Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 292(24): 10048-10060, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28432122

ABSTRACT

IFNs are effective in inhibiting angiogenesis in preclinical models and in treating several angioproliferative disorders. However, the detailed mechanisms of IFNα-mediated anti-angiogenesis are not completely understood. Stat1/2/3 and PML are IFNα downstream effectors and are pivotal regulators of angiogenesis. Here, we investigated PML's role in the regulation of Stat1/2/3 activity. In Pml knock-out (KO) mice, ablation of Pml largely reduces IFNα angiostatic ability in Matrigel plug assays. This suggested an essential role for PML in IFNα's anti-angiogenic function. We also demonstrated that PML shared a large cohort of regulatory genes with Stat1 and Stat3, indicating an important role of PML in regulating Stat1 and Stat3 activity. Using molecular tools and primary endothelial cells, we demonstrated that PML positively regulates Stat1 and Stat2 isgylation, a ubiquitination-like protein modification. Accordingly, manipulation of the isgylation system by knocking down USP18 altered IFNα-PML axis-mediated inhibition of endothelial cell migration and network formation. Furthermore, PML promotes turnover of nuclear Stat3, and knockdown of PML mitigates the effect of LLL12, a selective Stat3 inhibitor, on IFNα-mediated anti-angiogenic activity. Taken together, we elucidated an unappreciated mechanism in which PML, an IFNα-inducible effector, possess potent angiostatic activity, doing so in part by forming a positive feedforward loop with Stat1/2 and a negative feedback loop with Stat3. The interplay between PML, Stat1/Stat2, and Stat3 contributes to IFNα-mediated inhibition of angiogenesis, and disruption of this network results in aberrant IFNα signaling and altered angiostatic activity.


Subject(s)
Endothelium, Vascular/metabolism , Interferon-alpha/metabolism , Neovascularization, Pathologic/prevention & control , Promyelocytic Leukemia Protein/metabolism , STAT1 Transcription Factor/agonists , STAT2 Transcription Factor/agonists , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line , Cells, Cultured , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interferon-alpha/genetics , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Promyelocytic Leukemia Protein/antagonists & inhibitors , Promyelocytic Leukemia Protein/genetics , Protein Processing, Post-Translational , RNA Interference , Recombinant Proteins/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
2.
Mol Biol Cell ; 25(16): 2485-98, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24943846

ABSTRACT

Oxidative stress is a consequence of an imbalance between reactive oxygen species (ROS) production and the ability of the cytoprotective system to detoxify the reactive intermediates. The tumor suppressor promyelocytic leukemia protein (PML) functions as a stress sensor. Loss of PML results in impaired mitochondrial complex II activity, increased ROS, and subsequent activation of nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidative pathway. We also demonstrate that sulforaphane (SFN), an antioxidant, regulates Nrf2 activity by controlling abundance and subcellular distribution of PML and that PML is essential for SFN-mediated ROS increase, Nrf2 activation, antiproliferation, antimigration, and antiangiogenesis. Taking the results together, we have uncovered a novel antioxidative mechanism by which PML regulates cellular oxidant homeostasis by controlling complex II integrity and Nrf2 activity and identified PML as an indispensable mediator of SFN activity.


Subject(s)
NF-E2-Related Factor 2/metabolism , Nuclear Proteins/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells , Humans , Isothiocyanates/pharmacology , Mice , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , RNA, Small Interfering/genetics , Signal Transduction , Sulfoxides , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
3.
J Biol Chem ; 288(41): 29746-59, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23986437

ABSTRACT

The promyelocytic leukemia protein is a well known tumor suppressor, but its role in metabolism is largely unknown. Mice with a deletion in the gene for PML (KO mice) exhibit altered gene expression in liver, adipose tissue, and skeletal muscle, an accelerated rate of fatty acid metabolism, abnormal glucose metabolism, constitutive AMP-activating kinase (AMPK) activation, and insulin resistance in skeletal muscle. Last, an increased rate of energy expenditure protects PML KO mice from the effects of obesity induced by a Western diet. Collectively, our study uncovers a previously unappreciated role of PML in the regulation of metabolism and energy balance in mice.


Subject(s)
Energy Metabolism/genetics , Nuclear Proteins/genetics , Obesity/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , AMP-Activated Protein Kinases/metabolism , Adipokines/genetics , Adipose Tissue/metabolism , Animals , Blotting, Western , Body Temperature/genetics , CD36 Antigens/genetics , Diet/adverse effects , Fatty Acids/metabolism , Gene Expression , Glucose Transporter Type 4/genetics , Liver/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Muscle, Skeletal/metabolism , Nuclear Proteins/deficiency , Obesity/etiology , Obesity/metabolism , Oxidation-Reduction , Promyelocytic Leukemia Protein , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/deficiency , Tumor Suppressor Proteins/deficiency
4.
PLoS One ; 8(4): e60213, 2013.
Article in English | MEDLINE | ID: mdl-23593176

ABSTRACT

Glomerular podocytes are highly differentiated epithelial cells that are key components of the kidney filtration units. Podocyte damage or loss is the hallmark of nephritic diseases characterized by severe proteinuria. Recent studies implicate that hormones including glucocorticoids (ligand for glucocorticoid receptor) and vitamin D3 (ligand for vitamin D receptor) protect or promote repair of podocytes from injury. In order to elucidate the mechanisms underlying hormone-mediated podocyte-protecting activity from injury, we carried out microarray gene expression studies to identify the target genes and corresponding pathways in response to these hormones during podocyte differentiation. We used immortalized human cultured podocytes (HPCs) as a model system and carried out in vitro differentiation assays followed by dexamethasone (Dex) or vitamin D3 (VD3) treatment. Upon the induction of differentiation, multiple functional categories including cell cycle, organelle dynamics, mitochondrion, apoptosis and cytoskeleton organization were among the most significantly affected. Interestingly, while Dex and VD3 are capable of protecting podocytes from injury, they only share limited target genes and affected pathways. Compared to VD3 treatment, Dex had a broader and greater impact on gene expression profiles. In-depth analyses of Dex altered genes indicate that Dex crosstalks with a broad spectrum of signaling pathways, of which inflammatory responses, cell migration, angiogenesis, NF-κB and TGFß pathways are predominantly altered. Together, our study provides new information and identifies several new avenues for future investigation of hormone signaling in podocytes.


Subject(s)
Cholecalciferol/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Podocytes/drug effects , Podocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Molecular Sequence Annotation , Podocytes/cytology , Transcriptome
5.
BMC Genomics ; 13: 453, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22947142

ABSTRACT

BACKGROUND: Promyelocytic leukemia protein (PML) is a tumor suppressor that is highly expressed in endothelial cells nonetheless its role in endothelial cell biology remains elusive. Tumor necrosis factor alpha (TNFα) is an important cytokine associated with many inflammation-related diseases. We have previously demonstrated that TNFα induces PML protein accumulation. We hypothesized that PML may play a role in TNFα signaling pathway. To identify potential PML target genes and investigate the putative crosstalk between PML's function and TNFα signaling in endothelial cells, we carried out a microarray analysis in human primary umbilical endothelial cells (HUVECs). RESULTS: We found that PML and TNFα regulate common and distinct genes involved in a similar spectrum of biological processes, pathways and human diseases. More importantly, we found that PML is required for fine-tuning of TNFα-mediated immune and inflammatory responses. Furthermore, our data suggest that PML and TNFα synergistically regulate cell adhesion by engaging multiple molecular mechanisms. Our biological functional assays exemplified that adhesion of U937 human leukocytes to HUVECs is co-regulated by PML and TNFα signaling. CONCLUSIONS: Together, our study identified PML as an essential regulator of TNFα signaling by revealing the crosstalk between PML knockdown-mediated effects and TNFα-elicited signaling, thereby providing novel insights into TNFα signaling in endothelial cells.


Subject(s)
Gene Expression Regulation/genetics , Nuclear Proteins/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/metabolism , Cell Adhesion/genetics , Human Umbilical Vein Endothelial Cells , Humans , Microarray Analysis , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Suppressor Proteins/genetics , U937 Cells
6.
J Biol Chem ; 287(28): 23356-67, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22589541

ABSTRACT

Promyelocytic leukemia protein (PML) is a tumor suppressor that is highly expressed in vascular endothelium and inflamed tissues, yet its role in inflammation-associated cytokine-regulated angiogenesis and underlying mechanism remains largely unclear. We show that tumor necrosis factor α (TNFα) and interferon α (IFNα) stimulate PML expression while suppressing EC network formation and migration, two key events during angiogenesis. By a knockdown approach, we demonstrate that PML is indispensable for TNFα- and IFNα-mediated inhibition of EC network formation. We further demonstrate that signal transducer and activator of transcription 1 (STAT1) binds PML promoter and that is an important regulator of PML expression. Knockdown of STAT1 reduces endogenous PML and blocks TNFα- and IFNα-induced PML accumulation and relieves TNFα- and IFNα-mediated inhibition of EC network formation. Our data also indicate that PML regulates EC migration, in part, by modulating expression of downstream genes, such as negatively regulating integrin ß1 (ITGB1). In addition, knockdown of STAT1 or PML alleviates TNFα- and IFNα-mediated inhibition of ITGB1 expression. Antibody blockade demonstrates that ITGB1 is functionally important for PML- and STAT1-regulated EC migration. Taken together, our data provide novel mechanistic insights that PML functions as a negative regulator in EC network formation and migration.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/metabolism , Interferon-alpha/pharmacology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/metabolism , Cells, Cultured , Endothelial Cells/cytology , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Microscopy, Fluorescence , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Promyelocytic Leukemia Protein , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
7.
Front Oncol ; 2: 210, 2012.
Article in English | MEDLINE | ID: mdl-23316480

ABSTRACT

The tumor suppressor promyelocytic leukemia protein (PML) predominantly resides in a structurally distinct sub-nuclear domain called PML nuclear bodies. Emerging evidences indicated that PML actively participates in many aspects of cellular processes, but the molecular mechanisms underlying PML regulation in response to stress and environmental cues are not complete. Post-translational modifications, such as SUMOylation, phosphorylation, acetylation, and ubiquitination of PML add a complex layer of regulation to the physiological function of PML. In this review, we discuss the fast-moving horizon of post-translational modifications targeting PML.

8.
J Biol Chem ; 286(3): 1850-9, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21078666

ABSTRACT

Alpha actinins (ACTNs) are known for their ability to modulate cytoskeletal organization and cell motility by cross-linking actin filaments. We show here that ACTN4 harbors a functional LXXLL receptor interaction motif, interacts with nuclear receptors in vitro and in mammalian cells, and potently activates transcription mediated by nuclear receptors. Whereas overexpression of ACTN4 potentiates estrogen receptor α (ERα)-mediated transcription in transient transfection reporter assays, knockdown of ACTN4 decreases it. In contrast, histone deacetylase 7 (HDAC7) inhibits estrogen receptor α (ERα)-mediated transcription. Moreover, the ACTN4 mutant lacking the CaM (calmodulin)-like domain that is required for its interaction with HDAC7 fails to activate transcription by ERα. Chromatin immunoprecipitation (ChIP) assays demonstrate that maximal associations of ACTN4 and HDAC7 with the pS2 promoter are mutually exclusive. Knockdown of ACTN4 significantly decreases the expression of ERα target genes including pS2 and PR and also affects cell proliferation of MCF-7 breast cancer cells with or without hormone, whereas knockdown of HDAC7 exhibits opposite effects. Interestingly, overexpression of wild-type ACTN4, but not the mutants defective in interacting with ERα or HDAC7, results in an increase in pS2 and PR mRNA accumulation in a hormone-dependent manner. In summary, we have identified ACTN4 as a novel, atypical coactivator that regulates transcription networks to control cell growth.


Subject(s)
Actinin/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Estrogen Receptor alpha/metabolism , Histone Deacetylases/metabolism , Neoplasm Proteins/metabolism , Actinin/genetics , Amino Acid Motifs , Breast Neoplasms/genetics , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Female , Gene Knockdown Techniques , Histone Deacetylases/genetics , Humans , Neoplasm Proteins/genetics , Presenilin-2/biosynthesis , Presenilin-2/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary
9.
J Biol Chem ; 284(52): 36395-36404, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19858209

ABSTRACT

We have identified G protein suppressor 2 (GPS2) as a stable component of the SMRT corepressor complexes. GPS2 potently represses basal transcription, with the repression domain mapped to the N-terminal silencing mediator of retinoic acid and thyroid hormone receptor (SMRT)-interacting domain. Knockdown of GPS2 abrogates, whereas overexpression potentiates, SMRT-mediated repression activity. The SMRT complexes are involved in 4-hydroxyl-tamoxifen (4OHT)-mediated gene repression by estrogen receptor alpha (ERalpha). We show that 4OHT recruits SMRT and GPS2 to the promoter of pS2, an ERalpha target gene, in a dynamic manner. Unexpectedly, we also found that estradiol (E2) promotes promoter recruitment of the SMRT complexes. While knockdown of GPS2 compromised 4OHT-mediated repression, it enhanced E2-induced expression of a reporter gene and several endogenous ERalpha target genes, including pS2, cyclin D1 (CCND1), progesterone receptor (PR), and c-MYC. Finally, we show that depletion of GPS2 or SMRT by siRNA promotes cell proliferation in MCF-7 breast cancer cells. Thus, we concluded that GPS2 is an integral component of the SMRT complexes, important for ligand-dependent gene regulations by ERalpha and a suppressor for MCF-7 cell proliferation.


Subject(s)
Estrogen Receptor alpha/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Cyclin D1/biosynthesis , Cyclin D1/genetics , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Repressor Proteins/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Transcription, Genetic/drug effects
10.
Mol Cell Biol ; 28(18): 5658-67, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18625722

ABSTRACT

Promyelocytic leukemia protein (PML) sumoylation has been proposed to control the formation of PML nuclear bodies (NBs) and is crucial for PML-dependent cellular processes, including apoptosis and transcriptional regulation. However, the regulatory mechanisms of PML sumoylation and its specific roles in the formation of PML NBs remain largely unknown. Here, we show that histone deacetylase 7 (HDAC7) knockdown reduces the size and the number of the PML NBs in human umbilical vein endothelial cells (HUVECs). HDAC7 coexpression stimulates PML sumoylation independent of its HDAC activity. Furthermore, HDAC7 associates with the E2 SUMO ligase, Ubc9, and stimulates PML sumoylation in vitro, suggesting that it possesses a SUMO E3 ligase-like activity to promote PML sumoylation. Importantly, HDAC7 knockdown inhibits tumor necrosis factor alpha-induced PML sumoylation and the formation of PML NBs in HUVECs. These results demonstrate a novel function of HDAC7 and provide a regulatory mechanism of PML sumoylation.


Subject(s)
Histone Deacetylases/metabolism , Intranuclear Inclusion Bodies/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , HeLa Cells , Histone Deacetylases/genetics , Humans , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/genetics , Umbilical Veins/cytology
11.
Mol Biol Cell ; 19(7): 3020-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18463162

ABSTRACT

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) are dynamic subnuclear compartments that play roles in several cellular processes, including apoptosis, transcriptional regulation, and DNA repair. Histone deacetylase (HDAC) 7 is a potent corepressor that inhibits transcription by myocyte enhancer factor 2 (MEF2) transcription factors. We show here that endogenous HDAC7 and PML interact and partially colocalize in PML NBs. Tumor necrosis factor (TNF)-alpha treatment recruits HDAC7 to PML NBs and enhances association of HDAC7 with PML in human umbilical vein endothelial cells. Consequently, TNF-alpha promotes dissociation of HDAC7 from MEF2 transcription factors and the promoters of MEF2 target genes such as matrix metalloproteinase (MMP)-10, leading to accumulation of MMP-10 mRNA. Conversely, knockdown of PML enhances the association between HDAC7 and MEF2 and decreases MMP-10 mRNA accumulation. Accordingly, ectopic expression of PML recruits HDAC7 to PML NBs and leads to activation of MEF2 reporter activity. Notably, small interfering RNA knockdown of PML decreases basal and TNF-alpha-induced MMP-10 mRNA accumulation. Our results reveal a novel mechanism by which PML sequesters HDAC7 to relieve repression and up-regulate gene expression.


Subject(s)
Gene Expression Regulation, Enzymologic , Histone Deacetylases/metabolism , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Animals , Apoptosis , HeLa Cells , Humans , MEF2 Transcription Factors , Matrix Metalloproteinase 10/metabolism , Mice , Microscopy, Confocal , Myogenic Regulatory Factors/metabolism , Protein Interaction Mapping , RNA, Small Interfering/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Microbiol Immunol ; 49(8): 703-9, 2005.
Article in English | MEDLINE | ID: mdl-16113499

ABSTRACT

The high mutation rate of HIV-1 (human immunodeficiency virus-1) is a major obstacle to developing an effective vaccine. The mutation of ELDKWA-(aa669-674) to ELDEWA-epitope on HIV-1 gp41 caused the immune escape from neutralization by potent anti-HIV-1 human monoclonal antibody (mAb) 2F5. In this study, we suggested and evaluated a multi-epitope vaccine as a new strategy to develop HIV-1 vaccines. A glutathione S-transferase (GST) fusion protein (GST-K8E8) containing 8 copies of ELDKWA-and mutated ELDEWA-epitopes was constructed and used to immunize mice or rabbits. Analysis of the antisera (rAS3) induced by GST-K8E8 suggested that multi-epitope vaccine immunogen could raise antibodies in mice and rabbits against either the original ELDKWA-epitope or the mutated ELDEWA-epitope that resulted in immune escape. Briefly, ELDKWA-epitope-specific antibodies, directly purified from rAS3 by ELDKWA-epitope-peptide affinity chromatography, recognized either original gp41 protein (ELDKWA, rgp41K) or mutated gp41 (ELDEWA, rgp41E) in immunoblotting assay; in contrast, the existing ELDKWA-epitope antibodies recognized only rgp41K but not rgp41E, which were purified by ELDKWA-epitope-peptide affinity chromatography from rAS3 that were firstly completely pre-absorbed by ELDEWA-epitope-peptide affinity beads. And the same results were also observed when detecting the ELDEWA-epitope-specific antibodies in rAS3 by a means similar to the above. All the data presented here demonstrated that a high density multi-epitope vaccine could be an interesting strategy against HIV-1 mutation.


Subject(s)
Antibody Specificity/immunology , Epitopes/immunology , HIV Antibodies/biosynthesis , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Animals , Epitopes/genetics , HIV Antibodies/immunology , HIV Envelope Protein gp41/genetics , HIV-1/chemistry , Mice , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...