Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(9): 4969-4984, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38452206

ABSTRACT

Proteasome-mediated degradation of chromatin-bound NF-κB is critical in terminating the transcription of pro-inflammatory genes and can be triggered by Set9-mediated lysine methylation of the RelA subunit. However, the E3 ligase targeting methylated RelA remains unknown. Here, we find that two structurally similar substrate-recognizing components of Cullin-RING E3 ligases, WSB1 and WSB2, can recognize chromatin-bound methylated RelA for polyubiquitination and proteasomal degradation. We showed that WSB1/2 negatively regulated a subset of NF-κB target genes via associating with chromatin where they targeted methylated RelA for ubiquitination, facilitating the termination of NF-κB-dependent transcription. WSB1/2 specifically interacted with methylated lysines (K) 314 and 315 of RelA via their N-terminal WD-40 repeat (WDR) domains, thereby promoting ubiquitination of RelA. Computational modeling further revealed that a conserved aspartic acid (D) at position 158 within the WDR domain of WSB2 coordinates K314/K315 of RelA, with a higher affinity when either of the lysines is methylated. Mutation of D158 abolished WSB2's ability to bind to and promote ubiquitination of methylated RelA. Together, our study identifies a novel function and the underlying mechanism for WSB1/2 in degrading chromatin-bound methylated RelA and preventing sustained NF-κB activation, providing potential new targets for therapeutic intervention of NF-κB-mediated inflammatory diseases.


Subject(s)
Chromatin , Proteasome Endopeptidase Complex , Transcription Factor RelA , Ubiquitination , Humans , Chromatin/metabolism , HEK293 Cells , Lysine/metabolism , Methylation , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Transcription Factor RelA/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
2.
Cell Death Dis ; 14(6): 382, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37380643

ABSTRACT

The transcriptional repressor Snail induces EMT during embryonic development and tumor metastasis. Growing evidence indicates that Snail functions as a trans-activator to induce gene expression; however, the underlying mechanism remains elusive. Here, we report that Snail cooperates with GATA zinc finger protein p66ß to transactivate genes in breast cancer cells. Biologically, depletion of p66ß reduces cell migration and lung metastasis in BALB/c mice. Mechanistically, Snail interacts with p66ß and cooperatively induces gene transcription. Notably, a group of genes induced by Snail harbor conserved G-rich cis-elements (5'-GGGAGG-3', designated as G-box) in their proximal promoter regions. Snail directly binds to G-box via its zinc fingers and transactivates the G-box-containing promoters. p66ß enhances Snail binding affinity to G-box, whereas depletion of p66ß results in a decreased binding affinity of Snail to the endogenous promoters and concomitantly reduces the transcription of Snail-induced genes. Taken together, these data demonstrated that p66ß is critical for Snail-mediated cell migration by acting as a co-activator of Snail to induce genes containing G-box elements in the promoters.


Subject(s)
Lung Neoplasms , Transcription Factors , Female , Pregnancy , Animals , Mice , Transcription Factors/genetics , Cell Movement/genetics , Embryonic Development , Lung Neoplasms/genetics , Mice, Inbred BALB C , Zinc Fingers
3.
Cell Death Dis ; 14(4): 288, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37095090

ABSTRACT

Snail is a denoted transcriptional repressor that plays key roles in epithelial-mesenchymal transition (EMT) and metastasis. Lately, a plethora of genes can be induced by stable expression of Snail in multiple cell lines. However, the biological roles of these upregulated genes are largely elusive. Here, we report identification of a gene encoding the key GlcNAc sulfation enzyme CHST2 is induced by Snail in multiple breast cancer cells. Biologically, CHST2 depletion results in inhibition of breast cancer cell migration and metastasis, while overexpression of CHST2 promotes cell migration and lung metastasis in nude mice. In addition, the expression level of MECA79 antigen is elevated and blocking the cell surface MECA79 antigen with specific antibodies can override cell migration mediated by CHST2 upregulation. Moreover, the sulfation inhibitor sodium chlorate effectively inhibits the cell migration induced by CHST2. Collectively, these data provide novel insights into the biology of Snail/CHST2/MECA79 axis in breast cancer progression and metastasis as well as potential therapeutic strategy for the diagnosis and treatment of breast cancer metastasis.


Subject(s)
Epithelial-Mesenchymal Transition , Signal Transduction , Animals , Mice , Snail Family Transcription Factors/metabolism , Cell Line, Tumor , Mice, Nude , Cell Movement/physiology , Neoplasm Metastasis , Gene Expression Regulation, Neoplastic
4.
Front Oncol ; 12: 900166, 2022.
Article in English | MEDLINE | ID: mdl-36185184

ABSTRACT

DLC1 (deleted in liver cancer-1) is downregulated or deleted in colorectal cancer (CRC) tissues and functions as a potent tumor suppressor, but the underlying molecular mechanism remains elusive. We found that the conditioned medium (CM) collected from DLC1-overexpressed SW1116 cells inhibited the migration of colon adenocarcinoma cells HCT116 and SW1116, but had no effect on proliferation, which suggested DLC1-mediated secretory components containing a specific inhibitor for colon adenocarcinoma cell migration. Analysis by mass spectrometry identified mesencephalic astrocyte-derived neurotrophic factor (MANF) as a candidate. More importantly, exogenous MANF significantly inhibited the migration of colon adenocarcinoma cells HCT116 and SW1116, but did not affect proliferation. Mechanistically, DLC1 reduced the retention of MANF in ER by competing the interaction between MANF and GRP78. Taken together, these data provided new insights into the suppressive effects of DLC1 on CRC, and revealed the potential of MANF in the treatment of CRC.

5.
Mol Cell Endocrinol ; 539: 111485, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34619292

ABSTRACT

Adipogenesis is regulated by a complicated network of transcription factors among which PPARγ and C/EBP family members are the major regulators. During adipogenesis, C/EBPß is induced early and then transactivates PPARγ and C/EBPα, which cooperatively induce genes whose expressions give rise to the mature adipocyte phenotype. Identifying the factors that influence the expression and activity of C/EBPß should provide additional insight into the mechanisms regulating adipogenesis. Here, we demonstrate that depletion of Ajuba in 3T3-L1 cells significantly decreases mRNA and protein levels of PPARγ and C/EBPα and impairs adipocyte differentiation, while overexpression increases expression of these genes and promotes adipocyte differentiation. Moreover, restoration of C/EBPα or PPARγ expression in Ajuba-deficient 3T3-L1 cells improves the impaired lipid accumulation. Mechanistically, Ajuba interacts with C/EBPß and recruits CBP to facilitate the binding of C/EBPß to the promoter of PPARγ and C/EBPα, resulting in increased H3 histone acetylation and target gene expression. Collectively, these data indicate that Ajuba functions as a co-activator of C/EBPß, and may be an important therapeutic target for combating obesity-related diseases.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , LIM Domain Proteins/genetics , PPAR gamma/metabolism , 3T3-L1 Cells , Adipogenesis , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation , Gene Expression Regulation , HEK293 Cells , Humans , LIM Domain Proteins/metabolism , Lipid Metabolism , Mice , PPAR gamma/genetics
6.
Cells ; 10(12)2021 12 20.
Article in English | MEDLINE | ID: mdl-34944103

ABSTRACT

p66α is a GATA zinc finger domain-containing transcription factor that has been shown to be essential for gene silencing by participating in the NuRD complex. Several studies have suggested that p66α is a risk gene for a wide spectrum of diseases such as diabetes, schizophrenia, and breast cancer; however, its biological role has not been defined. Here, we report that p66α functions as a tumor suppressor to inhibit breast cancer cell growth and migration, evidenced by the fact that the depletion of p66α results in accelerated tumor growth and migration of breast cancer cells. Mechanistically, immunoprecipitation assays identify p66α as a p53-interacting protein that binds the DNA-binding domain of p53 molecule predominantly via its CR2 domain. Depletion of p66α in multiple breast cells results in decreased expression of p53 target genes, while over-expression of p66α results in increased expression of these target genes. Moreover, p66α promotes the transactivity of p53 by enhancing p53 binding at target promoters. Together, these findings demonstrate that p66α is a tumor suppressor by functioning as a co-activator of p53.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Prognosis , Promoter Regions, Genetic , Protein Binding , Protein Domains , Repressor Proteins/chemistry , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/chemistry
7.
J Cell Mol Med ; 25(16): 8006-8014, 2021 08.
Article in English | MEDLINE | ID: mdl-34173718

ABSTRACT

Ajuba is a multiple LIM domain-containing protein and functions as a transcriptional coregulator to modulate many gene expressions in various cellular processes. Here, we describe that the LIM domain of Ajuba interacts with Twist, and the Twist box is a pivotal motif for the interaction. Biologically, Ajuba enhances transcription of target gene N-cadherin as an obligate coactivator of Twist. The enhancement is achieved by binding to the E-box element within N-cadherin promoter as revealed by luciferase reporter and chromatin immunoprecipitation assays. Mechanistic investigation demonstrates that Ajuba recruits CBP and Twist to form a ternary complex at the Twist target promoter region and concomitantly enhances histone acetylation at these sites. These findings identify that Twist is a new interacting protein of Ajuba and Ajuba/Twist/CBP ternary complex may be a potential treatment strategy for Twist-related tumour metastasis.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation , LIM Domain Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Promoter Regions, Genetic , Sialoglycoproteins/metabolism , Twist-Related Protein 1/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Protein Interaction Domains and Motifs
8.
EMBO Rep ; 22(8): e51780, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34184805

ABSTRACT

Snail is a dedicated transcriptional repressor and acts as a master inducer of EMT and metastasis, yet the underlying signaling cascades triggered by Snail still remain elusive. Here, we report that Snail promotes colorectal cancer (CRC) migration by preventing non-coding RNA LOC113230-mediated degradation of argininosuccinate synthase 1 (ASS1). LOC113230 is a novel Snail target gene, and Snail binds to the functional E-boxes within its proximal promoter to repress its expression in response to TGF-ß induction. Ectopic expression of LOC113230 potently suppresses CRC cell growth, migration, and lung metastasis in xenograft experiments. Mechanistically, LOC113230 acts as a scaffold to facilitate recruiting LRPPRC and the TRAF2 E3 ubiquitin ligase to ASS1, resulting in enhanced ubiquitination and degradation of ASS1 and decreased arginine synthesis. Moreover, elevated ASS1 expression is essential for CRC growth and migration. Collectively, these findings suggest that TGF-ß and Snail promote arginine synthesis via inhibiting LOC113230-mediated LRPPRC/TRAF2/ASS1 complex assembly and this complex can serve as potential target for the development of new therapeutic approaches to treat CRC.


Subject(s)
Argininosuccinate Synthase , Epithelial-Mesenchymal Transition , Arginine , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Ubiquitination
9.
Oncogene ; 39(44): 6802-6815, 2020 10.
Article in English | MEDLINE | ID: mdl-32973332

ABSTRACT

Snail is a master inducer of epithelial-mesenchymal transition (EMT) and metastasis, however, Snail protein is labile and is quickly degraded through the predominate ubiquitination-mediated proteasome pathway. Deubiquitinases (DUBs) can counteract the Snail degradation process to maintain high level of Snail protein in cancer cells. In this study, we screened a cDNA library containing 79 DUBs, and discovered that a panel of DUBs consisting of USP13, USP28, USP29, USP37, OTUD6A, and DUB3 can markedly stabilize Snail protein, with USP29 displaying the strongest activity to prevent Snail degradation. Mechanistically, USP29 enhances the interaction of Snail and SCP1, resulting in simultaneous dephosphorylation and deubiquitination of Snail and thereafter cooperative prevention of Snail degradation. Biologically, ectopic expression of USP29 promotes gastric cancer cell migration, and depletion of Snail abolishes USP29-mediated cell migration; and USP29 can be induced by major EMT and metastatic inducing factors such as TGFß, TNFα, and hypoxia. More importantly, high expression levels of Snail, USP29, and SCP1 are associated with poor survival and prognosis. Collectively, these data indicate that Snail is a crucial substrate for USP29 to promote cell migration and USP29/SCP1 complex may be new therapeutic targets to treat metastatic cancer.


Subject(s)
Cell Movement/genetics , Phosphoprotein Phosphatases/metabolism , Snail Family Transcription Factors/metabolism , Stomach Neoplasms/pathology , Ubiquitin-Specific Proteases/metabolism , Cell Line, Tumor , Datasets as Topic , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kaplan-Meier Estimate , Phosphorylation , Prognosis , Protein Binding , Protein Stability , Proteolysis , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Ubiquitin-Specific Proteases/genetics , Ubiquitination
10.
Cell Death Differ ; 26(3): 565-579, 2019 03.
Article in English | MEDLINE | ID: mdl-29955124

ABSTRACT

The zinc finger protein Snail is a master regulator of epithelial-mesenchymal transition (EMT) and a strong inducer of tumor metastasis, yet the signal cascades triggered by Snail have not been completely revealed. Here, we report the discovery of the sulfation program that can be induced by Snail in breast cancer cells, and which plays an essential role in cell migration and metastasis. Specifically, Snail induces the expression of PAPSS2, a gene that encodes a rate-limiting enzyme in sulfation pathway, and VCAN, a gene that encodes the chondroitin sulfate proteoglycan Versican in multiple breast cancer cells. Depletion of PAPSS2 in MCF7 and MDA-MB-231 cells results in reduced cell migration, while overexpression of PAPSS2 promotes cell migration. Moreover, MDA-MB-231-shPAPSS2 cells display a significantly lower rate of lung metastasis and lower number of micrometastatic nodules in nude mice, and conversely, MDA-MB-231-PAPSS2 cells increase lung metastasis. Similarly, depletion of VCAN dampens the cell migration activity induced by Snail or PAPSS2 in MCF 10A cells. Moreover, PAPSS inhibitor sodium chlorate effectively decreases cell migration induced by Snail and PAPSS2. More importantly, the expression of Snail, PAPSS2, and VCAN is positively correlated in breast cancer tissues. Together, these findings are important for understanding the genetic programs that control tumor metastasis and may identify previously undetected therapeutic targets to treat metastatic disease.


Subject(s)
Breast Neoplasms/metabolism , Multienzyme Complexes/metabolism , Snail Family Transcription Factors/metabolism , Sulfate Adenylyltransferase/metabolism , Versicans/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , HEK293 Cells , Heterografts , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Multienzyme Complexes/genetics , Neoplasm Metastasis , Signal Transduction , Snail Family Transcription Factors/genetics , Sulfate Adenylyltransferase/genetics , Sulfates/metabolism , Versicans/genetics
11.
Mol Endocrinol ; 29(11): 1608-18, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26389695

ABSTRACT

The liver X receptors (LXRs) are important regulators of lipid, cholesterol, and glucose homeostasis by transcriptional regulation of many key genes in these processes, and the transcriptional activities of LXRs are finely controlled by cooperating with retinoid X receptors and many other coregulators. Here, we report that the LIM protein Ajuba binds to the hinge and the ligand binding domains of LXRα via its C-terminal tandem LIM motifs and enhances LXR target gene expression in liver cells. Depletion of Ajuba in HepG2 cells and in mouse primary hepatocytes decreases LXR target gene expression, whereas stable expression of Ajuba in HepG2 cells results in increased expression of these genes. Mechanistic investigations found that Ajuba selectively interacts with LXRα/retinoid X receptor-γ heterodimer to form a ternary complex, which displays a higher transactivation activity to LXR target genes. Moreover, Ajuba and LXR mutually affect their DNA binding activity at endogenous target chromatins and the cooperation between Ajuba and LXRα is dependent on the functional LXR response elements located in the target promoters. Together, our studies demonstrate that Ajuba is a novel coactivator for LXRs and may play important role in lipid and glucose metabolism.


Subject(s)
Gene Expression Regulation/genetics , LIM Domain Proteins/metabolism , Orphan Nuclear Receptors/metabolism , Retinoid X Receptor gamma/metabolism , Transcriptional Activation/genetics , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/metabolism , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Humans , LIM Domain Proteins/genetics , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding
12.
J Biol Chem ; 290(5): 2689-98, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25516596

ABSTRACT

Surface expression of voltage-gated Ca(2+) (Cav) channels is important for their function in calcium homeostasis in the physiology of excitable cells, but whether or not and how the α1 pore-forming subunits of Cav channels are trafficked to plasma membrane in the absence of the known Cav auxiliary subunits, ß and α2δ, remains mysterious. Here we showed that 14-3-3 proteins promoted functional surface expression of the Cav2.2 α1B channel in transfected tsA-201 cells in the absence of any known Cav auxiliary subunit. Both the surface to total ratio of the expressed α1B protein and the current density of voltage step-evoked Ba(2+) current were markedly suppressed by the coexpression of a 14-3-3 antagonist construct, pSCM138, but not its inactive control, pSCM174, as determined by immunofluorescence assay and whole cell voltage clamp recording, respectively. By contrast, coexpression with 14-3-3τ significantly enhanced the surface expression and current density of the Cav2.2 α1B channel. Importantly, we found that between the two previously identified 14-3-3 binding regions at the α1B C terminus, only the proximal region (amino acids 1706-1940), closer to the end of the last transmembrane domain, was retained by the endoplasmic reticulum and facilitated by 14-3-3 to traffic to plasma membrane. Additionally, we showed that the 14-3-3/Cav ß subunit coregulated the surface expression of Cav2.2 channels in transfected tsA-201 cells and neurons. Altogether, our findings reveal a previously unidentified regulatory function of 14-3-3 proteins in promoting the surface expression of Cav2.2 α1B channels.


Subject(s)
14-3-3 Proteins/metabolism , Calcium Channels, N-Type/metabolism , 14-3-3 Proteins/genetics , Animals , Blotting, Western , Calcium Channels, N-Type/genetics , Cell Membrane/metabolism , Cells, Cultured , Electrophysiology , Endoplasmic Reticulum/metabolism , Immunoprecipitation , Protein Binding , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
13.
Cancer Res ; 74(16): 4353-63, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24903147

ABSTRACT

Transcriptional repressor Snail is a master regulator of epithelial-mesenchymal transition (EMT), yet the epigenetic mechanism governing Snail to induce EMT is not well understood. Here, we report that in pancreatic ductal adenocarcinoma (PDAC), elevated levels of the ubiquitin E3 ligase Ring1B and Snail, along with elevated monoubiquitination of H2A at K119 (H2AK119Ub1), are highly correlated with poor survival. Mechanistic investigations identified Ring1B as a Snail-interacting protein and showed that the carboxyl zinc fingers of Snail recruit Ring1B and its paralog Ring1A to repress its target promoters. Simultaneous depletion of Ring1A and Ring1B in pancreatic cancer cells decreased Snail binding to the target chromatin, abolished H2AK119Ub1 modification, and thereby compromised Snail-mediated transcriptional repression and cell migration. We found that Ring1B and the SNAG-associated chromatin modifier EZH2 formed distinct protein complexes with Snail and that EZH2 was required for Snail-Ring1A/B recruitment to the target promoter. Collectively, our results unravel an epigenetic mechanism underlying transcriptional repression by Snail, suggest Ring1A/B as a candidate therapeutic target, and identify H2AK119Ub1 as a potential biomarker for PDAC diagnosis and prognosis.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Movement/physiology , Pancreatic Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , Transcription Factors/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/genetics , Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition , Female , HEK293 Cells , Humans , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding , Snail Family Transcription Factors , Tissue Array Analysis , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Pancreatic Neoplasms
14.
Biochem J ; 434(2): 275-85, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21133853

ABSTRACT

The tumour suppressor ARF (alternative reading frame) is one of the most important oncogenic stress sensors. ARF provides an 'oncogenic checkpoint' function through both p53-dependent and p53-independent mechanisms. In the present study, we demonstrate a novel p53-independent interaction between p14(ARF) and the adenovirus oncoprotein E1A. p14(ARF) inhibits E1A transcriptional function and promotes ubiquitination-dependent degradation of E1A. p14(ARF) overexpression relocalizes E1A into the nucleolus and inhibits E1A-induced cellular DNA replication independent of p53. Knockdown of endogenous p14(ARF) increases E1A transactivation. In addition, E1A can competitively inhibit ARF-Mdm2 (murine double minute 2) complex formation. These results identify a novel binding partner of p14(ARF) and reveal a mutually inhibitory interaction between p14(ARF) and E1A. We speculate that the ARF-E1A interaction may represent an additional host defence mechanism to limit viral replication. Alternatively, the interaction may allow adenovirus to sense the functional state of p53 in host cells, and fine-tune its own replication activity to prevent the triggering of a detrimental host response.


Subject(s)
Adenovirus E1A Proteins/antagonists & inhibitors , Adenovirus E1A Proteins/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Binding Sites , Cell Line , DNA Replication , Gene Silencing , HeLa Cells , Humans , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...