Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Heliyon ; 9(6): e16962, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37484408

RESUMO

Exosomes are released by various cells, including natural killer (NK) cells and transport signaling molecules for the intercellular communication. Hepatocellular carcinoma (HCC), also known as primary liver cancer, is often inoperable and difficult to accurate diagnosis. Notably, the prognosis and underlying mechanisms of HCC are not fully understood. Exosomes-derived NK cells (NK-exos) express unique cytotoxic proteins with a killing ability in tumors and can easily penetrate tumor tissues to improve their targeting ability. NK cell functions, inducing cellular cytotoxicity are modulated by cytokines such as interleukin (IL)-15 and IL-21. However, the mechanisms and effects of cytokines-stimulated NK-exos for the treatment of liver cancer, including HCC, are not well known. In this study, we aimed to investigate the synergistic anti-tumor effects of NK-exos stimulated with IL-15 and IL-21 (NK-exosIL-15/21) in Hep3B cells. Our findings revealed that NK-exosIL-15/21 expressed cytotoxic proteins (perforin and granzyme B) and contained typical exosome markers (CD9 and CD63) within the size range of 100-150 nm. Moreover, we demonstrated that NK-exosIL-15/21 induced the enhancement of cytotoxicity and apoptotic activity in Hep3B cells by activating the specific pro-apoptotic proteins (Bax, cleaved caspase 3, cleaved PARP, perforin, and granzyme B) and inhibiting the anti-apoptotic protein (Bcl-2). In summary, our results suggest that NK-exosIL-15/21 regulate strong anti-tumor effects of HCC cells, by increasing the cytotoxicity and apoptosis through the activation of specific cytotoxic molecules.

2.
Drug Deliv ; 29(1): 2897-2911, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36068970

RESUMO

Exosomes are nanosized extracellular vesicles secreted by various cell types, including those of the immune system, such as natural killer (NK) cells. They play a role in intercellular communication by transporting signal molecules between the cells. Recent studies have reported that NK cell-derived exosomes (NK-exo) contain cytotoxic proteins-induced cell death. However, the characteristics and potential functions of NK-exo, especially for the liver cancer are poorly understood. In this study, we investigated the anti-tumor effects of NK-exo in the primary liver cancer, hepatocellular carcinoma (HCC), using the orthotopic and subcutaneous tumor model. We found that NK-exo expressed both typical exosomal markers (e.g. CD63, CD81, and Alix) and cytotoxic proteins (e.g. perforin, granzyme B, FasL, and TRAIL). NK-exo were selectively taken up by HCC cells (e.g. Hep3B, HepG2, and Huh 7). Interestingly, Hep3B cells induced the highest cytotoxicity compared with HepG2 and Huh7 cells, and substantially enhanced the apoptosis by NK-exo. Furthermore, we demonstrated that NK-exo inhibited the phosphorylation of serine/threonine protein kinases (e.g. AKT and ERK1/2), and enhanced the activation of specific apoptosis markers (e.g. caspase-3, -7, -8, -9, and PARP) in Hep3B cells. NK-exo also exhibit the active targeting ability and potent therapeutic effects in both orthotopic and subcutaneous HCC mouse models. Overall, these results suggest that NK-exo indicate strong anti-tumor effects in HCC, which are mediated by novel regulatory mechanisms involved in serine/threonine kinase pathway-associated cell proliferation and caspase activation pathway-associated apoptosis.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Exossomos , Neoplasias Hepáticas , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Exossomos/metabolismo , Humanos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Neoplasias Hepáticas/metabolismo , Camundongos , Modelos Animais , Serina/metabolismo
3.
Drug Deliv ; 29(1): 2621-2631, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35941835

RESUMO

Colorectal cancer remains one of the main causes of cancer-related deaths worldwide. Although numerous nanomedicine formulations have been developed to tackle the disease, their low selectivity still limits effective therapeutic outcomes. In this study, we isolated extracellular vesicles (EVs) from CT26 colorectal cancer cells and 4T1 murine mammary carcinoma cells, loaded them with the chemotherapeutic agent (doxorubicin, DOX). Then we evaluated the cellular uptake of the extracellular vesicles both in 2D monolayer and 3D tumor spheroid setups using confocal laser scanning microscope and flow cytometry. In vivo tumor homing of the extracellular vesicles was verified on CT26 tumor bearing BALB/c mice using in vivo imaging system. Finally, in vivo therapeutic effects were evaluated and compared using the same animal models treated with five doses of EV formulations. CT26-EV-DOX exhibited excellent biocompatibility, a high drug-loading capacity, controlled drug release behavior, and a high capability for targeting colorectal cancer cells. In particular, we verified that CT26-EV-DOX could preferentially be up taken by their parent cells and could effectively target and penetrate 3D tumor spheroids resembling colorectal tumors in vivo in comparison with their 4T1 derived EV partner. Additionally, treatment of colorectal tumor-bearing BALB/c mice with of CT26-EV-DOX significantly inhibited the growth of the tumors during the treatment course. The developed CT26-EV-DOX nanoparticles may present a novel and effective strategy for the treatment of colorectal cancer.


Assuntos
Neoplasias Colorretais , Vesículas Extracelulares , Nanopartículas , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C
4.
Pharmaceutics ; 13(10)2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34683982

RESUMO

Various cell therapy strategies, including chimeric antigen receptor-expressing T or natural killer (NK) cells and cell-mediated drug delivery, have been developed for tumor eradication. However, the efficiency of these strategies against solid tumors remains unclear. We hypothesized that real-time control and visualization of therapeutic cells, such as NK cells, would improve their therapeutic efficacy against solid tumors. In this study, we engineered Sonazoid microbubble-conjugated NK (NK_Sona) cells and demonstrated that they were detectable by ultrasound imaging in real-time and maintained their functions. The Sonazoid microbubbles on the cell membrane did not affect the cytotoxicity and viability of the NK cells in vitro. Additionally, the NK_Sona cells could be visualized by ultrasound imaging and inhibited tumor growth in vivo. Taken together, our findings demonstrate the feasibility of this new approach in the use of therapeutic cells, such as NK cells, against solid tumors.

5.
ACS Nano ; 15(5): 8492-8506, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-33973786

RESUMO

Macrophages (MΦs) have the capability to sense chemotactic cues and to home tumors, therefore presenting a great approach to engineer these cells to deliver therapeutic agents to treat diseases. However, current cell-based drug delivery systems usually use commercial cell lines that may elicit an immune response when injected into a host animal. Furthermore, premature off-target drug release also remains an enormous challenge. Here, we isolated and differentiated MΦs from the spleens of BALB/c mice and developed dual-targeting MΦ-based microrobots, regulated by chemotaxis and an external magnetic field, and had a precise spatiotemporal controlled drug release at the tumor sites in response to the NIR laser irradiation. These microrobots were prepared by coloading citric acid (CA)-coated superparamagnetic nanoparticles (MNPs) and doxorubicin (DOX)-containing thermosensitive nanoliposomes (TSLPs) into the MΦs. CA-MNPs promoted a magnetic targeting function to the microrobots and also permitted photothermal heating in response to the NIR irradiation, triggering drug release from TSLPs. In vitro experiments showed that the microrobots effectively infiltrated tumors in 3D breast cancer tumor spheroids, particularly in the presence of the magnetic field, and effectively induced tumor cell death, further enhanced by the NIR laser irradiation. In vivo experiments confirmed that the application of the magnetic field and NIR laser could markedly inhibit the growth of tumors with a subtherapeutic dose of DOX and a single injection of the microrobots. In summary, the study proposes a strategy for the effective anticancer treatment using the developed microrobots.


Assuntos
Doxorrubicina , Nanopartículas , Animais , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Fototerapia
6.
J Cell Biochem ; 119(1): 748-757, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28657691

RESUMO

Pseudoshikonin I (PSI), a novel biomaterial isolated from Lithospermi radix, has been recognized as an herbal medicine for the treatment of infectious and inflammatory diseases. Bone remodeling maintains a balance through bone resorption (osteoclastogenesis) and bone formation (osteoblastogenesis). Bone formation is generally attributed to osteoblasts. However, the effects of PSI on the bone are not well known. In this study, we found that the ethanol extracts of PSI induced osteoblast differentiation by increasing the expression of bone morphogenic protein 4 (BMP 4). PSI positively regulates the transcriptional expression and osteogenic activity of osteoblast-specific transcription factors such as Runx2 and Osterix. To identify the signaling pathways that mediate PSI-induced osteoblastogenesis, we examined the effects of serine-threonine kinase inhibitors that are known regulators of Osterix and Runx2. PSI-induced upregulation of Osterix and Runx2 was suppressed by treatment with AKT and PKA inhibitors. These results suggest that PSI enhances osteoblast differentiation by stimulating Osterix and Runx2 via the AKT and PKA signaling pathways. Thus, the activation of Runx2 and Osterix is modulated by PSI, thereby demonstrating its potential as a treatment target for bone disease.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Etanol/farmacologia , Lithospermum/química , Osteoblastos/citologia , Fator de Transcrição Sp7/genética , Animais , Proteína Morfogenética Óssea 4/metabolismo , Remodelação Óssea , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos , Naftoquinonas/química , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Extratos Vegetais/farmacologia , Fator de Transcrição Sp7/metabolismo , Transcrição Gênica/efeitos dos fármacos
7.
Int J Mol Med ; 38(2): 610-8, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27353217

RESUMO

Bone remodeling and homeostasis are largely the result of the coordinated action of osteoblasts and osteoclasts. Osteoblasts are responsible for bone formation. The differentiation of osteoblasts is regulated by the transcription factors, Runx2 and Osterix. Natural products of plant origin are still a major part of traditional medicinal systems in Korea. The root of Lithospermum erythrorhizon Sieb. et Zucc. (LR), the purple gromwell, is an herbal medicine used for inflammatory and infectious diseases. LR is an anti-inflammatory and exerts anticancer effects by inducing the apoptosis of cancer cells. However, the precise molecular signaling mechanisms of osteoblastogenesis as regards LR and osteoblast transcription are not yet known. In this study, we investigated the effects of ethanol (EtOH) extract of LR (LES) on the osteoblast differentiation of C2C12 myoblasts induced by bone morphogenetic protein 4 (BMP4) and the potential involvement of Runx2 and Osterix in these effects. We found that the LES exhibited an ability to induce osteoblast differentiation. LES increased the expression of the osteoblast marker, alkaline phosphatase (ALP), as well as its activity, as shown by ALP staining and ALP activity assay. LES also increased mineralization, as shown by Alizarin Red S staining. Treatment with LES increased the protein levels (as shown by immunoblotting), as well as the transcriptional activity of Runx2 and Osterix and enhanced osteogenic activity. These results suggest that LES modulates osteoblast differentiation at least in part through Runx2 and Osterix.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Etanol/química , Regulação da Expressão Gênica/efeitos dos fármacos , Lithospermum/química , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Extratos Vegetais/farmacologia , Fatores de Transcrição/genética , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células HEK293 , Humanos , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteogênese/genética , Fator de Transcrição Sp7 , Transcrição Gênica/efeitos dos fármacos
8.
Bioorg Med Chem ; 24(4): 789-801, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26780832

RESUMO

Several androgen receptor (AR) antagonists are clinically prescribed to treat prostate cancer. Unfortunately, many patients become resistant to the existing AR antagonists. To overcome this, a novel AR antagonist candidate called DIMN was discovered by our research group in 2013. In order to develop compounds with improved potency, we designed novel DIMN derivatives based on a docking study and substituted carbons with heteroatom moieties. Encouraging in vitro results for compounds 1b, 1c, 1e, 3c, and 4c proved that the new design was successful. Among the newly synthesized compounds, 1e exhibited the strongest inhibitory effect on LNCaP cell growth (IC50=0.35µM) and also acted as a competitive AR antagonist with selectivity over the estrogen receptor (ER) and the glucocorticoid receptor (GR). A docking study of compound 1e fully supported these biological results. Compound 1e is considered to be a novel, potent and AR-specific antagonist for treating prostate cancer. Thus, our study successfully applied molecular modeling and bioisosteric replacement for hit optimization. The methods here provide a guide for future development of drug candidates through structure-based drug discovery and chemical modifications.


Assuntos
Antagonistas de Receptores de Andrógenos/síntese química , Antineoplásicos/síntese química , Desenho de Fármacos , Próstata/efeitos dos fármacos , Receptores Androgênicos/química , Motivos de Aminoácidos , Antagonistas de Receptores de Andrógenos/farmacologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Expressão Gênica , Genes Reporter , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Niacinamida/química , Próstata/metabolismo , Estrutura Secundária de Proteína , Pirazinamida/química , Pirimidinas/química , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Relação Estrutura-Atividade
9.
Mol Cells ; 39(2): 156-62, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26674964

RESUMO

Estrogen receptor α (ER-α), which is involved in bone metabolism and breast cancer, has been shown to have transcriptional targets. Dlx3 is essential for the skeletal development and plays an important role in osteoblast differentiation. Various osteogenic stimulators and transcription factors can induce the protein expression of Dlx3. However, the regulatory function of ER-α in the Dlx3 mediated osteogenic process remains unknown. Therefore, we investigated the regulation of Dlx3 and found that ER-α is a positive regulator of Dlx3 transcription in BMP2-induced osteoblast differentiation. We also found that ER-α interacts with Dlx3 and increases its transcriptional activity and DNA binding affinity. Furthermore, we demonstrated that the regulation of Dlx3 activity by ER-α is independent of the ligand (estradiol) binding domain. These results indicate that Dlx3 is a novel target of ER-α, and that ER-α regulates the osteoblast differentiation through modulation of Dlx3 expression and/or interaction with Dlx3.


Assuntos
Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Proteínas de Homeodomínio/genética , Mioblastos/metabolismo , Osteoblastos/metabolismo , Fatores de Transcrição/genética , Animais , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Proteínas de Homeodomínio/metabolismo , Humanos , Ligantes , Luciferases/genética , Luciferases/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcrição Gênica
10.
Bone ; 75: 201-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25744063

RESUMO

E3 ubiquitin ligase Cbl-b and c-Cbl play important roles in bone formation and maintenance. Cbl-b and c-Cbl regulate the activity of various receptor tyrosine kinases and intracellular protein tyrosine kinases mainly by regulating the degradation of target proteins. However, the precise mechanisms of how Cbl-b and c-Cbl regulate osteoblast differentiation are not well known. In this study, we investigated potential targets of Cbl-b and c-Cbl. We found that Cbl-b and c-Cbl inhibit BMP2-induced osteoblast differentiation in mesenchymal cells. Among various osteogenic transcription factors, we identified that Cbl-b and c-Cbl suppress the protein stability and transcriptional activity of Osterix. Our results suggest that Cbl-b and c-Cbl inhibit the function of Osterix by enhancing the ubiquitin-proteasome-mediated degradation of Osterix. Taken together, we propose novel regulatory roles of Cbl-b and c-Cbl during osteoblast differentiation in which Cbl-b and c-Cbl regulate the degradation of Osterix through the ubiquitin-proteasome pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular/fisiologia , Osteoblastos/citologia , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Osteogênese/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp7 , Transfecção , Ubiquitinação
11.
Mol Cell Endocrinol ; 407: 85-97, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25802190

RESUMO

Osterix, a zinc-finger transcription factor, is required for osteoblast differentiation and new bone formation during embryonic development. The c-Src of tyrosine kinase is involved in a variety of cellular signaling pathways, leading to the induction of DNA synthesis, cell proliferation, and cytoskeletal reorganization. Src activity is tightly regulated and its dysregulation leads to constitutive activation and cellular transformation. The function of Osterix can be also modulated by post-translational modification. But the precise molecular signaling mechanisms between Osterix and c-Src are not known. In this study we investigated the potential regulation of Osterix function by c-Src in osteoblast differentiation. We found that c-Src activation increases protein stability, osteogenic activity and transcriptional activity of Osterix. The siRNA-mediated knockdown of c-Src decreased the protein levels and transcriptional activity of Osterix. Conversely, Src specific inhibitor, SU6656, decreased the protein levels and transcriptional activity of Osterix. The c-Src interacts with and phosphorylates Osterix. These results suggest that c-Src signaling modulates osteoblast differentiation at least in part through Osterix.


Assuntos
Células Epiteliais/metabolismo , Mioblastos/metabolismo , Osteoblastos/metabolismo , Fatores de Transcrição/genética , Quinases da Família src/genética , Animais , Proteína Tirosina Quinase CSK , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , DNA/biossíntese , DNA/genética , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Indóis/farmacologia , Luciferases/genética , Luciferases/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteogênese/genética , Inibidores de Proteínas Quinases/farmacologia , Estabilidade Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Transcrição Sp7 , Sulfonamidas/farmacologia , Fatores de Transcrição/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
12.
Biochem Biophys Res Commun ; 453(3): 557-62, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25285640

RESUMO

SIRT2 is a member of the sirtuin family of NAD(+)-dependent protein deacetylases. It is involved in metabolic homeostasis and has been linked to the progression of age-related diseases. Casitas B-lineage lymphoma (Cbl) proteins regulate signal transduction through many pathways and, consequently, regulate cell function and development. Cbl proteins are ubiquitin ligases that ubiquitinate and target many signaling molecules for degradation. The function of SIRT2 is modulated by post-translational modifications. However, the precise molecular signaling mechanism of SIRT2 through interactions with Cbl proteins has not yet been established. In this study, we investigated the potential regulation of SIRT2 function by the Cbl mammalian family members Cbl-b and c-Cbl. We found that Cbl-b and c-Cbl increased the protein level and stability of SIRT2 and that Cbl-b and c-Cbl interact with SIRT2. They were also found to regulate the deacetylase activity of SIRT2. Further investigation revealed that Cbl-mediated SIRT2 regulation occurred via ubiquitination of SIRT2.


Assuntos
Proteínas Proto-Oncogênicas c-cbl/fisiologia , Sirtuína 2/fisiologia , Linhagem Celular , Humanos , Processamento de Proteína Pós-Traducional , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação
13.
Biochem Biophys Res Commun ; 450(2): 1120-5, 2014 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-24996174

RESUMO

SIRT2 is a mammalian member of the Sirtuin family of NAD(+)-dependent protein deacetylases. The tyrosine kinase Src is involved in a variety of cellular signaling pathways, leading to the induction of DNA synthesis, cell proliferation, and cytoskeletal reorganization. The function of SIRT2 is modulated by post-translational modifications; however, the precise molecular signaling mechanism of SIRT2 through interactions with c-Src has not yet been established. In this study, we investigated the potential regulation of SIRT2 function by c-Src. We found that the protein levels of SIRT2 were decreased by c-Src, and subsequently rescued by the addition of a Src specific inhibitor, SU6656, or by siRNA-mediated knockdown of c-Src. The c-Src interacts with and phosphorylates SIRT2 at Tyr104. c-Src also showed the ability to regulate the deacetylation activity of SIRT2. Investigation on the phosphorylation of SIRT2 suggested that this was the method of c-Src-mediated SIRT2 regulation.


Assuntos
Sirtuína 2/metabolismo , Quinases da Família src/metabolismo , Proteína Tirosina Quinase CSK , Células HEK293 , Humanos , Células MCF-7 , Fosforilação
14.
J Cell Biochem ; 115(10): 1808-15, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24905700

RESUMO

Osterix belongs to the SP gene family and is a core transcription factor responsible for osteoblast differentiation and bone formation. Activation of protein kinase A (PKA), a serine/threonine kinase, is essential for controlling bone formation and BMP-induced osteoblast differentiation. However, the relationship between Osterix and PKA is still unclear. In this report, we investigated the precise role of the PKA pathway in regulating Osterix during osteoblast differentiation. We found that PKA increased the protein level of Osterix; PKA phosphorylated Osterix, increased protein stability, and enhanced the transcriptional activity of Osterix. These results suggest that Osterix is a novel target of PKA, and PKA modulates osteoblast differentiation partially through the regulation of Osterix.


Assuntos
Remodelação Óssea/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Osteoblastos/citologia , Osteogênese/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Fosforilação , Fator de Transcrição Sp7 , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional
15.
FEBS J ; 281(16): 3656-66, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24961731

RESUMO

Runx2 plays essential roles in bone formation and chondrocyte maturation. Akt promotes osteoblast differentiation induced by the bone morphogenetic proteins BMP2 and enhances the function and transcriptional activity of Runx2. However, the precise molecular mechanism underlying the relationship between Runx2 and Akt is not well understood. In this study, we examined the role of Akt in regulating Runx2 function. We found that Akt increases the stability of Runx2 protein. However, the level of Runx2 mRNA was not affected by Akt, and we did not find any evidence for direct modification of Runx2 by Akt. Instead, we found evidence that Akt induces the phosphorylation of the Smad ubiquitination regulatory factor Smurf2 and decreases the level of Smurf2 protein through ubiquitin/proteasome-mediated degradation of Smurf2. Akt also alleviates Smurf2-mediated suppression of Runx2 transcriptional activity. Taken together, our results suggest that Akt regulates osteoblast differentiation, at least in part, by enhancing the protein stability and transcriptional activity of Runx2 through regulation of ubiquitin/proteasome-mediated degradation of Smurf2.


Assuntos
Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Células HEK293 , Humanos , Camundongos , Fosforilação , Ligação Proteica , Mapas de Interação de Proteínas , Estabilidade Proteica , Proteólise , Transcrição Gênica , Ubiquitinação
16.
J Cell Biochem ; 115(11): 2004-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24924519

RESUMO

Protein kinase A (PKA), a serine/threonine kinase, regulates bone formation, and enhances Bone morphogenetic protein (BMP)-induced osteoblast differentiation. However, the mechanisms of how PKA controls the cellular response to BMP are not well known. We investigated the effects of modulating PKA activity during BMP2-induced osteoblast differentiation, and found that PKA regulates the function of Dlx3. Dlx3 plays crucial roles in osteoblast differentiation and it is expressed in most skeletal elements during development. We found that PKA activation increases BMP2-induced expression of Dlx3 protein, and enhances the protein stability, DNA binding, and transcriptional activity of Dlx3. In addition, PKA activation induces the phosphorylation of Dlx3 at consensus PKA phosphorylation target site(s). Lastly, substitution of serine 10 in Dlx3 to alanine significantly reduces, if not completely abolishes, the phosphorylation of Dlx3 and the regulation of Dlx3 function by PKA. These results suggest that Dlx3 is a novel target of PKA, and that PKA mediates BMP signaling during osteoblast differentiation, at least in part, by phosphorylating Dlx3 and modulating the protein stability and function of Dlx3.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Osteoblastos/fisiologia , Serina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Substituição de Aminoácidos , Animais , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular , Linhagem Celular , Colforsina/farmacologia , Células HEK293 , Proteínas de Homeodomínio/química , Humanos , Isoquinolinas/farmacologia , Camundongos , Fosforilação , Estabilidade Proteica , Sulfonamidas/farmacologia , Fatores de Transcrição/química
17.
Mol Cell Endocrinol ; 383(1-2): 103-10, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24325869

RESUMO

Runx2 is a major transcription factor that induces osteoblast differentiation by bone morphogenetic proteins (BMPs). Conversely, YY1 is a transcription factor that inhibits BMP2-induced cell differentiation. Until now, there has been no understanding of how osteoblast differentiation by Runx2 and YY1 is regulated. In this study we focused on the relationship between Runx2 and YY1. We confirmed that alkaline phosphatase staining is repressed by YY1. Runx2 interacted with YY1 through Runt and the C-terminus domain of Runx2. YY1 markedly repressed the Runx2-mediated enhancement of transcriptional activity on the osteocalcin and alkaline phosphatase promoters. Knockdown of YY1 enhanced BMP2- and Runx2-induced osteoblast differentiation. YY1 decreased Runx2 DNA binding affinity. The results indicate that YY1 represses osteoblast differentiation by an interaction with Runx2 and inhibits the transcriptional activity of Runx2.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica , Osteoblastos/metabolismo , Transcrição Gênica , Fator de Transcrição YY1/genética , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/genética , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Células HEK293 , Humanos , Camundongos , Mioblastos/citologia , Mioblastos/metabolismo , Osteoblastos/citologia , Osteocalcina/genética , Osteocalcina/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Fator de Transcrição YY1/metabolismo
18.
Arch Pharm Res ; 37(2): 276-83, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23775473

RESUMO

Bromopropane (BP) is a halogenated alkan compound used in various industries as chemical intermediates, extraction solvents, and degreasing compounds. Halogenated alkan compounds can damage the nervous system, immune system, and hematopoietic and reproductive functions in animals and humans. However, the effect of BPs on bone formation has not yet been examined. This study examined the effects of BPs on osteoblast differentiation and analyzed the mechanisms involved in C2C12, mesenchymal stem cells. BPs dose dependently reduced the alkaline phosphatase activity, expression levels and promoter activity of bone marker genes. Additionally, 1,2-dibromopropane (1,2-DBP) significantly reduced the levels and transcriptional activity of Runx2 and Osterix, major bone transcription factors, in BMP2 induced C2C12 cells. Furthermore, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) were significantly inhibited by 1,2-DBP. These results demonstrate that BPs inhibit osteoblast differentiation by suppressing Runx2 and Osterix through the ERK/JNK pathway.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hidrocarbonetos Bromados/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Animais , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Propano/análogos & derivados , Propano/farmacologia , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos
19.
FEBS Lett ; 587(22): 3640-7, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24113655

RESUMO

Peptidyl-prolyl isomerase 1 (Pin1) is the only enzyme known to catalyze isomerization of the pSer/Thr-Pro peptide bond. Pin1 induces conformational change of substrates and subsequently regulates diverse cellular processes. However, its role in osteoblast differentiation is not well understood. Here we show that Pin1 enhances osteoblast differentiation. Pin1 interacts and affects the protein stability and transcriptional activity of an important osteogenic transcriptional factor Runx2. Our results indicate that this regulation is likely due to suppression of poly-ubiquitination-mediated proteasomal degradation of Runx2. Our current finding suggests that Pin1 is a novel regulator of osteoblast differentiation that acts through the regulation of Runx2 function.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos/enzimologia , Peptidilprolil Isomerase/fisiologia , Animais , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/química , Regulação da Expressão Gênica , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Peptidilprolil Isomerase de Interação com NIMA , Osteoblastos/fisiologia , Peptidilprolil Isomerase/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Proteólise , Transcrição Gênica
20.
Biochem Biophys Res Commun ; 425(4): 800-5, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22885182

RESUMO

Distal-less 3 (DLX3) is a highly conserved homeobox containing transcription factor. DLX3 is specifically expressed in osteoblasts and osteocytes of all developing bones. DLX3 is essential for osteoblast differentiation and skeletal morphogenesis and acts as a scaffold for nucleic acids and regulatory factors involved in skeletal gene expression. Akt can be activated by several osteogenic signaling molecules, but its precise function and downstream targets in bone development are unknown. In this report, we investigated a potential regulation of Dlx3 function by Akt1. We found that Akt1 phosphorylates Dlx3 and Akt1 activation increases protein stability, osteogenic activity and transcriptional activity of Dlx3. Also, BMP2 was shown to increase the protein level of Dlx3 in an Akt1 activity-dependent manner. Conversely, inhibition of Akt1 by the Akt inhibitor decreases the protein levels of Dlx3. These results suggest that Dlx3 is a novel target of Akt1 and the activity of Dlx3 could be modulated by a novel mechanism involving Akt1 during osteoblast differentiation.


Assuntos
Proteínas de Homeodomínio/metabolismo , Osteogênese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células HEK293 , Humanos , Camundongos , Osteoblastos/metabolismo , Fosforilação , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA