Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Curr Protein Pept Sci ; 19(2): 145-154, 2018.
Article in English | MEDLINE | ID: mdl-28901250

ABSTRACT

X-box binding protein 1 (XBP-1) is a key regulator of the unfolded protein response (UPR), which is activated in response to endoplasmic reticulum (ER) stress. Cells contain two protein isoforms of XBP-1, the active isoform (XBP-1S) and the inactive isoform (XBP-1U). Induction of UPR leads to the generation of XBP-1S while XBP-1U is dominant in ER stress-free cells. XBP-1S is a transcriptional activator and regulates the expression of a subset of UPR genes. Importantly, recent studies have demonstrated the essential role of XBP-1S in various human diseases, such as viral infections. Many viruses have evolved to manipulate UPR/XBP-1 of the infected cells to promote viral survival and replication. In this review, we will summarize the current findings on the involvement of XBP-1 in viral infection/ replication and discuss the potential anti-viral strategies by targeting XBP-1.


Subject(s)
Antiviral Agents/pharmacology , Virus Diseases/drug therapy , X-Box Binding Protein 1/metabolism , Antiviral Agents/therapeutic use , Endoplasmic Reticulum Stress , Humans , Protein Folding , Transcription Factors/metabolism , Unfolded Protein Response , Virus Diseases/metabolism , Virus Diseases/virology , Virus Replication
2.
Oncotarget ; 6(1): 271-87, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25426559

ABSTRACT

Cellular unfolded protein response (UPR) is induced when endoplasmic reticulum (ER) is under stress. XBP-1S, the active isoform of X-box binding protein 1 (XBP-1), is a key regulator of UPR. Previously, we showed that a histone acetyltransferase (HAT), p300/CBP-associated factor (PCAF), binds to XBP-1S and functions as an activator of XBP-1S. Here, we identify general control nonderepressible 5 (GCN5), a HAT with 73% identity to PCAF, as a novel XBP-1S regulator. Both PCAF and GCN5 bind to the same domain of XBP-1S. Surprisingly, GCN5 potently blocks the XBP-1S-mediated transcription, including cellular UPR genes and latent membrane protein 1 of Epstein-Barr virus. Unlike PCAF, GCN5 acetylates XBP-1S and enhances nuclear retention and protein stability of XBP-1S. However, such GCN5-mediated acetylation of XBP-1S shows no effects on XBP-1S activity. In addition, the HAT activity of GCN5 is not required for repression of XBP-1S target genes. We further demonstrate that GCN5 inhibits XBP-1S-mediated transcription by disrupting the PCAF-XBP-1S interaction and preventing the recruitment of XBP-1S to its target genes. Taken together, our results represent the first work demonstrating that GCN5 and PCAF exhibit different functions and antagonistically regulate the XBP-1S-mediated transcription.


Subject(s)
DNA-Binding Proteins/biosynthesis , Transcription Factors/biosynthesis , Transcriptional Activation/physiology , p300-CBP Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Immunoprecipitation , Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Transcription, Genetic , Transfection , X-Box Binding Protein 1
3.
Antiviral Res ; 102: 11-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316029

ABSTRACT

Expression of the human cytomegalovirus (HCMV) major immediate-early (MIE) genes is regulated by a strong enhancer-containing promoter with multiple binding sites for various transcription factors, including cyclic AMP response element binding protein 1 (CREB1). Here we show that overexpression of CREB1 potently blocked MIE transcription and HCMV replication. Surprisingly, CREB1 still exhibited strong inhibition of the MIE promoter when all five CREB binding sites within the enhancer were mutated, suggesting that CREB1 regulated the MIE gene expression indirectly. Promoter deletion analysis and site-directed mutagenesis identified the region between -130 and -50 upstream of the transcription start site of the MIE gene as the "CREB1 responsive region". Mutations of SP1/3 and NF-κB binding sites within this region interrupted the inhibitory effect induced by CREB1 overexpression. Our findings suggest that overexpression of CREB1 can cause repression of HCMV replication and may contribute to the development of new anti-HCMV strategies.


Subject(s)
Cyclic AMP Response Element-Binding Protein/biosynthesis , Cytomegalovirus/physiology , Gene Expression , Host-Pathogen Interactions , Virus Replication , Cell Line , Cytomegalovirus/genetics , DNA Mutational Analysis , DNA, Viral/genetics , Genes, Immediate-Early , Humans , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Sequence Deletion
4.
Cancers (Basel) ; 5(3): 838-56, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-24202322

ABSTRACT

Hexamethylene bisacetamide-inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which controls transcription elongation of RNA polymerase II and Tat transactivation of human immunodeficiency virus. Besides P-TEFb, several proteins have been identified as HEXIM1 binding proteins. It is noteworthy that more than half of the HEXIM1 binding partners are involved in cancers. P53 and two key regulators of the p53 pathway, nucleophosmin (NPM) and human double minute-2 protein (HDM2), are among the factors identified. This review will focus on the functional importance of the interactions between HEXIM1 and p53/NPM/HDM2. NPM and the cytoplasmic mutant of NPM, NPMc+, were found to regulate P-TEFb activity and RNA polymerase II transcription through the interaction with HEXIM1. Importantly, more than one-third of acute myeloid leukemia (AML) patients carry NPMc+, suggesting the involvement of HEXIM1 in tumorigenesis of AML. HDM2 was found to ubiquitinate HEXIM1. The HDM2-mediated ubiquitination of HEXIM1 did not lead to protein degradation of HEXIM1 but enhanced its inhibitory activity on P-TEFb. Recently, HEXIM1 was identified as a novel positive regulator of p53. HEXIM1 prevented p53 ubiquitination by competing with HDM2 in binding to p53. Taken together, the new evidence suggests a role of HEXIM1 in regulating the p53 pathway and tumorigenesis.

5.
PLoS One ; 8(8): e72823, 2013.
Article in English | MEDLINE | ID: mdl-23977357

ABSTRACT

Hexamethylene bisacetamide inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which is composed of cyclin-dependent kinase 9 (CDK9)/cyclin T1. P-TEFb is an essential regulator for the transcriptional elongation by RNA polymerase II. A genome-wide study using human embryonic stem cells shows that most mRNA synthesis is regulated at the stage of transcription elongation, suggesting a possible role for P-TEFb/HEXIM1 in the gene regulation of stem cells. In this report, we detected a marked increase in HEXIM1 protein levels in the differentiated human pluripotent stem cells (hPSCs) induced by LY294002 treatment. Since no changes in CDK9 and cyclin T1 were observed in the LY294002-treated cells, increased levels of HEXIM1 might lead to inhibition of P-TEFb activity. However, treatment with a potent P-TEFb inhibiting compound, flavopiridol, failed to induce hPSC differentiation, ruling out the possible requirement for P-TEFb kinase activity in hPSC differentiation. Conversely, differentiation was observed when hPSCs were incubated with hexamethylene bisacetamide, a HEXIM1 inducing reagent. The involvement of HEXIM1 in the regulation of hPSCs was further supported when overexpression of HEXIM1 concomitantly induced hPSC differentiation. Collectively, our study demonstrates a novel role of HEXIM1 in regulating hPSC fate through a P-TEFb-independent pathway.


Subject(s)
Cell Differentiation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Acetamides/pharmacology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Cell Lineage/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , Ectoderm/cytology , Flavonoids/pharmacology , Humans , Mesoderm/cytology , Piperidines/pharmacology , Pluripotent Stem Cells/drug effects , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , Positive Transcriptional Elongation Factor B/metabolism , Transcription Factors , Up-Regulation/drug effects , Up-Regulation/genetics
6.
Cell Cycle ; 12(6): 944-52, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23421996

ABSTRACT

Pin1 isomerizes the phosphorylated Ser/Thr-Pro peptide bonds and regulates the functions of its binding proteins by inducing conformational changes. Involvement of Pin1 in the aging process has been suggested based on the phenotype of Pin1-knockout mice and its interaction with lifespan regulator protein, p66 (Shc) . In this study, we utilize a proteomic approach and identify peroxiredoxin 1 (PRDX1), another regulator of aging, as a novel Pin1 binding protein. Pin1 binds to PRDX1 through interacting with the phospho-Thr ( 90) -Pro ( 91) motif of PRDX1, and this interaction is abolished when the Thr ( 90) of PRDX1 is mutated. The Pin1 binding motif, Thr-Pro, is conserved in the 2-Cys PRDXs, PRDX1-4 and the interactions between Pin1 and PRDX2-4 are also demonstrated. An increase in hydrogen peroxide buildup and a decrease in the peroxidase activity of 2-Cys PRDXs were observed in Pin1 (-/-) mouse embryonic fibroblasts (MEFs), with the activity of PRDXs restored when Pin1 was re-introduced into the cells. Phosphorylation of PRDX1 at Thr ( 90) has been shown to inhibit its peroxidase activity; however, how exactly the activity of PRDX1 is regulated by phosphorylation still remains unknown. Here, we demonstrate that Pin1 facilitates the protein phosphatase 2A-mediated dephosphorylation of PRDX1, which helps to explain the accumulation of the inactive phosphorylated form of PRDX1 in Pin1 (-/-) MEFs. Collectively, we identify Pin1 as a novel PRDX1 binding protein and propose a mechanism for Pin1 in regulating the metabolism of reactive oxygen species in cells.


Subject(s)
Hydrogen Peroxide/metabolism , Peptidylprolyl Isomerase/metabolism , Peroxiredoxins/metabolism , Aging , Animals , HEK293 Cells , HeLa Cells , Humans , Mice , NIMA-Interacting Peptidylprolyl Isomerase , Oxidation-Reduction , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
J Biol Chem ; 287(43): 36443-54, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22948151

ABSTRACT

Hexamethylene bisacetamide-inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which regulates the transcription elongation of RNA polymerase II and controls 60-70% of mRNA synthesis. Our previous studies show that HEXIM1 interacts with two key p53 regulators, nucleophosmin and human double minute-2 protein (HDM2), implying a possible connection between HEXIM1 and the p53 signaling pathway. Here we report the interaction between p53 and HEXIM1 in breast cancer, acute myeloid leukemia, and colorectal carcinoma cells. The C-terminal regions of p53 and HEXIM1 are required for the protein-protein interaction. Overexpression of HEXIM1 prevents the ubiquitination of p53 by HDM2 and enhances the protein stability of p53, resulting in up-regulation of p53 target genes, such as Puma and p21. Induction of p53 can be achieved by several means, such as UV radiation and treatment with anti-cancer agents (including doxorubicin, etoposide, roscovitine, flavopiridol, and nutlin-3). Under all the conditions examined, elevated protein levels of p53 are found to associate with the increased p53-HEXIM1 interaction. In addition, knockdown of HEXIM1 significantly inhibits the induction of p53 and releases the cell cycle arrest caused by p53. Finally, the transcription of the p53 target genes is regulated by HEXIM1 in a p53-dependent fashion. Our results not only identify HEXIM1 as a positive regulator of p53, but also propose a novel molecular mechanism of p53 activation caused by the anti-cancer drugs and compounds.


Subject(s)
RNA-Binding Proteins/metabolism , Signal Transduction/physiology , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitination/physiology , Up-Regulation/physiology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA-Binding Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Transcription Factors , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitination/drug effects , Ubiquitination/radiation effects , Ultraviolet Rays , Up-Regulation/drug effects , Up-Regulation/radiation effects
8.
Cell Cycle ; 10(12): 1978-87, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21558800

ABSTRACT

Nucleophosmin (NPM), an important regulator in p53 signaling pathway, is one of the most frequently mutated genes in acute myeloid leukemia (AML). In our previous study, we found that hexamethylene bisacetamide inducible protein 1 (HEXIM1) interacted with both wild-type NPM and cytoplasmic-misallocated NPMc(+) mutant, leading to an increase in RNA polymerase II transcription. Here, we examine the protein expression in wild-type NPM (AML2) and NPMc(+) mutant (AML3) AML cell lines. Significant lower levels of NPM, HEXIM1 and p53 proteins are detected in AML3 cells, and such differential protein expression is not regulated at transcriptional or post-translational stages. Effects of several anticancer compounds on cell viability of AML2 and AML3 cells are investigated. Compared to AML3 cells, AML2 cells are more sensitive to the treatment of the DNA-damaging compounds (doxorubicin and etoposide) and a specific p53-inducing compound (nutlin-3). However, no significant difference in cytotoxicity was observed when AML2 and AML3 cells were treated with cyclin-dependent kinase inhibitors, flavopiridol and CYC202. Our results provide a novel insight into the functional impact of the NPMc(+) mutation on protein expression and the potential approaches for selective therapy of AML.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/pathology , Nuclear Proteins/analysis , Tumor Suppressor Protein p53/analysis , Cell Line, Tumor , DNA Damage/drug effects , Drug Resistance, Neoplasm , Gene Expression , Humans , Mutation , Nucleophosmin , RNA-Binding Proteins/analysis , Transcription Factors
9.
Nucleic Acids Res ; 39(2): 429-39, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20817929

ABSTRACT

X-box binding protein 1 (XBP-1) is a key regulator required for cellular unfolded protein response (UPR) and plasma cell differentiation. In addition, involvement of XBP-1 in host cell-virus interaction and transcriptional regulation of viruses, such as human T-lymphotropic virus type 1 (HTLV-1), has been revealed recently. Two XBP-1 isoforms, XBP-1U and XBP-1S, which share an identical N-terminal domain, are present in cells. XBP-1S is a transcription activator while XBP-1U is the inactive isoform. Although the transactivation domain of XBP-1S has been identified within the XBP-1S-specific C-terminus, molecular mechanism of the transcriptional activation by XBP-1S still remains unknown. Here we report the interaction between p300/CBP-associated factor (PCAF) and XBP-1S through the C-terminal domain of XBP-1S. No binding between XBP-1U and PCAF is detected. In a cell-based reporter assay, overexpression of PCAF further stimulates the XBP-1S-mediated cellular and HTLV-1 transcription while knockdown of PCAF exhibits the opposite effect. Expression of endogenous XBP-1S cellular target genes, such as BiP and CHOP, is significantly inhibited when PCAF is knocked down. Furthermore, PCAF is recruited to the promoters of XBP-1S target genes in vivo, in a XBP-1S-dependent manner. Collectively, our results demonstrate that PCAF mediates the XBP-1S-dependent transcription through the interaction with XBP-1S.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , p300-CBP Transcription Factors/metabolism , DNA-Binding Proteins/chemistry , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Human T-lymphotropic virus 1/genetics , Humans , Protein Interaction Domains and Motifs , Regulatory Factor X Transcription Factors , Transcription Factors/chemistry , Unfolded Protein Response/genetics , X-Box Binding Protein 1
10.
J Biotechnol ; 146(4): 160-8, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20188772

ABSTRACT

Development of high-throughput functional genomic screening, including siRNA screening, provides a novel approach for quick identification of critical factors involved in biological processes. Here, we apply this strategy to search for cellular genes involved in recombinant protein production. Since most of biopharmaceutical proteins are secreted proteins, we develop a cell-based reporter assay using a secreted luciferase, Gaussia luciferase (Gluc), as the reporter. Human embryonic kidney 293 (HEK293) cells transiently transfected with the Gluc reporter plasmid are used to screen our siRNA panel. Three cellular genes, CCAAT/enhancer binding protein gamma (CEBPG), potassium channel tetramerisation domain containing 2 (KCTD2), transmembrane protein 183A (TMEM183A), were isolated from the screening. Production of erythropoietin (EPO) was significantly inhibited when CEBPG, KCTD2, and TMEM183A were knocked down. Furthermore, overexpression of CEBPG is shown to significantly improve production of recombinant EPO, interferon gamma, and monoclonal antibody in HEK293 and Chinese hamster ovary cells. Collectively, this novel Gluc-based siRNA screening system is proven to be a useful tool for investigation of secreted protein production in mammalian cells.


Subject(s)
Luciferases/genetics , Protein Engineering/methods , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CHO Cells , Cell Growth Processes/physiology , Cell Line , Cricetinae , Cricetulus , Erythropoietin/genetics , Erythropoietin/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Luciferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Recombinant Proteins/genetics
11.
Protein Pept Lett ; 16(5): 490-8, 2009.
Article in English | MEDLINE | ID: mdl-19442228

ABSTRACT

Human chitinases (EC.3.2.1.14) are classified into family 18 of glycosyl hydrolase (GH18) superfamily based on their amino acid sequence similarities. Active chitinase hydrolyzes chitin, a beta-1,4-linked N-acetyl-D-glucosamine oligosaccharide. Chitin is a major structural component of the insect exoskeletons and fungal cell walls, but is not found in vertebrates. In human, eight GH18 chitinases have been identified including active chitotriosidase and acidic mammalian chitinase. Most of the human chitinases lack chitinolytic activity due to mutation of an essential glutamic acid residue at the catalytic domain, and they are termed chitolectin. This review highlights some characteristics of human chitinases, compares structural differences among some human GH18 members, and discusses their cellular regulation and function. Finally, we summarize current views on the role of human chitinases in a variety of human diseases.


Subject(s)
Chitinases/metabolism , Disease , Amino Acid Sequence , Animals , Biomarkers/chemistry , Biomarkers/metabolism , Chitinases/chemistry , Humans , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...