Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Stem Cells ; 42(3): 251-265, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38051601

RESUMO

Human periodontal ligament cells (hPDLCs) cultured from periodontal ligament (PDL) tissue contain postnatal stem cells that can be differentiated into PDL fibroblasts. We obtained PDL fibroblasts from hPDLCs by treatment with low concentrations of TGF-ß1. Since the extracellular matrix and cell surface molecules play an important role in differentiation, we had previously developed a series of monoclonal antibodies against PDL fibroblast-specific cell surface molecules. One of these, the anti-PDL51 antibody, recognized a protein that was significantly upregulated in TGF-ß1-induced PDL fibroblasts and highly accumulated in the PDL region of the tooth root. Mass spectrometry revealed that the antigen recognized by the anti-PDL51 antibody was leucine-rich repeat containing 15 (LRRC15), and this antibody specifically recognized the extracellular glycosylated moiety of LRRC15. Experiments presented here show that as fibroblastic differentiation progresses, increased amounts of LRRC15 localized at the cell surface and membrane. Inhibition of LRRC15 by siRNA-mediated depletion and by antibody blocking resulted in downregulation of the representative PDL fibroblastic markers. Moreover, following LRRC15 inhibition, the directed and elongated cell phenotypes disappeared, and the long processes of the end of the cell body were no longer found. Through a specific interaction between integrin ß1 and LRRC15, the focal adhesion kinase signaling pathway was activated in PDL fibroblasts. Furthermore, it was shown that increased LRRC15 was important for the activation of the integrin-mediated cell adhesion signal pathway for regulation of cellular functions, including fibroblastic differentiation, proliferation, and cell migration arising from the expression of PDL-related genes in TGF-ß1-induced PDL fibroblastic differentiation.


Assuntos
Ligamento Periodontal , Fator de Crescimento Transformador beta1 , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Adesão Celular , Leucina/metabolismo , Proliferação de Células , Diferenciação Celular , Transdução de Sinais , Fibroblastos/metabolismo , Integrinas/metabolismo , Células Cultivadas , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
2.
Stem Cells ; 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32930424

RESUMO

TGF-ß and Wnt/ß-catenin signaling pathways are known to be essential for the development of periodontal tissue. In this study, we examined the crosstalk between TGF-ß and Wnt/ß-catenin signaling in ligament-fibroblastic differentiation of human periodontal ligament cells (hPDLCs). TGF-ß1 treatment significantly increased the expression of ligament-fibroblastic markers, but such expression was preventing by treatment with SB431542, a TGF-ß type I receptor inhibitor. As well as phosphorylation of Smad3, TGF-ß1 increased ß-catenin activation. The depletion of ß-catenin reduced the expression of ligament-fibroblastic markers, suggesting that ß-catenin is essential for ligament differentiation. The effect of TGF-ß1 on ß-catenin activation did not seem to be much correlated with Wnt stimuli, but endogenous DKK1 was suppressed by TGF-ß1, indicating that ß-catenin activation could be increased much more by TGF-ß1. In addition to DKK1 suppression, Smad3 phosphorylation by TGF-ß1 facilitated the nuclear translocation of cytoplasmic ß-catenin. In contrast to ligament-fibroblastic differentiation, inhibition of TGF-ß1 signaling was needed for cementoblastic differentiation of hPDLCs. BMP7 treatment accompanied by inhibition of TGF-ß1 signaling had a synergistic effect on cementoblastic differentiation. In conclusion, ß-catenin activation by TGF-ß1 caused ligament-fibroblastic differentiation of hPDLCs, and the presence of TGF-ß1 stimuli basically determined whether hPDLCs are differentiated into ligament progenitor or cementoblasts.

3.
J Cell Physiol ; 232(10): 2818-2828, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27861885

RESUMO

Polo-like kinase 1 (Plk1), a conserved Ser/Thr mitotic kinase, has been identified as a promising target for anticancer drug development because its overexpression is correlated with malignancy. Here, we found that genistein, an isoflavone, inhibits Plk1 kinase activity directly. Previously the mitotic disturbance phenomenon induced by treatment with genistein was not fully explained by its inhibitory effect on EGFR. In kinase profiling assays, it showed selectivity relative to a panel of kinases, including EGFR. Treatment with genistein induced cell death in a concentration-dependent manner in cancer cells from diverse tissue origins, but not in non-transformed cells such as hTERT-RPE or MCF10A cells. We also observed that genistein tended to be more selective against cancer cells with mutations in the TP53 gene. TP53-depeleted LNCaP and NCI-H460 cells using shRNA targeting human TP53 were more sensitive to cell death by treatment of genistein. Furthermore, genistein induced mitotic arrest by inhibiting Plk1 activity and, consequently, led to mitotic catastrophe and apoptosis. These data suggest that genistein may be a promising anticancer drug candidate due to its inhibitory activity against Plk1 as well as EGFR and effectiveness toward cancer cells, especially those with p53-mutation. J. Cell. Physiol. 232: 2818-2828, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Antineoplásicos Hormonais/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Genisteína/farmacologia , Mutação , Neoplasias/tratamento farmacológico , Fitoestrógenos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células HCT116 , Células Hep G2 , Humanos , Concentração Inibidora 50 , Células MCF-7 , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Quinase 1 Polo-Like
4.
Protein Expr Purif ; 135: 37-44, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28487257

RESUMO

Fibroblast growth factor (FGF) is a multifunctional growth factor that induces cell proliferation, survival, migration, and differentiation in various cell types and tissues. With these biological functions, FGF-2 has been evaluated for clinical use in the regeneration of damaged tissues. The expression of hFGF-2 in Escherichia coli and a purification system using the immobilized metal affinity chromatography (IMAC) is well established to generate a continuous supply of FGF-2. Although hexa-histidine tag (H6) is commonly used for IMAC purification, hexa-histidine-asparagine tag (HN6) is also efficient for purification as it is easily exposed on the surface of the protein. In this study, four different tagging constructs of hFGF-2 based on tag positions and types (H6-FGF2, FGF2-H6, HN6-FGF2, and FGF2-HN6) were designed and expressed under the inducible T7 expression system in E. coli. The experimental conditions of expression and purification of each recombinant protein were optimized. The effective dosages of the recombinant proteins were determined based on the increase of cell proliferation in human gingival fibroblast. ED50s of H6-FGF2, FGF2-H6, HN6-FGF2, and FGF2-HN6 were determined (4.42 ng/ml, 3.55 ng/ml, 3.54 ng/ml, and 4.14 ng/ml, respectively) and found to be comparable to commercial FGF-2 (3.67 ng/ml). All the recombinant hFGF-2s inhibit the osteogenic induction and mineralization in human periodontal ligament-derived cells. Our data suggested that biological activities of the recombinant hFGF-2 are irrelevant to types and positions of tags, but may have an influence on the expression efficiency and solubility.


Assuntos
Escherichia coli/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fibroblastos/efeitos dos fármacos , Vetores Genéticos/metabolismo , Osteoblastos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/biossíntese , Asparagina/metabolismo , Bacteriófago T7/genética , Bacteriófago T7/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cromatografia de Afinidade , Clonagem Molecular , Cemento Dentário/citologia , Cemento Dentário/efeitos dos fármacos , Cemento Dentário/metabolismo , Escherichia coli/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Vetores Genéticos/química , Gengiva/citologia , Gengiva/efeitos dos fármacos , Gengiva/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Ligamento Periodontal/citologia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Cultura Primária de Células , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
5.
Biochem Biophys Res Commun ; 478(2): 631-6, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27476491

RESUMO

ELP3, a core component of Elongator, has been implicated in translational regulation via modification of tRNA at the wobble position. However, the precise biological function of ELP3 in early mouse development has not yet been defined. We here provide evidence that ELP3 plays crucial roles in mouse embryonic stem cell (ESC) maintenance and early development. ELP3 was detected ubiquitously in blastocysts and E10.5 embryos and shown to be increased during ESC differentiation. Depletion of ELP3 in ESC led to aberrant cell cycle progression, along with reduced expression of genes for pluripotency. Interestingly, our analyses revealed that, although the mRNA levels of the genes related to cell cycle were increased, protein levels were diminished in knockdown (KD) ESCs. The data, therefore, suggest that ELP3 function is critical for translational efficiency of the genes. Consistent with a proliferation defect in KD cells, Elp3 knockout (KO) embryos suffered from severe growth retardation and failed to develop beyond E12.5. In conclusion, we have demonstrated that ELP3 plays an indispensable role in ESC survival, differentiation and embryonic development in mouse.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histona Acetiltransferases/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Animais , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Perda do Embrião/genética , Perda do Embrião/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Técnicas de Silenciamento de Genes , Histona Acetiltransferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Embrionárias Murinas/metabolismo
6.
Biochem Cell Biol ; 93(4): 298-305, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25789782

RESUMO

Basic fibroblast growth factor (bFGF or FGF-2) is widely used to modulate the proliferation and differentiation of certain cell types. An expression and purification system for recombinant human FGF-2 in Escherichia coli was established for the purpose of securing a continuous supply of this protein. The purified recombinant FGF-2 significantly increased the population of human embryonic stem cells. The optimal concentrations of FGF-2 for cell proliferative induction in various adult stem cells including human dental pulp stem cells, full term human periodontal ligament stem cells, human gingival fibroblasts, mesenchymal stem cells, and osteogenic oseosarcoma were established in a dose-dependent manner. When cells were treated with recombinant FGF-2 for 6 days before osteogenic induction, the mRNA expression of the bone markers was upregulated in cells originated from human dental pulp tissue, indicating that pretreatment with FGF-2 during culture increase stem cell/progenitor population and osteogenic potential.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Polpa Dentária/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Proteínas Recombinantes/farmacologia , Células-Tronco/citologia
7.
Stem Cells ; 32(10): 2626-41, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24898727

RESUMO

B-Cell receptor-associated protein 31 (BAP31) regulates the export of secreted membrane proteins from the endoplasmic reticulum (ER) to the downstream secretory pathway. Previously, we generated a monoclonal antibody 297-D4 against the surface molecule on undifferentiated human embryonic stem cells (hESCs). Here, we found that 297-D4 antigen was localized to pluripotent hESCs and downregulated during early differentiation of hESCs and identified that the antigen target of 297-D4 was BAP31 on the hESC-surface. To investigate the functional role of BAP31 in hESCs, BAP31 expression was knocked down by small interfering RNA. BAP31 depletion impaired hESC self-renewal and pluripotency and drove hESC differentiation into multicell lineages. BAP31 depletion hindered hESC proliferation by arresting cell cycle at G0/G1 phase and inducing caspase-independent cell death. Interestingly, BAP31 depletion reduced hESC adhesion to extracellular matrix (ECM). Analysis of cell surface molecules showed decreased expression of epithelial cell adhesion molecule (EpCAM) in BAP31-depleted hESCs, while ectopic expression of BAP31 elevated the expression of EpCAM. EpCAM depletion also reduced hESC adhesion to ECM, arrested cell cycle at G0/G1 phase and induced cell death, producing similar effects to those of BAP31 depletion. BAP31 and EpCAM were physically associated and colocalized at the ER and cell surface. Both BAP31 and EpCAM depletion decreased cyclin D1 and E expression and suppressed PI3K/Akt signaling, suggesting that BAP31 regulates hESC stemness and survival via control of EpCAM expression. These findings provide, for the first time, mechanistic insights into how BAP31 regulates hESC stemness and survival via control of EpCAM expression.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Antígenos/metabolismo , Caspases/metabolismo , Adesão Celular , Morte Celular , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula , Proliferação de Células , Sobrevivência Celular , Regulação para Baixo , Molécula de Adesão da Célula Epitelial , Matriz Extracelular/metabolismo , Fase G1 , Humanos , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fase de Repouso do Ciclo Celular , Transdução de Sinais
8.
Stem Cells ; 31(12): 2647-58, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23495120

RESUMO

Self-renewal and pluripotency of human embryonic stem cells (hESCs) are a complex biological process for maintaining hESC stemness. However, the molecular mechanisms underlying these special properties of hESCs are not fully understood. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) is a multifunctional RNA-binding protein whose expression is related to cell proliferation and carcinogenesis. In this study, we found that hnRNP A2/B1 expression was localized to undifferentiated hESCs and decreased upon differentiation of hESCs. hnRNP A2/B1 knockdown reduced the number of alkaline phosphatase-positive colonies in hESCs and led to a decrease in the expression of pluripotency-associated transcription factors OCT4, NANOG, and SOX2, indicating that hnRNP A2/B1 is essential for hESC self-renewal and pluripotency. hnRNP A2/B1 knockdown increased the expression of gene markers associated with the early development of three germ layers, and promoted the process of epithelial-mesenchymal transition, suggesting that hnRNP A2/B1 is required for maintaining the undifferentiated and epithelial phenotypes of hESCs. hnRNP A2/B1 knockdown inhibited hESC proliferation and induced cell cycle arrest in the G0/G1 phase before differentiation via degradation of cyclin D1, cyclin E, and Cdc25A. hnRNP A2/B1 knockdown increased p27 expression and induced phosphorylation of p53 and Chk1, suggesting that hnRNP A2/B1 also regulates the G1/S transition of hESC cell cycle through the control of p27 expression and p53 and Chk1 activity. Analysis of signaling molecules further revealed that hnRNP A2/B1 regulated hESC proliferation in a PI3K/Akt-dependent manner. These findings provide for the first time mechanistic insights into how hnRNP A2/B1 regulates hESC self-renewal and pluripotency.


Assuntos
Células-Tronco Embrionárias/citologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Técnicas de Cocultura , Regulação para Baixo , Células-Tronco Embrionárias/metabolismo , Fibroblastos/citologia , Fase G1/fisiologia , Técnicas de Silenciamento de Genes , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Humanos , Camundongos , Células-Tronco Pluripotentes/metabolismo , Fase S/fisiologia , Transfecção
9.
Nephron Exp Nephrol ; 126(3): 127-40, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24863135

RESUMO

BACKGROUND: Endoplasmic reticulum (ER) stress has been implicated in inducing epithelial-mesenchymal transition (EMT). ER stress is also known to induce autophagy. However, it is unclear whether ER stress-induced autophagy contributes to EMT. We hypothesized that ER stress might induce EMT through autophagy via activation of c-Src kinase in tubular epithelial cells. METHOD: All experiments were performed using HK-2 cells. Protein expression was measured by Western blot analysis. Immunofluorescence and small interfering RNA (siRNA) experiments were performed. RESULTS: Chemical ER stress inducers such as tunicamycin (TM, 0.2 µM) and thapsigargin (TG, 0.2 µM) induced EMT, as shown by upregulation of α-smooth muscle actin and downregulation of E-cadherin. ER stress inhibitors such as 4-PBA and salubrinal suppressed both TM- and TG-induced EMT. TM and TG also induced autophagy, as evidenced by upregulation of LC3-II and beclin-1, which were abolished by pretreatment with ER stress inhibitors. Transfection with siRNA targeting ER stress protein (IRE-1) blocked the TM- or TG-induced EMT and autophagy. Autophagy inhibitors such as 3-methyladenine and bafilomycin inhibited the TM- or TG-induced EMT. Transfection with siRNA targeting autophagy protein (beclin-1) also blocked the TM- or TG-induced EMT. Both TM and TG induced activation of c-Src kinase. Inhibitor of c-Src kinase (PP2) suppressed the TM- or TG-induced autophagy and EMT. CONCLUSION: ER stress by TM or TG induced EMT through autophagy via activation of c-Src kinase in tubular epithelial cells.


Assuntos
Autofagia/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Túbulos Renais Proximais/enzimologia , Quinases da Família src/metabolismo , Autofagia/efeitos dos fármacos , Proteína Tirosina Quinase CSK , Linhagem Celular , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Quinases da Família src/antagonistas & inibidores
10.
Biochem Biophys Res Commun ; 436(1): 12-8, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23643811

RESUMO

Cdc20 is an activator of the anaphase-promoting complex (APC/C), and APC/C(Cdc20) is essential for metaphase-anaphase transition. To allow progression beyond mitosis, Cdc20 is degraded through KEN-box-dependent APC/C(Cdh1) activity. Mammalian Cdc20 contains the CRY box, a second APC/C(Cdh1)-dependent degron, but the molecular mechanism in degradation process remains undefined. Polo-like kinase-1 (Plk1) is an essential mitotic kinase regulating various targets in kinetochore, centrosome, and midbody for proper mitotic progression. Plk1 directly bound to Cdc20 and phosphorylates it on serine-170 located in CRY-box. Whereas wild-type Cdc20 was degraded according to progress cell cycle beyond mitosis, the phosphorylation-defective mutant, which serine-170 was changed into alanine, was not destroyed in early G1 phase. The phosphorylation on serine-170 by Plk1 was important for ubiquitination and Cdh1-dependent proteolysis. However, this modification by Plk1 on CRY box had no effect on the subcellular localization of Cdc20 and the formation of APC/C-inhibitory checkpoint complexes under spindle assembly checkpoint. This mechanism will be the first finding of inhibitory phosphorylation related to Cdc20 instability.


Assuntos
Caderinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Antígenos CD , Proteínas Cdc20 , Ciclo Celular/fisiologia , Fase G1 , Regulação Enzimológica da Expressão Gênica , Inativação Gênica , Células HeLa , Humanos , Microscopia Confocal/métodos , Mitose , Mutação , Fosforilação , Ubiquitina/metabolismo , Quinase 1 Polo-Like
11.
Yeast ; 30(10): 379-94, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23640764

RESUMO

Under nutritional deprivation caused by prolonged culture, actively growing cells prepare to enter stationary phase. We showed here that Sds23/Psp1/Moc1 was phosphorylated by both cAMP-dependent kinase and stress-activated MAP kinase Sty1 upon entry into stationary phase. Overexpression of the phosphorylation-defective mutant Sds23/Psp1/Moc1 induced cell death in prolonged culture and blocked re-entry into the cell division cycle. These phosphorylations are likely to be required for cell survival during stationary phase and for binding of Ufd2, a Schizosaccharomyces pombe homologue of multi-ubiquitin chain assembly factor E4. Deletion of the Ufd2 gene and overexpression of Sds23/Psp1/Moc1 increased cell viability in prolonged stationary phase. These results suggested that Ufd2 induces cell death in prolonged nutrient deprivation, that Sds23/Psp1/Moc1 may be a target protein of the ubiquitin-fusion degradation pathway for regulation of cell viability under this stress condition, and that Sty1 and PKA activity in stationary phase is essential for interaction between Sds23/Psp1/Moc1 and Ufd2.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Viabilidade Microbiana , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/citologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Deleção de Genes , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Serina/metabolismo , Estresse Fisiológico
12.
Stem Cells Int ; 2023: 5924286, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37396953

RESUMO

Human periodontal ligament stem cells (hPDLSCs) contain multipotent postnatal stem cells that differentiate into PDL progenitors, osteoblasts, and cementoblasts. Previously, we obtained cementoblast-like cells from hPDLSCs using bone morphogenetic protein 7 (BMP7) treatment. Differentiation into appropriate progenitor cells requires interactions and changes between stem or progenitor cells and their so-called environment niches, and cell surface markers play an important role. However, cementoblast-specific cell surface markers have not yet been fully studied. Through decoy immunization with intact cementoblasts, we developed a series of monoclonal antibodies against cementoblast-specific membrane/extracellular matrix (ECM) molecules. One of these antibodies, the anti-CM3 antibody, recognized an approximate 30 kDa protein in a mouse cementoblast cell line, and the CM3 antigenic molecule accumulated in the cementum region of human tooth roots. Using mass spectrometric analysis, we found that the antigenic molecules recognized by the anti-CM3 antibody were galectin-3. As cementoblastic differentiation progressed, the expression of galectin-3 increased, and it localized at the cell surface. Inhibition of galectin-3 via siRNA and a specific inhibitor showed the complete blockage of cementoblastic differentiation and mineralization. In contrast, ectopic expression of galectin-3 induced cementoblastic differentiation. Galectin-3 interacted with laminin α2 and BMP7, and these interactions were diminished by galectin-3 inhibitors. These results suggested that galectin-3 participates in binding to the ECM component and trapping BMP7 to induce, in a sustained fashion, the upregulation of cementoblastic differentiation. Finally, galectin-3 could be a potential cementoblast-specific cell surface marker, with functional importance in cell-to-ECM interactions.

13.
J Biol Chem ; 286(38): 33012-20, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21813642

RESUMO

Polo-like kinase-1 (Plk1) is essential for progression of mitosis and localizes to centrosomes, central spindles, midbody, and kinetochore. Ran, a small GTPase of the Ras superfamily, plays a role in microtubule dynamics and chromosome segregation during mitosis. Although Ran-binding protein-1 (RanBP1) has been reported as a regulator of RanGTPase for its mitotic functions, the action mechanism between Ran and RanBP1 during mitosis is still unknown. Here, we demonstrated in vitro and in vivo phosphorylation of RanBP1 by Plk1 as well as the importance of phosphorylation of RanBP1 in the interaction between Plk1 and Ran during early mitosis. Both phosphorylation-defective and N-terminal deletion mutant constructs of RanBP1 disrupted the interaction with Ran, and depletion of Plk1 also disrupted the formation of a complex between Ran and RanBP1. In addition, the results from both ectopic expression of phosphorylation-defective mutant construct and a functional complementation on RanBP1 deficiency with this mutant indicated that phosphorylation of RanBP1 by Plk1 might be crucial to microtubule nucleation and spindle assembly during mitosis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mitose , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Ciclina B1/metabolismo , Células HeLa , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Nucleares/química , Fosforilação , Fosfotreonina/metabolismo , Ligação Proteica , Transporte Proteico , Quinase 1 Polo-Like
14.
Biochem Biophys Res Commun ; 423(3): 593-9, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22704936

RESUMO

DNA damage induces multiple checkpoint pathways to arrest cell cycle progression until damage is repaired. In our previous reports, when DNA damage occurred in prometaphase, cells were accumulated in 4 N-DNA G1 phase, and mitosis-specific kinases were inactivated in dependent on ATM/Chk1 after a short incubation for repair. We investigated whether or not mitotic DNA damage causes cells to skip-over late mitotic periods under prolonged incubation in a time-lapse study. 4 N-DNA-damaged cells re-replicated without cell division and accumulated in 8 N-DNA content, and the activities of apoptotic factors were increased. The inhibition of DNA replication reduced the 8 N-DNA cell population dramatically. Induction of replication without cell division was not observed upon depletion of Chk1 or ATM. Finally, mitotic DNA damage induces mitotic slippage and that cells enter G1 phase with 4 N-DNA content and then DNA replication is occurred to 8 N-DNA content before completion of mitosis in the ATM/Chk1-dependent manner, followed by caspase-dependent apoptosis during long-term repair.


Assuntos
Dano ao DNA , Replicação do DNA/genética , Mitose/genética , Neoplasias/genética , Neoplasias/patologia , Apoptose/genética , Divisão Celular , Linhagem Celular Tumoral , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Células HeLa , Humanos
15.
Genes (Basel) ; 13(4)2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35456465

RESUMO

Ligament-fibroblastic cells and cementoblasts, two types of progenitor cells that differentiate from periodontal ligament stem cells (hPDLSCs), are responsible for the formation of the adhesive tissues in the tooth root. Since one of the factors that determines the fate of stem cell differentiation is the change in the microenvironment of the stem/progenitor cells, this study attempted to compare and analyze the molecular differences in the membrane and ECM of the two progenitor cells. Single cells derived from hPDLSCs were treated with TGF-ß1 and BMP7 to obtain ligament-fibroblastic and cementoblastic cells, respectively. The transcriptome profiles of three independent replicates of each progenitor were evaluated using next-generation sequencing. The representative differentially expressed genes (DEGs) were verified by qRT-PCR, Western blot analysis, and immunohistochemistry. Among a total of 2245 DEGs identified, 142 and 114 DEGs related to ECM and cell membrane molecules were upregulated in ligament-fibroblastic and cementoblast-like cells, respectively. The major types of integrin and cadherin were found to be different between the two progenitor cells. In addition, the representative core proteins for each glycosaminoglycan-specific proteoglycan class were different between the two progenitors. This study provides a detailed understanding of cell-cell and cell-ECM interactions through the specific components of the membrane and ECM for ligament-fibroblastic and cementoblastic differentiation of hPDLSCs.


Assuntos
Cemento Dentário , Ligamento Periodontal , Diferenciação Celular/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Ligamentos , Ligamento Periodontal/metabolismo , Transcriptoma/genética
16.
Stem Cells Int ; 2022: 3273779, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35879965

RESUMO

Human periodontal ligament stem cells (hPDLSCs) can be differentiated into periodontal ligament- (PDL-) fibroblastic progenitors by treatment with low concentrations of transforming growth factor beta 1 (TGF-ß1). Although much is known about the profibrotic effects of TGF-ß1, the molecular mechanisms mediating the activation of fibroblasts in periodontal ligament-fibroblastic differentiation are not well known. Our study was to investigate the mechanism of the fibroblastic process in the periodontal ligament differentiation of hPDLSCs through the discovery of novel markers. One of the monoclonal antibodies previously established through decoy immunization was the anti-LG11 antibody, which recognized Golgi subfamily A member 5 (GOLGA5) as a PDL-fibroblastic progenitor-specific antigen. GOLGA5/LG11 was significantly upregulated in TGF-ß1-induced PDL-fibroblastic progenitors and accumulated in the PDL region of the tooth root. GOLGA5 plays a role in vesicle tethering and docking between the endoplasmic reticulum and the Golgi apparatus. siRNA-mediated depletion of endogenous GOLGA5 upregulated in TGF-ß1-induced PDL-fibroblastic progenitors resulted in downregulation of representative PDL-fibroblastic markers and upregulation of osteoblast markers. When the TGF-ß1 signaling pathway was blocked or GOLGA5 was depleted by siRNA, the levels of extracellular matrix (ECM) proteins, such as type I collagen and fibronectin, decreased in PDL-fibroblastic progenitors. In addition, Golgi structures in the perinuclear region underwent fragmentation under these conditions. These results suggest that GOLGA5/LG11 is a PDL-fibroblastic marker with functional importance in ECM protein production and secretion, which are important processes in PDL-fibroblastic differentiation.

17.
Stem Cells Dev ; 31(21-22): 684-695, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35859453

RESUMO

Primary dental pulp cells can be differentiated into odontoblast-like cells, which are responsible for dentin formation and mineralization. Successful differentiation of primary dental pulp cells can be verified using a few markers. However, odontoblast-specific cell surface markers have not been fully studied yet. LEucine PRoline-Enriched Proteoglycan 1 (LEPRE1) is a basement membrane-associated proteoglycan. LEPRE1 protein levels are increased during odontoblastic differentiation of human dental pulp cells (hDPCs). Intracellular and cell surface accumulation of this protein completely disappeared during dentin maturation and mineralization. Cell surface binding of an anti-LEPRE1 monoclonal antibody that could recognize an extracellular region was gradually increased in the odontoblastic stage. Overexpression and knockdown experiments showed that accumulation of intracellular LEPRE1 could lead to inefficient odontoblastic differentiation and that the movement of LEPRE1 from intracellular region to the cell surface was required for odontoblastic differentiation. Indeed, when LEPRE1 already located on the cell surface was blocked by the anti-LEPRE1 monoclonal antibody, odontoblastic differentiation of hDPCs was inhibited. In this study, we looked at other aspects of LEPRE1 function as a cell surface molecule rather than its known intracellular hydroxylase activity. Our results indicate that this protein has potential as a specific cell surface marker in odontoblastic differentiation.


Assuntos
Polpa Dentária , Glicoproteínas de Membrana , Prolil Hidroxilases , Proteoglicanas , Humanos , Anticorpos Monoclonais/farmacologia , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas da Matriz Extracelular/metabolismo , Odontoblastos , Fosfoproteínas/metabolismo , Proteoglicanas/metabolismo , Células-Tronco , Glicoproteínas de Membrana/metabolismo , Prolil Hidroxilases/metabolismo
18.
Stem Cells Int ; 2021: 9326298, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34512768

RESUMO

Human dental pulp stem cells (hDPSCs) are the primary cells responsible for dentin regeneration. Typically, in order to allow for odontoblastic differentiation, hDPSCs are cultured over weeks with differentiation-inducing factors in a typical monolayered culture. However, monolayered cultures have significant drawbacks including inconsistent differentiation efficiency, require a higher BMP concentration than should be necessary, and require periodic treatment with BMPs for weeks to see results. To solve these problems, we developed a 3D-cell spheroid culture system for odontoblastic differentiation using microparticles with leaf-stacked structure (LSS), which allow for the sustained release of BMPs and adequate supply of oxygen in cell spheroids. BMPs were continuously released and maintained an effective concentration over 37 days. hDPSCs in the spheroid maintained their viability for 5 weeks, and the odontoblastic differentiation efficiency was increased significantly compared to monolayered cells. Finally, dentin-related features were detected in the spheroids containing BMPs-loaded microparticles after 5 weeks, suggesting that these hDPSC-LSS spheroids might be useful for dentin tissue regeneration.

19.
Stem Cell Res Ther ; 11(1): 210, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32493499

RESUMO

BACKGROUND: The differentiation of human mesenchymal stem cells (hMSCs) into osteoblasts (OBs) is a prerequisite for bone formation. However, little is known about the definitive surface markers for OBs during osteogenesis. METHODS: To study the surface markers on OBs, we generated and used monoclonal antibodies (MAbs) against surface molecules on transforming growth factor-ß1 (TGF-ß1)-treated cancer cells. The generated MAbs were further selected toward expression changes on hMSCs cultured with TGF-ß1/bone morphogenetic protein-2 (BMP-2) or osteogenic differentiation medium (ODM) by flow cytometry. Immunoprecipitation and mass spectrometry were performed to identify target antigens of selected MAbs. Expression changes of the target antigens were evaluated in hMSCs, human periodontal ligament cells (hPDLCs), and human dental pulp cells (hDPCs) during osteogenic and adipogenic differentiation by quantitative polymerase chain reaction (qPCR) and flow cytometry. hMSCs were also sorted by the MAbs using magnetic-activated cell sorting system, and osteogenic potential of sorted cells was evaluated via Alizarin Red S (ARS) staining and qPCR. RESULTS: The binding reactivity of MR14-E5, one of the MAbs, was downregulated in hMSCs with ODM while the binding reactivity of ER7-A7, ER7-A8, and MR1-B1 MAbs was upregulated. Mass spectrometry and overexpression identified that MR14-E5, ER7-A7/ER7-A8, and MR1-B1 recognized integrin α2, α3, and αV, respectively. Upon osteogenic differentiation of hMSCs, the expression of integrin α2 was drastically downregulated, but the expression of integrin α3 and αV was upregulated in accordance with upregulation of osteogenic markers. Expression of integrin α3 and αV was also upregulated in hPDLCs and hDPCs during osteogenic differentiation. Cell sorting showed that integrin αV-high hMSCs have a greater osteogenic potential than integrin αV-low hMSCs upon the osteogenic differentiation of hMSCs. Cell sorting further revealed that the surface expression of integrin αV is more dramatically induced even in integrin αV-low hMSCs. CONCLUSION: These findings suggest that integrin α3 and αV induction is a good indicator of OB differentiation. These findings also shed insight into the expression dynamics of integrins upon osteogenic differentiation of hMSCs and provide the reason why different integrin ligands are required for OB differentiation of hMSCs.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Diferenciação Celular , Células Cultivadas , Humanos , Integrina alfa2 , Osteoblastos
20.
J Oral Pathol Med ; 38(7): 591-6, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19453842

RESUMO

BACKGROUND: The human oral cells have different physiological properties and different origins in developmental stages. Mechanical, physiological, and chemical stress can cause damage and irritation during clinical treatment in various oral tissues. PURPOSE: The effects of DNA damage response and gene silencing of checkpoint kinases (Chk1/2) is not unclear in oral primary and cancer cells. METHOD: Treatment with doxorubicin involving DNA damage and gene silencing of Chk1/2 by shRNA constructs was performed in pulp, periodontal ligament, gingival tissues (HGF), and mouth epithelial carcinoma cells (KB). RESULTS: The KB cells were more sensitive to genotoxic stress response than oral primary cells. Endogenous levels of Chk1/2 in KB cell were higher than in pulp cells. When doxorubicin was administered, Chk2 activation was induced in KB cell, but not in pulp cells. However, viability in KB cells did not decrease by the suppression of the checkpoint proteins, whereas primary cells were defective in gene silencing. When doxorubicin treatment and gene silencing were combined, both primary cells and KB cell were defective. Moreover, in case of KB cell, cell death was increased and activation of Chk2 was increased in doxorubicin dose-dependent. CONCLUSION: These data indicate that not only stress response mechanism may be different in oral primary and cancer cells but also Chk1/2 proteins may have essential roles in oral primary cells. Based on these data, checkpoint proteins may be crucial drug targets for oral cancer therapy.


Assuntos
Carcinoma/enzimologia , Citotoxinas/toxicidade , Doxorrubicina/toxicidade , Fibroblastos/enzimologia , Neoplasias Bucais/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Carcinoma/tratamento farmacológico , Carcinoma/genética , Carcinoma/patologia , Sobrevivência Celular , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Dano ao DNA/fisiologia , Polpa Dentária/citologia , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/enzimologia , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Inativação Gênica , Genes cdc/efeitos dos fármacos , Genes cdc/fisiologia , Gengiva/efeitos dos fármacos , Gengiva/enzimologia , Humanos , Boca/citologia , Boca/efeitos dos fármacos , Boca/enzimologia , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/enzimologia , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA