Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Physiol Biochem ; 79(1): 83-105, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36194366

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging regulators of vascular diseases, yet their role in diabetic vascular calcification/aging remains poorly understood. In this study, we identified a down-expressed lncRNA SNHG1 in high glucose (HG)-induced vascular smooth muscle cells (HA-VSMCs), which induced excessive autophagy and promoted HA-VSMCs calcification/senescence. Overexpression of SNHG1 alleviated HG-induced HA-VSMCs calcification/senescence. The molecular mechanisms of SNHG1 in HA-VSMCs calcification/senescence were explored by RNA pull-down, RNA immunoprecipitation, RNA stability assay, luciferase reporter assay, immunoprecipitation and Western blot assays. In one mechanism, SNHG1 directly interacted with Bhlhe40 mRNA 3'-untranslated region and increased Bhlhe40 mRNA stability and expression. In another mechanism, SNHG1 enhanced Bhlhe40 protein SUMOylation by serving as a scaffold to facilitate the binding of SUMO E3 ligase PIAS3 and Bhlhe40 protein, resulting in increased nuclear translocation of Bhlhe40 protein. Moreover, Bhlhe40 suppressed the expression of Atg10, which is involved in the process of autophagosome formation. Collectively, the protective effect of SNHG1 on HG-induced HA-VSMCs calcification/senescence is accomplished by stabilizing Bhlhe40 mRNA and promoting the nuclear translocation of Bhlhe40 protein. Our study could provide a novel approach for diabetic vascular calcification/aging.


Subject(s)
Autophagy-Related Proteins , Basic Helix-Loop-Helix Transcription Factors , MicroRNAs , RNA, Long Noncoding , Vascular Calcification , Humans , Autophagy , Autophagy-Related Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Glucose/metabolism , Homeodomain Proteins , MicroRNAs/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/pharmacology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/pharmacology , RNA, Long Noncoding/metabolism
2.
J Nanobiotechnology ; 19(1): 379, 2021 Nov 21.
Article in English | MEDLINE | ID: mdl-34802438

ABSTRACT

Matrix metalloproteinase (MMP) 2 and 9 are the family members of proteases normally up-regulated in tumor to enhance the invasion and metastatic of tumor cells, and are associated with poor outcome of head and neck squamous cell carcinomas (HNSCCs). In the present work, MMPs-degradable gelatin nanoparticles (GNPs) are simultaneously loaded with photosensitizer indocyanine green (ICG) along with signal transducer activator of transcription 3 (STAT3) inhibitor NSC74859 (NSC, N) for efficient photothermal therapy (PTT) and immunotherapy of HNSCCs. In the tumor tissue, Gel-N-ICG nanoparticle was degraded and encapsulated ICG and NSC were effectively released. Under near-infrared (NIR) irradiation, the released ICG nanoparticles enabled effective photothermal destruction of tumors, and the STAT3 inhibitor NSC elicited potent antitumor immunity for enhanced cancer therapy. Based on two HNSCC mouse models, we demonstrated that Gel-N-ICG significantly delayed tumor growth without any appreciable body weight loss. Taken together, the strategy reported here may contribute that the stimuli-responsive proteases triggered nanoplatform could reduce tumor size more effectively in complex tumor microenvironment (TME) through combination of PTT and immunotherapy.


Subject(s)
Gelatinases/metabolism , Nanoparticles , Photosensitizing Agents , Protein Inhibitors of Activated STAT , Animals , Cell Line, Tumor , Cell Survival/drug effects , Immunotherapy , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Photothermal Therapy , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/pharmacokinetics , Protein Inhibitors of Activated STAT/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors
3.
Clin Cancer Res ; 20(19): 5124-32, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25124686

ABSTRACT

PURPOSE: Deregulation of STAT3 activation is a hallmark of many cancer cells, and the underlying mechanisms are subject to intense investigation. We examined the extent of PIAS3 expression in mesothelioma cells and human tumor samples and determined the functional effects of PIAS3 expression on STAT3 signaling. EXPERIMENTAL DESIGN: We evaluated the expression of PIAS3 in mesothelioma tumors from patients and correlated the expression levels with the course of the disease. We also measured the effects of enhanced PIAS3 activity on STAT3 signaling, cellular growth, and viability in cultured mesothelioma cells. RESULTS: Gene expression databases revealed that mesotheliomas have the lowest levels of PIAS3 transcripts among solid tumors. PIAS3 expression in human mesothelioma tumors is significantly correlated with overall survival intervals (P = 0.058). The high expression of PIAS3 is predictive of a favorable prognosis and decreases the probability of death within one year after diagnosis by 44%. PIAS3 expression is functionally linked to STAT3 activation in mesothelioma cell lines. STAT3 downregulation with siRNA or enhanced expression of PIAS3 both inhibited mesothelioma cell growth and induced apoptosis. Mesothelioma cells are sensitive to curcumin and respond by the induction of PIAS3. Corroborative evidence has been obtained from STAT3 inhibition experiments. Exposure of the cells to a peptide derived from the PIAS3 protein that interferes with STAT3 function resulted in apoptosis induction and the inhibition of cell growth. CONCLUSION: These results suggest that PIAS3 protein expression impacts survival in patients with mesothelioma and that PIAS3 activation could become a therapeutic strategy. Clin Cancer Res; 20(19); 5124-32. ©2014 AACR.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mesothelioma/genetics , Mesothelioma/metabolism , Molecular Chaperones/genetics , Protein Inhibitors of Activated STAT/genetics , STAT3 Transcription Factor/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/pharmacology , Gene Expression , Humans , Lung Neoplasms/mortality , Mesothelioma/mortality , Mesothelioma, Malignant , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Chaperones/pharmacology , Peptide Fragments/pharmacology , Prognosis , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics
4.
PLoS One ; 7(5): e37636, 2012.
Article in English | MEDLINE | ID: mdl-22649547

ABSTRACT

Negative regulation of the NF-κB transcription factor is essential for tissue homeostasis in response to stress and inflammation. NF-κB activity is regulated by a variety of biochemical mechanisms including phosphorylation, acetylation, and ubiquitination. In this study, we provide the first experimental evidence that NF-κB is regulated by SUMOylation, where the RelA subunit of NF-κB is SUMOylated by PIAS3, a member of the PIAS (protein inhibitor of activated STAT) protein family with E3 SUMO ligase activity. PIAS3-mediated NF-κB repression was compromised by either RelA mutant resistant to SUMOylation or PIAS3 mutant defective in SUMOylation. PIAS3-mediated SUMOylation of endogenous RelA was induced by NF-κB activation thus forming a negative regulatory loop. The SUMOylation of endogenous RelA was enhanced in IκBα null as compared with wild type fibroblasts. The RelA SUMOylation was induced by TNFα but not leptomycin B mediated RelA nuclear translocation. Furthermore, RelA mutants defective in DNA binding were not SUMOylated by PIAS3, suggesting that RelA DNA binding is a signal for PIAS3-mediated SUMOylation. These results support a novel negative feedback mechanism for NF-κB regulation by PIAS3-mediated RelA SUMOylation.


Subject(s)
Feedback, Physiological/physiology , Ligases/metabolism , NF-kappa B/antagonists & inhibitors , Protein Inhibitors of Activated STAT/metabolism , Sumoylation/drug effects , Active Transport, Cell Nucleus , Cloning, Molecular , Fatty Acids, Unsaturated/metabolism , Feedback, Physiological/drug effects , Fibroblasts , HEK293 Cells , Humans , Immunoblotting , Lentivirus , Ligases/genetics , Luciferases , Plasmids/genetics , Protein Inhibitors of Activated STAT/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
Brain Behav Immun ; 25(8): 1716-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21798341

ABSTRACT

IFN-alpha has been found to inhibit glucocorticoid receptor (GR) function by activating janus kinase-signal transducer and activator of transcription (JAK-STAT) inflammatory signaling pathways. In contrast, through stimulation of protein kinase A (PKA), cAMP has been shown to enhance GR function and can inhibit inflammatory signaling. We therefore examined whether increased cAMP-PKA pathway activation could reverse IFN-alpha-induced inhibition of GR function and whether decreased cAMP-PKA activity might exacerbate IFN-alpha effects on the GR. Activation of cAMP by forskolin (10 µM) reversed the inhibitory effects of mIFN-alpha (1000 U/ml) on dexamethasone (DEX)-induced MMTV-luciferase activity in hippocampal HT22 cells. Forskolin treatment also blocked both IFN-alpha-induced activation of phosphorylated STAT5 (pSTAT5) and inhibitory protein-protein interactions between pSTAT5 and GR in the nucleus of HT22 cells treated with IFN-alpha and DEX. These effects of forskolin were reversed by co-administration of the PKA inhibitor, H89. Conversely, the combination of IFN-alpha and treatment with either H89 or siRNA directed against the alpha and beta catalytic subunit isoforms of PKA led to an additive inhibitory effect on DEX-induced GR activity in HT22 cells. Taken together, these findings suggest that inhibition of GR signaling by mIFN-alpha and STAT5 can be reversed by activation of cAMP-PKA pathways, whereas decreased PKA activity increases the inhibitory effect of IFN-alpha on GR function. Given decreased PKA activity found in patients with major depression, these data suggest that depressed patients may be vulnerable to cytokine effects on GR, and cAMP-PKA agonists may serve to reverse glucocorticoid resistance in patients with depression and increased inflammation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Interferon-alpha/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , STAT5 Transcription Factor/physiology , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/drug effects , Cytosol/metabolism , Dexamethasone/pharmacology , Enzyme Activation/physiology , Immunoprecipitation , Indicators and Reagents , Luciferases/metabolism , Mice , Phosphorylation , Protein Inhibitors of Activated STAT/pharmacology , RNA, Small Interfering/pharmacology , Receptors, Glucocorticoid/physiology
6.
Biochem J ; 419(1): 201-9, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19067654

ABSTRACT

The orphan nuclear receptor LRH-1 (liver receptor homologue-1; NR5A2) plays a critical role in development, bile acid synthesis and cholesterol metabolism. LRH-1 is also expressed in the ovary where it is implicated in the regulation of steroidogenic genes for steroid hormone synthesis. In the present study, we investigated the molecular mechanisms of the transcriptional regulation of CYP11A1 by LRH-1 and found that LRH-1-mediated transactivation was markedly repressed by PIASy [protein inhibitor of activated STAT (signal transducer and activator of transcription) y], the shortest member of the PIAS family. The suppression of LRH-1 activity requires the N-terminal repression domain. Although PIAS proteins also function as E3 SUMO (small ubiquitin-related modifier) ligases and enhance SUMO conjugation, PIASy-mediated repression was independent of LRH-1 SUMOylation status. In addition, histone deacetylase activity was not involved in the inhibition of LRH-1 by PIASy. Immunoprecipitation and mammalian two-hybrid analyses indicated that PIASy interacted with LRH-1 through the C-terminal region, including the AF-2 (activation function-2) motif, which was also involved in the interaction between LRH-1 and the co-activator SRC-1 (steroid receptor co-activator-1). PIASy inhibited the binding of SRC-1 to LRH-1, although overexpression of SRC-1 partially overcame the PIASy inhibition of LRH-1 induction of the CYP11A1 promoter. The results of the present study suggest that competition with co-activators may be an important mechanism underlying the PIASy repression of LRH-1-mediated transactivation.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme/genetics , DNA-Binding Proteins/pharmacology , Histone Acetyltransferases/metabolism , Protein Inhibitors of Activated STAT/pharmacology , Transcription Factors/metabolism , Transcription Factors/pharmacology , Animals , Blotting, Western , Cell Line , DNA-Binding Proteins/metabolism , Humans , Immunoprecipitation , Mice , Nuclear Receptor Coactivator 1 , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Inhibitors of Activated STAT/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transfection , Two-Hybrid System Techniques
7.
J Biochem ; 139(2): 305-14, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16452319

ABSTRACT

Duplin was originally isolated as a negative regulator of beta-catenin-dependent T-cell factor (Tcf) transcriptional activity in the Wnt signaling pathway. However, Duplin knockout mice exhibit embryonic lethality at 5.5-em day, suggesting that Duplin has important roles other than as a negative regulator of the Wnt signal. To identify new roles of Duplin, the Duplin-binding proteins were screened. PIAS3, which is a SUMO E3 ligase and acts as an inhibitor of signal transducer and activator of transcription (STAT3), was identified as a Duplin-binding protein. Duplin was sumoylated, but PIAS3 affected neither the sumoylation of Duplin nor its ability to inhibit Tcf-4 activity. Like PIAS3, Duplin suppressed the leukemia-inhibitory factor (LIF)-induced STAT3 transcriptional activity. Duplin did not affect the LIF-dependent tyrosine phosphorylation or nuclear localization of STAT3 but inhibited the formation of complex between STAT3 and DNA. Although STAT3 is not modified with SUMO, PIAS3 inhibited the STAT3 activity in a manner partially depending on its SUMO E3 ligase activity. Duplin suppressed the LIF-dependent STAT3 activity independently of sumoylation. These results demonstrate that Duplin inhibits not only Tcf-4 but also STAT3, suggesting that Duplin may act as a repressor for multiple transcriptional factors.


Subject(s)
DNA-Binding Proteins/physiology , STAT3 Transcription Factor/antagonists & inhibitors , Wnt Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , DNA/drug effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/pharmacology , HeLa Cells , Humans , Mice , Protein Binding , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Wnt Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...