neck Neoplasms/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics
4.
Am J Cancer Res ; 12(3): 1433-1434, 2022.
Article in English | MEDLINE | ID: mdl-35411236

ABSTRACT

[This corrects the article on p. 514 in vol. 5, PMID: 25973294.].

5.
J Tissue Eng ; 13: 20417314221084095, 2022.
Article in English | MEDLINE | ID: mdl-35321254

ABSTRACT

Apical periodontitis (AP) causes arrest of tooth root development, which is associated with impaired odontoblastic differentiation of stem cells from apical papilla (SCAPs), but the underlying mechanism remains unclear. Here, we investigated roles of extracellular vesicle (EV) in AP and odontoblastic differentiation of SCAPs, moreover, a novel nuclear factor I/C (NFIC)-encapsulated EV was developed to promote dentin regeneration. We detected a higher expression of EV marker CD63 in inflamed apical papilla, and found that EVs from LPS-stimulated dental pulp cells suppressed odontoblastic differentiation of SCAPs through downregulating NFIC. Furthermore, we successfully constructed the NFIC-encapsulated EV by overexpressing NFIC in HEK293FT cells, which could upregulate cellular NFIC level in SCAPs, promoting the proliferation and migration of SCAPs, as well as dentinogenesis both in vitro and in vivo. Collectively, based on pathological roles of EV in AP, our study provides a novel strategy for dentin regeneration by exploiting EV to deliver NFIC.

6.
Int Endod J ; 55(3): 263-274, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34807471

ABSTRACT

AIM: To evaluate the effects of hsa-miRNA-143-3p on the cytodifferentiation of human stem cells from the apical papilla (hSCAPs) and the post-transcriptional regulation of Nuclear factor I-C (NFIC). METHODOLOGY: miRNA expression profiles in human immature permanent teeth and during hSCAP differentiation were examined. hSCAPs were treated with miR-143-3p overexpression or silencing viruses, and the proliferation and odontogenic and osteogenic differentiation of these stem cells, and the involvement of the NFIC pathway, were investigated. Luciferase reporter and NFIC mutant plasmids were used to confirm NFIC mRNA as a direct target of miR-143-3p. NFIC expression analysis in the miR-143-3p overexpressing hSCAPs was used to investigate whether miR-143-3p functioned by targeting NFIC. Student's t-test and chi-square tests were used for statistical analysis. RESULTS: miR-143-3p expression was screened by microarray profiling and was found to be significantly reduced during hSCAP differentiation (p < .05). Overexpression of miR-143-3p inhibited the mineralization of hSCAPs significantly (p < .05) and downregulated the levels of odontogenic differentiation markers (NFIC [p < .05], DSP [p < .01] and KLF4 [p < .01]), whereas silencing of miR-143-3p had the opposite effect. The luciferase reporter gene detection and bioinformatic approaches identified NFIC mRNA as a potential target of miR-143-3p. NFIC overexpression reversed the inhibitory effect of miR-143-3p on the odontogenic differentiation of hSCAPs. CONCLUSIONS: miR-143-3p maintained the stemness of hSCAPs and modulated their differentiation negatively by directly targeting NFIC. Thus, inhibition of this miRNA represents a potential strategy to promote the regeneration of damaged tooth roots.


Subject(s)
Cell Differentiation , Dental Papilla/cytology , MicroRNAs , NFI Transcription Factors , Cells, Cultured , Humans , MicroRNAs/genetics , NFI Transcription Factors/genetics , Osteogenesis , Stem Cells
7.
Cell Biochem Funct ; 39(6): 702-712, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33929054

ABSTRACT

Autophagy is an evolutionarily conserved cellular process, in which damaged organelles and proteins are engulfed in autophagic vesicles and subsequently fuse with lysosomes for degradation. Autophagy is widely involved in different physiologic or pathologic processes in human. Accumulating evidence indicates that autophagy operates as a critical quality control mechanism to maintain pulp homeostasis and structural integrity of the dentin-pulp complex. Autophagy is activated during stresses and is involved in the pathogenesis of pulpitis and periapical infection. Recent discoveries have also provided intriguing insights into the roles of autophagy in tooth development, pulp aging and stress adaptation. In this review, we provide an update on the multifaceted functions of autophagy in physiology and pathophysiology of tooth. We also discuss the therapeutic implications of autophagy modulation in diseases and the regeneration of dentin-pulp complex.


Subject(s)
Autophagy , Dental Implants , Periapical Diseases/therapy , Pulpitis/therapy , Animals , Humans , Periapical Diseases/pathology , Pulpitis/pathology
8.
Stem Cell Res Ther ; 11(1): 517, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33256846

ABSTRACT

BACKGROUND: To investigate the odonto-immunomodulatory properties of dental pulp stem cell-derived small extracellular vesicles (DPSCs-sEV), which promote odontogenesis by switching macrophages toward the pro-healing M2 phenotype. METHODS: MicroRNA sequencing was carried out for microRNA profiling of DPSCs-sEV. Automated Western blot, qPCR, ELISA, and flow cytometry were performed to identify the functions of microRNA-enriched DPSCs-sEV in macrophages. A luciferase reporter gene assay was carried out to confirm exosomal miR-125a-3p's direct target gene. DPSCs-sEV-stimulated macrophage-conditioned media were used to promote odontogenesis in DPSCs and explore the mechanism of immune response in DPSCs-SEV-stimulated odontogenesis. DPSCs-sEV were injected into the exposed pulp tissue of rat incisor to investigate the odonto-immunomodulatory properties of DPSCs-sEV in vivo. RESULTS: DPSCs-sEV switched macrophages to the pro-healing M2 phenotype by inhibiting TLR and NFκΒ signaling. MicroRNA sequencing found 81 microRNAs significantly altered in DPSCS-sEV, with miR-125a-3p showing a 12-fold upregulation. Exosomal miR-125a-3p switched macrophages toward the M2 phenotype via inhibiting NFκΒ and TLR signaling via direct IKBKB targeting. Interestingly, DPSCs-sEV and the encapsulated miR-125a-3p enhanced BMP2 release in macrophages, promoting odontogenesis in DPSCs through BMP2 pathway activation. The rat study confirmed that DPSCs-sEV could be used as ideal biomimetic tools to enhance odontogenesis by switching macrophages toward pro-healing M2 cells. CONCLUSIONS: We firstly defined the odonto-immunomodulatory properties of microRNA-enriched DPSCs-sEV, which could be used as ideal biomimetic tools to enhance odontogenesis by switching macrophages toward the pro-healing M2 phenotype.


Subject(s)
Extracellular Vesicles , MicroRNAs , Animals , Cell Differentiation , Dental Pulp , Immunity , Macrophages , MicroRNAs/genetics , Odontogenesis , Rats , Stem Cells
9.
ACS Appl Mater Interfaces ; 12(40): 45504-45515, 2020 Oct 07.
Article in English | MEDLINE | ID: mdl-32911929

ABSTRACT

A soft body area sensor network presents a promising direction in wearable devices to integrate on-body sensors for physiological signal monitoring and flexible printed circuit boards (FPCBs) for signal conditioning/readout and wireless transmission. However, its realization currently relies on various sophisticated fabrication approaches such as lithography or direct printing on a carrier substrate before attaching to the body. Here, we report a universal fabrication scheme to enable printing and room-temperature sintering of the metal nanoparticle on paper/fabric for FPCBs and directly on the human skin for on-body sensors with a novel sintering aid layer. Consisting of polyvinyl alcohol (PVA) paste and nanoadditives in the water, the sintering aid layer reduces the sintering temperature. Together with the significantly decreased surface roughness, it allows for the integration of a submicron-thick conductive pattern with enhanced electromechanical performance. Various on-body sensors integrated with an FPCB to detect health conditions illustrate a system-level example.


Subject(s)
Monitoring, Physiologic , Skin/chemistry , Temperature , Wearable Electronic Devices , Humans , Metal Nanoparticles/chemistry , Nickel/chemistry , Paper , Particle Size , Silver/chemistry , Surface Properties
10.
Dalton Trans ; 48(22): 7659-7665, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31049511

ABSTRACT

The development of flexible current collectors as an indispensable component in energy storage devices has been in strong demand for the ever-growing market of flexible and wearable electronics. Herein, flexible and conductive paper-based current collectors are fabricated by directly depositing a metallic Ni layer composed of spiny Ni nanospheres of 400 nm diameter on the surface of filter paper via electroless deposition. The metallic paper shows excellent electric and mechanical properties: the sheet resistance is 2.7 Ω cm-2 (R0 = 0.8 Ω cm-2) after 5000 bending cycles and the mass density is only 0.35 g cm-3. MnO2 is selected as an electrode active material to explore the role of flexible and conductive paper-based current collectors in supercapacitors. Electrochemical results reveal that the largest areal specific capacitance is 1095 mF cm-2 at 1 mA cm-2 and the excellent electrochemical performance can be attributed to the hierarchical porous fibre structure of paper and the lower contact resistance between the active material and the current collector. Note that the approach can be applied to an enlarged size of metallic conductive paper or textile, presenting a simple and feasible method to fabricate flexible current collectors in a large scale.

11.
Stem Cell Res Ther ; 10(1): 48, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30704530

ABSTRACT

BACKGROUND: Bone marrow mesenchymal stem cells (BMMSCs) are suitable cell sources for dental pulp regeneration, but the mechanism of BMMSCs differentiation into odontogenic lineage remains unknown. The aim of the present study was to reveal the role of magnesium transporter protein 1 (MagT1) and MAPK pathways in the odontogenic differentiation of BMMSCs. METHODS: The RNA sequencing (RNA-seq) was performed to explore the altered transcriptome of BMMSCs undergoing odontogenic differentiation induced by tooth germ cell-condition medium (TGC-CM). Pathway analysis was conducted to explore enriched pathways of the differential expression signature. Automated western blot, real-time PCR, shRNA lentivirus, and flow cytometry were used to detect the function of MagTl and MAPK pathway in odontogenic differentiation of BMMSCs. RESULTS: RNA-seq identified 622 differentially expressed genes associated with odontogenic differentiation of BMMSCs induced by TGC-CM, some of which were responsible for MAPK pathway. Consistently, we verified that TGC-CM induced odontogenic differentiation of BMMSCs through activating ERK/MAPK pathway, and the inactivation of ERK/MAPK pathway inhibited the odontogenic differentiation induced by TGC-CM. We also found MagT1 protein was significantly increased during odontogenic differentiation of BMMSCs induced by TGC-CMM, in accordance, MagT1 knockdown significantly decreased the extent of mineralized nodules and the protein levels of alkaline phosphatase (ALP), dentin matrix protein 1 (DMP-1), and dentin sialophosphoprotein (DSP). Flow cytometry showed that intracellular Mg2+ was significantly reduced in MagT1-knockdown BMMSCs, indicating the suppression of MagT1 inhibited odontogenic differentiation of BMMSCs by decreasing intracellular Mg2+. Finally, we performed RNA-seq to explore the altered transcriptome of MagT1-knockdown BMMSCs undergoing odontogenic differentiation and identified 281 differentially expressed genes, some of which were involved in MAPK pathway. Consistently, automated western blot analysis found the ERK/MAPK pathway was inhibited in MagT1-knockdown BMMSCs during odontogenic differentiation, indicating that suppression of MagT1 inhibited odontogenic differentiation of BMMSCs via ERK/MAPK pathway. CONCLUSIONS: This study identified the significant alteration of transcriptome in BMMSCs undergoing odontogenic differentiation induced by TGC-CM. We clarified the pivotal role of MagT1 and ERK/MAPK pathway in odontogenic differentiation of BMMSCs, and suppression of MagT1 inhibited the odontogenic differentiation of BMMSCs by decreasing the intracellular Mg2+ and inactivating ERK/MAPK pathway.


Subject(s)
Cation Transport Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Cation Transport Proteins/genetics , Cell Differentiation/physiology , Culture Media, Conditioned , MAP Kinase Signaling System , Odontogenesis , Rats , Rats, Sprague-Dawley , Signal Transduction
12.
EBioMedicine ; 36: 209-220, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30243489

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) play an important role in regulating tumor progression by transferring exosomes to neighboring cells. Our aim was to clarify the role of microRNA encapsulated in the exosomes derived from CAFs in oral squamous cell carcinoma (OSCC). METHODS: We examined the microRNA expression profiles of exosomes derived from CAFs and donor-matched normal fibroblasts (NFs) from patients with OSCC. We used confocal microscopy to examine the transportation of exosomal miR-34a-5p between CAFs and OSCC cells. Next, luciferase reporter and its mutant plasmids were used to confirm direct target gene of miR-34a-5p. Phenotypic assays and in vivo tumor growth experiments were used to investigate the functional significance of exosomal miR-34a-5p. FINDINGS: We found that the expression of miR-34a-5p in CAF-derived exosomes was significantly reduced, and fibroblasts could transfer exosomal miR-34a-5p to OSCC cells. In xenograft experiments, miR-34a-5p overexpression in CAFs could inhibit the tumorigenesis of OSCC cells. We further revealed that miR-34a-5p binds to its direct downstream target AXL to suppress OSCC cell proliferation and metastasis. Stable ectopic expression of AXL in OSCC cells overexpressing miR-34a-5p restored proliferation and motility abolished by the miRNA. The miR-34a-5p/AXL axis promoted OSCC progression via the AKT/GSK-3ß/ß-catenin signaling pathway, which could induce the epithelial-mesenchymal transition (EMT) to promote cancer cells metastasis. The miR-34a-5p/AXL axis enhanced nuclear translocation of ß-catenin and then induced transcriptional upregulation of SNAIL, which in turn activated both MMP-2 and MMP-9. INTERPRETATION: The miR-34a-5p/AXL axis confers aggressiveness in oral cancer cells through the AKT/GSK-3ß/ß-catenin/Snail signaling cascade and might represent a therapeutic target for OSCC. FUND: National Natural Science Foundation of China.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Exosomes/metabolism , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Paracrine Communication , Animals , Biomarkers , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Models, Biological , Mouth Neoplasms/pathology , Proto-Oncogene Proteins/genetics , RNA Interference , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Axl Receptor Tyrosine Kinase
13.
Exp Cell Res ; 369(1): 43-53, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29719198

ABSTRACT

Podoplanin is upregulated in the invasive front of oral squamous cell carcinoma (OSCC). Carcinoma-associated fibroblasts (CAFs) may mediate podoplanin expression. However, the role of podoplanin in OSCC cell and fibroblast interaction remains elusive. In the present study, we found that positive podoplanin expression in OSCC cells correlated with smooth muscle actin (α-SMA) expression in CAFs. Using CAFs and normal mucosal fibroblasts (NFs), we established indirect and direct co-culture systems mimicking the structure of OSCC. Podoplanin-overexpressing OSCC cells promoted NF activation; in direct co-culture, but not in indirect co-culture, podoplanin-overexpressing OSCC cells increased fibroblast invasion via matrix metalloproteinase 2 (MMP-2), MMP-14, and αv/ß6 integrin receptor (ITGA5/ITGB6) signaling. CAFs also induced podoplanin expression through the transforming growth factor-ß1 (TGF-ß1)/Smad pathway. TGF-ß1 increased the podoplanin-dependent activation of epidermal growth factor receptor (EGFR), AKT, and extracellular signal-regulated kinase (ERK) signaling. Additionally, CAFs promoted OSCC cell invasion by upregulating MMP-2 and MMP-14 expression in both indirect and direct co-culture. Taken together, our findings indicate that podoplanin regulates the interaction between OSCC cells and CAFs via the mutual paracrine effects of TGF-ß1.


Subject(s)
Cancer-Associated Fibroblasts/physiology , Carcinoma, Squamous Cell/pathology , Fibroblasts/physiology , Membrane Glycoproteins/physiology , Mouth Neoplasms/pathology , Transforming Growth Factor beta1/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/genetics , Cell Communication/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/genetics , Mouth Neoplasms/genetics , Paracrine Communication/genetics , Signal Transduction/genetics , Transforming Growth Factor beta1/genetics
14.
Oral Oncol ; 51(10): 935-43, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26194050

ABSTRACT

OBJECTIVE: The present study aimed to clarify the role of Moesin in oral squamous cell carcinoma (OSCC) progression, especially in regulation of cell motility. MATERIALS AND METHODS: Immunohistochemistry and western blotting were used to investigate the expression of Moesin, E-cadherin, p120-catenin and MT1-MMP in normal epithelia, dysplasia and OSCCs. Then, Moesin was knockdown by siRNA in OSCC cell lines, WSU-HN6 and CAL27, and the biological role of Moesin in cell adhesion and motility was evaluated by transwell system, cell spreading and aggregation assays. The interactions between Moesin, MT1-MMP and E-cadherin/p120-catenin complex were determined by co-immunoprecipitation and immunofluorescence. RESULTS: Moesin expression was found decreased in the membrane and increased in cytoplasm during the malignant transformation of oral epithelia, and cytoplasmic overexpression of Moesin correlated with nodal metastasis and poor prognosis of OSCCs. Furthermore, Moesin-silencing induced an increased cell-cell adhesion but decreased invasiveness, which was subsequently demonstrated might due to Moesin-mediated E-cadherin and p120-catenin interaction. Meantime, Moesin-silencing significantly down-regulated MT1-MMP expression, accompanied by reduced cell motility and impaired filopodia formation, which was also observed when MT1-MMP knockdown by RNAi or tissue inhibitor (TIMP2), indicating the involvement of MT1-MMP in Moesin-mediated cell motility. Finally, the relationship between Moesin, E-cadherin and MT1-MMP was confirmed in OSCC tissue samples. CONCLUSION: Taken together, our results indicate Moesin may regulate cell motility through its interactions with MT1-MMP and E-cadherin/p120-catenin adhesion complex and cytoplasmic expression of Moesin correlates with nodal metastasis and poor prognosis of OSCCs, indicating Moesin may be a potential candidate for targeted gene therapy for OSCCs.


Subject(s)
Cadherins/metabolism , Carcinoma, Squamous Cell/pathology , Catenins/metabolism , Matrix Metalloproteinase 14/metabolism , Microfilament Proteins/physiology , Mouth Neoplasms/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Humans , Mouth Neoplasms/enzymology , Mouth Neoplasms/metabolism , Delta Catenin
15.
Am J Cancer Res ; 5(2): 514-29, 2015.
Article in English | MEDLINE | ID: mdl-25973294

ABSTRACT

Podoplanin overexpression has been reported in various cancers, however, the precise mechanism for podoplanin to promote tumor progression remains elusive. In the present study, podoplanin overexpression was found associated with invasiveness both in OSCC tissues and cell lines. Moreover, the cell invasiveness increased with forced podoplanin expression and decreased when podoplanin was knockdown, indicating podoplanin-mediated cell invasion during OSCC progression. To further identify the role of podoplanin in tumor invasion, cell spreading and immunofluorescence assay were performed firstly. It was found that podoplanin knockdown caused an impaired cell spreading with reduced filopodia and the premature assembly of stress fibers while podoplanin overexpression induced an increase in cellular protrusions and stress fibers with extensive parallel bundles. Then, pull-down assays revealed forced podoplanin expression increased Cdc42 activity and reduced RhoA activity while podoplanin knockdown decreased Cdc42 and increased RhoA markedly. Moreover, a hierarchy of crosstalk between RhoA and Cdc42 was confirmed in podoplanin-mediated cell motility. On the other hand, a significant correlation between podoplanin and MT1-MMP expression in OSCCs was found both in vivo and in vitro, co-located in invasive cells and cellular protrusions. Furthermore, our data showed MT1-MMP knockdown significantly blocked the upregulation of cell motility by forced podoplanin expression, indicating that MT1-MMP played a role in podoplanin-mediated tumor invasion. To further confirm the interaction between RhoA/Cdc42 complex, MT1-MMP and podoplanin, co-precipitation experiments were performed. Both the co-precipitation of podoplanin with MT1-MMP and the podoplanin-induced specific binding of MT1-MMP to Cdc42 were found, and immunofluorescence revealed the co-location of podoplanin, MT1-MMP and Cdc42 at the plasma membrane and filopodia induced an increase in cellular protrusion and stress fibers formation. Moreover, MT1-MMP inhibition could partly rescue the increase of Cdc42 activity caused by forced podoplanin expression. Taken together, our data demonstrated a hierarchy of crosstalk between RhoA and Cdc42 was involved in podoplanin-mediated cytoskeleton remodeling and invasion; the co-location and co-ordination of podoplanin, Cdc42 and MT1-MMP in the invadopodia might induce cytoskeleton remodeling, ECM degradation and tumor invasion, while podoplanin-induced EMT may not be indispensible during OSCC progression.

16.
Biochem Biophys Res Commun ; 452(3): 490-6, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25172658

ABSTRACT

The transcriptional factor Snail has been reported to possess properties related to cancer progression; however, the mechanism for it is not fully understood. Our data showed that Snail knockdown by small interfering RNA in two OSCC cell lines, WSU-HN6 and CAL27, significantly inhibited cell migration and invasion which also resulted in decreased cell motility, such as impaired cell spreading on type I collagen substrate, reduced filopodia, and premature assembly of stress fibers. In addition, Snail-silencing decreased Cdc42 activity but increased RhoA activity, accompanied by the downregulation in both p-ERM expression and cell motility. Meanwhile, endogenous p-ERM was found specifically co-precipitated with activated Cdc42, but not RhoA, and this co-association was decreased by Snail-silencing. The small molecule inhibitors of Rho-associated kinase (Y27632) markedly enhanced Cdc42 activity and the association of p-ERM with activated Cdc42, increasing cell motility remarkably. Using immunohistochemistry, Snail and p-ERM overexpressions were found in OSCC tissues correlated with nodal metastasis and shorter survival. Taken together, these results demonstrate that Snail regulates cell motility through RhoA/Cdc42/p-ERM pathway and may serve as a biomarker to predict prognosis for OSCC patients. Although RhoA and Cdc42 are concurrently regulated downstream of Snail, there is a direct interplay between them, which indicates RhoA has to be inactivated at some point in cell motility cycle.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/genetics , Transcription Factors/genetics , cdc42 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , Aged , Amides/pharmacology , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Mouth Neoplasms/diagnosis , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Phosphorylation , Prognosis , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Snail Family Transcription Factors , Survival Analysis , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
17.
Dermatology ; 228(2): 115-20, 2014.
Article in English | MEDLINE | ID: mdl-24577280

ABSTRACT

The skin excretes substances primarily through sweat glands. Several conditions have been demonstrated to be associated with diminished sweating. However, few studies have concentrated on the metabolism and excretion of sweat. This review focuses on the relationship between temperature and the thermoregulatory efficacy of sweat, and then discusses the excretion of sweat, which includes the metabolism of water, minerals, proteins, vitamins as well as toxic substances. The potential role of sweat secretion in hormone homeostasis and the effects on the defense system of the skin are also clarified.


Subject(s)
Sweat Glands/metabolism , Sweat/metabolism , Sweating/physiology , Body Temperature Regulation/physiology , Humans , Immunoglobulin A, Secretory/metabolism , Immunologic Factors/metabolism , Skin/metabolism , Sweat/chemistry , Sweat/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...