Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Acta Pharmacol Sin ; 41(4): 581-582, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31383989

RESUMO

During re-read of our previously article Plumbagin attenuates cancer cell growth and osteoclast formation in the bone microenvironment of micepublished in Acta Pharmacologica Sinica, we were regretted to point out a mistake shown in Fig. 2a. The representative figure chosen to indicate the inhibitory effect of 4 mg/kg of plumbagin treatment at 1 week against MDA-MB-231SArfp cells localization within bone environment was incorrect due to the mishandling in manuscript preparation. Although this correction does not affect the results and conclusion of the paper, all the authors agree on the correction of our negligence as providing the corrected Fig. 2a presented below. We feel sorry and apologize for all the inconvenience it caused.An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Acta Pharmacol Sin ; 40(8): 1127-1128, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31097762

RESUMO

The authors regretted to find the mis-representative images in Fig. 3a, c and Fig. 4a, c when re-read our previously published article Synergistic suppression of human breast cancer cells by combination of plumbagin and zoledronic acid In vitro (DOI: 10.1038/aps.2015.42) in the journal of Acta Pharmacologica Sinica. This mistake occurred due to the careless compilation when the authors tried to show the synergistic effect against tumor apoptosis during figure presentation process. The right Fig. 3a, c and Fig. 4a, c were provided below. Despite that this correction does not affect the results and conclusions of the aforementioned paper, all the authors still consent on the correction of this negligence. We apologize to the Editor and the readership of the journal for any inconvenience caused. Your thoughtful understanding is highly appreciated.

3.
Cell Death Dis ; 9(7): 714, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29915309

RESUMO

The loss of appropriate cell adhesion normally induces apoptosis via a process termed anoikis. The aim of this study was to investigate the effects of mesenchymal stem cells (MSCs) in the cancer microenvironment on the anoikis resistance and pulmonary metastasis of osteosarcoma (OS) cells, and to evaluate the critical role of the interleukin (IL)-8/C-X-C chemokine receptor (CXCR) 1/Akt-signaling pathway in these processes. Metastatic OS subtype cells, which did or did not interact with MSC-conditioned medium (MSC-CM) in vitro, were isolated from the pulmonary site and named Saos2-lung-M. Both MSC-CM and IL-8 treatment increased the anoikis resistance of Saos2 cells in vitro. Moreover, exogenous MSC-CM promoted the survival and metastasis of Saos2 cells in nude mice. Saos2-lung-M cells were more malignant and resistant to anoikis than parental cells. MSCs secreted IL-8, thereby protecting OS cells from anoikis. Blocking the IL-8/CXCR1/Akt pathway via CXCR1 knockdown inhibited the pulmonary metastasis of Saos2-lung-MSCs and prolonged the survival of tumor-bearing mice. In conclusion, MSCs enhanced OS cell resistance to anoikis and pulmonary metastasis via regulation of the IL-8/CXCR1/Akt pathway. These findings suggest that MSCs can "select for" OS cells with high metastatic potential in vivo, and highlight CXCR1 as a key target in the regulation of pulmonary metastasis of OS cells.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Pulmonares/secundário , Células-Tronco Mesenquimais/fisiologia , Osteossarcoma/patologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores de Interleucina-8A/fisiologia , Animais , Anoikis/efeitos dos fármacos , Anoikis/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-8/metabolismo , Interleucina-8/farmacologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Osteossarcoma/genética , Osteossarcoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
4.
Front Genet ; 9: 135, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29731768

RESUMO

Osteosarcoma is the most common bone cancer in children and adolescents. Tissue inhibitors of metalloproteinases (TIMPs)-3 inhibit matrix metalloproteinases to limit extracellular matrix degradation. Cisplatin is a widely used chemotherapeutic drug used to cure osteosarcoma. Interleukin (IL)-6 and TIMP3 play important roles in the drug resistance of osteosarcoma; however, their relationship in this process remains unclear. This study aimed to explore the role of TIMP3 in the cisplatin sensitivity of osteosarcoma and its underlying molecular mechanisms in vitro and in vivo. We compared TIMP3 expression levels between patients with cisplatin-sensitive and -insensitive osteosarcoma. TIMP3 was overexpressed or knocked down in the Saos2-lung cell line, which is a Saos2 subtype isolated from pulmonary metastases that has higher cisplatin chemoresistance than Saos2 cells. IL-6 expression, cell proliferation, sensitivity to cisplatin, migration, and invasion after TIMP3 overexpression or knockdown were determined. The same experiments were performed using MG63 and U2OS cells. Subsequently, luciferase-labeled Saos2-lung cells overexpressing TIMP3 were injected into the tibiae of nude mice treated with cisplatin. The results showed that IL-6 inhibited TIMP3 expression in Saos2 and Saos2-lung cells via signal transducer and activator of transcription 3 (STAT3) activation. STAT3 knockdown reversed the effect of IL-6. The expression of TIMP3 was higher in patients with cisplatin-sensitive osteosarcoma than in those with insensitive osteosarcoma. IL-6 expression was downregulated upon TIMP3 overexpression, and upregulated by TIMP3 knockdown. TIMP3 overexpression suppressed cell proliferation and enhanced cisplatin sensitivity by activating apoptosis-related signal pathways and inhibiting IL-6 expression in vitro and in vivo. In conclusion, cisplatin sensitivity correlated positively with TIMP3 expression, which is regulated by the IL-6/TIMP3/caspase pathway. The TIMP3 pathway could represent a target for new therapies to treat osteosarcoma.

5.
Sci Rep ; 6: 32771, 2016 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-27600021

RESUMO

The endocrine role of the skeleton-which is impaired in human diseases including osteoporosis, obesity and diabetes-has been highlighted previously. In these diseases, the role of AMPK, a sensor and regulator of energy metabolism, is of biological and clinical importance. Since AMPK's main catalytic subunit α has two isoforms, it is unclear whether functional differences between them exist in the skeletal system. The current study overexpressed AMPKα1 and α2 in MC3T3-E1 cells, primary osteoblasts and mouse BMSCs by lentiviral transduction. Cells overexpressing AMPKα2 showed higher osteogenesis potential than AMPKα1, wherein androgen receptor (AR) and osteoactivin played important roles. RANKL and M-CSF were secreted at lower levels from cells overexpressing α2 than α1, resulting in decreased osteoblast-associated osteoclastogenesis. Adipogenesis was inhibited to a greater degree in 3T3-L1 cells overexpressing α2 than α1, which was modulated by AR. An abnormal downregulation of AMPKα2 was observed in human BMSCs exhibiting the fibrous dysplasia (FD) phenotype. Overexpression of AMPKα2 in these cells rescued the defect in osteogenesis, suggesting that AMPKα2 plays a role in FD pathogenesis. These findings highlight functional differences between AMPKα1 and α2, and provide a basis for investigating the molecular mechanisms of diseases associated with impaired functioning of the skeletal system.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Adipogenia , Osteoblastos/citologia , Osteoclastos/citologia , Osteogênese , Células 3T3-L1 , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Proteínas do Olho/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores Androgênicos/metabolismo
6.
Cell Signal ; 28(9): 1270-1282, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27283242

RESUMO

Several metabolic, genetic and oncogenic bone diseases share the common pathological phenotype of defective bone marrow stromal cell (BMSC) differentiation. Many reports in bone science in the past several years have suggested that the skeleton also has an endocrine role. The role of AMP-activated protein kinase (AMPK) as an energy metabolism sensor and how it regulates BMSC differentiation is largely unknown. In the current study, we used AMPK agonists to activate AMPK in MC3T3-E1 cells to investigate the functional roles of AMPK in osteogenesis. However, metformin and AICAR failed to activate AMPK consistently. Therefore, we established MC3T3-E1 and 3T3-L1 cell models of AMPK α subunit overexpression through lentivirus vector, in which AMPK was overactivated. AMPK hyperactivation stimulated MC3T3-E1 cell osteogenesis and inhibited 3T3-L1 cell adipogenesis. Osteopontin (OPN) mediated AMPK regulation of osteogenesis and adipogenesis. Furthermore, we provided evidence that the transcriptional repressor growth factor independence-1 (Gfi1) was downregulated and disassociated from the OPN promoter in response to AMPK activation, resulting in the upregulation of OPN. Overexpression of wild-type and dominant-negative Gfi1 modulated MC3T3-E1 osteogenesis and 3T3-L1 adipogenesis. Further evidence suggested that AMPK enhanced ectopic bone formation of MC3T3-E1 cells through the AMPK-Gfi1-OPN axis. In conclusion, AMPK was sufficient to stimulate osteogenesis of MC3T3-E1 cells and inhibit adipogenesis of 3T3-L1 cells through the AMPK-Gfi1-OPN axis. These findings helped elucidate the molecular mechanisms underlying AMPK regulation of osteogenesis and adipogenesis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adipogenia , Proteínas de Ligação a DNA/metabolismo , Osteogênese , Osteopontina/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Biomarcadores/metabolismo , Osso e Ossos/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Metformina/farmacologia , Camundongos , Camundongos Nus , Osteogênese/efeitos dos fármacos , Osteopontina/genética , Regiões Promotoras Genéticas/genética , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos
7.
Cancer Lett ; 369(2): 405-15, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26391645

RESUMO

Chemotherapy resistance is a major cause of poor prognoses for osteosarcoma patients. This study aimed to determine whether CXCR1 gene knockdown improves the sensitivity of osteosarcomas to chemotherapy. Both CXCR1 expression and cisplatin sensitivity were investigated and compared in two osteosarcoma cell lines. Sensitivity to the chemotherapy drug cisplatin and apoptosis were investigated with or without stimulation via Interleukin-8 (IL-8), which is a ligand of CXCR1. Furthermore, activation of the Akt signaling pathway was determined. Finally, luciferase-labeled CXCR1-knockdown Saos2-lung cells were injected into the tibiae of nude mice that were treated with cisplatin thereafter. We found that CXCR1 expression and cisplatin sensitivity were negatively correlated in osteosarcoma cell lines. IL-8-induced reduction in sensitivity could be blocked by silencing CXCR1, and CXCR1 knockdown suppressed the Akt signaling pathway. Moreover, CXCR1-knockdown tumors were significantly smaller than control tumors, which was consistent with the luciferase intensity results. The expression levels of IL-8, CXCR1 and p-Akt were suppressed in CXCR1-knockdown cells. Taken together, these data indicate that CXCR1 gene knockdown in osteosarcoma cells improved the sensitivity to chemotherapy and that this process might be regulated in part by the IL-8/CXCR1/Akt signaling pathway.


Assuntos
Cisplatino/farmacologia , Interleucina-8/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Receptores de Interleucina-8A/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Humanos , Interleucina-8/genética , Camundongos , Camundongos Nus , Osteossarcoma/tratamento farmacológico , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Interleucina-8A/genética , Transfecção
8.
Acta Pharmacol Sin ; 36(9): 1085-98, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26235741

RESUMO

AIM: Zoledronic acid (ZA), a bisphosphonate, is currently used in combination with chemotherapeutic agents to suppress breast cancer cell proliferation or breast cancer-induced osteolysis. The aim of this study was to investigate the effects of ZA combined with a natural anticancer compound plumbagin (PL) against human breast cancer cells in vitro. METHODS: Human breast cancer MDA-MB-231SArfp cells were treated with ZA, PL or a combination of ZA and PL. The cell growth, apoptosis and migration were evaluated using CCK-8 assay, flow cytometry and transwell assay, respectively. The expression of apoptosis-related proteins was measured using real-time PCR and Western blotting. Synergism was evaluated using Compusyn software, and the combination index (CI) and drug reduction index (DRI) values were determined. RESULTS: PL or ZA alone caused mild cytotoxicity (the IC50 value at 24 h was 12.18 and above 100 µmol/L, respectively). However, the combination of ZA and PL caused a synergistic cytotoxicity (CI=0.26). The DRI values also showed a synergistic effect between PL and ZA, with actual values of 5.52 and 3.59, respectively. Furthermore, PL and ZA synergistically induced apoptosis and inhibited migration of the breast cancer cells. Moreover, the combination of ZA and PL decreased the expression of Notch-1, cleaved PARP, Bcl-2 and Bcl-xl, and increased the expression of cleaved caspase-3, CDKN1A and ID1. When the breast cancer cells were transfected with specific siRNA against Notch-1, the combination of ZA and PL markedly increased the expression of Bcl-2. CONCLUSION: Combination of ZA and PL synergistically suppresses human breast cancer MDA-MB-231SArfp cells in vitro. PL can inhibit ZA-induced activation of the Notch-1 signaling pathway and subsequently reduce the expression of Bcl-2, thus potentiating cancer cell apoptosis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Conservadores da Densidade Óssea/farmacologia , Neoplasias da Mama/tratamento farmacológico , Difosfonatos/farmacologia , Imidazóis/farmacologia , Naftoquinonas/farmacologia , Antineoplásicos Fitogênicos/administração & dosagem , Apoptose/efeitos dos fármacos , Conservadores da Densidade Óssea/administração & dosagem , Mama/efeitos dos fármacos , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Difosfonatos/administração & dosagem , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imidazóis/administração & dosagem , Naftoquinonas/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Interferente Pequeno/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ácido Zoledrônico
9.
J Biochem ; 158(6): 445-57, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26078467

RESUMO

Many skeletal diseases have common pathological phenotype of defective osteogenesis of bone marrow stromal cells (BMSCs), in which histone modifications play an important role. However, few studies have examined the dynamics of distinct histone modifications during osteogenesis. In this study, we examined the dynamics of H3K9/K14 and H4K12 acetylation; H3K4 mono-, di- and tri-methylation; H3K9 di-methylation and H3K27 tri-methylation in osteogenic genes, runt-related transcription factor 2 (Runx2), osterix (Osx), alkaline phosphatase, bone sialoprotein and osteocalcin, during C3H10T1/2 osteogenesis. H3 and H4 acetylation and H3K4 di-methylation were elevated, and H3K9 di-methylation and H3K27 tri-methylation were reduced in osteogenic genes during C3H10T1/2 osteogenesis. C3H10T1/2 osteogenesis could be modulated by altering the patterns of H3 and H4 acetylation and H3K27 tri-methylation. In a glucocorticoid-induced osteoporosis mouse model, we observed the attenuation of osteogenic potential of osteoporotic BMSCs in parallel with H3 and H4 hypo-acetylation and H3K27 hyper-tri-methylation in Runx2 and Osx genes. When H3 and H4 acetylation was elevated, and H3K27 tri-methylation was reduced, the attenuated osteogenic potential of osteoporotic BMSCs was rescued effectively. These observations provide a deeper insight into the mechanisms of osteogenic differentiation and the pathophysiology of osteoporosis and can be used to design new drugs and develop new therapeutic methods to treat skeletal diseases.


Assuntos
Diferenciação Celular/genética , Código das Histonas , Histonas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Acetilação , Fosfatase Alcalina/metabolismo , Animais , Doenças Ósseas/genética , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Metilação , Camundongos , Osteocalcina/metabolismo , Osteopontina/metabolismo , Processamento de Proteína Pós-Traducional , Fator de Transcrição Sp7 , Fatores de Transcrição/metabolismo
10.
Acta Pharmacol Sin ; 35(1): 124-34, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24384612

RESUMO

AIM: To investigate the effects of plumbagin, a naphthoquinone derived from the medicinal plant Plumbago zeylanica, on human breast cancer cell growth and the cancer cell-induced osteolysis in the bone microenvironment of mice. METHODS: Human breast cancer cell subline MDA-MB-231SA with the ability to spread and grow in the bone was tested. The cell proliferation was determined using the CCK-8 assay. Apoptosis was detected with Annexin V/PI double-labeled flow cytometry. Red fluorescent protein-labeled MDA-MB-231SArfp cells were injected into the right tibia of female BALB/c-nu/nu mice. Three days after the inoculation, the mice were injected with plumbagin (2, 4, or 6 mg/kg, ip) 5 times per week for 7 weeks. The growth of the tumor cells was monitored using an in vivo imaging system. After the mice were sacrificed, the hind limbs were removed for radiographic and histological analyses. RESULTS: Plumbagin (2.5-20 µmol/L) concentration-dependently inhibited the cell viability and induced apoptosis of MDA-MB-231SA cells in vitro (the IC50 value of inhibition of cell viability was 14.7 µmol/L). Administration of plumbagin to breast cancer bearing mice delayed the tumor growth by 2-3 weeks and reduced the tumor volume by 44%-74%. The in vivo imaging study showed that plumbagin dose-dependently inhibited MDA-MB-231SArfp cell growth in bone microenvironment. Furthermore, X-ray images and micro-CT study demonstrated that plumbagin reduced bone erosion area and prevented a decrease in bone tissue volume. Histological studies showed that plumbagin dose-dependently inhibited the breast cancer cell growth, enhanced the cell apoptosis and reduced the number of TRAcP-positive osteoclasts. CONCLUSION: Plumbagin inhibits the cell growth and induces apoptosis in human breast cancer cells in mice bone microenvironment, leading to significant reduction in osteolytic lesions caused by the tumor cells.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Neoplasias da Mama/prevenção & controle , Naftoquinonas/uso terapêutico , Osteoclastos/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos/farmacologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Naftoquinonas/farmacologia , Osteoclastos/patologia , Microambiente Tumoral/fisiologia
11.
Exp Cell Res ; 320(1): 164-73, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24183998

RESUMO

Mesenchymal stem cells (MSCs) are among the most important components of the osteosarcoma microenvironment and are reported to promote tumor progression. However, the means by which osteosarcoma cells modulate MSC behavior remains unclear. The aim of this study was to determine the effects of osteosarcoma cells on both the production of pro-tumor cytokines by mesenchymal stem cells (MSCs) and the osteogenic differentiation of MSCs. High level of transforming growth factor-ß (TGF-ß) was detected in three osteosarcoma cell lines. Conditioned media (CM) from the osteosarcoma cell lines Saos-2 and U2-OS were used to stimulate the cultured MSCs. We found that osteosarcoma cells promoted the production of IL-6 and VEGF in MSCs by inhibiting their osteogenic differentiation. Furthermore, TGF-ß in tumor CM was proved to be an important factor. The TGF-ß neutralizing antibody antagonized the effects induced by osteosarcoma CM. The inhibition of Smad2/3 by siRNA significantly decreased the production of IL-6 and VEGF in MSCs and induced their osteogenic differentiation. We also found that Smad2/3 enhanced the expression of ß-catenin in MSCs by decreasing the level of Dickkopf-1 (DKK1). Although the inhibition of ß-catenin did not affect the production of IL-6 or VEGF, or the gene expression of the early osteogenic markers Runx2 and ALP, it did enhance the gene expression of osteocalcin. Taken together, our data indicate that osteosarcoma cells secrete TGF-ß to maintain the stemness of MSCs and promote the production of pro-tumor cytokines by these cells.


Assuntos
Citocinas/biossíntese , Células-Tronco Mesenquimais/metabolismo , Osteossarcoma/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/patologia , Osteogênese , Osteossarcoma/patologia , RNA Interferente Pequeno/farmacologia , Proteína Smad2/antagonistas & inibidores , Proteína Smad3/antagonistas & inibidores , Relação Estrutura-Atividade
12.
FEBS J ; 280(22): 5801-14, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23981481

RESUMO

The balance between osteogenesis and adipogenesis of bone marrow stromal cells is impaired in many human diseases. Knowledge of how to fine-tune this balance is of medical importance. CCAAT/enhancer binding protein α (C/EBPα) has been shown to regulate the balance between osteogenesis and adipogenesis of C3H10T1/2 cells, with epigenetic modifications of the C/EBPα promoter playing an important role. The present study aimed to elucidate the underlying molecular mechanisms. The results showed that peroxisome proliferator-activated receptor γ (PPARγ) binds the -1286 bp/-1065 bp region of the C/EBPα promoter to activate C/EBPα expression during osteogenesis and adipogenesis of C3H10T1/2 cells. DNA hypermethylation in the -1286 bp/-1065 bp region, observed at the terminal stage of osteogenesis, prevented PPARγ binding, and then histone deacetylase 1 (HDAC1) occupied this region to reduce the level of histone acetylation. We regulated the balance between osteogenesis and adipogenesis of mouse bone marrow stromal cells through modulation of DNA methylation and histone acetylation status. In addition, in bone marrow stromal cells from the glucocorticoid-induced osteoporosis (GIO) mouse, hypomethylation of CpG sites, higher binding of PPARγ, acetylated histones 3 and 4, and reduced binding of HDAC1 in the -1286 bp/-1065 bp region of C/EBPα promoter were observed, compared with normal mice. This study provides a deeper insight into the molecular mechanisms underlying the balance between osteogenesis and adipogenesis regulated by C/EBPα in synergy with PPARγ, and suggests a molecular model for how DNA methylation and histone acetylation are linked by PPARγ to regulate differentiation of bone marrow stromal cells.


Assuntos
Adipogenia/genética , Adipogenia/fisiologia , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Osteogênese/genética , Osteogênese/fisiologia , PPAR gama/metabolismo , Acetilação , Animais , Linhagem Celular , Ilhas de CpG , Metilação de DNA , Regulação da Expressão Gênica , Histona Desacetilase 1/metabolismo , Histonas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , PPAR gama/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
Chin Med J (Engl) ; 125(22): 4022-30, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23158137

RESUMO

BACKGROUND: Osteosarcoma (OS) is the most common primary malignant tumor of bone. Mouse models of human OS can invariably provide greater insight into the complex mechanisms that underlie the development and pathogenesis of this aggressive tumor. Bioluminescence technology favored tracing cancer cells in vivo. In this study, an OS model was described and evaluated using human OS cell line, Saos2, labeled with luciferase (Saos2-luc). METHODS: Saos2 cells were infected by lentivirus loading a firefly luciferase gene. Luciferase expression of Saos2-luc cells was characterized both in vitro and in vivo. Specific biologic and oncologic features of Saos2-luc cells were analyzed. The OS was established as orthotopic xenografts in nude mice. Both orthotopic tumors and spontaneous lung metastasis were analyzed. RESULTS: Tumorigenesis and spontaneous lung metastasis in nude mice could be monitored in vivo through in vivo imaging system. The enhancement in proliferation, migration and invasion abilities and the attenuation in adhesion ability were observed in Saos2-luc cells compared with Saos2 cells. Furthermore, there were the up-regulation of Osteocalcin, CCR10, CXCR1 and ID1 and the down-regulation of ALP, collagen I, CCR1, CCR3, CXCR3, NID and N-cadherin in Saos2-luc cells compare to Saos2 cells. The rate of spontaneous lung metastasis in Saos2-luc cells was higher than that in Saos2 cells, although without significant difference. CONCLUSIONS: Lentivirus transfection may cause alteration of gene expression profiles and further biological functions. This model can be used in the elucidation of molecular mechanisms of tumorigenesis and the screening of new therapeutic agents.


Assuntos
Carcinogênese/patologia , Metástase Neoplásica/patologia , Osteossarcoma/patologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
14.
Cancer Lett ; 325(1): 80-8, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22743617

RESUMO

We previously demonstrated that human mesenchymal stem cells (MSCs) promote the growth of osteosarcoma in the bone microenvironment. The aim of the present study was to further determine the effect of IL-6/STAT3 signaling on the progression of osteosarcoma. First, conditioned medium from MSCs was used to stimulate the growth of osteosarcoma cells (Saos-2) in vitro. We found that STAT3 was activated and that the activation could be blocked by an IL-6-neutralizing antibody. The inhibition of STAT3 in Saos-2 cells by siRNA or AG490 decreased cell proliferation, migration and invasion, down-regulated the mRNA expression of Cyclin D, Bcl-xL and Survivin and enhanced the apoptotic response. Furthermore, a nude mouse osteosarcoma model was established by injecting luciferase-labeled Saos-2 cells into the tibia, and the effect of STAT3 on tumor growth was determined by treating the mice with AG490. In vivo bioluminescence images showed that tumor growth was dramatically reduced in the AG490 group. In addition, STAT3 inhibition decreased the lung metastasis rate and prolonged the survival of these mice. After treatment with AG490, the protein levels of IL-6, p-STAT3 and PCNA were decreased, and the level of apoptosis in the tumor was increased. Altogether, these data indicate that MSCs in the bone microenvironment might promote the progression of osteosarcoma and protect tumor cells from drug-induced apoptosis through IL-6/STAT3 signaling.


Assuntos
Neoplasias Ósseas/patologia , Interleucina-6/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteossarcoma/patologia , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose/fisiologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células , Meios de Cultivo Condicionados/metabolismo , Ciclina D/genética , Ciclina D/metabolismo , Progressão da Doença , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Interleucina-6/genética , Masculino , Células-Tronco Mesenquimais/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Osteossarcoma/genética , Osteossarcoma/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fator de Transcrição STAT3/genética , Transdução de Sinais , Survivina , Transfecção , Transplante Heterólogo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
15.
J Pathol ; 228(1): 45-55, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22450860

RESUMO

Fibrous dysplasia (FD) is characterized by the replacement of normal bone with abnormal fibro-osseous tissue. This disorder is due to activating missense mutations in the GNAS gene and resultant over-production of cAMP. However, the signalling pathways that contribute to FD pathogenesis remain unknown. In the current study, bone marrow stromal cells (BMSCs) carrying GNAS R201H mutation were isolated from lesion site of FD patients. cAMP accumulation, enhanced proliferation and impaired osteogenesis potential were observed. Two cell models, BMSCs treated with excess exogenous cAMP and BMSCs infected with lentivirus GNAS R201H, were established to model the pathological conditions of FD and used to investigate its pathogenesis. The results suggest that the CREB-Smad6-Runx2 axis is involved in osteogenesis dysfunction of BMSCs with the FD phenotype. We confirmed the results in FD lesion-derived BMSCs and observed that the impaired osteogenesis potential of BMSCs infected with lentivirus GNAS (R201H) was recovered in vitro through modulation of the CREB-Smad6-Runx2 axis. This study provides useful insight into the signalling pathways involved in the FD phenotype and facilitates dissection of the molecular pathogenesis of FD and testing of novel therapies.


Assuntos
Proteína de Ligação a CREB/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Displasia Fibrosa Óssea/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Proteína Smad6/metabolismo , Adolescente , Proteína Morfogenética Óssea 2/farmacologia , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cromograninas , AMP Cíclico/farmacologia , Feminino , Displasia Fibrosa Óssea/genética , Displasia Fibrosa Óssea/patologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Vetores Genéticos , Humanos , Lentivirus , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/patologia , Mutação de Sentido Incorreto , Osteogênese/efeitos dos fármacos , Proteínas Recombinantes , Transdução de Sinais , Transdução Genética , Adulto Jovem
16.
Cancer Sci ; 101(12): 2554-60, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20874851

RESUMO

Our previous study showed that exogenous human mesenchymal stem cells (hMSCs) targeted established osteosarcoma and promoted its growth and pulmonary metastasis in vivo. As a follow-up, the present study aimed to investigate how hMSCs would interact with Saos-2 through autocrine/paracrine communication. The results showed that co-injection of hMSCs with Saos-2 into the proximal tibia of nude mice could promote tumor growth and progression. In vitro, the proliferation of Saos-2 and hMSCs was promoted by each other's conditioned medium, in which interleukin-6 (IL-6) played an important role. Osteogenic differentiation of hMSCs could be inhibited by conditioned medium of Saos-2, in which IL-6 was also involved. Furthermore, decreased IL-6 secretion by hMSCs during its osteogenesis and increased IL-6 secretion in response to conditioned medium of Saos-2 were observed. Based on these data, we suggest that there was a positive feedback loop of IL-6 in the interaction between hMSCs and Saos-2.


Assuntos
Neoplasias Ósseas/metabolismo , Comunicação Celular/fisiologia , Interleucina-6/biossíntese , Células-Tronco Mesenquimais/metabolismo , Osteossarcoma/metabolismo , Animais , Neoplasias Ósseas/patologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Osteossarcoma/patologia , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Cancer Lett ; 281(1): 32-41, 2009 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-19342158

RESUMO

In an effort to study the interaction between MSCs and osteosarcoma, we established an animal model of primary osteosarcoma in nude mice using Saos-2 cells. hMSCs, labeled with adv-GFP, were injected through the caudal vein. We observed that exogenous hMSCs targeted the osteosarcoma site and promoted its growth and pulmonary metastasis in vivo. To elucidate the underlying mechanisms, we employed transwell, neutralization antibody and MTT assays in vitro. hMSCs migrated toward the conditioned medium from Saos-2 cells, and SDF-1 was involved in this migration. Likewise, Saos-2 cells migrated toward the conditioned medium from hMSCs and CCL5 played an important role in this migration. Furthermore, proliferation of Saos-2 cells was enhanced by the conditioned medium from hMSCs and CCL5 was at least partly responsible for this enhancement.


Assuntos
Neoplasias Ósseas/patologia , Quimiocina CCL5/fisiologia , Quimiocina CXCL12/fisiologia , Neoplasias Pulmonares/secundário , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Osteossarcoma/patologia , Animais , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/transplante , Movimento Celular , Quimiocina CCL5/metabolismo , Quimiocina CXCL12/metabolismo , Meios de Cultivo Condicionados/farmacologia , Progressão da Doença , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Osteossarcoma/metabolismo , Osteossarcoma/secundário
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA