Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Connect Tissue Res ; 62(6): 689-697, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33334200

RESUMO

Purpose: In our previous study, we demonstrated that hyaluronan induces odontoblastic differentiation of dental pulp stem cells via interactions with CD44. However, it remains unclear whether CD44 expression by dental pulp stem cells is required for odontoblastic differentiation.Methods: We searched for a compound other than hyaluronan that induces odontoblastic differentiation of dental pulp stem cells and used western blotting to determine whether CD44 is involved in the induction of odontoblastic differentiation by the compound. We further validated the cell signaling details of the compound-induced expression of dentin sialophosphoprotein (DSPP), which is known as a marker of odontoblastic differentiation.Results: We investigated shikonin, which is one of the derivatives of naphthoquinone, the skeleton of vitamin K. Shikonin-induced expression of DSPP was inhibited by PI3K, AKT, and mTOR inhibitors. Additionally, shikonin-induced expression of DSPP was inhibited in dental pulp stem cells transfected with siRNA against CD44.Conclusions: Shikonin can stimulate dental pulp stem cells to undergo odontoblastic differentiation through a mechanism involving the AKT-mTOR signaling pathway and CD44. Although expression of CD44 is important for inducing odontoblastic differentiation of dental pulp stem cells, the relationship between the AKT-mTOR signaling pathway and CD44 expression, in the context of shikonin stimulation, has not yet been elucidated. This study suggests that shikonin may be useful for inducing odontoblastic differentiation of dental pulp stem cells, and that it may have clinical applications, including protection of the dental pulp.


Assuntos
Naftoquinonas , Odontoblastos , Diferenciação Celular , Células Cultivadas , Polpa Dentária/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Ácido Hialurônico/metabolismo , Naftoquinonas/metabolismo , Naftoquinonas/farmacologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células-Tronco , Serina-Treonina Quinases TOR/metabolismo
2.
Cancer Sci ; 108(11): 2273-2280, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28869796

RESUMO

We previously reported that eribulin mesylate (eribulin), a tubulin-binding drug (TBD), could remodel tumor vasculature (i.e. increase tumor vessels and perfusion) in human breast cancer xenograft models. However, the role of this vascular remodeling in antitumor effects is not fully understood. Here, we investigated the effects of eribulin-induced vascular remodeling on antitumor activities in multiple human cancer xenograft models. Microvessel densities (MVD) were evaluated by immunohistochemistry (CD31 staining), and antitumor effects were examined in 10 human cancer xenograft models. Eribulin significantly increased MVD compared to the controls in six out of 10 models with a correlation between enhanced MVD levels and antitumor effects (R2  = 0.54). Because of increased MVD, we next used radiolabeled liposomes to examine whether eribulin treatment would result in increased tumoral accumulation levels of these macromolecules and, indeed, we found that eribulin, unlike vinorelbine (another TBD) enhanced them. As eribulin increased accumulation of radiolabeled liposomes, we postulated that this treatment might enhance the antitumor effect of Doxil (a liposomal anticancer agent) and facilitate recruitment of immune cells into the tumor. As expected, eribulin enhanced antitumor activity of Doxil in a post-erlotinib treatment H1650 (PE-H1650) xenograft model. Furthermore, infiltrating CD11b-positive immune cells were significantly increased in multiple eribulin-treated xenografted tumors, and natural killer (NK) cell depletion reduced the antitumor effects of eribulin. These findings suggest a contribution of the immune cells for antitumor activities of eribulin. Taken together, our results suggest that vascular remodeling induced by eribulin acts as a microenvironment modulator and, consequently, this alteration enhanced the antitumor effects of eribulin.


Assuntos
Furanos/administração & dosagem , Cetonas/administração & dosagem , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Remodelação Vascular/efeitos dos fármacos , Animais , Doxorrubicina/administração & dosagem , Doxorrubicina/análogos & derivados , Feminino , Células HCT116 , Humanos , Camundongos , Neoplasias/patologia , Polietilenoglicóis/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cancer Sci ; 106(2): 201-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25458359

RESUMO

Almost all cancers show intrinsic and/or evasive resistance to vascular endothelial growth factor (VEGF) inhibitors by multiple mechanisms. Serum angiopoietin-2 (Ang2) level has been proposed as a potential biomarker of VEGF inhibitor response in several cancers. From these clinical observations, the Ang2 and Tie2 (its receptor) axis has been focused on as a promising target. Here, we show a novel strategy to circumvent the resistance by combining multi-tyrosine kinase inhibitors lenvatinib (VEGF receptor, fibroblast growth factor receptor, and RET inhibitor) and golvatinib (E7050; c-Met, Tie2, and EphB4 inhibitor). Tie2 identifies a highly pro-angiogenic macrophage subset, Tie2-expressing macrophages (TEM). Angi-Tie2 and EphB4-EphrinB2 signaling plays critical roles in pericyte-mediated vessel stabilization. In vitro analyses suggested that golvatinib combined with lenvatinib inhibited pericyte-mediated vessel stabilization and TEM differentiation. In thyroid and endometrial cancer models, golvatinib and lenvatinib inhibited pericyte network development and TEM infiltration, resulting in severe perfusion disorder and massive apoptosis. Body weight loss was tolerable, and no macroscopic change was observed. These preclinical studies suggest that modulation of the tumor microenvironment by a strategic and well-tolerated combination of multi-targeting tyrosine kinase inhibitors may sensitize cancer to VEGF inhibitors.


Assuntos
Inibidores da Angiogênese/farmacologia , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/farmacologia , Angiopoietina-2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Efrina-B2/metabolismo , Feminino , Humanos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor EphB4/metabolismo , Receptor TIE-2/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Cancer Sci ; 105(6): 723-30, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24689876

RESUMO

Vascular endothelial growth factor receptor (VEGFR) inhibitors are approved for the treatment of several tumor types; however, some tumors show intrinsic resistance to VEGFR inhibitors, and some patients develop acquired resistance to these inhibitors. Therefore, a strategy to overcome VEGFR inhibitor resistance is urgently required. Recent reports suggest that activation of the hepatocyte growth factor (HGF) pathway through its cognate receptor, Met, contributes to VEGFR inhibitor resistance. Here, we explored the effect of the HGF/Met signaling pathway and its inhibitors on resistance to lenvatinib, a VEGFR inhibitor. In in vitro experiments, addition of VEGF plus HGF enhanced cell growth and tube formation of HUVECs when compared with stimulation by either factor alone. Lenvatinib potently inhibited the growth of HUVECs induced by VEGF alone, but cells induced by VEGF plus HGF showed lenvatinib resistance. This HGF-induced resistance was cancelled when the Met inhibitor, golvatinib, was added with lenvatinib. Conditioned medium from tumor cells producing high amounts of HGF also conferred resistance to inhibition by lenvatinib. In s.c. xenograft models based on various tumor cell lines with high HGF expression, treatment with lenvatinib alone showed weak antitumor effects, but treatment with lenvatinib plus golvatinib showed synergistic antitumor effects, accompanied by decreased tumor vessel density. These results suggest that HGF from tumor cells confers resistance to tumor endothelial cells against VEGFR inhibitors, and that combination therapy using VEGFR inhibitors with Met inhibitors may be effective for overcoming resistance to VEGFR inhibitors. Further evaluation in clinical trials is warranted.


Assuntos
Aminopiridinas/farmacologia , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Compostos de Fenilureia/farmacologia , Piperazinas/farmacologia , Quinolinas/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Feminino , Fator de Crescimento de Hepatócito/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Sci ; 105(10): 1334-42, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25060424

RESUMO

Eribulin mesylate is a synthetic macrocyclic ketone analog of the marine sponge natural product halichondrin B and an inhibitor of microtubule dynamics. Some tubulin-binding drugs are known to have antivascular (antiangiogenesis or vascular-disrupting) activities that can target abnormal tumor vessels. Using dynamic contrast-enhanced MRI analyses, here we show that eribulin induces remodeling of tumor vasculature through a novel antivascular activity in MX-1 and MDA-MB-231 human breast cancer xenograft models. Vascular remodeling associated with improved perfusion was shown by Hoechst 33342 staining and by increased microvessel density together with decreased mean vascular areas and fewer branched vessels in tumor tissues, as determined by immunohistochemical staining for endothelial marker CD31. Quantitative RT-PCR analysis of normal host cells in the stroma of xenograft tumors showed that eribulin altered the expression of mouse (host) genes in angiogenesis signaling pathways controlling endothelial cell-pericyte interactions, and in the epithelial-mesenchymal transition pathway in the context of the tumor microenvironment. Eribulin also decreased hypoxia-associated protein expression of mouse (host) vascular endothelial growth factor by ELISA and human CA9 by immunohistochemical analysis. Prior treatment with eribulin enhanced the anti-tumor activity of capecitabine in the MDA-MB-231 xenograft model. These findings suggest that eribulin-induced remodeling of abnormal tumor vasculature leads to a more functional microenvironment that may reduce the aggressiveness of tumors due to elimination of inner tumor hypoxia. Because abnormal tumor microenvironments enhance both drug resistance and metastasis, the apparent ability of eribulin to reverse these aggressive characteristics may contribute to its clinical benefits.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Furanos/farmacologia , Cetonas/farmacologia , Moduladores de Tubulina/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Remodelação Vascular/efeitos dos fármacos , Animais , Neoplasias da Mama/patologia , Capecitabina , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Feminino , Fluoruracila/análogos & derivados , Fluoruracila/farmacologia , Humanos , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Exp Ther Med ; 27(2): 75, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38264427

RESUMO

Cluster of differentiation (CD)44 is a marker of dental pulp stem cells and is involved in odontoblast differentiation and calcification. Chemokine-like receptor 1 (CMKLR1), also known as chemerin receptor 23 (ChemR23) is also expressed in odontoblasts and dental pulp stem cells and is involved in inflammation suppression and tooth regeneration. Resolvin E1, a bioactive lipid, is a CMKLR1 ligand that mediates the chemerin-CMKLR1 interaction and suppresses pulpal inflammation. The present study clarified the intracellular and tissue localization of CD44 and CMKLR1 by immunohistochemical staining of normal pulp and pulp with pulpitis from 12-week-old male Wistar rat teeth or human teeth. In addition, the localization of CD44 and CMKLR1 in human dental pulp stem cells was observed by immunofluorescence staining. The present study also examined the involvement of resolvin E1 in inhibiting inflammation and calcification by western blotting. CD44- and CMKLR1-positive cells were confirmed in the odontoblast layer in normal dental pulp of rats and humans. CD44 was mainly localized in the cell membrane and CMKLR1 was mainly found in the cytoplasm of human dental pulp stem cells. CMKLR1 was also confirmed in the odontoblast layer in rats and humans with pulpitis but CD44 was not present. Following treatment of dental pulp stem cells with lipoteichoic acid, which imitates Gram-positive bacterial infection, resolvin E1 did not suppress the expression of cyclooxygenase-2 or of the odontoblast differentiation marker, dentin sialophosphoprotein. Furthermore, resolvin E1 induced the differentiation of dental pulp stem cells into odontoblasts even in the presence of the inflammatory stimulus.

7.
Anticancer Res ; 44(6): 2393-2406, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38821585

RESUMO

BACKGROUND/AIM: Cholangiocarcinoma (CCA) is an aggressive tumor with limited treatment options especially in 2nd line or later treatments. Targeting fibroblast growth factor receptor (FGFR) 2 has recently emerged as a promising treatment option for patients with CCA harboring FGFR2-fusion. This study investigated the antitumor activities of tasurgratinib as an orally available FGFR1-3 inhibitor, in preclinical FGFR2-driven CCA models. MATERIALS AND METHODS: Antitumor activities of tasurgratinib were examined in vitro and in vivo using NIH/3T3 cells expressing FGFR2-fusion as FGFR2-driven CCA models, and in vivo using a CCA patient-derived xenograft model. The molecular mechanism of action of tasurgratinib was elucidated through co-crystal structure analysis with FGFR1, manual complex model analysis with FGFR2, and binding kinetics analysis with FGFR2. Furthermore, the cell-based inhibitory activities against acquired resistant FGFR2 mutations in patients with CCA treated with FGFR inhibitors were evaluated. RESULTS: Tasurgratinib showed antitumor activity in preclinical FGFR2-driven CCA models by inhibiting the FGFR signaling pathway in vitro and in vivo. Furthermore, cell-based target engagement assays indicated that tasurgratinib had potent inhibitory activities against FGFR2 mutations, such as N549H/K, which are the major acquired mutations in CCA. We also confirmed that tasurgratinib exhibited fast association and slow dissociation kinetics with FGFR2, binding to the ATP-binding site and the neighboring region, and adopting an Asp-Phe-Gly (DFG)-"in" conformation. CONCLUSION: These data demonstrate the therapeutic potential of tasurgratinib in FGFR2-driven CCA and provide molecular mechanistic insights into its unique inhibitory profile against secondary FGFR2 resistance mutations in patients with CCA treated with FGFR inhibitors.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Ensaios Antitumorais Modelo de Xenoenxerto , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Colangiocarcinoma/metabolismo , Animais , Humanos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Camundongos , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Administração Oral , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Células NIH 3T3 , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Pirimidinas/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas de Fusão Oncogênica/antagonistas & inibidores
8.
J Biol Chem ; 286(42): 36667-76, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21880726

RESUMO

In normal epithelial cells, integrin α(6)ß(4) is abundantly expressed and forms hemidesmosomes, which is a cellular structure that mediates cell-extracellular matrix binding. In many types of cancer cells, integrin α(6)ß(4) is up-regulated, laminin is cleaved, and hemidesmosomes are disrupted, eventually causing an enhancement of cancer cell movement and facilitation of their invasion. We previously showed that the immunoglobulin-like cell adhesion molecule Necl-2 (Nectin-like molecule 2), known as a tumor suppressor, inhibits cancer cell movement by suppressing the ErbB3/ErbB2 signaling. We show here that Necl-2 interacts in cis with integrin α(6)ß(4). The binding of Necl-2 with integrin ß(4) was mediated by its extracellular region. In human colorectal adenocarcinoma Caco-2 cells, integrin α(6)ß(4) was localized at hemidesmosomes. Small interfering RNA-mediated suppression of Necl-2 expression enhanced the phorbol ester-induced disruption of the integrin α(6)ß(4) complex at hemidesmosomes, whereas expression of Necl-2 suppressed the disruption of this structure. These results indicate that tumor-suppressive functions of Necl-2 are mediated by the stabilization of the hemidesmosome structure in addition to the inhibition of the ErbB3/ErbB2 signaling.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hemidesmossomos/metabolismo , Imunoglobulinas/metabolismo , Integrina alfa6beta4/metabolismo , Células CACO-2 , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/genética , Células HEK293 , Hemidesmossomos/genética , Humanos , Imunoglobulinas/genética , Integrina alfa6beta4/genética , Integrina beta4/genética , Integrina beta4/metabolismo , Laminina/biossíntese , Laminina/genética , Ligação Proteica , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
9.
J Cell Sci ; 122(Pt 23): 4319-29, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19887591

RESUMO

Afadin is an actin-filament-binding protein that binds to nectin, an immunoglobulin-like cell-cell adhesion molecule, and plays an important role in the formation of adherens junctions. Here, we show that afadin, which did not bind to nectin and was localized at the leading edge of moving cells, has another role: enhancement of the directional, but not random, cell movement. When NIH3T3 cells were stimulated with platelet-derived growth factor (PDGF), afadin colocalized with PDGF receptor, alphavbeta3 integrin and nectin-like molecule-5 at the leading edge and facilitated the formation of leading-edge structures and directional cell movement in the direction of PDGF stimulation. However, these phenotypes were markedly perturbed by knockdown of afadin, and were dependent on the binding of afadin to active Rap1. Binding of Rap1 to afadin was necessary for the recruitment of afadin and the tyrosine phosphatase SHP-2 to the leading edge. SHP-2 was previously reported to tightly regulate the activation of PDGF receptor and its downstream signaling pathway for the formation of the leading edge. These results indicate that afadin has a novel role in PDGF-induced directional cell movement, presumably in cooperation with active Rap1 and SHP-2.


Assuntos
Movimento Celular/efeitos dos fármacos , Proteínas dos Microfilamentos/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Western Blotting , Bovinos , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Imunoprecipitação , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Células NIH 3T3 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo
10.
Genes Cells ; 15(9): 995-1001, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20695903

RESUMO

Integrin alpha(6)beta(4) is abundantly expressed in normal epithelial cells and forms hemidesmosomes, one of cell-extracellular matrix junctions. In many types of cancer cells, integrin alpha(6)beta(4) is up-regulated, laminin, an integrin alpha(6)beta(4)-binding extracellular matrix protein, is cleaved, and hemidesmosomes are disrupted, eventually causing an enhancement of cancer cell movement and a facilitation of their invasion. It was previously shown that integrin alpha(6)beta(4) interacts with ErbB1 and ErbB2 and enhances cell proliferation and motility. Here we show that integrin alpha(6)beta(4) interacts with ErbB3 but not with ErbB1, ErbB2 or ErbB4, and enhances the heregulin-induced, ErbB3/ErbB2 heterodimer-mediated DNA synthesis, but not cell motility, in A549 cells.


Assuntos
DNA/biossíntese , Integrina alfa6beta4/metabolismo , Neuregulina-1/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA/genética , Células HEK293 , Humanos , Integrina alfa6beta4/genética , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Interferência de RNA , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-3/química , Receptor ErbB-3/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção
11.
Materials (Basel) ; 14(16)2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34443162

RESUMO

Mineral trioxide aggregate (MTA) is an alternative endodontic material that predicts conductive or inductive calcified tissue formation from immature pulp mesenchymal stem cells (IPMSCs). The purpose of this study was to investigate whether MTA could promote reparative odontoblast differentiation via IPMSCs in the early phase of regeneration and compare with calcium hydroxide (CH). Direct pulp capping using calcium hydroxide (CH), MTA, and MTA with platelet-rich plasma (MTA + PRP) was performed on maxillary first molars of 8-week-old male Wistar rats (n = 36). After 3, 7, or 14 days, the teeth were analyzed for mineral density (MD) and volume of MD (VMD) via micro-focusing computed tomography (µCT), nestin, dentin matrix acidic phosphoprotein 1 (DMP1) immunohistochemistry, and real-time PCR for DMP1 mRNA expression. MTA stimulated the early phase differentiation of the IPMSCs into odontoblasts, with positive results for nestin and DMP1 compared with CH. Moreover, MTA + PRP stimulated calcified granule and dentin bridge formation through calcium mineral deposition, following the induction of DMP1 mRNA expression in IPMSCs. Our results suggested that the combination of MTA and PRP is an effective and clinically applicable method for activating endogenous dental pulp stem cells into odontoblasts in the early stages of pulp regeneration.

12.
Dent Mater J ; 40(4): 863-869, 2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-33642445

RESUMO

Mineral trioxide aggregate (MTA) cement is widely used in the field of endodontic treatment. We herein synthesized calcium silicates from calcium carbonate and silicon dioxide, with the aim of reducing the cost associated with the MTA. Additionally, we prepared gypsum-containing calcium silicate cement to reduce the setting time while enhancing the mechanical strength. We evaluated the physical properties of this cement and investigated the response of human dental pulp stem cells (hDPSCs) grown in culture media containing cement eluate. Our results revealed that calcium silicates could be easily synthesized in lab-scale. Furthermore, we demonstrate that gypsum addition helps shorten the setting time while increasing the compressive strength of dental cements. The synthesized gypsum-containing calcium silicate cement showed minimal cytotoxicity and did not inhibit the proliferation of hDPSCs. These results suggested that the newly developed calcium silicate material could be a promising pulp capping material.


Assuntos
Sulfato de Cálcio , Cimentos Dentários , Compostos de Alumínio , Cálcio , Compostos de Cálcio , Combinação de Medicamentos , Humanos , Teste de Materiais , Óxidos , Cimento de Silicato , Silicatos
13.
Eur J Pharm Biopharm ; 155: 77-87, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32781024

RESUMO

We previously reported that oral and intestinal absorption of insulin in rats and mice is significantly enhanced in vivo by coadministration with cell-penetrating peptides (CPPs). To evaluate the clinical use of CPPs as absorption enhancers, it is imperative to clarify the mechanisms associated with the permeation-stimulatory effect of CPPs in vitro. The confirmation experiment revealed a discrepancy between in vivo and in vitro effects of CPPs, such as D-octaarginine (D-R8) and L-penetratin, on epithelial permeation of insulin. The present study was designed to determine the factors that work in vivo but are deficient in an in vitro system consisting of Caco-2 cells. The effects of D-R8 and L-penetratin on permeation of insulin through the Caco-2 cell monolayer were partially boosted in fasted-state simulated intestinal fluid (FaSSIF). Contrary to expectation, the effects of CPPs on cellular uptake of insulin and the binding ratio of CPPs to insulin analyzed by surface plasmon resonance in normal buffer and FaSSIF were similar. Also, the effects of CPPs, especially D-R8, on cellular uptake of insulin, were stronger in Caco-2 cell monolayers with microfold cell (M cell)-like properties. These results suggested a key role of intestinal lipids and M cells in the stimulatory effect of CPPs on net epithelial permeation of insulin in vivo.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Peptídeos Penetradores de Células/metabolismo , Insulina/metabolismo , Absorção Intestinal/fisiologia , Mucosa Intestinal/metabolismo , Sequência de Aminoácidos , Linfoma de Burkitt/metabolismo , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/administração & dosagem , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Humanos , Insulina/administração & dosagem , Insulina/genética , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
J Endod ; 46(8): 1149-1154, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32437788

RESUMO

Although bisphosphonates are widely used to treat conditions such as osteoporosis, they may cause osteonecrosis of the jaw. We treated a patient with no history of tooth extraction or other surgical treatment who developed medication-related osteonecrosis of the jaw (MRONJ) with secondary pulpal disease. A 79-year-old woman presented with purulent discharge from the gum at the incisor region. She had been using bisphosphonates for 9 years. Tooth #6 had undertaken root canal treatment at a general practice. All teeth other than tooth #6 reacted to electric pulp testing. Computed tomographic imaging revealed signs suggestive of necrotic bone, and MRONJ was diagnosed. Teeth #7 and #8, which had initially exhibited vital reactions, also subsequently ceased to react to thermal and electric pulp testing. Root canal treatment was performed on teeth #6-8, and their condition was monitored. Computed tomographic imaging at 9 months after the first presentation revealed that the bone defect had greatly enlarged with separation of the necrotic bone; therefore, excision of the necrotic bone and curettage were performed in the department of oral and maxillofacial surgery. The loss of pulp reaction in teeth that had exhibited a vital reaction at the first presentation was considered to indicate that teeth #6-8 had developed dental pulp pathosis as a result of MRONJ.


Assuntos
Osteonecrose da Arcada Osseodentária Associada a Difosfonatos , Conservadores da Densidade Óssea/efeitos adversos , Difosfonatos/efeitos adversos , Idoso , Feminino , Humanos , Extração Dentária
15.
Genes Cells ; 13(3): 269-84, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18298801

RESUMO

It was previously shown that platelet-derived growth factor (PDGF) receptor physically and functionally interacts with integrin alpha(v)beta(3), effectively inducing cell movement. We previously showed that Necl-5, originally identified as a poliovirus receptor, interacts with integrin alpha(v)beta(3) and enhances its clustering and the formation of focal complexes at the leading edges of moving cells, resulting in an enhancement of cell movement. We showed here that Necl-5 additionally interacts with PDGF receptor in NIH3T3 cells and regulates the interaction between PDGF receptor and integrin alpha(v)beta(3), effectively inducing directional cell movement. PDGF receptor co-localized with Necl-5 and integrin alpha(v)beta(3) at peripheral ruffles over lamellipodia, which were formed at the leading edges of moving cells in response to PDGF, but not at the focal complexes under these ruffles, whereas Necl-5 and integrin alpha(v)beta(3) co-localized at these focal complexes. The clustering of these three molecules at peripheral ruffles required the activation of integrin alpha(v)beta(3) by vitronectin and the PDGF-induced activation of the small G protein Rac and subsequent re-organization of the actin cytoskeleton. These results indicate a key role of Necl-5 in directional cell movement by physically and functionally interacting with both integrin alpha(v)beta(3) and PDGF receptor.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Proteínas de Neoplasias/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Humanos , Integrina alfaVbeta3/metabolismo , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/genética , Ligação Proteica/fisiologia , Pseudópodes/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas rac de Ligação ao GTP/metabolismo
16.
Sci Rep ; 9(1): 8656, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-31209263

RESUMO

Despite their outstanding antitumour activity in mice, the limited supply from the natural sources has prevented drug discovery/development based on intact halichondrins. We achieved a total synthesis of C52-halichondrin-B amine (E7130) on a >10 g scale with >99.8% purity under GMP conditions. Interestingly, E7130 not only is a novel microtubule dynamics inhibitor but can also increase intratumoural CD31-positive endothelial cells and reduce α-SMA-positive cancer-associated fibroblasts at pharmacologically relevant compound concentrations. According to these unique effects, E7130 significantly augment the effect of antitumour treatments in mouse models and is currently in a clinical trial. Overall, our work demonstrates that a total synthesis can address the issue of limited material supply in drug discovery/development even for the cases of complex natural products.


Assuntos
Antineoplásicos Fitogênicos/síntese química , Neoplasias da Mama/tratamento farmacológico , Carcinoma de Células Escamosas/tratamento farmacológico , Éteres Cíclicos/síntese química , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Macrolídeos/síntese química , Moduladores de Tubulina/síntese química , Actinas/genética , Actinas/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica , Produtos Biológicos/síntese química , Produtos Biológicos/farmacologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Cetuximab/farmacologia , Descoberta de Drogas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Éteres Cíclicos/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/mortalidade , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Macrolídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Análise de Sobrevida , Moduladores de Tubulina/farmacologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Endod ; 44(1): 80-86, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29079051

RESUMO

INTRODUCTION: Pulp tissue regeneration is becoming a reality after discovery of mesenchymal stem cells (MSCs) residing in the pulp tissues through various clinical innovations, although MSC transplantation into the pulp space has met with challenges of in vitro cell expansion and cultures. As a way to circumvent the regulatory and technical complexities of in vitro MSC culture, we investigated the use of minced pulp tissues as a source of pulpal MSCs for tissue regeneration. METHODS: We characterized the phenotype of cells explanted from minced pulp (MP), namely MP-derived MSCs (MP-MSCs), compared with dental pulp stem cells (DPSCs) established from pulp tissues by enzyme digestion. Phenotypic characterization included replication kinetics, immunophenotyping, and multilineage differentiation. Using the tooth slice model, we assessed odonto/osteogenic differentiation of DPSCs, MP-MSCs, and minced pulp tissues in situ. RESULTS: In vitro replication of MP-MSCs occurred more rapidly during the initial phase of subcultures compared with DPSCs; however, MP-MSCs arrived at senescence at population doubling 47, whereas DPSCs replicated until population doubling 64, indicating shorter replicative lifespan. MP-MSCs also demonstrated stronger odonto/osteogenic differentiation than DPSCs by alkaline phosphatase activity and the protein expression. Both MP-MSCs and DPSCs demonstrated odonto/osteogenic and adipogenic differentiation capacities. Both cell types also showed mineralized tissue formation in the tooth slice model. Seeding minced pulp tissue on poly-L-lactic acid scaffold allowed for migration of MP-MSCs from the tissues and odontogenic differentiation with dentin sialophosphoprotein expression in the tooth slice model. CONCLUSIONS: These data indicated that MP may be an alternative source of pulpal MSCs that may allow de novo pulp-dentin regeneration without the need for in vitro culture and expansion.


Assuntos
Diferenciação Celular , Polpa Dentária/citologia , Células-Tronco Mesenquimais , Odontogênese , Adolescente , Células Cultivadas , Feminino , Humanos , Masculino , Adulto Jovem
19.
J Endod ; 32(11): 1102-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17055917

RESUMO

The purpose of this study was to evaluate the cleaning effect of root canal walls using strong acid electrolytic water (SAEW) as a root canal irrigant, and to investigate the influence of SAEW on the root canal dentin by micro-hardness test. Forty-three single-rooted, single-canaled teeth were instrumented using standard step-back technique with K-files. Irrigation was performed using distilled water, 5.25% NaOCl and 3% H(2)O(2), SAEW, or 15% EDTA solution in five groups. Samples were prepared to be examined under a scanning electron microscope (SEM) and micro Vickers hardness (H(V)) test machine. Our results showed that the root cleaning effects of the combined use of SAEW and NaOCl solution as root canal irrigants were equivalent to those in the group with NaOCl and 15% EDTA. When SAEW was used for 1 min under ultrasonic vibration, no decreases in the hardness of dentin inside the root canal were detected.


Assuntos
Condicionamento Ácido do Dente/métodos , Cavidade Pulpar/ultraestrutura , Dentina/ultraestrutura , Irrigantes do Canal Radicular/química , Água/química , Quelantes/química , Ácido Edético/química , Dureza , Humanos , Peróxido de Hidrogênio/química , Teste de Materiais , Microscopia Eletrônica de Varredura , Preparo de Canal Radicular/métodos , Hipoclorito de Sódio/química , Estresse Mecânico , Terapia por Ultrassom
20.
Anticancer Res ; 36(4): 1553-61, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27069131

RESUMO

BACKGROUND: Eribulin mesilate (eribulin), a first-in-class halichondrin B-based microtubule dynamics inhibitor, has been shown to promote vascular remodeling and reversal of epithelial-mesenchymal transition (EMT) apart from its antimitotic activity in breast cancer models. MATERIALS AND METHODS: Anti-proliferative activity of eribulin was examined in vitro and in vivo in several human soft tissue sarcoma (STS) cell lines. To assess tumor blood perfusion and phenotypic changes, eribulin was investigated in a leiomyosarcoma xenograft and in vitro in liposarcoma and leiomyosarcoma cell lines. RESULTS: Eribulin showed anti-proliferative activity in vitro against all six cell lines investigated, with 50% inhibitory concentration values of around 1 nmol/l, as well as significant antitumor activity against four xenografts in vivo. In addition, eribulin significantly enhanced tumor blood perfusion in xenografts and induced morphological changes and up-regulation of differentiation marker genes. CONCLUSION: In pre-clinical models, eribulin showed anti-proliferative activity against a variety of histopathological subtypes of STS. Eribulin might also cause tumor vasculature remodeling to enhance tumor blood perfusion and induce tumor cell differentiation.


Assuntos
Antimitóticos/uso terapêutico , Furanos/uso terapêutico , Cetonas/uso terapêutico , Sarcoma/tratamento farmacológico , Animais , Antimitóticos/farmacologia , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Furanos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Cetonas/farmacologia , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/genética , Sarcoma/genética , Sarcoma/patologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Calponinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA