Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 52(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38874019

ABSTRACT

2',3',4'­trihydroxyflavone (2­D08), a SUMO E2 inhibitor, has several biological functions, including anticancer activity, but its effects on uterine leiomyosarcoma (Ut­LMS) are unknown. The anticancer activity of 2­D08 was explored in an in vitro model using SK­LMS­1 and SK­UT­1B cells (human Ut­LMS cells). Treatment with 2­D08 inhibited cell viability in a dose­ and time­dependent manner and significantly inhibited the colony­forming ability of Ut­LMS cells. In SK­UT­1B cells treated with 2­D08, flow cytometric analysis revealed a slight increase in apoptotic rates, while cell cycle progression remained unaffected. Western blotting revealed elevated levels of RIP1, indicating induction of necrosis, but LC3B levels remained unchanged, suggesting no effect on autophagy. A lactate dehydrogenase (LDH) assay confirmed increased LDH release, further supporting the induction of apoptosis and necrosis by 2­D08 in SK­UT­1B cells. 2­D08­induced production of reactive oxygen species and apoptosis progression were observed in SK­LMS­1 cells. Using Ki67 staining and bromodeoxyuridine assays, it was found that 2­D08 suppressed proliferation in SK­LMS­1 cells, while treatment for 48 h led to cell­cycle arrest. 2­D08 upregulated p21 protein expression in SK­LMS­1 cells and promoted apoptosis through caspase­3. Evaluation of α­SM­actin, calponin 1 and TAGLN expression indicated that 2­D08 did not directly initiate smooth muscle phenotypic switching in SK­LMS­1 cells. Transcriptome analysis on 2­D08­treated SK­LMS­1 cells identified significant differences in gene expression and suggested that 2­D08 modulates cell­cycle­ and apoptosis­related pathways. The analysis identified several differentially expressed genes and significant enrichment for biological processes related to DNA replication and molecular functions associated with the apoptotic process. It was concluded that 2­D08 exerts antitumor effects in Ut­LMS cells by modulating multiple signaling pathways and that 2­D08 may be a promising candidate for the treatment of human Ut­LMS. The present study expanded and developed knowledge regarding Ut­LMS management and indicated that 2­D08 represents a notable finding in the exploration of fresh treatment options for such cancerous tumors.


Subject(s)
Apoptosis , Cell Proliferation , Leiomyosarcoma , Uterine Neoplasms , Humans , Leiomyosarcoma/drug therapy , Leiomyosarcoma/pathology , Leiomyosarcoma/metabolism , Female , Uterine Neoplasms/drug therapy , Uterine Neoplasms/pathology , Uterine Neoplasms/metabolism , Cell Line, Tumor , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Flavones/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Autophagy/drug effects
2.
J Muscle Res Cell Motil ; 42(2): 193-202, 2021 06.
Article in English | MEDLINE | ID: mdl-34142311

ABSTRACT

SUMOylation is one of the post-translational modifications that involves the covalent attachment of the small ubiquitin-like modifier (SUMO) to the substrate. SUMOylation regulates multiple biological processes, including myoblast proliferation, differentiation, and apoptosis. 2-D08 is a synthetically available flavone, which acts as a potent cell-permeable SUMOylation inhibitor. Its mechanism of action involves preventing the transfer of SUMO from the E2 thioester to the substrate without influencing SUMO-activating enzyme E1 (SAE-1/2) or E2 Ubc9-SUMO thioester formation. However, both the effects and mechanisms of 2-D08 on C2C12 myoblast cells remain unclear. In the present study, we found that treatment with 2-D08 inhibits C2C12 cell proliferation and differentiation. We confirmed that 2-D08 significantly hampers the viability of C2C12 cells. Additionally, it inhibited myogenic differentiation, decreasing myosin heavy chain (MHC), MyoD, and myogenin expression. Furthermore, we confirmed that 2-D08-mediated anti-myogenic effects impair myoblast differentiation and myotube formation, reducing the number of MHC-positive C2C12 cells. In addition, we found that 2-D08 induces the activation of ErK1/2 and the degradation of MyoD and myogenin in C2C12 cells. Taken together, these results indicated that 2-D08 treatment results in the deregulated proliferation and differentiation of myoblasts. However, further research is needed to investigate the long-term effects of 2-D08 on skeletal muscles.


