Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Heliyon ; 9(6): e16962, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37484408

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-36068970

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Exosomes , Liver Neoplasms , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Exosomes/metabolism , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Liver Neoplasms/metabolism , Mice , Models, Animal , Serine/metabolism
3.
Drug Deliv ; 29(1): 2621-2631, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35941835

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Extracellular Vesicles , Nanoparticles , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Mice , Mice, Inbred BALB C
4.
Chem Commun (Camb) ; 58(12): 1978-1981, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35045150

ABSTRACT

CRISPR-Cas12a has been widely used in genome editing and nucleic acid detection. In both of these applications, Cas12a cleaves target DNA in a divalent metal ion-dependent manner. However, when and how metal ions contribute to the cleavage reaction is unclear. Here, using a single-molecule FRET assay, we reveal that these metal ions are necessary for stabilising cleavage-competent conformations and that they are easily exchangeable, suggesting that they are dynamically coordinated.


Subject(s)
CRISPR-Cas Systems , DNA/genetics , Metals/chemistry , Cations, Divalent , Fluorescence Resonance Energy Transfer/methods , Gene Editing/methods , Nucleic Acid Conformation
5.
Pharmaceutics ; 13(10)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34683982

ABSTRACT

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.

6.
ACS Nano ; 15(5): 8492-8506, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33973786

ABSTRACT

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.


Subject(s)
Doxorubicin , Nanoparticles , Animals , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Delivery Systems , Drug Liberation , Macrophages , Mice , Mice, Inbred BALB C , Phototherapy
7.
Nat Commun ; 9(1): 2777, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30018371

ABSTRACT

Cas12a (also called Cpf1) is a representative type V-A CRISPR effector RNA-guided DNA endonuclease, which provides an alternative to type II CRISPR-Cas9 for genome editing. Previous studies have revealed that Cas12a has unique features distinct from Cas9, but the detailed mechanisms of target searching and DNA cleavage by Cas12a are still unclear. Here, we directly observe this entire process by using single-molecule fluorescence assays to study Cas12a from Acidaminococcus sp. (AsCas12a). We determine that AsCas12a ribonucleoproteins search for their on-target site by a one-dimensional diffusion along elongated DNA molecules and induce cleavage in the two DNA strands in a well-defined order, beginning with the non-target strand. Furthermore, the protospacer-adjacent motif (PAM) for AsCas12a makes only a limited contribution of DNA unwinding during R-loop formation and shows a negligible role in the process of DNA cleavage, in contrast to the Cas9 PAM.


Subject(s)
Acidaminococcus/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , DNA/genetics , RNA, Guide, Kinetoplastida/genetics , Acidaminococcus/enzymology , Base Pairing , Base Sequence , CRISPR-Associated Protein 9/metabolism , Cloning, Molecular , DNA/metabolism , DNA Cleavage , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Editing , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes , RNA, Guide, Kinetoplastida/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
J Cell Biochem ; 119(1): 748-757, 2018 01.
Article in English | MEDLINE | ID: mdl-28657691

ABSTRACT

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.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Ethanol/pharmacology , Lithospermum/chemistry , Osteoblasts/cytology , Sp7 Transcription Factor/genetics , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Remodeling , Cell Differentiation/drug effects , Cell Line , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mice , Naphthoquinones/chemistry , Osteoblasts/drug effects , Osteoblasts/metabolism , Plant Extracts/pharmacology , Sp7 Transcription Factor/metabolism , Transcription, Genetic/drug effects
9.
Int J Mol Med ; 38(2): 610-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27353217

ABSTRACT

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.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Ethanol/chemistry , Gene Expression Regulation/drug effects , Lithospermum/chemistry , Osteoblasts/metabolism , Osteogenesis/drug effects , Plant Extracts/pharmacology , Transcription Factors/genetics , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , HEK293 Cells , Humans , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/genetics , Sp7 Transcription Factor , Transcription, Genetic/drug effects
10.
Bioorg Med Chem ; 24(4): 789-801, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26780832

ABSTRACT

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.


Subject(s)
Androgen Receptor Antagonists/chemical synthesis , Antineoplastic Agents/chemical synthesis , Drug Design , Prostate/drug effects , Receptors, Androgen/chemistry , Amino Acid Motifs , Androgen Receptor Antagonists/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , Gene Expression , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Molecular Docking Simulation , Molecular Sequence Data , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Niacinamide/chemistry , Prostate/metabolism , Protein Structure, Secondary , Pyrazinamide/chemistry , Pyrimidines/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship
11.
Mol Cells ; 39(2): 156-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26674964

ABSTRACT

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.


Subject(s)
Estradiol/metabolism , Estrogen Receptor alpha/genetics , Homeodomain Proteins/genetics , Myoblasts/metabolism , Osteoblasts/metabolism , Transcription Factors/genetics , Animals , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Cell Line , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Ligands , Luciferases/genetics , Luciferases/metabolism , Mice , Myoblasts/cytology , Myoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
12.
Mol Cell Endocrinol ; 413: 217-27, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26159900

ABSTRACT

Yin Yang 1 (YY1) is an ubiquitously distributed transcription factor that belongs to the GLI-Kruppel class of zinc finger proteins. The mechanism by which YY1 regulates adipocyte differentiation remains unclear. In this study, we investigated the functional role of YY1 during adipocyte differentiation. During the early stage, YY1 gene and protein expression was transiently downregulated upon the induction of differentiation, however, it was consistently induced during the later stage. YY1 overexpression decreased adipocyte differentiation and blocked cell differentiation at the preadipocyte stage, while YY1 knockdown by RNA interference increased adipocyte differentiation. YY1 physically interacted with PPARγ (Peroxisome proliferator-activated receptor gamma) and C/EBPß (CCAAT/enhancer-binding protein beta) respectively in 3T3-L1 cells. Through its interaction with PPARγ, YY1 directly decreased PPARγ transcriptional activity. YY1 ectopic expression prevented C/EBPß from binding to the PPARγ promoter, resulting in the downregulation of PPARγ transcriptional activity. These results indicate that YY1 repressed adipocyte differentiation by repressing the activity of adipogenic transcriptional factors in 3T3-L1 cells.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/physiology , Gene Expression Regulation/physiology , Transcription, Genetic/physiology , YY1 Transcription Factor/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , YY1 Transcription Factor/genetics
13.
Bone ; 75: 201-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25744063

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/physiology , Osteoblasts/cytology , Proto-Oncogene Proteins c-cbl/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Humans , Immunoblotting , Immunoprecipitation , Mice , Osteogenesis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sp7 Transcription Factor , Transfection , Ubiquitination
14.
Mol Cell Endocrinol ; 407: 85-97, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25802190

ABSTRACT

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.


Subject(s)
Epithelial Cells/metabolism , Myoblasts/metabolism , Osteoblasts/metabolism , Transcription Factors/genetics , src-Family Kinases/genetics , Animals , CSK Tyrosine-Protein Kinase , Cell Differentiation , Cell Line , Cell Proliferation , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , DNA/biosynthesis , DNA/genetics , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , Humans , Indoles/pharmacology , Luciferases/genetics , Luciferases/metabolism , Mice , Myoblasts/cytology , Myoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/genetics , Protein Kinase Inhibitors/pharmacology , Protein Stability , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sp7 Transcription Factor , Sulfonamides/pharmacology , Transcription Factors/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
15.
Biochem Biophys Res Commun ; 453(3): 557-62, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25285640

ABSTRACT

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.


Subject(s)
Proto-Oncogene Proteins c-cbl/physiology , Sirtuin 2/physiology , Cell Line , Humans , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Ubiquitination
16.
Biochem Biophys Res Commun ; 450(2): 1120-5, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24996174

ABSTRACT

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.


Subject(s)
Sirtuin 2/metabolism , src-Family Kinases/metabolism , CSK Tyrosine-Protein Kinase , HEK293 Cells , Humans , MCF-7 Cells , Phosphorylation
17.
J Cell Biochem ; 115(10): 1808-15, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24905700

ABSTRACT

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.


Subject(s)
Bone Remodeling/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Osteoblasts/cytology , Osteogenesis/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Line , HEK293 Cells , Humans , Mice , Phosphorylation , Sp7 Transcription Factor , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation
18.
FEBS J ; 281(16): 3656-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24961731

ABSTRACT

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.


Subject(s)
Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/physiology , Proto-Oncogene Proteins c-akt/physiology , Ubiquitin-Protein Ligases/physiology , Animals , HEK293 Cells , Humans , Mice , Phosphorylation , Protein Binding , Protein Interaction Maps , Protein Stability , Proteolysis , Transcription, Genetic , Ubiquitination
19.
J Cell Biochem ; 115(11): 2004-11, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24924519

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Osteoblasts/physiology , Serine/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Substitution , Animals , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation , Cell Line , Colforsin/pharmacology , HEK293 Cells , Homeodomain Proteins/chemistry , Humans , Isoquinolines/pharmacology , Mice , Phosphorylation , Protein Stability , Sulfonamides/pharmacology , Transcription Factors/chemistry
20.
Mol Cell Endocrinol ; 383(1-2): 103-10, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24325869

ABSTRACT

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.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation , Osteoblasts/metabolism , Transcription, Genetic , YY1 Transcription Factor/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Cell Line , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , HEK293 Cells , Humans , Mice , Myoblasts/cytology , Myoblasts/metabolism , Osteoblasts/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Signal Transduction , YY1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...