Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell Mol Life Sci ; 79(7): 367, 2022 Jun 19.
Article in English | MEDLINE | ID: mdl-35718818

ABSTRACT

Upregulation of death-domain-associated protein (Daxx) is strongly associated with diverse cancer types. Among these, the clinicopathological significance and molecular mechanisms of Daxx overexpression in colorectal cancer (CRC) remain unknown. Here, we showed that Daxx expression was increased in both clinical CRC samples and CRC cell lines. Daxx knockdown significantly reduced proliferation activity in CRC cells and tumor growth in a xenograft model. Further studies revealed that Daxx expression could be attenuated by either treatment with the PIK3CA inhibitor PIK-75 or PIK3CA depletion in CRC cells. Conversely, expression of PIK3CA constitutively active mutants could increase Daxx expression. These data suggest that PIK3CA positively regulates Daxx expression. Consistently, the expression levels of PIK3CA and Daxx were positively correlated in sporadic CRC samples. Interestingly, Daxx knockdown or overexpression yielded decreased or increased levels of PIK3CA, respectively, in CRC cells. We further demonstrated that Daxx activates the promoter activity and expression of PIK3CA. Altogether, our results identify a mechanistic pathway of Daxx overexpression in CRC and suggest a reciprocal regulation between Daxx and PIK3CA for CRC cell growth.


Subject(s)
Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , Cell Line, Tumor , Cell Proliferation/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism
2.
Biochem Biophys Res Commun ; 584: 32-38, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34763165

ABSTRACT

Melatonin is a hormone majorly secreted by the pineal gland and contributes to a various type of physiological functions in mammals. The melatonin production is tightly limited to the AANAT level, yet the most known molecular mechanisms underlying AANAT gene transcription is limited in the pinealocyte. Here, we find that c-Fos and cAMP-response element-binding protein (CREB) decreases and increases the AANAT transcriptional activity in renal tubular epithelial cell, respectively. Notably, c-Fos knockdown significantly upregulates melatonin levels in renal tubular cells. Functional results indicate that AANAT expression is decreased by c-Fos and resulted in enhancement of cell damage in albumin-injury cell model. We further find an inverse correlation between c-Fos and AANAT levels in renal tubular cells from experimental membranous nephropathy (MN) samples and clinical MN specimens. Our finding provides the molecular basis of c-Fos in transcriptionally downregulating expression of AANAT and melatonin, and elucidate the protective role of AANAT in preventing renal tubular cells death in albumin-injury cell model and MN progression.


Subject(s)
Arylalkylamine N-Acetyltransferase/genetics , Down-Regulation , Epithelial Cells/metabolism , Glomerulonephritis, Membranous/genetics , Proto-Oncogene Proteins c-fos/genetics , Animals , Arylalkylamine N-Acetyltransferase/metabolism , Cell Line , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , HEK293 Cells , Humans , Kidney Tubules/cytology , Melatonin/metabolism , Mice , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation
3.
Nat Commun ; 12(1): 645, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510150

ABSTRACT

Hepatocellular carcinoma (HCC) is the most predominant primary malignancy in the liver. Genotoxic and genetic models have revealed that HCC cells are derived from hepatocytes, but where the critical region for tumor foci emergence is and how this transformation occurs are still unclear. Here, hyperpolyploidization of hepatocytes around the centrilobular (CL) region is demonstrated to be closely linked with the development of HCC cells after diethylnitrosamine treatment. We identify the CL region as a dominant lobule for accumulation of hyperpolyploid hepatocytes and preneoplastic tumor foci formation. We also demonstrate that upregulation of Aurkb plays a critical role in promoting hyperpolyploidization. Increase of AURKB phosphorylation is detected on the midbody during cytokinesis, causing abscission failure and hyperpolyploidization. Pharmacological inhibition of AURKB dramatically reduces nucleus size and tumor foci number surrounding the CL region in diethylnitrosamine-treated liver. Our work reveals an intimate molecular link between pathological hyperpolyploidy of CL hepatocytes and transformation into HCC cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Hepatocytes/metabolism , Liver Neoplasms/genetics , Liver/metabolism , Polyploidy , Precancerous Conditions/genetics , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Diethylnitrosamine/toxicity , Female , Hepatocytes/drug effects , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Confocal , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism
4.
J Cell Physiol ; 236(3): 2023-2035, 2021 03.
Article in English | MEDLINE | ID: mdl-32730662

ABSTRACT

The downregulation of melatonin receptor 1A (MTNR1A) is associated with a range of pathological conditions, including membranous nephropathy. Knowledge of the mechanism underlying MTNR1A expression has been limited to the transcriptional regulation level. Here, RNA interference screening in human kidney cells revealed that heterogeneous nuclear ribonucleoprotein L (hnRNPL) upregulated MTNR1A RNA post-transcriptionally. hnRNPL knockdown or overexpression led to increased or decreased levels of cyclic adenosine monophosphate-responsive element-binding protein phosphorylation, respectively. Molecular studies showed that cytoplasmic hnRNPL exerts a stabilizing effect on the MTNR1A transcript through CA-repeat elements in its coding region. Further studies revealed that the interaction between hnRNPL and MTNR1A serves to protect MNTR1A RNA degradation by the exosome component 10 protein. MTNR1A, but not hnRNPL, displays a diurnal rhythm in mouse kidneys. Enhanced levels of MTNR1A recorded at midnight correlated with robust binding activity between cytoplasmic hnRNPL and the MTNR1A transcript. Both hnRNPL and MTNR1A were decreased in the cytoplasm of tubular epithelial cells from experimental membranous nephropathy kidneys, supporting their clinical relevance. Collectively, our data identified cytoplasmic hnRNPL as a novel player in the upregulation of MTNR1A expression in renal tubular epithelial cells, and as a potential therapeutic target.


Subject(s)
Cytoplasm/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Kidney Tubules/metabolism , Receptor, Melatonin, MT1/genetics , Animals , Cell Line , Circadian Rhythm/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Epithelial Cells/metabolism , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Humans , Kidney Tubules/pathology , Mice, Inbred BALB C , Models, Biological , Open Reading Frames/genetics , Phosphorylation , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Melatonin, MT1/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Up-Regulation/genetics
5.
Sci Rep ; 10(1): 9350, 2020 Jun 04.
Article in English | MEDLINE | ID: mdl-32494003

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Sci Rep ; 8(1): 9786, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29955155

ABSTRACT

Smad4, a common-mediator of Smads, plays a central role in forming complexes with receptor-phosphorylated Smads, and then transduces transforming growth factor (TGF)-ß signals into the nuclei. Although many cellular factors are involved in TGF-ß induced epithelial-to-mesenchymal transition (EMT) and cell migration, very little is known with the mechanism of Smad4 regulation on pro-oncogenes response by TGF-ß. Herein, we demonstrate the interaction of Sentrin-specific protease 2 (SENP2) with Smad4 through SENP2 residue 363~400. The same segment is also important for desumoylation of Smad4, and able to relieve sumoylation-mediated TGF-ß repression. The SENP2363~400 segment is critical for TGF-ß-induced cell migration, which is correlated with SENP2363~400 deletion mutant failed to increase matrix metalloproteinase (MMP)-9 and EMT marker gene expression. Moreover, our results suggest that the interaction and desumoylation between SENP2 and Smad4 promote cell migration in triple-negative breast cancer cells. Altogether, our data show how SENP2 regulates its substrate for desumoylation, and also the role of SENP2 in TGF-ß induced cancer cell migration.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Cysteine Endopeptidases/metabolism , Cell Movement , Humans , Protein Binding , Signal Transduction/drug effects , Smad4 Protein/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Substrate Specificity , Sumoylation , Transforming Growth Factor beta
7.
Hum Mol Genet ; 27(13): 2306-2317, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29668892

ABSTRACT

XRCC1 is an essential scaffold protein for base excision repair (BER) and helps to maintain genomic stability. XRCC1 has been indicated as a substrate for small ubiquitin-like modifier modification (SUMOylation); however, how XRCC1 SUMOylation is regulated in cells and how SUMOylated XRCC1 regulates BER activity are not well understood. Here, we show that SUMOylation of XRCC1 is regulated in cells under methyl-methanesulfonate (MMS) treatment and facilitates BER. Poly(ADP-ribose) polymerase 1 (PARP1) is activated by MMS immediately and synthesizes poly(ADP-ribose) (PAR), which in turn promotes recruitment of SUMO E3 TOPORS to XRCC1 and facilitates XRCC1 SUMOylation. A SUMOylation-defective mutant of XRCC1 had lower binding activity for DNA polymerase beta (POLB) and was linked to a lower capacity for repair of MMS-induced DNA damages. Our study therefore identified a pathway in which DNA damage-induced poly(ADP-ribosyl)ation (PARylation) promotes SUMOylation of XRCC1, which leads to more efficient recruitment of POLB to complete BER.


Subject(s)
DNA Polymerase beta/genetics , Poly ADP Ribosylation/genetics , Sumoylation/genetics , X-ray Repair Cross Complementing Protein 1/genetics , Alcohol Oxidoreductases/genetics , DNA Damage/drug effects , DNA Repair/genetics , DNA-Binding Proteins/genetics , Genomic Instability/genetics , Humans , Methyl Methanesulfonate/pharmacology , Poly (ADP-Ribose) Polymerase-1/genetics , Protein Binding/genetics
8.
J Pineal Res ; 65(1): e12482, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29480949

ABSTRACT

Membranous nephropathy (MN), a type of glomerular nephritis, is one of the most common causes of nephrotic syndrome in adults. Although it is known that melatonin plays a protective role in MN, the role of melatonin receptors in the pathophysiology of MN is unclear. Using an experimental MN model and clinical MN specimens, we studied melatonin receptor expression and found that melatonin receptor 1A (MTNR1A) expression was significantly downregulated in renal tubular epithelial cells. Molecular studies showed that the transcription factor pituitary homeobox-1 (PITX1) promoted MTNR1A expression via direct binding to its promoter. Treatment of a human tubular cell line with albumin to induce injury resulted in the stable reduction in MTNR1A and PITX1 expression. PITX1 levels were significantly downregulated in tubular epithelial cells from mice MN kidneys and MN renal specimens. Knockdown of MTNR1A, PITX1, or cyclic adenosine monophosphate-responsive element-binding protein (CREB) decreased E-cadherin (CDH1) expression, but upregulated Per2 and α-smooth muscle actin (αSMA) expression. Blockade of the MTNR1A receptor with luzindole in MN mice further impaired renal function; this was accompanied by CDH1 downregulation and Per2 and αSMA upregulation. Together, our results suggest that in injured tissue, decreased PITX1 expression at the MTNR1A promoter regions leads to decreased levels of MTNR1A in renal tubular epithelial cells, which increases the future risk of MN.


Subject(s)
Epithelial Cells/metabolism , Glomerulonephritis, Membranous/metabolism , Kidney Tubules/metabolism , Paired Box Transcription Factors/metabolism , Receptor, Melatonin, MT1/metabolism , Animals , Chromatin Immunoprecipitation , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Glomerulonephritis, Membranous/genetics , Immunohistochemistry , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic/genetics , RNA Interference
9.
Oncotarget ; 8(67): 111631-111641, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29340080

ABSTRACT

Idiopathic membranous nephropathy (MN) is an autoimmune-mediated glomerulonephritis and the most common cause of idiopathic nephrotic syndrome in adult humans. A tumor necrosis factor α (TNF-α)-mediated inflammatory response via TNF receptor 1 (TNFR1) and TNFR2 has been proposed as a pathogenic factor. In this study, we assessed the therapeutic response to blocking TNF signaling in experimental MN. Murine MN was induced experimentally by cationic bovine serum albumin (cBSA); phosphate-buffered saline was used in control mice. In MN mice, TNF was inhibited by etanercept blocking of TNFR1/TNFR2 or the preligand assembly domain fusion protein (PLAD.Fc), a small fusion protein that can preferentially block TNFR1 signaling. Disease severity and possible mechanisms were assessed by analyzing the metabolic and histopathology profiles, lymphocyte subsets, immunoglobulin production, oxidative stress, and apoptosis. cBSA-induced MN mice exhibited typical nephrotic syndrome and renal histopathology. MN mice given etanercept or PLAD.Fc did not exhibit significant reduction of proteinuria, amelioration of glomerular lesions, or attenuation of immune complex deposition. Immune cell subsets, serum immunoglobulin levels, production of reactive oxygen species, and cell apoptosis in the kidney were not altered by TNF inhibition. By contrast, MN mice receiving etanercept or PLAD.Fc exhibited significantly decreased infiltration of immune cells into the kidney. These results show that the therapeutic effects of blocking TNFR1 and/or TNFR2 signaling in experimental MN are not clinically effective. However, TNF signaling inhibition significantly attenuated renal immune cell infiltration in experimental MN.

10.
Oncotarget ; 7(28): 43256-43266, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27248326

ABSTRACT

Long noncoding RNAs (lncRNAs) dysregulated in cancer potentially play oncogenic or tumor-suppressive roles. While the X inactivate-specific transcript (Xist) lncRNA is important for X-chromosome inactivation in female cells, very little is known about the role of Xist in human breast cancer in modulating cellular pathway(s). Here, we show that Xist expression is significantly reduced in breast tumor samples and cancer cell lines. Xist knockdown or overexpression resulted in increased or decreased levels, respectively, of AKT phosphorylation and cell viability. Further studies revealed an inverse correlation between Xist and phospho-AKT levels in breast cancer samples. Additionally, Xist knockdown-elicited increase of cell viability was attenuated by AKT inhibitor. These results suggest that Xist negatively regulates cell viability via inhibition of AKT activation. Interestingly, decreased Xist expression in breast cancer samples was associated with reduced levels of Jpx RNA, an lncRNA that positively regulates Xist promoter activity. Accordingly, Jpx knockdown enhanced AKT activation and cell viability. We also demonstrate that knockdown of Xist or SPEN, an intermediator protein to link Xist, SMRT co-repressor and HDAC3 complexes for X-chromosome inactivation, decreased expression of PHLPP1, a phosphatase to remove AKT phosphorylation, via increased HDAC3 recruitment to the PHLPP1 promoter, correlating with increased AKT phosphorylation. Our findings elucidate the tumor suppressor role of Xist in breast cancer and provide the molecular basis of Xist in downregulating AKT activation.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylases/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , Animals , Breast Neoplasms/pathology , Cell Survival/genetics , DNA-Binding Proteins , Datasets as Topic , Down-Regulation , Female , Gene Knockdown Techniques , Genes, Tumor Suppressor , Histone Deacetylases/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MCF-7 Cells , Mice , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Phosphorylation , Promoter Regions, Genetic , RNA Interference , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Signal Transduction/genetics , Tissue Array Analysis , Up-Regulation , X Chromosome Inactivation/genetics
11.
Clin Oral Investig ; 20(3): 533-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26205068

ABSTRACT

OBJECTIVES: Death domain-associated protein (Daxx) has been recently implicated as a positive factor in ovarian cancer and prostate cancer, but the role of Daxx in oral squamous cell carcinoma (OSCC) has never been addressed. Herein, we investigate the expression and function of Daxx in OSCC. MATERIALS AND METHODS: RT-quantitative PCR, Western blotting, and immunohistochemistry were used to evaluation of the expression of Daxx in human OSCC cell lines and clinical surgical specimens. Short hairpin RNA targeting Daxx was transduced by lentivirus infection to knockdown the expression of Daxx in SAS and SCC25 cell lines, and the influence of this knockdown was evaluated by analyzing the growth and the cell cycle in transduced cells. Immunoprecipitation and sequential chromatin immunoprecipitation-quantitative PCR were used to analyze the associations between Daxx, TCF4, and cyclin D1 promoter. Xenograft tumor model was used to evaluate the in vivo tumorigenicity of Daxx in OSCC. RESULTS: Daxx mRNA and protein expression are elevated in several OSCC cell lines and human OSCC samples in comparison to those in normal tissue. We further find that depletion of Daxx decreases OSCC cell growth activity through G1 cell cycle arrest. Daxx silencing reduces cyclin D1 expression via a Daxx-TCF4 interaction, whereas the Daxx depletion-mediated G1 arrest can be relieved by ectopic expression of cyclin D1. Moreover, we show that in OSCC clinical samples, the expression of Daxx is significantly correlated with that of cyclin D1. CONCLUSION: Our data demonstrate the importance of Daxx in regulation of cyclin D1 expression and provide the first evidence that Daxx exhibits tumor-promoting activity in OSCC. CLINICAL RELEVANCE: Daxx plays an important role in malignant transformation of OSCC and may serves as a target for cancer prevention and treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Squamous Cell/metabolism , Cyclin D1/metabolism , Mouth Neoplasms/metabolism , Nuclear Proteins/physiology , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Animals , Blotting, Western , Carcinoma, Squamous Cell/pathology , Cell Cycle , Cell Line, Tumor , Co-Repressor Proteins , Female , Heterografts , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Molecular Chaperones , Mouth Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Transcription Factor 4
12.
PLoS One ; 10(5): e0125726, 2015.
Article in English | MEDLINE | ID: mdl-25954969

ABSTRACT

BACKGROUND: Idiopathic membranous nephropathy (MN) is an autoimmune-mediated glomerulonephritis and a common cause of nephrotic syndrome in adults. There are limited available treatments for MN. We assessed the efficacy of resveratrol (RSV) therapy for treatment of MN in a murine model of this disease. METHODS: Murine MN was experimentally induced by daily subcutaneous administration of cationic bovine serum albumin, with phosphate-buffered saline used in control mice. MN mice were untreated or given RSV. Disease severity and pathogenesis was assessed by determination of metabolic and histopathology profiles, lymphocyte subsets, immunoglobulin production, oxidative stress, apoptosis, and production of heme oxygenase-1 (HO1). RESULTS: MN mice given RSV had significantly reduced proteinuria and a marked amelioration of glomerular lesions. RSV also significantly attenuated immunofluorescent staining of C3, although there were no changes of serum immunoglobulin levels or immunocomplex deposition in the kidneys. RSV treatment of MN mice also reduced the production of reactive oxygen species (ROS), reduced cell apoptosis, and upregulated heme oxygenase 1 (HO1). Inhibition of HO1 with tin protoporphyrin IX partially reversed the renoprotective effects of RSV. The HO1 induced by RSV maybe via Nrf2 signaling. CONCLUSION: Our results show that RSV increased the expression of HO1 and ameliorated the effects of membranous nephropathy in a mouse model due to its anti-complement, anti-oxidative, and anti-apoptotic effects. RSV appears to have potential as a treatment for MN.


Subject(s)
Glomerulonephritis, Membranous/drug therapy , Heme Oxygenase-1/biosynthesis , Kidney/drug effects , Stilbenes/administration & dosage , Animals , Apoptosis/drug effects , Cattle , Complement System Proteins/drug effects , Disease Models, Animal , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Heme Oxygenase-1/genetics , Kidney/injuries , Kidney/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Metalloporphyrins/administration & dosage , Mice , Oxidative Stress/drug effects , Protoporphyrins/administration & dosage , Reactive Oxygen Species/metabolism , Resveratrol , Serum Albumin/administration & dosage
13.
Cell Cycle ; 13(19): 3132-42, 2014.
Article in English | MEDLINE | ID: mdl-25486572

ABSTRACT

Promyelocytic leukemia protein (PML) is emerging as an important tumor suppressor. Its expression is lost during the progression of several types of cancer, including lung cancer. The EGF receptor (EGFR), a membrane-bound receptor tyrosine kinase, transduces intracellular signals responsible for cell proliferation, differentiation and migration. EGFR activity is frequently abnormally upregulated in lung adenocarcinoma (LAC) and thus is considered to be a driving oncogene for LAC. EGFR translocates into the nucleus and transcriptionally activates genes, such as CCND1, that promote cell growth. Recently, we demonstrated that PML interacted with nuclear EGFR (nEGFR) and suppressed the nEGFR-mediated transcriptional activation of CCND1 in lung cancer cells, thereby restraining cell growth. When we further investigated the interplay between PML and EGFR in lung cancer metastasis, we found that the matrix metalloprotease-2 gene (MMP2) was a novel nEGFR target gene and was repressed by PML. We provide evidence that nEGFR bound to the AT-rich sequence (ATRS) in the MMP2 promoter and enhanced its transcriptional activity. In addition, we demonstrated that PML repressed nEGFR-induced MMP2 transcription and reduced cell invasion. PML was recruited by nEGFR to the MMP2 promoter where it reduced histone acetylation, leading to the transcriptional repression of MMP2. Finally, we demonstrated that PML upregulation by interferon-ß (IFNß) in lung cancer cells decreased MMP2 expression and cell invasion. Together, our results suggested that IFNß induced PML to inhibit lung cancer metastasis by repressing the nEGFR-mediated transcriptional activation of MMP2.


Subject(s)
ErbB Receptors/metabolism , Matrix Metalloproteinase 2/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Epidermal Growth Factor/pharmacology , HEK293 Cells , Histones/metabolism , Humans , Interferon-beta/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Promoter Regions, Genetic , Promyelocytic Leukemia Protein , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Activation/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Up-Regulation/drug effects
14.
Biochem Biophys Res Commun ; 452(3): 415-21, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25157805

ABSTRACT

Membranous nephropathy (MN), a type of glomerular nephritis, is the most common cause of nephrotic syndrome in human adults. Changes in gene expression as a result of epigenetic dysregulation through long noncoding RNAs (lncRNAs) are increasingly being recognized as important factors in disease. Using an experimental MN mouse model, we identify the first dysregulated lncRNAs, Xist and NEAT1, whose levels are significantly upregulated in both tubular epithelial and glomerular cells. MN is also often characterized by glomerular podocyte injury. Treatment of a mouse podocyte cell line with lipopolysaccharides to induce injury resulted in the stable elevation of Xist, but not NEAT1 levels. In mice, the observed changes in Xist levels are specific: Xist can be effectively detected in urine, with a strong correlation to disease severity, but not serum in MN samples. We find that regulation of Xist may be controlled by post-translational modifications. H3K27me3 levels are significantly downregulated in mouse MN kidney, where chromatin immunoprecipitation experiments also showed decreased H3K27me3 at Xist promoter regions. Finally, we show that our findings in mice can be extended to human clinical samples. Urinary Xist is significantly elevated in urine samples from patients with different types of glomerular nephritis, including MN, compared to normal counterparts. Together, our results suggest that a reduction of H3K27me3 at Xist promoter regions leads to elevated levels of urinary Xist, which may be used as a biomarker to detect MN.


Subject(s)
Glomerulonephritis, Membranous/genetics , Histones/genetics , Podocytes/metabolism , RNA, Long Noncoding/genetics , Animals , Biomarkers/urine , Cell Line , Disease Models, Animal , Female , Gene Expression Regulation , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/pathology , Glomerulonephritis, Membranous/urine , Histones/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Podocytes/drug effects , Podocytes/pathology , Promoter Regions, Genetic , Protein Binding , RNA, Long Noncoding/agonists , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/urine , Severity of Illness Index
15.
Cell Cycle ; 11(1): 99-108, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22185778

ABSTRACT

The phosphorylation of cAMP response element-binding protein (CREB) induced by the cAMP-dependent protein kinase A (PKA) elicits the recruitment of CREB-binding protein (CBP) for activating cAMP responsive gene expression. Several reports indicate that proteins binding to CREB and/or CBP play important roles in modulating the CREB-dependent transactivation. Here, we show that Daxx interacts with CREB and modulates CREB-mediated transcription. Daxx was identified as a CREB-interacting protein by a yeast two-hybrid screen. Depletion of endogenous Daxx by specific shRNA or overexpression of Daxx resulted in decreased or increased levels of the cAMP/PKA-induced reporter activity and target gene expression, respectively. In vitro and in vivo binding studies revealed that Daxx C-terminal domain binds to CREB basic leucine zipper domain. The binding of Daxx to CREB correlates with its repressive effect on a CRE-mediated reporter activity induced by forskolin or PKA. Furthermore, the results of electrophoresis mobility shift assays and chromatin immunoprecipitation experiments showed that Daxx attenuated the DNA binding potential of the CREB. Our study provides a previously undescribed role of Daxx in repressing cAMP-responsive gene expression and also a mechanism underlying the repressive effect of Daxx on CREB transcriptional potential.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , CREB-Binding Protein/metabolism , Cell Line , Chromatin Immunoprecipitation , Co-Repressor Proteins , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/metabolism , Humans , Leucine Zippers , Mice , Molecular Chaperones , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation
16.
Mol Cell ; 42(1): 62-74, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474068

ABSTRACT

Small ubiquitin-like modifier (SUMO) conjugation and interaction are increasingly associated with various cellular processes. However, little is known about the cellular signaling mechanisms that regulate proteins for distinct SUMO paralog conjugation and interactions. Using the transcriptional coregulator Daxx as a model, we show that SUMO paralog-selective binding and conjugation are regulated by phosphorylation of the Daxx SUMO-interacting motif (SIM). NMR structural studies show that Daxx (732)E-I-I-V-L-S-D-S-D(740) is a bona fide SIM that binds to SUMO-1 in a parallel orientation. Daxx-SIM is phosphorylated by CK2 kinase at residues S737 and S739. Phosphorylation promotes Daxx-SIM binding affinity toward SUMO-1 over SUMO-2/3, causing Daxx preference for SUMO-1 conjugation and interaction with SUMO-1-modified factors. Furthermore, Daxx-SIM phosphorylation enhances Daxx to sensitize stress-induced cell apoptosis via antiapoptotic gene repression. Our findings provide structural insights into the Daxx-SIM:SUMO-1 complex, a model of SIM phosphorylation-enhanced SUMO paralog-selective modification and interaction, and phosphorylation-regulated Daxx function in apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/physiology , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Apoptosis/genetics , Carrier Proteins/genetics , Casein Kinase II/metabolism , Cell Line , Co-Repressor Proteins , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Models, Molecular , Molecular Chaperones , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , SUMO-1 Protein/metabolism , Stress, Physiological
17.
Biochem Biophys Res Commun ; 386(4): 762-8, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19563778

ABSTRACT

Constitutive activation of the transcription factor TCF4 activity by mutated APC or beta-catenin contributes to cell neoplastic transformation. While numerous proteins were identified to activate TCF4-dependent activity via beta-catenin interaction, little is known about factors directly acting on TCF4. Here we report that Daxx binds to TCF4 and potentiates beta-catenin/TCF4-mediated transcriptional activation and target gene expression. Binding studies revealed that Daxx-TCF4 interaction is through the C-terminal domain of Daxx and TCF4 segment containing amino acid residue 269-327. Alteration of Daxx levels in cells by overexpression or RNA interference resulted in an increase or decrease of the beta-catenin/TCF4-dependent transactivation activity and target gene expression, respectively. Furthermore, TCF4-(269-327) segment acts as a dominantly negative mutant by blocking Daxx-TCF4 interaction and TCF4-mediated transactivation potential. Together, our results suggest that Daxx functions as a positive coregulator in modulating the beta-catenin/TCF4-dependent transcriptional potential via TCF4 interaction.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Co-Repressor Proteins , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Humans , Immunoprecipitation , Molecular Chaperones , Nuclear Proteins/genetics , Protein Structure, Tertiary , Transcription Factor 4 , Transcription Factors/genetics , Transcription, Genetic , Two-Hybrid System Techniques , beta Catenin/genetics
18.
Biochem Biophys Res Commun ; 384(4): 444-9, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19422794

ABSTRACT

Small ubiquitin-like modifier (SUMO) modification has been shown to be involved in the regulation of various cellular processes including gene transcription, nucleocytoplasmic transport, cell cycle, DNA repair, stress response, and signal transduction. However, very little is known about the process of cell migration being modulated by SUMO modification. Here, we show that calpain-2, a protease involved in cell motility, can be SUMO modified at lysine residue 390. Converting the SUMO acceptor lysine residue to arginine residue significantly attenuated calpain-2 activity, correlating well with a loss of calpain-2-elicited cell motility. Accordingly, expression of SENP1 could abrogate calpain-2 sumoylation, causing an inhibition on calpain-2-dependent activity and cell motility. These results not only identify calpain-2 as a substrate for sumoylation but also provide an important role of sumoylation in regulating cell migration.


Subject(s)
Calpain/metabolism , Cell Movement , SUMO-1 Protein/metabolism , Animals , COS Cells , Calpain/genetics , Chlorocebus aethiops , Cysteine Endopeptidases , Endopeptidases/metabolism , Humans , Lysine/genetics , Lysine/metabolism , Mutation
19.
Cancer Sci ; 100(3): 537-45, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19134007

ABSTRACT

Strategies to increase antitumor efficacy of oncolytic adenoviruses are actively investigated. We have previously shown that E1B-55 kDa-deleted adenovirus, designated Ad5WS1, has therapeutic potential for treating hepatocellular carcinoma (HCC). To achieve HCC-restricted replication of oncolytic adenovirus, we generated Ad5WS2, an E1B-55 kDa-deleted adenovirus with its E1A gene driven by the liver-specific transthyretin promoter. Our results showed that Ad5WS2 could replicate within tumor cells where the transthyretin gene was expressed. Mouse transthyretin promoter was active in murine and human HCC cells, but relatively quiescent in cells of non-liver origin. Ad5WS2 caused severe cytolytic effect on HCC cells, but was much attenuated in non-HCC cells. Peritoneal administration of Ad5WS2 into mice bearing liver tumors grown in ascites resulted in enhanced survival. In an orthotopic HCC model, Ad5WS2, when systemically administered, exerted higher antitumor effects than Ad5WS1. Lack of viral replication in normal organs and minimal hepatic toxicity was noted after Ad5WS2 treatment. Furthermore, the antitumor effect of Ad5WS2 could be enhanced when combined with chemotherapeutic agent cisplatin in the ascites tumor model. These results suggest that E1B-55 kDa-deleted adenovirus driven by the transthyretin promoter may be a safer and more efficacious oncolytic agent for the treatment of primary and metastatic HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Oncolytic Virotherapy/methods , Prealbumin/genetics , Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E1B Proteins/deficiency , Adenovirus E1B Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Ascites/pathology , Ascites/virology , Carcinoma, Hepatocellular/virology , Cisplatin/pharmacology , Combined Modality Therapy , Female , Humans , Liver Neoplasms/virology , Mice , Oncolytic Viruses/genetics , Promoter Regions, Genetic
20.
Mol Cell ; 24(3): 341-54, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-17081986

ABSTRACT

Small ubiquitin-like modifier (SUMO) modification has emerged as an important posttranslational control of protein functions. Daxx, a transcriptional corepressor, was reported to repress the transcriptional potential of several transcription factors and target to PML oncogenic domains (PODs) via SUMO-dependent interactions. The mechanism by which Daxx binds to sumoylated factors mediating transcriptional and subnuclear compartmental regulation remains unclear. Here, we define a SUMO-interacting motif (SIM) within Daxx and show it to be crucial for targeting Daxx to PODs and for transrepression of several sumoylated transcription factors, including glucocorticoid receptor (GR). In addition, the capability of Daxx SIM to bind SUMO also controls Daxx sumoylation. We further demonstrate that arsenic trioxide-induced sumoylation of PML correlates with a change of endogenous Daxx partitioning from GR-regulated gene promoter to PODs and a relief of Daxx repression on GR target gene expression. Our results provide mechanistic insights into Daxx in SUMO-dependent transcriptional control and subnuclear compartmentalization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Arsenic Trioxide , Arsenicals/pharmacology , COS Cells , Carrier Proteins/chemistry , Chlorocebus aethiops , Co-Repressor Proteins , Dexamethasone/pharmacology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Molecular Chaperones , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/chemistry , Oxides/pharmacology , Promyelocytic Leukemia Protein , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...