Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 19: 3513-3536, 2024.
Article in English | MEDLINE | ID: mdl-38623081

ABSTRACT

Purpose: Proliferative vitreoretinal diseases (PVDs) represent a heterogeneous group of pathologies characterized by the presence of retinal proliferative membranes, in whose development retinal pigment epithelium (RPE) is deeply involved. As the only effective treatment for PVDs at present is surgery, we aimed to investigate the potential therapeutic activity of Nutlin-3a, a small non-genotoxic inhibitor of the MDM2/p53 interaction, on ARPE-19 cell line and on human RPE primary cells, as in vitro models of RPE and, more importantly, to formulate and evaluate Nutlin-3a loaded liposomes designed for ophthalmic administration. Methods: Liposomes were produced using an innovative approach by a microfluidic device under selection of different conditions. Liposome size distribution was evaluated by photon correlation spectroscopy and centrifugal field flow fractionation, while the liposome structure was studied by transmission electron microscopy and Fourier-transform infrared spectroscopy. The Nutlin-3a entrapment capacity was evaluated by ultrafiltration and HPLC. Nutlin-3a biological effectiveness as a solution or loaded in liposomes was evaluated by viability, proliferation, apoptosis and migration assays and by morphological analysis. Results: The microfluidic formulative study enabled the selection of liposomes composed of phosphatidylcholine (PC) 5.4 or 8.2 mg/mL and 10% ethanol, characterized by roundish vesicular structures with 150-250 nm mean diameters. Particularly, liposomes based on the lower PC concentration were characterized by higher stability. Nutlin-3a was effectively encapsulated in liposomes and was able to induce a significant reduction of viability and migration in RPE cell models. Conclusion: Our results lay the basis for a possible use of liposomes for the ocular delivery of Nutlin-3a.


Subject(s)
Eye Diseases , Imidazoles , Liposomes , Piperazines , Humans , Liposomes/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Microfluidics , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Apoptosis
2.
J Pept Sci ; 30(1): e3535, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37580909

ABSTRACT

Intracellular protein-protein interactions provide a major therapeutic target for the development of peptide-based anticancer therapeutic agents. MDM2 is the 491-residue protein encoded by the MDM2 oncogene. Being a ubiquitin-protein ligase, MDM2 represses the transcription ability of the tumor suppressor p53 by proteasome-mediated degradation. Under typical cellular circumstances, a sustained p53 expression level is maintained by negative regulation of MDM2, whereas under stress conditions, this is alleviated to increase the p53 level. Modulation of MDM2-p53 interaction via fabrication of an MDM2-interacting peptide could be a useful strategy to inhibit subsequent proteasomal degradation of p53 and initiation of p53 signaling leading to the initiation of p53-mediated apoptosis of tumor cells. Here, in this research work, a novel anticancer peptide mPNC-NLS targeting the nucleus and the MDM2 protein (p53 negative regulator) was designed to promote the p53 protein activity for the prevention of cancer. It induces effective apoptosis in both A549 and U87 cells and remains non-cytotoxic to normal lung fibroblast cells (WI38). Further, immunocytochemistry and Western blot results confirm that the designed mPNC-NLS peptide induces the apoptotic death of lung cancer cells via activation of p53 and p21 proteins and remarkably stifled the in vitro growth of 3D multicellular spheroids composed of A549 cells.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Peptides/pharmacology , Peptides/metabolism
3.
Anticancer Agents Med Chem ; 23(16): 1829-1837, 2023.
Article in English | MEDLINE | ID: mdl-37259217

