Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Braz Oral Res ; 38: e037, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38747824

RESUMO

Dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) can differentiate into osteoblasts, indicating that both are potential candidates for bone tissue engineering. Osteogenesis is influenced by many environmental factors, one of which is lipopolysaccharide (LPS). LPS-induced NF-κB activity affects the osteogenic potencies of different types of MSCs differently. This study evaluated the effect of LPS-induced NF-κB activity and its inhibition in DPSCs and PDLSCs. DPSCs and PDLSCs were cultured in an osteogenic medium, pretreated with/without NF-κB inhibitor Bay 11-7082, and treated with/without LPS. Alizarin red staining was performed to assess bone nodule formation, which was observed under an inverted light microscope. NF-κB and alkaline phosphatase (ALP) activities were measured to examine the effect of Bay 11-7082 pretreatment and LPS supplementation on osteogenic differentiation of DPSCs and PDLSCs. LPS significantly induced NF-κB activity (p = 0.000) and reduced ALP activity (p = 0.000), which inhibited bone nodule formation in DPSCs and PDLSCs. Bay 11-7082 inhibited LPS-induced NF-κB activity, and partially maintained ALP activity and osteogenic potency of LPS-supplemented DPSCs and PDLSCs. Thus, inhibition of LPS-induced NF-κB activity can maintain the osteogenic potency of DPSCs and PDLSCs.


Assuntos
Fosfatase Alcalina , Diferenciação Celular , Polpa Dentária , Lipopolissacarídeos , NF-kappa B , Nitrilas , Osteogênese , Ligamento Periodontal , Células-Tronco , Humanos , Lipopolissacarídeos/farmacologia , Ligamento Periodontal/citologia , Ligamento Periodontal/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Polpa Dentária/citologia , Polpa Dentária/efeitos dos fármacos , NF-kappa B/metabolismo , Fosfatase Alcalina/análise , Diferenciação Celular/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Células Cultivadas , Nitrilas/farmacologia , Sulfonas/farmacologia , Reprodutibilidade dos Testes , Fatores de Tempo , Adulto Jovem , Adolescente
2.
Braz. oral res. (Online) ; 38: e037, 2024. graf
Artigo em Inglês | LILACS-Express | LILACS, BBO - Odontologia | ID: biblio-1557359

RESUMO

Abstract Dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) can differentiate into osteoblasts, indicating that both are potential candidates for bone tissue engineering. Osteogenesis is influenced by many environmental factors, one of which is lipopolysaccharide (LPS). LPS-induced NF-κB activity affects the osteogenic potencies of different types of MSCs differently. This study evaluated the effect of LPS-induced NF-κB activity and its inhibition in DPSCs and PDLSCs. DPSCs and PDLSCs were cultured in an osteogenic medium, pretreated with/without NF-κB inhibitor Bay 11-7082, and treated with/without LPS. Alizarin red staining was performed to assess bone nodule formation, which was observed under an inverted light microscope. NF-κB and alkaline phosphatase (ALP) activities were measured to examine the effect of Bay 11-7082 pretreatment and LPS supplementation on osteogenic differentiation of DPSCs and PDLSCs. LPS significantly induced NF-κB activity (p = 0.000) and reduced ALP activity (p = 0.000), which inhibited bone nodule formation in DPSCs and PDLSCs. Bay 11-7082 inhibited LPS-induced NF-κB activity, and partially maintained ALP activity and osteogenic potency of LPS-supplemented DPSCs and PDLSCs. Thus, inhibition of LPS-induced NF-κB activity can maintain the osteogenic potency of DPSCs and PDLSCs.

3.
Eur Arch Otorhinolaryngol ; 279(4): 1805-1812, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34008035

RESUMO

PURPOSE: Sensorineural hearing loss (SNHL) is commonly caused by the death or dysfunction of cochlear cell types as a result of their lack of regenerative capacity. However, regenerative medicine, such as stem cell therapy, has become a promising tool to cure many diseases, including hearing loss. In this study, we determined whether DPSCs could differentiate into cochlear hair cell in vitro. METHODS: DPSCs derived from human third molar dental pulp were induced into NSCs using a medium containing basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) for 7 days, and then into cochlear hair cell using a medium containing EGF and IGF-1 for the next 14 days. We used the neuroepithelial protein marker nestin and cochlear hair cell marker myosin VIIa as the markers for cells differentiation. Cells expressing the positive markers under the microscope were confirmed to have differentiated into cochlear hair cell. RESULTS: DPSCs were successfully induced to differentiate into NSCs, with mean 24% nestin-positive cells. We found that DPSC-derived NSCs have a great capacity in differentiating into inner ear hair cell-like cells with an average of 81% cells presenting myosin VIIa. Thus, DPSCs have high potential to serve as a good resource for SNHL treatment. CONCLUSION: We found the high potential of DPSCs to differentiate into NSC. The ability of DPSCs in differentiating into neural lineage cell made them a good candidate for regenerative therapy in neural diseases, such as SNHL.


Assuntos
Polpa Dentária , Perda Auditiva Neurossensorial , Diferenciação Celular , Fator de Crescimento Epidérmico/metabolismo , Células Ciliadas Auditivas , Perda Auditiva Neurossensorial/terapia , Humanos , Células-Tronco
4.
F1000Res ; 9: 537, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34394921

RESUMO

Background: Human umbilical cord blood-mesenchymal stem cell (hUCB-MSC)-derived secretome is known to be able to promote neovascularization and angiogenesis, so it is also thought to have a capability to modulate endothelial progenitor cell (EPC) functions. Atorvastatin is the cornerstone of coronary artery disease (CAD) treatment which can enhance EPCs proliferation and migration. This study aims to analyze the effect of the hUCB-MSC-derived secretome and its combination with atorvastatin toward EPCs proliferation and migration. Methods: EPCs were isolated from a CAD patient's peripheral blood. Cultured EPCs were divided into a control group and treatment group of 2.5 µM atorvastatin, hUCB-MSC-derived secretome (2%, 10%, and 20% concentration) and its combination. EPCs proliferation was evaluated using an MTT cell proliferation assay, and EPC migration was evaluated using a Transwell migration assay kit. Results: This research showed that hUCB-MSC-derived secretomes significantly increase EPC proliferation and migration in a dose-dependent manner. The high concentration of hUCB-MSC-derived secretome were shown to be superior to atorvastatin in inducing EPC proliferation and migration (p<0.001). A combination of the hUCB-MSC-derived secretome and atorvastatin shown to improve EPCs proliferation and migration compared to hUCB-MSC-derived secretome treatment or atorvastatin alone (p<0.001). Conclusions: This study concluded that the hUCB-MSC-derived secretome work synergistically with atorvastatin treatment in improving EPCs proliferation and migration.


Assuntos
Células Progenitoras Endoteliais , Células-Tronco Mesenquimais , Atorvastatina/farmacologia , Proliferação de Células , Sangue Fetal , Humanos
5.
Iran J Basic Med Sci ; 21(7): 745-752, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30140415

RESUMO

OBJECTIVES: This study aimed to determine the collagen type II (COL2) and SOX9 expression in interleukin growth factor (IGF-1)-induced Wharton's Jelly mesenchymal stem cells (WJMSCs) and the level of chondrogenic markers in co-culture IGF1-WJMSCs and IL1ß-CHON002 as osteoarthritis (OA) cells model. MATERIALS AND METHODS: WJMSCs were induced with IGF1 (75, 150, and 300 ng/ml) to enhance their chondrogenesis capability. The gene expression of SOX9 and COL2 was evaluated with quantitative RT-PCR. Furthermore, IGF1-WJMSCs were co-cultured with IL1ß-CHON002 cells in varied ratios (1:2, 1:1, 2:1). Chondrogenic markers ADAMTS1, ADAMTS5, MMP3, MMP1, and RANKL were measured with ELISA. RESULTS: The IGF1-WJMSCs had an increased expression of COL2 and SOX9. ADAMTS1, ADAMTS5, MMP1, MMP3, and RANKL levels were decreased in the co-culture IGF1-WJMSCs and IL1ß-CHON002. CONCLUSION: The IGF1-induced WJMSCs were capable to enhance chondrogenesis, indicated by increased expression of SOX9 and COL2 and decreased expression of ADAMTS1, ADAMTS5, MMP3, MMP1, and RANKL. These findings can be further used in the osteoarthritis treatment.

6.
Mol Cell Biochem ; 400(1-2): 201-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25421411

RESUMO

Endothelial progenitor cells (EPCs) clinical applications have been well reported. However, due to low number of EPCs that could be isolated, EPCs expansion study became one of the main focuses. Some optimized mediums to culture EPCs were currently available. However, the proliferation signaling pathway is not clearly disclosed yet. Peripheral blood was collected from eight healthy subjects, followed by mononuclear cells (MNCs) isolation. MNCs were then prepared and cultured for 2 days. After that, non-adherent cells were harvested and further cultured for 3 days. Resulted colony-forming unit (CFU)-Hill colonies were documented and enumerated under an inverted light microscope. To detect membrane markers, immunofluorescence was performed to detect CD34, VEGFR-2, and CD133. Cell documentation was conducted under a fluorescence microscope. To check cell proliferation, XTT Cell Proliferation Assay Kit was used according to kit insert. To detect possible activation of p44/42 MAPK, western blot was performed to detect p44/42 MAPK and phosphorylated p44/42 MAPK. All visualized bands were captured and quantified. Our results showed that EPCs markers (CD34, CD133 and VEGFR-2) were detected in 3 days culture. From XTT cell proliferation assay and CFU enumeration results, we found that EPCs proliferated significantly (p = 0.012) with addition of supplement. Phosphorylated-p42 MAPK expression of EPCs treated with supplement was significantly higher than the one of EPCs without treatment. Significant inhibition of p42 MAPK phosphorylation by U0126 was observed (p = 0.012). By pretreatment of U0126, number of viable cells and CFUs treated with supplement was significantly decreased (p = 0.012). Our results showed that MEK-dependent p42 MAPK pathway might play an important role in EPCs proliferation.


Assuntos
Diferenciação Celular/genética , Proliferação de Células/genética , Células Progenitoras Endoteliais , Proteína Quinase 1 Ativada por Mitógeno/biossíntese , Adesão Celular/genética , Movimento Celular , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/genética
7.
Acta Med Indones ; 43(3): 178-84, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21979283

RESUMO

AIM: to validate isolation and cultivation methods of bone marrow mesenchymal stem cells (BM-MSCs) from iliac crest, and to compare biological characteristics of BM-MSCs from different age groups for preparation of autologous stem cell therapy in cartilage defect. METHODS: patients undergoing spinal surgery were selected and grouped according to age. Iliac crest bone marrow from the patients was aspirated. BM-MSCs were isolated from the bone marrow and then cultivated. Their biological characteristics including morphological appearances and surface biomarkers were evaluated. Growth curves were observed. Sterility and Mycoplasma tests were also performed for quality assessment of BM-MSCs culture procedure. RESULTS: in average, cultivated-BM-MSCs reached the number of 7.56-22.95 x 106 in 4-7 weeks period. BM-MSCs of all age groups showed the same quality of morphology, shape and surface biomarkers (CD105+, CD73+, CD34-, CD45-, CD14-, CD19-, HLA-DR-). CONCLUSION: our procedures in isolating and cultivating of BM-MSCs have reached required amount for implantation into the cartilage lesion. In addition, the cultivated-BM-MSCs' biological characteristics were also in accordance with International Society of Cell Therapy (ISCT) MSCs criteria.


Assuntos
Antígenos CD/análise , Células da Medula Óssea , Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Adolescente , Adulto , Fatores Etários , Cartilagem/lesões , Cartilagem/cirurgia , Proliferação de Células , Meios de Cultura , Feminino , Antígenos HLA-DR/análise , Humanos , Ílio , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/microbiologia , Pessoa de Meia-Idade , Mycoplasma/isolamento & purificação , Adulto Jovem
8.
BMC Res Notes ; 4: 371, 2011 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-21943045

RESUMO

BACKGROUND: The finding of human umbilical cord blood as one of the most likely sources of hematopoietic stem cells offers a less invasive alternative for the need of hematopoietic stem cell transplantation. Due to the once-in-a-life time chance of collecting it, an optimum cryopreservation method that can preserve the life and function of the cells contained is critically needed. METHODS: Until now, slow-cooling has been the routine method of cryopreservation; however, rapid-cooling offers a simple, efficient, and harmless method for preserving the life and function of the desired cells. Therefore, this study was conducted to compare the effectiveness of slow- and rapid-cooling to preserve umbilical cord blood of mononucleated cells suspected of containing hematopoietic stem cells. The parameters used in this study were differences in cell viability, malondialdehyde content, and apoptosis level. The identification of hematopoietic stem cells themselves was carried out by enumerating CD34+ in a flow cytometer. RESULTS: Our results showed that mononucleated cell viability after rapid-cooling (91.9%) was significantly higher than that after slow-cooling (75.5%), with a p value = 0.003. Interestingly, the malondialdehyde level in the mononucleated cell population after rapid-cooling (56.45 µM) was also significantly higher than that after slow-cooling (33.25 µM), with a p value < 0.001. The apoptosis level in rapid-cooling population (5.18%) was not significantly different from that of the mononucleated cell population that underwent slow-cooling (3.81%), with a p value = 0.138. However, CD34+ enumeration was much higher in the population that underwent slow-cooling (23.32 cell/µl) than in the one that underwent rapid-cooling (2.47 cell/µl), with a p value = 0.001. CONCLUSIONS: Rapid-cooling is a potential cryopreservation method to be used to preserve the umbilical cord blood of mononucleated cells, although further optimization of the number of CD34+ cells after rapid-cooling is critically needed.

9.
J Nat Med ; 64(4): 423-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20544395

RESUMO

In our screening projects for anticancer agents from natural resources, artocarpin [6-(3-methyl-1-butenyl)-5,2',4'-trihydroxy-3-isoprenyl-7-methoxyflavone] isolated from wood of jack fruit (Artocarpus heterophyllus) showed potent cytotoxic activity on human T47D breast cancer cells. The mode of action of artocarpin was evaluated by its effect on cell viability, nuclear morphology, cell cycle progression, expression of protein markers for apoptosis, and mitochondrial membrane potential (Delta psi m). These results showed that artocarpin caused a reduction of cell viability in a concentration-dependent manner and an alteration of cell and nuclear morphology. Moreover, the percentage of the sub-G1 phase formation was elevated dose-dependently. Artocarpin induced activation of caspase 8 and 10 as indicated by stronger signal intensity of cleaved-caspase 8 and weaker signal intensity of caspase 10 markers detected after artocarpin treatment. In addition, we also noticed the activation of caspase 3 by artocarpin. There were negligible changes in mitochondrial membrane potential (Delta psi m) due to artocarpin treatment. All together, these data indicated that artocarpin induced apoptosis in T47D cells possibly via an extrinsic pathway.


Assuntos
Artocarpus/toxicidade , Neoplasias da Mama/patologia , Citotoxinas/toxicidade , Inibidores do Crescimento/toxicidade , Lectinas de Ligação a Manose/toxicidade , Lectinas de Plantas/toxicidade , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Neoplasias da Mama/prevenção & controle , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citotoxinas/isolamento & purificação , Humanos , Lectinas de Ligação a Manose/isolamento & purificação , Lectinas de Plantas/isolamento & purificação
10.
J Acupunct Meridian Stud ; 2(4): 306-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20633507

RESUMO

We investigated the leaves of Kleinhovia hospita, a plant which has been traditionally used in Indonesia as phytotherapy to cure liver disease, to describe antioxidant materials from plant sources. K. hospita leaves were extracted with methanol and further partitioned into n-hexane, diethyl ether, and ethyl acetate. The antioxidant activity of each fraction and the residue was assessed using a 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging method and their cytotoxicity on HepG2 liver cancer cells was determined by a MTT assay. The K. hospita leaf methanol extract showed strong antioxidant activity (96%) compared with vitamin C (98%) by the DPPH method and the measured activity from the subsequent extracts of the methanol extract were 48.9% for n-hexane, 74.0% for diethyl ether, 84.3% for ethyl acetate, and 77.1% for the residue. The MTT assay showed the cytotoxicity of the methanol extract on HepG2 cells at 14%, 76%, and 80% at concentrations of 50 microg/mL, 87.5 microg/mL, and 125 microg/mL, respectively. Leaf extracts of the medicinal plant K. hospita showed potent antioxidant activity and moderate cytotoxicity on HepG2 liver cancer cells.


Assuntos
Antioxidantes/análise , Malvaceae/química , Extratos Vegetais/análise , Plantas Medicinais/química , Antioxidantes/toxicidade , Células Hep G2 , Humanos , Indonésia , Extratos Vegetais/toxicidade , Folhas de Planta/química
11.
Oral Oncol ; 42(4): 415-20, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16413220

RESUMO

Osteoprotegerin (OPG) is a useful receptor in inhibiting Receptor Activator of NFkappaB Ligand (RANKL) in inducing osteoclastogenesis. Tumor Necrosis Factor (TNF)-Related Apoptosis-Inducing Ligand (TRAIL) is a potent apoptosis-inducing ligand in ameloblastomas. Since OPG has been reported to bind to TRAIL as well, the effect of OPG in TRAIL's function in inducing apoptosis should also be investigated. To investigate on the expression of OPG in ameloblastomas, immuhistochemistry, immunofluorescence and Western blot were performed. From the immunohistochemistry results, we found that OPG was expressed in ameloblastoma tissues. Expression of OPG was clearly seen in AM-1 cells by immunofluorescence as well. Additionally, Western blot analysis confirmed OPG expression in ameloblastoma tissues and AM-1 cells. To investigate on the potential of TNFalpha, TRAIL and RANKL in inducing apoptosis, we performed an apoptosis assay. From the apoptosis assay, we found that TRAIL had the highest potential in inducing apoptosis compared to TNFalpha and RANKL. To investigate the binding of OPG with RANKL and TRAIL, we performed a binding assay. We noticed that OPG preferably bind with RANKL than TRAIL. However, at low levels of RANKL, marked binding of OPG with TRAIL was seen. As we suspected that the binding of OPG and TRAIL might cause the effect of TRAIL in inducing apoptosis in ameloblastomas, we combined the treatment of OPG and TRAIL in AM-1 cells. From the apoptosis assay, we found that under treatment of OPG, TRAIL's function in inducing apoptosis was suppressed. These data suggest that by binding with TRAIL, OPG suppressed TRAIL's function in inducing apoptosis in ameloblastomas.


Assuntos
Ameloblastoma/metabolismo , Neoplasias Maxilomandibulares/metabolismo , Osteoprotegerina/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ameloblastoma/fisiopatologia , Apoptose , Western Blotting , Feminino , Humanos , Neoplasias Maxilomandibulares/fisiopatologia , Masculino
12.
Histochem Cell Biol ; 125(3): 215-25, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16205940

RESUMO

Simon extracts are vitamin K(1)-rich food materials extracted from the leaves of the Simon sweet potato. Although vitamin K is known to stimulate bone formation, we postulated that Simon extracts also contain unknown biological compounds having the ability to regulate bone resorption. Here we prepared the vitamin K-free fraction from the Simon extracts and investigated the ability of this fraction on the differentiation of osteoclasts. A remarkable inhibitory effect of osteoclastogenesis was observed when osteoclast precursors were treated with this fraction in rat bone marrow culture systems as well as in a pure differentiation system using murine osteoclast precursor cell line. The vitamin K-free Simon extracts markedly suppressed severe bone destruction mediated by abundant osteoclasts associated with adjuvant-induced arthritis in rats. High performance liquid chromatography (HPLC) analysis revealed that the vitamin K-free Simon extracts contained three types of low molecular weight inhibitors for osteoclastogenesis; caffeic acid, chlorogenic acids and isochlorogenic acids. Among these substances, caffeic acid showed the most powerful inhibitory effects on osteoclastogenesis. Caffeic acid significantly suppressed expression of NFATc1, a key transcription factor for the induction of osteoclastogenesis. Our current study enlightened a high utility of the Simon extracts and their chemical components as effective regulators for bone resorption accompanied with inflammation and metabolic bone diseases.


Assuntos
Artrite Experimental/tratamento farmacológico , Ácidos Cafeicos/farmacologia , Osteoclastos/efeitos dos fármacos , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Sequência de Bases , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Catepsina K , Catepsinas/genética , Células Cultivadas , Primers do DNA/genética , Gliceraldeído-3-Fosfato Desidrogenases/genética , Técnicas In Vitro , Ipomoea batatas/química , Masculino , Osteoclastos/patologia , Fitoterapia , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Receptor Ativador de Fator Nuclear kappa-B/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vitamina K/isolamento & purificação
13.
Oral Oncol ; 42(1): 39-45, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16140562

RESUMO

Tumor necrosis factor alpha (TNFalpha) can trigger both cell survival and apoptosis. In the present study, from the flow cytometry results, we found that the prolonged-treatment of TNFalpha until 24 h, resulted apoptosis in AM-1 cells (ameloblastoma cell line). These results were confirmed by DAPI staining, which showed nuclear fragmentation feature of AM-1 cells under treatment of TNFalpha. Our further investigation using specific caspase inhibitors showed that caspase-3 played a crucial role in mediating TNFalpha-induced apoptosis in AM-1 cells. In addition, significant elevation of TNFalpha potential in inducing apoptosis was seen by applying LY294002, phosphatidylinositol-3-OH kinase (PI3K) inhibitor, or U0126, mitogen-activated extracellular-regulated kinase (MEK1/2) inhibitor, prior to the treatment of TNFalpha in AM-1 cells. These results suggested that TNFalpha induced both cell survival and apoptosis pathways in ameloblastoma and potential of TNFalpha in inducing apoptosis can be improved by inhibiting TNFalpha-induced Akt and p44/42 mitogen-activated protein kinase (MAPK) cell survival pathways.


Assuntos
Ameloblastoma/fisiopatologia , Apoptose/fisiologia , Neoplasias Maxilomandibulares/fisiopatologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Ameloblastoma/metabolismo , Caspases/farmacologia , Fragmentação do DNA , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes , Humanos , Neoplasias Maxilomandibulares/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilação , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Cancer Res ; 65(18): 8286-97, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16166305

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to have selective antitumor activity. TRAIL induces ubiquitous pathways of cell death in which caspase activation is mediated either directly or via the release of apoptogenic factors from mitochondria; however, the precise components of the mitochondrial signaling pathway have not been well defined. Notably, mitochondria constitute an important target in overcoming resistance to TRAIL in many types of tumors. Bid is considered to be fundamental in engaging mitochondria during death receptor-mediated apoptosis, but this action is dependent on mitochondrial lipids. Here, we report that TRAIL signaling induces an alteration in mitochondrial membrane lipids, particularly cardiolipin. This occurs independently of caspase activation and primes mitochondrial membranes to the proapoptotic action of Bid. We unveil a link between TRAIL signaling and alteration of membrane lipid homeostasis that occurs in parallel to apical caspase activation but does not take over the mode of cell death because of the concurrent activation of caspase-8. In particular, TRAIL-induced alteration of mitochondrial lipids follows an imbalance in the cellular homeostasis of phosphatidylcholine, which results in an elevation in diacylglycerol (DAG). Elevated DAG in turn activates the delta isoform of phospholipid-dependent serine/threonine protein kinase C, which then accelerates the cleavage of caspase-8. We also show that preservation of phosphatidylcholine homeostasis by inhibition of lipid-degrading enzymes almost completely impedes the activation of pro-caspase-9 while scarcely changing the activation of caspase-8.


Assuntos
Proteínas Reguladoras de Apoptose/farmacologia , Cardiolipinas/metabolismo , Glicoproteínas de Membrana/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspases/metabolismo , Diglicerídeos/biossíntese , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Células Jurkat , Potenciais da Membrana/efeitos dos fármacos , Fosfolipases/metabolismo , Proteína Quinase C-delta/metabolismo , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF
15.
Tumour Biol ; 26(5): 258-64, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16110259

RESUMO

Tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL/Apo-2L), a potent ligand in inducing apoptosis, has recently emerged as a novel anticancer agent based on its ability to induce apoptosis in tumor cells, while exhibiting no toxicity in most normal cells. Since no potent apoptosis-inducing factor has been found yet in ameloblastoma, the present study was conducted. In the present study, expressions of TRAIL receptors, death receptor 4 (DR4) and DR5, were detected in all ameloblastoma tissues by immunohistochemistry as well as in AM-1 cells by immunofluorescence. By applying TRAIL in AM-1 cells, ameloblastoma cell line, for 24 h, we found that TRAIL cleaved caspase-8, -9 and -3, and lowered mitochondrial membrane potential (Deltapsim), and markedly induced apoptosis in AM-1 cells (46%). These results suggested that TRAIL is a potent apoptosis-inducing ligand in ameloblastoma.


Assuntos
Apoptose/genética , Caspases/metabolismo , Glicoproteínas de Membrana/fisiologia , Neoplasias Bucais/genética , Neoplasias Bucais/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Ameloblastos , Proteínas Reguladoras de Apoptose , Caspase 3 , Caspase 8 , Caspase 9 , Feminino , Humanos , Imuno-Histoquímica , Ligantes , Masculino , Ligante Indutor de Apoptose Relacionado a TNF
16.
Oral Oncol ; 41(6): 637-44, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15935726

RESUMO

Ameloblastoma, a tumor located in bone, when neglected, can perforate the bone and, ultimately, spread into the soft tissues. To expand in the bone, ameloblastoma must have a mechanism of resorbing the surrounding bone. However, the mechanism for bone resorption is poorly understood. In the present study, we found that RANKL and TNFalpha were expressed and secreted by ameloblastoma cells, and was proven to induce osteoclastogenesis. Our present results also showed that phosphorylation of p38, SAPK, p44/42 and Akt were upregulated under treatment of 10xCM (concentrated conditioned media of AM-1 cells). We also noticed formation of resorption lacunae on dentin slice by 10xCM-induced osteoclast-like MNCs. These results suggested that ameloblastoma by secreting RANKL and TNFalpha could induce osteoclastogenesis.


Assuntos
Ameloblastoma/fisiopatologia , Reabsorção Óssea/fisiopatologia , Neoplasias Maxilomandibulares/fisiopatologia , Osteoclastos/fisiologia , Ameloblastoma/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados , Dentina/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Maxilomandibulares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Oral Oncol ; 41(4): 375-82, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15792609

RESUMO

Tumour necrosis factor alpha (TNFalpha) is known crucial in inducing cell survival, proliferation, differentiation, and apoptosis. In the present study, we found that TNFalpha as well as its receptors, TNFR1 (TNF Receptor 1) and TNFR2, were clearly expressed in ameloblastoma tissues and AM-1 cells. By stimulation of TNFalpha in AM-1 cells, the phosphorylation of Akt (Ser473) and p44/42 mitogen-activated protein kinase (MAPK) (Thr202/Tyr204) was markedly increased in TNFalpha concentration and time dependent manner. Pretreatment with U0126, mitogen-activated extracellular-regulated kinase (MEK) 1/2 inhibitor, prior to TNFalpha stimulation, specifically inhibited TNFalpha-induced phosphorylation of p44/42 MAPK (Thr202/Tyr204) in AM-1 cells. Meanwhile, pretreatment with LY294002, phosphatidylinositol-3-OH kinase (PI3K) inhibitor, could inhibit both TNFalpha-induced phosphorylation of Akt (Ser473) and p44/42 MAPK (Thr202/Tyr204). These results suggested that TNFalpha is expressed in ameloblastoma and it can induce Akt and p44/42 MAPK activation through PI3K, which later might induce cell survival and proliferation in ameloblastoma.


Assuntos
Ameloblastoma/metabolismo , Neoplasias Maxilomandibulares/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Relação Dose-Resposta a Droga , Humanos , Técnicas Imunoenzimáticas , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
18.
J Biol Chem ; 279(53): 55594-601, 2004 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-15485835

RESUMO

Here we show a novel mechanism by which FLICE-like inhibitory protein (c-FLIP) regulates apoptosis induced by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and one of its receptors, DR5. c-FLIP is a critical regulator of the TNF family of cytokine receptor signaling. c-FLIP has been postulated to prevent formation of the competent death-inducing signaling complex (DISC) in a ligand-dependent manner, through its interaction with FADD and/or caspase-8. In order to identify regulators of TRAIL function, we used the intracellular death domain (DD) of DR5 as a target to screen a phage-displayed combinatorial peptide library. The DD of DR5 selected from the library a peptide that showed sequence similarity to a stretch of amino acids in the C terminus of c-FLIP(L). The phage-displayed peptide selectively interacted with the DD of DR5 in in vitro binding assays. Similarly, full-length c-FLIP (c-FLIP(L)) and the C-terminal p12 domain of c-FLIP interacted with DR5 both in in vitro pull-down assays and in mammalian cells. This interaction was independent of TRAIL. To the contrary, TRAIL treatment released c-FLIP(L) from DR5, permitting the recruitment of FADD to the active DR5 signaling complex. By employing FADD-deficient Jurkat cells, we demonstrate that DR5 and c-FLIP(L) interact in a FADD-independent manner. Moreover, we show that a cellular membrane permeable version of the peptide corresponding to the DR5 binding domain of c-FLIP induces apoptosis in mammalian cells. Taken together, these findings indicate that c-FLIP interacts with the DD of DR5, thus preventing death (L)signaling by DR5 prior to the formation of an active DISC. Because TRAIL and DR5 are ubiquitously expressed, the interaction of c-FLIP(L) and DR5 indicates a mechanism by which tumor selective apoptosis can be achieved through protecting normal cells from undergoing death receptor-induced apoptosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores do Fator de Necrose Tumoral/química , Proteínas Adaptadoras de Transdução de Sinal/química , Alanina/química , Apoptose , Proteínas Reguladoras de Apoptose , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Caspase 8 , Caspases/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Proteína de Domínio de Morte Associada a Fas , Glutationa Transferase/metabolismo , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/química , Células Jurkat , Ligantes , Glicoproteínas de Membrana/metabolismo , Microscopia de Fluorescência , Biblioteca de Peptídeos , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Ligante Indutor de Apoptose Relacionado a TNF , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
19.
J Exp Med ; 200(7): 941-6, 2004 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-15452179

RESUMO

Osteoclasts are bone-resorbing, multinucleated giant cells that are essential for bone remodeling and are formed through cell fusion of mononuclear precursor cells. Although receptor activator of nuclear factor-kappaB ligand (RANKL) has been demonstrated to be an important osteoclastogenic cytokine, the cell surface molecules involved in osteoclastogenesis are mostly unknown. Here, we report that the seven-transmembrane receptor-like molecule, dendritic cell-specific transmembrane protein (DC-STAMP) is involved in osteoclastogenesis. Expression of DC-STAMP is rapidly induced in osteoclast precursor cells by RANKL and other osteoclastogenic stimulations. Targeted inhibition of DC-STAMP by small interfering RNAs and specific antibody markedly suppressed the formation of multinucleated osteoclast-like cells. Overexpression of DC-STAMP enhanced osteoclastogenesis in the presence of RANKL. Furthermore, DC-STAMP directly induced the expression of the osteoclast marker tartrate-resistant acid phosphatase. These data demonstrate for the first time that DC-STAMP has an essential role in osteoclastogenesis.


Assuntos
Proteínas de Transporte/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Osteoclastos/fisiologia , RNA Mensageiro/metabolismo , Fosfatase Ácida/genética , Fosfatase Ácida/metabolismo , Animais , Northern Blotting , Proteínas de Transporte/fisiologia , Células Cultivadas , Imuno-Histoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Oligonucleotídeos , Ligante RANK , RNA Interferente Pequeno/genética , Receptor Ativador de Fator Nuclear kappa-B , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fosfatase Ácida Resistente a Tartarato
20.
Lab Invest ; 84(9): 1092-102, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15195117

RESUMO

In the rat model of rheumatoid arthritis, a marked formation of osteoclasts is found in the distal tibia and the metatarsal bone. It was therefore postulated that osteoclast progenitors would be increased in the bone marrow cavities of rats with adjuvant-induced arthritis (AA rats). Bone marrow cells obtained from tibia of AA rats were cultured to form cells in the osteoclast lineage to access the number of osteoclast progenitors. Unexpectedly, only a suppressed level of osteoclast progenitors was detected in the diaphyseal bone marrow of tibia in AA rats. Distribution of osteoclast progenitors in the bone marrow cavity was examined, and it was shown that osteoclast progenitors accumulated in the distal tibia. Macrophage inflammatory protein (MIP)-1alpha, an osteoclastogenic CC chemokine, was expressed in ED-1-positive macrophages localizing in the distal tibia with marked bone destruction. Chemotaxis studies showed that MIP-1alpha expressed significant activity towards bone marrow cells. The suppressed level of osteoclastogenesis in bone marrow cells of AA rats was restored to a normal level by the addition of MIP-1alpha. It was suggested that MIP-1alpha is involved in the migration of osteoclast progenitors to the distal tibia as well as in osteoclastogenesis in AA rats. In these rats, in situ hybridization of the distal tibia with a high level of bone destruction showed significant expression of Receptor activator nuclear factor kappaB ligand (RANKL) messenger RNA in aggregates of multinucleated osteoclast-like cells present in the bone marrow cavity, a unique pathological feature for these rats. Migrated osteoclast progenitors are thought to be efficiently differentiated into osteoclasts in response to RANKL expressed by the aggregates of osteoclast-like cells under the influence of the MIP-1alpha. Such positive-feedback regulation of osteoclastogenesis could result in the highest recruitment of active osteoclasts in the area of marked bone destruction.


Assuntos
Artrite Experimental/patologia , Proteínas Inflamatórias de Macrófagos/metabolismo , Células-Tronco Mesenquimais/patologia , Osteoclastos/patologia , Osteogênese/fisiologia , Animais , Artrite Experimental/etiologia , Artrite Experimental/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Proteínas de Transporte/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Células Clonais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Gigantes/metabolismo , Células Gigantes/patologia , Proteínas Inflamatórias de Macrófagos/farmacologia , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Ligante RANK , Ratos , Ratos Endogâmicos Lew , Tíbia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...