Subject(s)
Biological Phenomena , Proteasome Endopeptidase Complex , Cell Differentiation , Cell Proliferation , MyoD Protein , Myoblasts/metabolism , Myogenin/metabolism , Signal Transduction
3.
Exp Mol Med ; 53(2): 250-263, 2021 02.
Article in English | MEDLINE | ID: mdl-33564100

ABSTRACT

The demethylation of histone lysine residues, one of the most important modifications in transcriptional regulation, is associated with various physiological states. KDM2B is a demethylase of histones H3K4, H3K36, and H3K79 and is associated with the repression of transcription. Here, we present a novel mechanism by which KDM2B demethylates serum response factor (SRF) K165 to negatively regulate muscle differentiation, which is counteracted by the histone methyltransferase SET7. We show that KDM2B inhibited skeletal muscle differentiation by inhibiting the transcription of SRF-dependent genes. Both KDM2B and SET7 regulated the balance of SRF K165 methylation. SRF K165 methylation was required for the transcriptional activation of SRF and for the promoter occupancy of SRF-dependent genes. SET7 inhibitors blocked muscle cell differentiation. Taken together, these data indicate that SRF is a nonhistone target of KDM2B and that the methylation balance of SRF as maintained by KDM2B and SET7 plays an important role in muscle cell differentiation.


Subject(s)
Cell Differentiation , F-Box Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Muscle, Skeletal/metabolism , Serum Response Factor/metabolism , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Line , Cells, Cultured , F-Box Proteins/genetics , Gene Expression Regulation , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Methylation , Models, Biological , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Protein Binding , Response Elements , Transcription, Genetic
4.
Mol Ther Nucleic Acids ; 22: 627-639, 2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33230462

ABSTRACT

Vascular calcification, the ectopic deposition of calcium in blood vessels, develops in association with various metabolic diseases and atherosclerosis and is an independent predictor of morbidity and mortality associated with these diseases. Herein, we report that reduction of microRNA-27a-3p (miR-27a-3p) causes an increase in activating transcription factor 3 (ATF3), a novel osteogenic transcription factor, in vascular smooth muscle cells. Both microRNA (miRNA) and mRNA microarrays were performed with rat vascular smooth muscle cells, and reciprocally regulated pairs of miRNA and mRNA were selected after bioinformatics analysis. Inorganic phosphate significantly reduced the expression of miR-27a-3p in A10 cells. The transcript level was also reduced in vitamin D3-administered mouse aortas. miR-27a-3p mimic reduced calcium deposition, whereas miR-27a-3p inhibitor increased it. The Atf3 mRNA level was upregulated in a cellular vascular calcification model, and miR-27a-3p reduced the Atf3 mRNA and protein levels. Transfection with Atf3 could recover the miR-27a-3p-induced reduction of calcium deposition. Our results suggest that reduction of miR-27a-3p may contribute to the development of vascular calcification by de-repression of ATF3.

5.
J Cell Mol Med ; 24(18): 10542-10550, 2020 09.
Article in English | MEDLINE | ID: mdl-32783377

ABSTRACT

Calcium deposition in vascular smooth muscle cells (VSMCs) is a form of ectopic ossification in blood vessels. It can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that the microRNA miR-134-5p potentiates inorganic phosphate (Pi)-induced calcium deposition in VSMCs by inhibiting histone deacetylase 5 (HDAC5). Using miRNA microarray analysis of Pi-treated rat VSMCs, we first selected miR-134-5p for further evaluation. Quantitative RT-PCR confirmed that miR-134-5p was increased in Pi-treated A10 cells, a rat VSMC line. Transfection of miR-134-5p mimic potentiated the Pi-induced increase in calcium contents. miR-134-5p increased the amounts of bone runt-related transcription factor 2 (RUNX2) protein and bone morphogenic protein 2 (BMP2) mRNA in the presence of Pi but decreased the expression of osteoprotegerin (OPG). Bioinformatic analysis showed that the HDAC5 3'untranslated region (3'UTR) was one of the targets of miR-134-5p. The luciferase construct containing the 3'UTR of HDAC5 was down-regulated by miR-134-5p mimic in a dose-dependent manner in VSMCs. Overexpression of HDAC5 mitigated the calcium deposition induced by miR-134-5p. Our results suggest that a Pi-induced increase of miR-134-5p may cause vascular calcification through repression of HDAC5.


Subject(s)
Calcium/metabolism , Histone Deacetylases/drug effects , MicroRNAs/physiology , Myocytes, Smooth Muscle/metabolism , Vascular Calcification/etiology , 3' Untranslated Regions , Animals , Aorta, Thoracic/cytology , Cell Line , Computer Simulation , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/immunology , Down-Regulation , Gene Expression Regulation , Genes, Reporter , Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , MicroRNAs/genetics , Microarray Analysis , Muscle, Smooth, Vascular/cytology , Osteoprotegerin/biosynthesis , Osteoprotegerin/genetics , Phosphates/toxicity , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/prevention & control
7.
Cardiovasc Res ; 115(13): 1850-1860, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-30596969

ABSTRACT

AIMS: Previously, we reported that phosphorylation of histone deacetylase 2 (HDAC2) and the resulting activation causes cardiac hypertrophy. Through further study of the specific binding partners of phosphorylated HDAC2 and their mechanism of regulation, we can better understand how cardiac hypertrophy develops. Thus, in the present study, we aimed to elucidate the function of one such binding partner, heat shock protein 70 (HSP70). METHODS AND RESULTS: Primary cultures of rat neonatal ventricular cardiomyocytes and H9c2 cardiomyoblasts were used for in vitro cellular experiments. HSP70 knockout (KO) mice and transgenic (Tg) mice that overexpress HSP70 in the heart were used for in vivo analysis. Peptide-precipitation and immunoprecipitation assay revealed that HSP70 preferentially binds to phosphorylated HDAC2 S394. Forced expression of HSP70 increased phosphorylation of HDAC2 S394 and its activation, but not that of S422/424, whereas knocking down of HSP70 reduced it. However, HSP70 failed to phosphorylate HDAC2 in the cell-free condition. Phosphorylation of HDAC2 S394 by casein kinase 2α1 enhanced the binding of HSP70 to HDAC2, whereas dephosphorylation induced by the catalytic subunit of protein phosphatase 2A (PP2CA) had the opposite effect. HSP70 prevented HDAC2 dephosphorylation by reducing the binding of HDAC2 to PP2CA. HSP70 KO mouse hearts failed to phosphorylate S394 HDAC2 in response to isoproterenol infusion, whereas Tg overexpression of HSP70 increased the phosphorylation and activation of HDAC2. 2-Phenylethynesulfonamide (PES), an HSP70 inhibitor, attenuated cardiac hypertrophy induced either by phenylephrine in neonatal ventricular cardiomyocytes or by aortic banding in mice. PES reduced HDAC2 S394 phosphorylation and its activation by interfering with the binding of HSP70 to HDAC2. CONCLUSION: These results demonstrate that HSP70 specifically binds to S394-phosphorylated HDAC2 and maintains its phosphorylation status, which results in HDAC2 activation and the development of cardiac hypertrophy. Inhibition of HSP70 has possible application as a therapeutic.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Histone Deacetylase 2/metabolism , Hypertrophy, Left Ventricular/enzymology , Myocytes, Cardiac/enzymology , Ventricular Function, Left , Ventricular Remodeling , Animals , Binding Sites , Cell Line , Disease Models, Animal , Enzyme Activation , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/deficiency , HSP70 Heat-Shock Proteins/genetics , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Sulfonamides/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
8.
Exp Mol Med ; 50(7): 1-14, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30050113

ABSTRACT

Cardiac hypertrophy occurs in response to increased hemodynamic demand and can progress to heart failure. Identifying the key regulators of this process is clinically important. Though it is thought that the phosphorylation of histone deacetylase (HDAC) 2 plays a crucial role in the development of pathological cardiac hypertrophy, the detailed mechanism by which this occurs remains unclear. Here, we performed immunoprecipitation and peptide pull-down assays to characterize the functional complex of HDAC2. Protein phosphatase (PP) 2 A was confirmed as a binding partner of HDAC2. PPP2CA, the catalytic subunit of PP2A, bound to HDAC2 and prevented its phosphorylation. Transient overexpression of PPP2CA specifically regulated both the phosphorylation of HDAC2 S394 and hypertrophy-associated HDAC2 activation. HDAC2 S394 phosphorylation was increased in a dose-dependent manner by PP2A inhibitors. Hypertrophic stresses, such as phenylephrine in vitro or pressure overload in vivo, caused PPP2CA to dissociate from HDAC2. Forced expression of PPP2CA negatively regulated the hypertrophic response, but PP2A inhibitors provoked hypertrophy. Adenoviral delivery of a phosphomimic HDAC2 mutant, adenovirus HDAC2 S394E, successfully blocked the anti-hypertrophic effect of adenovirus-PPP2CA, implicating HDAC2 S394 phosphorylation as a critical event for the anti-hypertrophic response. PPP2CA transgenic mice were protected against isoproterenol-induced cardiac hypertrophy and subsequent cardiac fibrosis, whereas simultaneous expression of HDAC2 S394E in the heart did induce hypertrophy. Taken together, our results suggest that PP2A is a critical regulator of HDAC2 activity and pathological cardiac hypertrophy and is a promising target for future therapeutic interventions.


Subject(s)
Cardiomegaly/metabolism , Histone Deacetylase 2/metabolism , Myocytes, Cardiac/metabolism , Protein Phosphatase 2/metabolism , Animals , Cell Line , Cells, Cultured , Histone Deacetylase 2/genetics , Mice , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
9.
Exp Mol Med ; 50(1): e427, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29328071

ABSTRACT

Sumoylation, the conjugation of a small ubiquitin-like modifier (SUMO) protein to a target, has diverse cellular effects. However, the functional roles of the SUMO modification during myogenesis have not been fully elucidated. Here, we report that basal sumoylation of histone deacetylase 1 (HDAC1) enhances the deacetylation of MyoD in undifferentiated myoblasts, whereas further sumoylation of HDAC1 contributes to switching its binding partners from MyoD to Rb to induce myocyte differentiation. Differentiation in C2C12 skeletal myoblasts induced new immunoblot bands above HDAC1 that were gradually enhanced during differentiation. Using SUMO inhibitors and sumoylation assays, we showed that the upper band was caused by sumoylation of HDAC1 during differentiation. Basal deacetylase activity was not altered in the SUMO modification-resistant mutant HDAC1 K444/476R (HDAC1 2R). Either differentiation or transfection of SUMO1 increased HDAC1 activity that was attenuated in HDAC1 2R. Furthermore, HDAC1 2R failed to deacetylate MyoD. Binding of HDAC1 to MyoD was attenuated by K444/476R. Binding of HDAC1 to MyoD was gradually reduced after 2 days of differentiation. Transfection of SUMO1 induced dissociation of HDAC1 from MyoD but potentiated its binding to Rb. SUMO1 transfection further attenuated HDAC1-induced inhibition of muscle creatine kinase luciferase activity that was reversed in HDAC1 2R. HDAC1 2R failed to inhibit myogenesis and muscle gene expression. In conclusion, HDAC1 sumoylation plays a dual role in MyoD signaling: enhancement of HDAC1 deacetylation of MyoD in the basally sumoylated state of undifferentiated myoblasts and dissociation of HDAC1 from MyoD during myogenesis.


Subject(s)
Histone Deacetylase 1/metabolism , MyoD Protein/metabolism , Myoblasts/metabolism , Acetylation , Animals , Cell Differentiation/physiology , Cell Line , Histone Deacetylase 1/genetics , Mice , Muscle Development , Muscle, Skeletal/cytology , Myogenin/genetics , Promoter Regions, Genetic , Signal Transduction , Sumoylation
10.
Nat Commun ; 7: 10492, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26832969

ABSTRACT

Vascular calcification (VC) is often associated with cardiovascular and metabolic diseases. However, the molecular mechanisms linking VC to these diseases have yet to be elucidated. Here we report that MDM2-induced ubiquitination of histone deacetylase 1 (HDAC1) mediates VC. Loss of HDAC1 activity via either chemical inhibitor or genetic ablation enhances VC. HDAC1 protein, but not mRNA, is reduced in cell and animal calcification models and in human calcified coronary artery. Under calcification-inducing conditions, proteasomal degradation of HDAC1 precedes VC and it is mediated by MDM2 E3 ubiquitin ligase that initiates HDAC1 K74 ubiquitination. Overexpression of MDM2 enhances VC, whereas loss of MDM2 blunts it. Decoy peptide spanning HDAC1 K74 and RG 7112, an MDM2 inhibitor, prevent VC in vivo and in vitro. These results uncover a previously unappreciated ubiquitination pathway and suggest MDM2-mediated HDAC1 ubiquitination as a new therapeutic target in VC.


Subject(s)
Histone Deacetylase 1/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Vascular Calcification/metabolism , Animals , Calcium , Gene Expression Regulation , Histone Deacetylase 1/genetics , Humans , Male , Mice , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins c-mdm2/genetics , Rats , Ubiquitination
11.
Cell Signal ; 26(10): 2240-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25025573

ABSTRACT

Skeletal muscle atrophy results from the net loss of muscular proteins and organelles and is caused by pathologic conditions such as nerve injury, immobilization, cancer, and other metabolic diseases. Recently, ubiquitination-mediated degradation of skeletal-muscle-specific transcription factors was shown to be involved in muscle atrophy, although the mechanisms have yet to be defined. Here we report that ret finger protein (RFP), also known as TRIM27, works as an E3 ligase in Pax7-induced degradation of MyoD. Muscle injury induced by sciatic nerve transection up-regulated RFP and RFP physically interacted with both Pax7 and MyoD. RFP and Pax7 synergistically reduced the protein amounts of MyoD but not the mRNA. RFP-induced reduction of MyoD protein was blocked by proteasome inhibitors. The Pax7-induced reduction MyoD was attenuated by RFP siRNA and by MG132, a proteasome inhibitor. RFPΔR, an RFP construct that lacks the RING domain, failed to reduce MyoD amounts. RFP ubiquitinated MyoD, but RFPΔR failed to do so. Forced expression of RFP, but not RFPΔR, enhanced Pax7-induced ubiquitination of MyoD, whereas RFP siRNA blocked the ubiquitination. Sciatic nerve injury-induced muscle atrophy as well the reduction in MyoD was attenuated in RFP knockout mice. Taken together, our results show that RFP works as a novel E3 ligase in the Pax7-mediated degradation of MyoD in response to skeletal muscle atrophy.


Subject(s)
DNA-Binding Proteins/metabolism , Muscular Atrophy/pathology , MyoD Protein/metabolism , Nuclear Proteins/metabolism , PAX7 Transcription Factor/metabolism , Animals , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Disease Models, Animal , HEK293 Cells , Humans , Leupeptins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Atrophy/metabolism , MyoD Protein/chemistry , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , PAX7 Transcription Factor/chemistry , Protease Inhibitors/pharmacology , Protein Binding , Proteolysis/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Regeneration , Ubiquitin-Protein Ligases , Ubiquitination/drug effects
12.
Circ Res ; 115(5): 493-503, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015078

ABSTRACT

RATIONALE: Small heterodimer partner (SHP; NR0B2) is an atypical orphan nuclear receptor that lacks a conventional DNA-binding domain. Through interactions with other transcription factors, SHP regulates diverse biological events, including glucose metabolism in liver. However, the role of SHP in adult heart diseases has not yet been demonstrated. OBJECTIVE: We aimed to investigate the role of SHP in adult heart in association with cardiac hypertrophy. METHODS AND RESULTS: The roles of SHP in cardiac hypertrophy were tested in primary cultured cardiomyocytes and in animal models. SHP-null mice showed a hypertrophic phenotype. Hypertrophic stresses repressed the expression of SHP, whereas forced expression of SHP blocked the development of hypertrophy in cardiomyocytes. SHP reduced the protein amount of Gata6 and, by direct physical interaction with Gata6, interfered with the binding of Gata6 to GATA-binding elements in the promoter regions of natriuretic peptide precursor type A. Metformin, an antidiabetic agent, induced SHP and suppressed cardiac hypertrophy. The metformin-induced antihypertrophic effect was attenuated either by SHP small interfering RNA in cardiomyocytes or in SHP-null mice. CONCLUSIONS: These results establish SHP as a novel antihypertrophic regulator that acts by interfering with GATA6 signaling. SHP may participate in the metformin-induced antihypertrophic response.


Subject(s)
Cardiomegaly/prevention & control , GATA6 Transcription Factor/metabolism , Myocytes, Cardiac/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Binding Sites , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Disease Models, Animal , GATA6 Transcription Factor/genetics , Gene Expression Regulation , Genotype , HEK293 Cells , Humans , Male , Metformin/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenotype , Promoter Regions, Genetic , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/drug effects , Transfection
13.
Atherosclerosis ; 225(1): 41-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22898620

ABSTRACT

OBJECTIVE: Sulforaphane, a naturally occurring organosulfur compound in broccoli, has chemopreventive properties in cancer. However, the effects of sulforaphane in vascular diseases have not been examined. We therefore aimed to investigate the effects of sulforaphane on vascular smooth muscle cell (VSMC) proliferation and neointimal formation and the related mechanisms. METHODS: The expression of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) was examined in VSMCs. The nuclear translocation of nuclear factor-κB (NF-κB) and GATA6 expression was examined in VSMCs and in a carotid artery injury model by Western blot and immunohistochemistry. We also investigated whether local delivery of sulforaphane affected neointimal formation. RESULTS: Sulforaphane inhibited the mRNA and protein expression of VCAM-1 induced by tumor necrosis factor (TNF)-α in VSMCs. Treatment of VSMCs with sulforaphane blocked TNF-α-induced IκBα degradation and NF-κB p65 and GATA6 expression. Furthermore, NF-κB p65 and GATA6 expression were reduced in sulforaphane-treated carotid injury sections. Notably, binding of GATA6 to the VCAM-1 promoter was dramatically reduced by sulforaphane. The MTT, BrdU incorporation, and in vitro scratch assays revealed that the proliferation and migration of VSMCs were reduced by sulforaphane. Furthermore, local administration of sulforaphane significantly reduced neointima formation 14 days after vascular injury in rats. CONCLUSIONS: Our results indicate that sulforaphane inhibits neointima formation via targeting of adhesion molecules through the suppression of NF-κB/GATA6. Furthermore, sulforaphane regulates migration and proliferation in VSMCs. Sulforaphane may be a potential therapeutic agent for preventing restenosis after vascular injury.


Subject(s)
Thiocyanates/pharmacology , Animals , GATA6 Transcription Factor/biosynthesis , GATA6 Transcription Factor/metabolism , I-kappa B Proteins/biosynthesis , Intercellular Adhesion Molecule-1/biosynthesis , Isothiocyanates , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Neointima/prevention & control , RNA, Messenger/metabolism , Rats , Sulfoxides , Vascular Cell Adhesion Molecule-1/biosynthesis
14.
Atherosclerosis ; 222(1): 84-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22398275

ABSTRACT

OBJECTIVE: Previously, we reported that enhancer of polycomb1 (Epc1) induces skeletal muscle differentiation through the serum response factor (SRF). Considering that SRF plays a critical role in vascular smooth muscle cell (VSMC) differentiation, we expected that Epc1 also works in VSMCs. Here we examined the effect of Epc1 on neointima formation after arterial balloon injury and the underlying mechanism. METHODS: Epc1 expression was examined in carotid artery injury or VSMC models. Interaction with myocardin (Myocd), a master regulator of smooth muscle differentiation, was examined by immunoprecipitation or promoter analysis with smooth muscle (SM) 22α promoter. Finally, we investigated whether local delivery of Epc1 regulated neointimal formation after injury. RESULTS: Epc1 expression was down-regulated during proliferation induced by platelet-derived growth factor BB, whereas it was upregulated during differentiation in VSMCs. Forced expression of Epc1 induced VSMC differentiation. Epc1 physically interacted with Myocd to synergistically activate SM22α promoter activity. Transient transfection of Epc1 enhanced the physical interaction between Myocd and SRF, whereas that interaction was reduced when A10 cells were treated with siRNA for Epc1. Local delivery of Epc1 significantly reduced neointima formation induced by balloon injury. CONCLUSIONS: Our results indicate that Epc1 induces VSMC differentiation by interacting with Myocd to induce SRF-dependent smooth muscle genes. We propose that Epc1 acts as a novel negative regulator of neointima formation after carotid injury.


Subject(s)
Cell Differentiation/drug effects , Neointima , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Angioplasty, Balloon , Animals , Becaplermin , Carotid Artery Injuries/metabolism , Cell Differentiation/physiology , Cell Proliferation/drug effects , Down-Regulation , Mice , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Neointima/drug therapy , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Rats , Rats, Sprague-Dawley , Serum Response Factor/metabolism , Up-Regulation
15.
Circulation ; 123(21): 2392-403, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21576649

ABSTRACT

BACKGROUND: Cardiac hypertrophy is characterized by transcriptional reprogramming of fetal gene expression, and histone deacetylases (HDACs) are tightly linked to the regulation of those genes. We previously demonstrated that activation of HDAC2, 1 of the class I HDACs, mediates hypertrophy. Here, we show that casein kinase-2α1 (CK2α1)-dependent phosphorylation of HDAC2 S394 is required for the development of cardiac hypertrophy. METHODS AND RESULTS: Hypertrophic stimuli phosphorylated HDAC2 S394, which was necessary for its enzymatic activation, and therefore the development of hypertrophic phenotypes in rat neonatal cardiomyocytes or in isoproterenol-administered mice hearts. Transgenic mice overexpressing HDAC2 wild type exhibited cardiac hypertrophy, whereas those expressing phosphorylation-resistant HDAC2 S394A did not. Compared with that in age-matched normal human hearts, phosphorylation of HDAC2 S394 was dramatically increased in patients with hypertrophic cardiomyopathy. Hypertrophy-induced phosphorylation of HDAC2 S394 and its enzymatic activity were completely blocked either by CK2 blockers or by CK2α1 short interfering RNA. Hypertrophic stimuli led CK2α1 to be activated, and its chemical inhibitors blocked hypertrophy in both phenylephrine-treated cardiomyocytes and isoproterenol-administered mice. CK2α1-transgenic mice developed hypertrophy, which was attenuated by administration of trichostatin A, an HDAC inhibitor. Overexpression of CK2α1 caused hypertrophy in cardiomyocytes, whereas chemical inhibitors of both CK2 and HDAC as well as HDAC2 S394A blunted it. Hypertrophy in CK2α1-transgenic mice was exaggerated by crossing these mice with wild-type-HDAC2-overexpressing mice. By contrast, however, it was blocked when CK2α1-transgenic mice were crossed with HDAC2 S394A-transgenic mice. CONCLUSIONS: We have demonstrated a novel mechanism in the development of cardiac hypertrophy by which CK2 activates HDAC2 via phosphorylating HDAC2 S394.


Subject(s)
Cardiomegaly/enzymology , Casein Kinase II/metabolism , Heart Ventricles/enzymology , Histone Deacetylase 2/metabolism , Serine/metabolism , Alanine/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomyopathy, Hypertrophic/enzymology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Casein Kinase II/genetics , Enzyme Activation/genetics , Heart Ventricles/pathology , Histone Deacetylase 2/biosynthesis , Histone Deacetylase 2/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/genetics , Serine/genetics
16.
J Biol Chem ; 284(24): 16308-16316, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19359245

ABSTRACT

Skeletal muscle differentiation is well regulated by a series of transcription factors. We reported previously that enhancer of polycomb1 (Epc1), a chromatin protein, can modulate skeletal muscle differentiation, although the mechanisms of this action have yet to be defined. Here we report that Epc1 recruits both serum response factor (SRF) and p300 to induce skeletal muscle differentiation. Epc1 interacted physically with SRF. Transfection of Epc1 to myoblast cells potentiated the SRF-induced expression of skeletal muscle-specific genes as well as multinucleation. Proximal CArG box in the skeletal alpha-actin promoter was responsible for the synergistic activation of the promoter-luciferase. Epc1 knockdown caused a decrease in the acetylation of histones associated with serum response element (SRE) of the skeletal alpha-actin promoter. The Epc1.SRF complex bound to the SRE, and the knockdown of Epc1 resulted in a decrease in SRF binding to the skeletal alpha-actin promoter. Epc1 recruited histone acetyltransferase activity, which was potentiated by cotransfection with p300 but abolished by si-p300. Epc1 directly bound to p300 in myoblast cells. Epc1+/- mice showed distortion of skeletal alpha-actin, and the isolated myoblasts from the mice had impaired muscle differentiation. These results suggest that Epc1 is required for skeletal muscle differentiation by recruiting both SRF and p300 to the SRE of muscle-specific gene promoters.


Subject(s)
Muscle, Skeletal/cytology , Myoblasts/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serum Response Factor/metabolism , Acetylation , Actins/genetics , Animals , Cell Differentiation/physiology , Cells, Cultured , E1A-Associated p300 Protein/metabolism , Embryonic Stem Cells/cytology , Heterozygote , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/cytology , Promoter Regions, Genetic/physiology
17.
Circ Res ; 103(11): 1259-69, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18849323

ABSTRACT

Diverse cardiac diseases induce cardiac hypertrophy, which leads to dilatation and heart failure. We previously reported that hypertrophy can be blocked by class I histone deacetylase (HDAC) inhibitor, which prompted us to investigate the regulatory mechanism of class I HDACs. Cardiac hypertrophy was introduced by aortic banding, by infusion of isoproterenol or angiotensin II, or by swimming. Hypertrophic stimuli transiently elevated the activity of histone deacetylase-2 (Hdac2), a class I HDAC. In cardiomyocytes, forced expression of Hdac2 simulated hypertrophy in an Akt-dependent manner, whereas enzymatically inert Hdac2 H141A failed to do so. Hypertrophic stimuli induced the expression of heat shock protein (Hsp)70. The induced Hsp70 physically associated with and activated Hdac2. Hsp70 overexpression produced a hypertrophic phenotype, which was blocked either by siHdac2 or by a dominant negative Hsp70DeltaABD. In Hsp70.1(-/-) mice, cardiac hypertrophy and Hdac2 activation were significantly blunted. Heat shock either to cardiomyocytes or to mice activated Hdac2 and induced hypertrophy. However, heat shock-induced Hdac2 activation was blunted in the cardiomyocytes isolated from Hsp70.1(-/-) mice. These results suggest that the induction of Hsp70 in response to diverse hypertrophic stresses and the ensuing activation of HDAC2 trigger cardiac hypertrophy, emphasizing HSP70/HDAC2 as a novel mechanism regulating hypertrophy.


Subject(s)
Cardiomegaly/physiopathology , Heat-Shock Response/physiology , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Echocardiography , Enzyme Activation , Gene Expression Regulation , HSP72 Heat-Shock Proteins/deficiency , Histone Deacetylase 2 , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Rats , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...