ABSTRACT

INTRODUCTION: Small cell lung cancer (SCLC) is featured by a high TP53 mutant rate. Our previous research found that arsenic trioxide (As2O3) could significantly inhibit the growth and metastasis of SCLC. Studies have shown that the degradation of mutant p53 mediated by murine double minute 2 (MDM2) can be induced by As2O3, which probably contributes to the inhibition of SCLC, but the detailed mechanism is still unclear. We aimed to testify that As2O3 can inhibit the growth of SCLC cells by degrading mutant p53 protein via binding to MDM2. METHODS: CCK-8 assay, cell cycle analysis, and western blot of apoptosis markers were used to evaluate the inhibitory effect of As2O3 on NCI-H446 cells (containing mutant p53) and NCI-H1299 cells (p53 null). The effects of As2O3 on p53 and its downstream proteins were identified by western blot using mut-p53-knockdown and overexpressed cell models. MDM2-knockdown cell models were constructed, and western blot, co-IP of mut-p53, and ubiquitin were carried out to explore the mediating effect of MDM2 in As2O3 induced mut-p53 degradation. RESULTS: As2O3 inhibited proliferation and induced cell cycle arrest and apoptosis of SCLC cells in a dose- and timedependent manner. After mut-p53 knockdown or overexpressed, the inhibitory effect of As2O3 was dampened or enhanced. Additionally, As2O3-induced mut-p53 ubiquitination was significantly weakened after MDM2 knockdown. CONCLUSION: As2O3 could inhibit SCLC cells by inhibiting proliferation and inducing cell cycle arrest and apoptosis. These inhibitory effects were achieved at least in part by upregulating MDM2, which, in turn, promotes ubiquitination and degradation of mut-p53.


Subject(s)
Antineoplastic Agents , Arsenicals , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Animals , Mice , Arsenic Trioxide/pharmacology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Oxides/pharmacology , Oxides/metabolism , Oxides/therapeutic use , Arsenicals/pharmacology , Arsenicals/metabolism , Arsenicals/therapeutic use , Cell Line, Tumor , Apoptosis , Lung Neoplasms/pathology , Cell Proliferation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Proto-Oncogene Proteins c-mdm2/therapeutic use
4.
Drug Dev Res ; 84(6): 1183-1203, 2023 09.
Article in English | MEDLINE | ID: mdl-37191966

ABSTRACT

The novel series of furan-bearing pyrazolo[3,4-b]pyridines were designed as cyclin-dependent kinase 2 (CDK2) inhibitors and as p53-murine double minute 2 (MDM2) inhibitors. The newly synthesized compounds were screened for their antiproliferative activity toward hepatocellular carcinoma (HepG2) and breast cancer (MCF7) cell lines. The most active compounds on both cell lines were additionally evaluated for their in vitro CDK2 inhibitory activity. Compounds 7b and 12f displayed enhanced activity (half-maximal inhibitory concentration [IC50 ] = 0.46 and 0.27 µM, respectively) in comparison to the standard roscovitine (IC50 = 1.41 ± 0.03 µM), in addition to, cell cycle arrest at S phase and G1/S transition phase in MCF7 cells treated with both compounds, respectively. Moreover, the most active spiro-oxindole derivative against MCF7 cell line, 16a, exhibited enhanced inhibitory activity against p53-MDM2 interaction in vitro (IC50 = 3.09 ± 0.12 µM) compared to nutlin, and increased the levels of both p53 and p21 by nearly fourfold in comparison to the negative control. Molecular docking studies demonstrated the plausible interaction patterns of the most potent derivatives 17b and 12f in the CDK2 binding pocket and the spiro-oxindole 16a with p53-MDM2 complex, respectively. Consequently, the new chemotypes 7b, 12f, and 16a can be presented as promising antitumor hits for further studies and optimization.


Subject(s)
Antineoplastic Agents , Tumor Suppressor Protein p53 , Humans , Animals , Mice , Cyclin-Dependent Kinase 2/metabolism , Molecular Docking Simulation , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Antineoplastic Agents/chemistry , Pyridines/pharmacology , Furans/pharmacology , Cell Proliferation , Cell Line, Tumor , Molecular Structure , Drug Screening Assays, Antitumor , Structure-Activity Relationship
5.
Diagn Microbiol Infect Dis ; 103(3): 115715, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35598409

ABSTRACT

Chlamydia (C.) abortus is an emerging zoonotic pathogen. Since data on its antimicrobial susceptibility are lacking, we aimed to determine minimal inhibitory (MIC) and minimal bactericidal concentrations (MBC) for azithromycin and doxycycline in HeLa-cells and primary human macrophages (M1). We also examined the MDM2-p53-inhibitor nutlin-3, an anti-infective imidazoline analog. Azithromycin and doxycycline demonstrated MICs and MBCs equal or below their peak serum concentrations (PSC) after standard dosing in both cell types. While doxycycline exhibited an MIC 64-fold and an MBC 4-fold below its PSC in HeLa-cells, the MIC of azithromycin was 4-fold below, the MBC equal to the PSC. However, azithromycin revealed lower MBCs in M1. The pharmacological advantage of azithromycin accumulation in phagocytes and their role as chlamydial reservoirs remain uncertain. However, our data suggest possible therapeutic advantages of doxycycline in epithelial cells and we provide first evidence for an anti-C. abortus effect of nutlin-3 in M1.


Subject(s)
Anti-Bacterial Agents , Chlamydia Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Azithromycin/pharmacology , Chlamydia , Chlamydia trachomatis , Doxycycline/pharmacology , Doxycycline/therapeutic use , Epithelial Cells , Humans , Imidazoles , Macrophages , Piperazines , Proto-Oncogene Proteins c-mdm2/pharmacology , Tumor Suppressor Protein p53/pharmacology , Tumor Suppressor Protein p53/therapeutic use
6.
Biofactors ; 48(4): 926-933, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35266593

ABSTRACT

Dysfunction of the blood-brain barrier (BBB) endothelium increases infiltration of lymphocytes and innate immune cells in the brain, leading to the development of neurological disorders. Heat shock protein 90 (Hsp90) inhibitors are anti-inflammatory agents and P53 inducers, which reduce the production of reactive oxygen species (ROS) in a diverse variety of human tissues. In this study, we investigate the effects of those compounds in LPS-induced brain endothelial inflammation, by utilizing human cerebral microvascular endothelial cells (hCMEC/D3). Our results suggest that Hsp90 inhibitors suppress inflammation by inhibiting the LPS-induced signal transducer and activator of transcription 3 (STAT3); and P38 activation. Moreover, those compounds reduce the P53 suppressors murine double minute 2 (MDM2) and murine double minute 4 (MDM4). Immunoglobulin heavy chain binding protein/glucose-regulated protein 78 (BiP/Grp78)-a key element of endothelial barrier integrity-was also increased by Hsp90 inhibition. Hence, we conclude that application of Hsp90 inhibitors in diseases related to BBB dysfunction may deliver a novel therapeutic possibility in the affected population.


Subject(s)
Endothelial Cells , Lipopolysaccharides , Animals , Brain/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Endothelium/metabolism , HSP90 Heat-Shock Proteins , Humans , Inflammation/metabolism , Lipopolysaccharides/metabolism , Mice , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Tumor Suppressor Protein p53/metabolism
8.
Mol Cancer Ther ; 20(6): 1161-1172, 2021 06.
Article in English | MEDLINE | ID: mdl-33850004

ABSTRACT

Neuroblastoma tumors frequently overexpress the anti-apoptotic protein B-cell lymphoma/leukemia 2 (BCL-2). We previously showed that treating BCL-2-dependent neuroblastoma cells with the BCL-2 inhibitor venetoclax results in apoptosis, but unfortunately partial therapy resistance is observed. The current study describes the identification of drugs capable of resensitizing venetoclax-resistant neuroblastoma cells to venetoclax. To examine these effects, venetoclax resistance was induced in BCL-2-dependent neuroblastoma cell lines KCNR and SJNB12 by continuous exposure to high venetoclax concentrations. Non-resistant and venetoclax-resistant neuroblastoma cell lines were exposed to a 209-compound library in the absence and presence of venetoclax to identify compounds that were more effective in the venetoclax-resistant cell lines under venetoclax pressure. Top hits were further validated in combination with venetoclax using BCL-2-dependent neuroblastoma model systems. Overall, high-throughput drug screening identified the MDM2 inhibitor idasanutlin as a promising resensitizing agent for venetoclax-resistant neuroblastoma cell lines. Idasanutlin treatment induced BAX-mediated apoptosis in venetoclax-resistant neuroblastoma cells in the presence of venetoclax, whereas it caused p21-mediated growth arrest in control cells. In vivo combination treatment showed tumor regression and superior efficacy over single-agent therapies in a BCL-2-dependent neuroblastoma cell line xenograft and a patient-derived xenograft. However, xenografts less dependent on BCL-2 were not sensitive to venetoclax-idasanutlin combination therapy. This study demonstrates that idasanutlin can overcome resistance to the BCL-2 inhibitor venetoclax in preclinical neuroblastoma model systems, which supports clinical development of a treatment strategy combining the two therapies.


Subject(s)
High-Throughput Screening Assays/methods , Neuroblastoma/drug therapy , Proto-Oncogene Proteins c-mdm2/therapeutic use , Pyrrolidines/therapeutic use , para-Aminobenzoates/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Proto-Oncogene Proteins c-mdm2/pharmacology , Pyrrolidines/pharmacology , para-Aminobenzoates/pharmacology
9.
Cells ; 9(6)2020 06 16.
Article in English | MEDLINE | ID: mdl-32560270

ABSTRACT

More than 50% of human cancers harbor TP53 mutations and increased expression of Mouse double minute 2 homolog(MDM2), which contribute to cancer progression and drug resistance. Renal cell carcinoma (RCC) has an unusually high incidence of wild-type p53, with a mutation rate of less than 4%. MDM2 is master regulator of apoptosis in cancer cells, which is triggered through proteasomal degradation of wild-type p53. Recently, we found that p53 protein levels in RCC are regulated by autophagic degradation. Transglutaminase 2 (TGase 2) was responsible for p53 degradation through this pathway. Knocking down TGase 2 increased p53-mediated apoptosis in RCC. Therefore, we asked whether depleting p53 from RCC cells occurs via MDM2-mediated proteasomal degradation or via TGase 2-mediated autophagic degradation. In vitro gene knockdown experiments revealed that stability of p53 in RCC was inversely related to levels of both MDM2 and TGase 2 protein. Therefore, we examined the therapeutic efficacy of inhibitors of TGase 2 and MDM2 in an in vivo model of RCC. The results showed that inhibiting TGase 2 but not MDM2 had efficient anticancer effects.


Subject(s)
Carcinoma, Renal Cell/drug therapy , GTP-Binding Proteins/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Piperazines/pharmacology , Transglutaminases/antagonists & inhibitors , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Humans , Protein Glutamine gamma Glutamyltransferase 2 , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology
11.
Biochem Pharmacol ; 170: 113675, 2019 12.
Article in English | MEDLINE | ID: mdl-31634457

ABSTRACT

Cellular stimuli that increase diacylglycerol levels activate several protein kinase C (PKC) isoforms; however, prolonged stimulation depletes cells of PKCs. Ubiquitination is a critical cellular event that mediates the degradation of numerous proteins, including PKCs, but little is known of the molecular mechanisms involved in PKC ubiquitination. PKCßII is the most widely expressed PKC isoform and regulates a variety of cellular functions. Here, we show that in response to stimulation of the Gq-coupled angiotensin II type 1 receptor or treatment with phorbol ester, Mdm2, E3 ubiquitin ligase, interacted with PKCßII isotype in the nucleus, resulting in ubiquitination of PKCßII at the C-terminal K668 and K672 residues and its subsequent downregulation. Ubiquitinated PKCßII mediated the clathrin-mediated endocytosis of G protein-coupled receptors like the D2 and D3 dopamine receptors; in contrast, non-ubiquitinated PKCßII mediated an as yet uncharacterized clathrin- and caveolar-independent endocytic pathway. In conclusion, we characterized the molecular mechanisms involved in the activity-dependent ubiquitination of PKCßII that determine its life span and endocytic roles. Considering that PKCßII plays an important role in the development of various diseases, including diabetic vasculitis, the results obtained in this study will contribute to better understanding the pathogenesis of PKCßII-related diseases.


Subject(s)
Cell Nucleus/metabolism , Clathrin/metabolism , Endocytosis/physiology , Protein Kinase C beta/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Ubiquitination/physiology , Amino Acid Sequence , Cell Nucleus/drug effects , Endocytosis/drug effects , HEK293 Cells , Humans , Ubiquitination/drug effects
12.
Prostate ; 68(6): 599-609, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18196567

ABSTRACT

BACKGROUND: Antisense MDM2 oligonucleotide (AS-MDM2) sensitizes androgen sensitive LNCaP cells to androgen deprivation (AD) in vitro and in vivo. In this study, we investigated the effects of AS-MDM2 combined with AD on androgen resistant LNCaP (LNCaP-Res) and moderately androgen resistant bcl-2 overexpressing LNCaP (LNCaP-BST) cells. METHODS: The LNCaP-Res cell line was generated by culturing LNCaP cells in medium containing charcoal-stripped serum for more than 1 year. Apoptosis was quantified in vitro by Annexin V staining and caspase 3 + 7 activity. For the in vivo studies, orthotopic tumor growth was monitored by magnetic resonance imaging (MRI). AS-MDM2 and the mismatch control were given by i.p. injection at doses of 25 mg/kg per day, 5 days/week for 15 days. RESULTS: LNCaP-Res cells expressed high levels of androgen receptor (AR) and bcl-2, and displayed no growth inhibition to AD. AS-MDM2 caused significant reductions in MDM2 and AR expression, and increases in p53 and p21 expression in both cell lines. AS-MDM2 + AD resulted in the highest levels of apoptosis in vitro and tumor growth inhibition in vivo in both cell lines; although, these effects were less pronounced in LNCaP-BST cells. CONCLUSIONS: AS-MDM2 + AD enhanced apoptotic cell death in vitro and tumor growth inhibition in vivo in androgen resistant cell lines. The action of AS-MDM2 + AD was influenced somewhat by bcl-2 expression as an isolated change (LNCaP-BST cells), but not when accompanied by other molecular changes associated with androgen insensitivity (LNCaP-Res cells). MDM2 knockdown has promise for the treatment of men with early hormone refractory disease.


Subject(s)
Androgen Antagonists/pharmacology , Apoptosis/drug effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Orchiectomy , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
13.
J Biomol Screen ; 12(8): 1050-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17989425

ABSTRACT

Tumor suppressor p53 is typically maintained at low levels in normal cells. In response to cellular stresses, such as DNA damage, p53 is stabilized and can stimulate responses leading to cell cycle arrest or apoptosis. Corresponding to its central role in preventing propagation of damaged cells, mutation or deletion of p53 is found in nearly 50% of all human tumors. Mdm2 (mouse-d-minute 2) and its human ortholog (hmdm2 or hdm2) catalyze the ubiquitination of p53, targeting it for degradation via the proteosome. Thus, the activity of mdm2 is inversely correlated with p53 levels. Based on this, inhibition of human mdm2 activity by a small-molecule therapeutic will lead to net stabilization of p53 and be the basis for development of a novel cancer therapeutic. Previous high-throughput screening assays of mdm2 measured the autoubiquitination activity of mdm2, which occurs in the absence of an acceptor substrate such as p53. The major drawback to this approach is that inhibitors of mdm2 autoubiquitination may lead to a net stabilization of mdm2 and thus have the opposite effect of inhibitors that interfere with p53 ubiquitination. The authors describe the development, validation, and execution of a high-throughput screening measuring the ubiquitination of p53 by mdm2, with p53 labeled with europium and the other substrate (Ub-UbcH5b) labeled with a Cy5 on the ubiquitin. After confirming that known inhibitors are detected with this assay, it was successfully automated and used to query >600,000 compounds from the GlaxoSmithKline collection for mdm2 inhibitors.


Subject(s)
Biological Assay/methods , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Catalysis/drug effects , Europium/pharmacology , Fluorescence Resonance Energy Transfer , Humans , Inhibitory Concentration 50 , Proto-Oncogene Proteins c-mdm2/pharmacology , Reproducibility of Results , Time Factors , Titrimetry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/drug effects
14.
Biotechnol Bioeng ; 97(3): 601-14, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17149774

ABSTRACT

The ability to regulate apoptosis in mammalian cell cultures represents one approach to developing more economical and efficient processes. Genetic modification of cells using anti-apoptotic genes is one method that may be used to improve cellular performance. This study investigates a method to inhibit upstream apoptosis pathways through the overexpression of MDM2, an E3 ubiquitin ligase for p53. Both 293 and CHO cells expressing MDM2 were examined under both batch and spent media conditions. For batch cultures, MDM2 overexpression increased viable cell densities and viabilities over control cells with the largest enhancements observed in CHO cells. When CHO cells were passaged without medium exchange, cells expressing MDM2 reached a viable cell density that was nearly double the control and survived for an extra day in culture. When exposed to spent media initially, both 293-MDM2 and CHO-MDM2 cells continued to grow for 2 days while the control cells stopped growing after the first day. DNA analysis using flow cytometry confirmed that while CHO controls were found to be undergoing DNA fragmentation, CHO-MDM2 cells exhibit DNA degradation at a much slower rate. When compared to Bcl-2-expressing cells, MDM2 expression showed greater protection against apoptosis in passaged culture, spent medium, and following transient p53 overexpression. However, expression of the RING sequence of MDM2 responsible for E3 ligase activity without the other components of the protein was found to be toxic to 293 cells in culture. These results suggest that the overexpression of heterologous MDM2 represents a promising method to delay apoptosis in mammalian cell cultures.


Subject(s)
Apoptosis/drug effects , Proto-Oncogene Proteins c-mdm2/pharmacology , Animals , Apoptosis/physiology , CHO Cells , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cricetinae , Cricetulus , Gene Expression Regulation, Neoplastic/drug effects , Mammals , Proto-Oncogene Proteins c-mdm2/chemistry
15.
Mol Cell ; 20(5): 699-708, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16337594

ABSTRACT

Inactivation of retinoblastoma protein (Rb) plays a critical role in the development of human malignancies. It has been shown that Rb is degraded through a proteasome-dependent pathway, yet the mechanism is largely unclear. MDM2 is frequently found amplified and overexpressed in a variety of human tumors. In this study, we find that MDM2 promotes Rb degradation in a proteasome-dependent and ubiquitin-independent manner. We show that Rb, MDM2, and the C8 subunit of the 20S proteasome interact in vitro and in vivo and that MDM2 promotes Rb-C8 interaction. Expression of wild-type MDM2, but not the mutant MDM2 defective either in Rb interaction or in RING finger domain, promotes cell cycle S phase entry independent of p53. Furthermore, MDM2 ablation results in Rb accumulation and inhibition of DNA synthesis. Taken together, these findings demonstrate that MDM2 is a critical negative regulator for Rb and suggest that MDM2 overexpression contributes to cancer development by destabilizing Rb.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Retinoblastoma Protein/metabolism , Ubiquitin/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/metabolism , DNA/biosynthesis , DNA/drug effects , Gamma Rays , Humans , In Vitro Techniques , Proto-Oncogene Proteins c-mdm2/pharmacology , Proto-Oncogene Proteins c-mdm2/radiation effects , Retinoblastoma Protein/drug effects , S Phase/physiology , S Phase/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...