Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Med Chem ; 65(7): 5317-5333, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35352560

ABSTRACT

Polycomb Repressive Complex 2 (PRC2) plays an important role in transcriptional regulation during animal development and in cell differentiation, and alteration of PRC2 activity has been associated with cancer. On a molecular level, PRC2 catalyzes methylation of histone H3 lysine 27 (H3K27), resulting in mono-, di-, or trimethylated forms of H3K27, of which the trimethylated form H3K27me3 leads to transcriptional repression of polycomb target genes. Previously, we have shown that binding of the low-molecular-weight compound EED226 to the H3K27me3 binding pocket of the regulatory subunit EED can effectively inhibit PRC2 activity in cells and reduce tumor growth in mouse xenograft models. Here, we report the stepwise optimization of the tool compound EED226 toward the potent and selective EED inhibitor MAK683 (compound 22) and its subsequent preclinical characterization. Based on a balanced PK/PD profile, efficacy, and mitigated risk of forming reactive metabolites, MAK683 has been selected for clinical development.


Subject(s)
Histones , Neoplasms , Animals , Enzyme Inhibitors , Histones/metabolism , Humans , Methylation , Mice , Neoplasms/drug therapy , Polycomb Repressive Complex 2
2.
Am J Transl Res ; 14(1): 197-210, 2022.
Article in English | MEDLINE | ID: mdl-35173838

ABSTRACT

BACKGROUND: Serine peptidase inhibitor Kazal type 2 (SPINK2) has been reported to be involved in certain cancers. We conducted an in-depth investigation on the role and mechanism of SPINK2 in acute myeloid leukemia (AML). METHODS: The relationship between SPINK2 expression and AML clinicopathologic characteristics was determined using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. Concomitantly, we used Kaplan-Meier survival analysis, as well as univariate and multivariate regression analyses to evaluate SPINK2 as a prognostic marker of AML. Additionally, we annotated the enrichment and function of SPINK2 using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Sets Enrichment Analysis (GSEA). The CIBERSORT algorithm was used to analyze the relationship between SPINK2 expression and immune infiltration. RESULTS: SPINK2 expression was significantly higher in AML patients compared to healthy individuals (P<0.001). The area under receiver operating characteristic curve in the GSE9476 dataset was 0.660, whereas that in the Genotype-Tissue Expression (GTEx) and TCGA datasets was 0.935. In addition, GSEA also showed that several pathways were enriched in the group with high SPINK2 expression, such as PI3K-AKT signaling, PD-L1 expression, and checkpoint pathways. Analysis of immune infiltration showed that SPINK2 expression was correlated with certain immune infiltrating cells. Cox multivariate analysis revealed that the level of SPINK2 was an independent risk factor for the progression of AML (P<0.001). Moreover, age, M1, M5, M6, and CytoRisk-Poor also affected the progression of AML (P<0.05). The C-index of the nomogram in our internal validation was 0.702. CONCLUSION: The high expression of SPINK2 in AML suggests that SPINK2 may play an important role in the immune microenvironment and thus could be a biomarker for diagnosis and prognosis of AML.

3.
Mol Med Rep ; 19(1): 508-514, 2019 01.
Article in English | MEDLINE | ID: mdl-30483794

ABSTRACT

MicroRNAs (miRNAs) have been demonstrated to regulate the progression of numerous types of cancer, including acute myeloid leukemia (AML). Previous studies demonstrated that miR­1271­5p functions as a tumor suppressor; however, the roles of miR­1271­5p in AML remain unknown. In the present study, miR­1271­5p was significantly downregulated in AML tissues compared with normal tissues by reverse transcription­quantitative polymerase chain reaction analysis. Furthermore, the expression levels of miR­1271­5p in patients with AML may function as a prognostic marker. In addition, overexpression of miR­1271­5p significantly suppressed the proliferation and induced apoptosis of AML cells by Cell Counting kit­8 and fluorescence activated cell sorter assays; miR­1271­5p downregulation exhibited opposing effects. Additionally, transcription factor ZIC2 may be a direct target of miR­1271­5p in AML cells, which was demonstrated by a luciferase reporter assay and RNA pulldown assay. Overexpression of miR­1271­5p significantly reduced the mRNA and protein expression levels of ZIC2 in AML193 and OCI­AML2 cells by reverse transcription­quantitative polymerase chain reaction analysis and western blotting. Furthermore, an inverse correlation between miR­1271­5p and ZIC2 expression in AML samples was observed. In summary, ZIC2 was upregulated in AML tissues, and restoration of ZIC2 expression was able to promote the proliferation and reduce the apoptosis of AML cells transfected with miR­1271­5p mimics. The results of the present study demonstrated that miR­1271­5p inhibited the progression of AML by targeting ZIC2.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MicroRNAs/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adolescent , Adult , Cell Line , Cell Line, Tumor , Child , Child, Preschool , Down-Regulation/genetics , Female , Gene Expression Regulation, Leukemic/genetics , HL-60 Cells , Humans , Male , Middle Aged , RNA, Messenger/genetics , Young Adult
4.
J Med Chem ; 60(6): 2215-2226, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28092155

ABSTRACT

Overexpression and somatic heterozygous mutations of EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), are associated with several tumor types. EZH2 inhibitor, EPZ-6438 (tazemetostat), demonstrated clinical efficacy in patients with acceptable safety profile as monotherapy. EED, another subunit of PRC2 complex, is essential for its histone methyltransferase activity through direct binding to trimethylated lysine 27 on histone 3 (H3K27Me3). Herein we disclose the discovery of a first-in-class potent, selective, and orally bioavailable EED inhibitor compound 43 (EED226). Guided by X-ray crystallography, compound 43 was discovered by fragmentation and regrowth of compound 7, a PRC2 HTS hit that directly binds EED. The ensuing scaffold hopping followed by multiparameter optimization led to the discovery of 43. Compound 43 induces robust and sustained tumor regression in EZH2MUT preclinical DLBCL model. For the first time we demonstrate that specific and direct inhibition of EED can be effective as an anticancer strategy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Sulfones/chemistry , Sulfones/pharmacology , Triazoles/chemistry , Triazoles/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Crystallography, X-Ray , Dogs , Female , Haplorhini , Histones/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lysine/metabolism , Male , Methylation/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Rats , Sulfones/pharmacokinetics , Sulfones/therapeutic use , Triazoles/pharmacokinetics , Triazoles/therapeutic use
5.
Nat Chem Biol ; 13(4): 381-388, 2017 04.
Article in English | MEDLINE | ID: mdl-28135235

ABSTRACT

Polycomb repressive complex 2 (PRC2) consists of three core subunits, EZH2, EED and SUZ12, and plays pivotal roles in transcriptional regulation. The catalytic subunit EZH2 methylates histone H3 lysine 27 (H3K27), and its activity is further enhanced by the binding of EED to trimethylated H3K27 (H3K27me3). Small-molecule inhibitors that compete with the cofactor S-adenosylmethionine (SAM) have been reported. Here we report the discovery of EED226, a potent and selective PRC2 inhibitor that directly binds to the H3K27me3 binding pocket of EED. EED226 induces a conformational change upon binding EED, leading to loss of PRC2 activity. EED226 shows similar activity to SAM-competitive inhibitors in blocking H3K27 methylation of PRC2 target genes and inducing regression of human lymphoma xenograft tumors. Interestingly, EED226 also effectively inhibits PRC2 containing a mutant EZH2 protein resistant to SAM-competitive inhibitors. Together, we show that EED226 inhibits PRC2 activity via an allosteric mechanism and offers an opportunity for treatment of PRC2-dependent cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Histones/metabolism , Lysine/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Sulfones/chemistry , Sulfones/pharmacology , Triazoles/chemistry , Triazoles/pharmacology , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/chemistry , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Histones/chemistry , Humans , Lysine/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Structure-Activity Relationship , Sulfones/metabolism , Triazoles/metabolism , Tumor Cells, Cultured
6.
Anal Biochem ; 522: 37-45, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28111304

ABSTRACT

The EED (embryonic ectoderm development) subunit of the Polycomb repressive complex 2 (PRC2) plays an important role in the feed forward regulation of the PRC2 enzymatic activity. We recently identified a new class of allosteric PRC2 inhibitors that bind to the H3K27me3 pocket of EED. Multiple assays were developed and used to identify and characterize this type of PRC2 inhibitors. One of them is a genetically encoded EED biosensor based on the EED[G255D] mutant and the split firefly luciferase. This EED biosensor can detect the compound binding in the transfected cells and in the in vitro biochemical assays. Compared to other commonly used cellular assays, the EED biosensor assay has the advantage of shorter compound incubation with cells. The in vitro EED biosensor is much more sensitive than other label-free biophysical assays (e.g. DSF, ITC). Based on the crystal structure, the DSF data as well as the biosensor assay data, it's most likely that compound-induced increase in the luciferase activity of the EED[G255D] biosensor results from the decreased non-productive interactions between the EED subdomain and other subdomains within the biosensor construct. This new insight of the mechanism might help to broaden the use of the split luciferase based biosensors.


Subject(s)
Biological Assay/methods , Luciferases, Firefly/metabolism , Luminescent Measurements/methods , Mutation, Missense , Polycomb Repressive Complex 2/metabolism , Amino Acid Substitution , Cell Line , Humans , Luciferases, Firefly/genetics , Polycomb Repressive Complex 2/genetics , Protein Binding , Protein Domains
7.
J Med Chem ; 60(1): 415-427, 2017 01 12.
Article in English | MEDLINE | ID: mdl-27992714

ABSTRACT

PRC2 is a multisubunit methyltransferase involved in epigenetic regulation of early embryonic development and cell growth. The catalytic subunit EZH2 methylates primarily lysine 27 of histone H3, leading to chromatin compaction and repression of tumor suppressor genes. Inhibiting this activity by small molecules targeting EZH2 was shown to result in antitumor efficacy. Here, we describe the optimization of a chemical series representing a new class of PRC2 inhibitors which acts allosterically via the trimethyllysine pocket of the noncatalytic EED subunit. Deconstruction of a larger and complex screening hit to a simple fragment-sized molecule followed by structure-guided regrowth and careful property modulation were employed to yield compounds which achieve submicromolar inhibition in functional assays and cellular activity. The resulting molecules can serve as a simplified entry point for lead optimization and can be utilized to study this new mechanism of PRC2 inhibition and the associated biology in detail.


Subject(s)
Enzyme Inhibitors/chemistry , Epigenesis, Genetic , Methyltransferases/antagonists & inhibitors , Polycomb Repressive Complex 2/chemistry , Allosteric Regulation , Caco-2 Cells , Chromatography, Liquid , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Molecular Structure , Proton Magnetic Resonance Spectroscopy , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 109(52): 21360-5, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236167

ABSTRACT

Ezh2 (Enhancer of zeste homolog 2) protein is the enzymatic component of the Polycomb repressive complex 2 (PRC2), which represses gene expression by methylating lysine 27 of histone H3 (H3K27) and regulates cell proliferation and differentiation during embryonic development. Recently, hot-spot mutations of Ezh2 were identified in diffused large B-cell lymphomas and follicular lymphomas. To investigate if tumor growth is dependent on the enzymatic activity of Ezh2, we developed a potent and selective small molecule inhibitor, EI1, which inhibits the enzymatic activity of Ezh2 through direct binding to the enzyme and competing with the methyl group donor S-Adenosyl methionine. EI1-treated cells exhibit genome-wide loss of H3K27 methylation and activation of PRC2 target genes. Furthermore, inhibition of Ezh2 by EI1 in diffused large B-cell lymphomas cells carrying the Y641 mutations results in decreased proliferation, cell cycle arrest, and apoptosis. These results provide strong validation of Ezh2 as a potential therapeutic target for the treatment of cancer.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Embryo, Mammalian/cytology , Enhancer of Zeste Homolog 2 Protein , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Methylation/drug effects , Mice , Mutation/genetics , Phenotype , Polycomb Repressive Complex 2/metabolism , Small Molecule Libraries/chemistry , Tumor Stem Cell Assay , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 29(2): 156-8, 2009 Feb.
Article in Chinese | MEDLINE | ID: mdl-19382479

ABSTRACT

OBJECTIVE: To investigate the clinical curative effect of Lianhuang Decoction (LHD) in treating fetomaternal ABO blood group incompatibility (FM-ABOI). METHODS: Sixty pregnancy women diagnosed as FM-ABOI were randomly assigned to two groups, Group A treated with LHD and Group B treated with Western medicine. The therapeutic efficacy in the two groups was observed. RESULTS: The antibody titer decreased after treatment in both groups to different extent; the effective rate was 90.0% in Group A and 56.7% in Group B. The bilirubin level in cord blood of Group A was lower than that in Group B significantly (P<0.01). No statistically significant difference was found between the two groups new-bom in terms of 5-minute Apgar score, body weight and hemoglobin content in cord blood (P>0.05). CONCLUSION: LHD has good clinical curative effect in treating FM-ABOI, could decrease the serum antibody titer, and prevent the occurrence of postpartum hemolytic disease in newborns.


Subject(s)
ABO Blood-Group System , Blood Group Incompatibility/drug therapy , Drugs, Chinese Herbal/therapeutic use , Phytotherapy , Adult , Female , Humans , Infant, Newborn , Jaundice, Neonatal/prevention & control , Pregnancy , Young Adult
10.
Nucleic Acids Res ; 36(1): e8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18086703

ABSTRACT

Knowledge of all binding sites for transcriptional activators and repressors is essential for computationally aided identification of transcriptional networks. The techniques developed for defining the binding sites of transcription factors tend to be cumbersome and not adaptable to high throughput. We refined a versatile yeast strategy to rapidly and efficiently identify genomic targets of DNA-binding proteins. Yeast expressing a transcription factor is mated to yeast containing a library of genomic fragments cloned upstream of the reporter gene URA3. DNA fragments with target-binding sites are identified by growth of yeast clones in media lacking uracil. The experimental approach was validated with the tumor suppressor protein p53 and the forkhead protein FoxI1 using genomic libraries for zebrafish and mouse generated by shotgun cloning of short genomic fragments. Computational analysis of the genomic fragments recapitulated the published consensus-binding site for each protein. Identified fragments were mapped to identify the genomic context of each binding site. Our yeast screening strategy, combined with bioinformatics approaches, will allow both detailed and high-throughput characterization of transcription factors, scalable to the analysis of all putative DNA-binding proteins.


Subject(s)
DNA-Binding Proteins/metabolism , Genomics/methods , Regulatory Elements, Transcriptional , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Computational Biology , Consensus Sequence , DNA/chemistry , Forkhead Transcription Factors/metabolism , Genomic Library , Mice , Plasmids/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 104(47): 18613-8, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-18003932

ABSTRACT

The evolutionary forces that establish and hone target gene networks of transcription factors are largely unknown. Transposition of retroelements may play a role, but its global importance, beyond a few well described examples for isolated genes, is not clear. We report that LTR class I endogenous retrovirus (ERV) retroelements impact considerably the transcriptional network of human tumor suppressor protein p53. A total of 1,509 of approximately 319,000 human ERV LTR regions have a near-perfect p53 DNA binding site. The LTR10 and MER61 families are particularly enriched for copies with a p53 site. These ERV families are primate-specific and transposed actively near the time when the New World and Old World monkey lineages split. Other mammalian species lack these p53 response elements. Analysis of published genomewide ChIP data for p53 indicates that more than one-third of identified p53 binding sites are accounted for by ERV copies with a p53 site. ChIP and expression studies for individual genes indicate that human ERV p53 sites are likely part of the p53 transcriptional program and direct regulation of p53 target genes. These results demonstrate how retroelements can significantly shape the regulatory network of a transcription factor in a species-specific manner.


Subject(s)
Endogenous Retroviruses/physiology , Gene Regulatory Networks/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Binding Sites , Cell Line, Tumor , Endogenous Retroviruses/classification , Evolution, Molecular , Gene Dosage/genetics , Gene Expression Regulation, Viral/genetics , Genome, Viral/genetics , Humans , Protein Binding , Regulatory Elements, Transcriptional/genetics
12.
Int J Oncol ; 31(5): 1205-11, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17912449

ABSTRACT

The tumor suppressor protein p53 is a transcription factor that is frequently mutated in human cancers. In response to DNA damage, unmutated or wild-type (wt) p53 protein is stabilized and activated by post-transcriptional modifications that enable it to induce either apoptosis or cell cycle arrest. Using a yeast p53-dissociator assay, we identified MAGED2 as a potential negative regulator of wt p53 activity. Subsequently, using co-immunoprecipitation and reporter gene assays in human cultured cells that are often adopted for functional analysis of p53 we demonstrated that MAGED2 interacted physically with p53 and modified its activity. Finally, we were able to illustrate expression of both p53 and MAGED2 within the same subcellular compartment, i.e. either nucleus or cytoplasm, in 2,682 human cancer tissue specimens using a common cancer tissue microarray and antibodies against MAGED2 and p53. The present results implicate MAGED2, a novel protein, as a p53-dissociator.


Subject(s)
Antigens, Neoplasm/physiology , Tumor Suppressor Protein p53/physiology , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Antigens, Neoplasm/analysis , Antigens, Neoplasm/chemistry , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/analysis , Humans , Molecular Sequence Data , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/physiology , Nuclear Proteins/analysis , Nuclear Proteins/physiology , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/chemistry
13.
Hepatogastroenterology ; 54(77): 1591-5, 2007.
Article in English | MEDLINE | ID: mdl-17708308

ABSTRACT

BACKGROUND/AIMS: The aim of this study was to evaluate the prognostic significance of some serum tumor marker level, extracellular matrix (ECM), matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF) in patients with gastric cancer. METHODOLOGY: The serum tumor markers included CEA, CA50 and CA19-9, ECM included laminin (LN), hyaluronic acid (HA), and collagen type III and IV were measured in 40 patients with gastric carcinoma and 20 matched healthy controls by radioimmunoassay. MMP-9, VEGF and MVD were measured with immunohistochemical methods and the computer image analyzer. Microvascular density (MVD) in tissues of patients with gastric carcinoma was detected. RESULTS: The levels of CEA, CA50, CA19-9, HA, LN and collagen type IV in the patients with metastasis were significantly higher than those in the patients without metastasis (p < 0.05). The expression of MMP-9 and collage type IV in the patients with metastasis and poorly differentiated carcinomas were significantly higher than those in the patients without metastasis whose tumors were well/moderately differentiated (p < 0.05). CONCLUSIONS: CEA, CA50, CA19-9, HA, LN and collagen type IV levels can be used as a signal of metastasis and disease progression in patients with gastric carcinoma. When a gastric carcinoma expresses a high level of MMP-9 and VEGF with high MVD, the power of infiltration and metastasis of the gastric carcinoma is enhanced.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma/blood , Carcinoma/pathology , Extracellular Matrix/pathology , Matrix Metalloproteinase 9/blood , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor A/blood , Adult , Aged , Female , Humans , Male , Middle Aged
14.
Bioinformatics ; 23(13): i104-14, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17646286

ABSTRACT

MOTIVATION: Many biomedical projects would benefit from reducing the time and expense of in vitro experimentation by using computer models for in silico predictions. These models may help determine which expensive biological data are most useful to acquire next. Active Learning techniques for choosing the most informative data enable biologists and computer scientists to optimize experimental data choices for rapid discovery of biological function. To explore design choices that affect this desirable behavior, five novel and five existing Active Learning techniques, together with three control methods, were tested on 57 previously unknown p53 cancer rescue mutants for their ability to build classifiers that predict protein function. The best of these techniques, Maximum Curiosity, improved the baseline accuracy of 56-77%. This article shows that Active Learning is a useful tool for biomedical research, and provides a case study of interest to others facing similar discovery challenges.


Subject(s)
Algorithms , Artificial Intelligence , Biomarkers, Tumor/metabolism , DNA Mutational Analysis/methods , Neoplasm Proteins/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Humans , Mutation , Neoplasms/metabolism
15.
Carcinogenesis ; 28(2): 289-98, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16861262

ABSTRACT

Over 1000 different mutants of the tumor suppressor protein p53 with one amino acid change in the core domain have been reported in human cancers. In mouse knock-in models, two frequent mutants displayed loss of wild-type (wt) p53 function, inhibition of wt p53 and wt p53-independent gain of function. The remaining mutants have been systematically characterized for loss of wt p53 function, but not other phenotypes. We report the concomitant assessment of loss of function and interference with wt p53 using URA3-based p53 yeast and confirmatory mammalian assays. We studied 76 mutants representing 54% of over 15 000 reported missense core domain mutations. The majority showed the expected complete loss of wt p53 function and dominant p53 inhibition. A few infrequent p53 mutants had wt p53-like activity. Remarkably, one-third showed no interference with wt p53 despite loss of wt p53 function at 37 degrees C. Half of this group consisted of temperature-sensitive p53 mutants, but the other half was surprisingly made up of mutants with complete loss of wt p53 function. Our findings illustrate the diverse behavior of p53 mutants and mechanisms of malignant transformation by p53 mutants. The identification of full-length p53 mutants without dominant inhibition of wt p53 highlights the importance of determining the status of the wt p53 allele in human cancers, in particular in the context of clinical studies. In the case of p53 mutants with no or weak dominant p53 inhibition, presence of the wt allele may indicate a good prognosis cancer, whereas loss of heterozygosity may spell an aggressive, therapy-resistant cancer.


Subject(s)
Genes, p53 , Loss of Heterozygosity , Mutation , Neoplasms/genetics , Cell Line, Tumor , Genes, Reporter , Humans , Neoplasms/pathology , Saccharomyces cerevisiae/genetics
16.
Article in English | MEDLINE | ID: mdl-17048398

ABSTRACT

Many biomedical problems relate to mutant functional properties across a sequence space of interest, e.g., flu, cancer, and HIV. Detailed knowledge of mutant properties and function improves medical treatment and prevention. A functional census of p53 cancer rescue mutants would aid the search for cancer treatments from p53 mutant rescue. We devised a general methodology for conducting a functional census of a mutation sequence space by choosing informative mutants early. The methodology was tested in a double-blind predictive test on the functional rescue property of 71 novel putative p53 cancer rescue mutants iteratively predicted in sets of three (24 iterations). The first double-blind 15-point moving accuracy was 47 percent and the last was 86 percent; r = 0.01 before an epiphanic 16th iteration and r = 0.92 afterward. Useful mutants were chosen early (overall r = 0.80). Code and data are freely available (http://www.igb.uci.edu/research/research.html, corresponding authors: R.H.L. for computation and R.K.B. for biology).


Subject(s)
Computational Biology/methods , Models, Statistical , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Artificial Intelligence , Binding Sites/genetics , Humans , Internet , Models, Molecular , Mutation/physiology , Mutation, Missense/genetics , Mutation, Missense/physiology , Neoplasms/drug therapy , Neoplasms/genetics , Protein Folding , Protein Structure, Tertiary , ROC Curve , Suppression, Genetic/genetics , Suppression, Genetic/physiology , Surface Properties , Tumor Suppressor Protein p53/chemistry
17.
Genetics ; 173(3): 1377-87, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16648584

ABSTRACT

Two dominant temperature-sensitive (DTS) lethal mutants of Drosophila melanogaster are Pros26(1) and Prosbeta2(1), previously known as DTS5 and DTS7. Heterozygotes for either mutant die as pupae when raised at 29 degrees , but are normally viable and fertile at 25 degrees . Previous studies have identified these as missense mutations in the genes encoding the beta6 and beta2 subunits of the 20S proteasome, respectively. In an effort to isolate additional proteasome-related mutants a screen for dominant suppressors of Pros26(1) was carried out, resulting in the identification of Pros25(SuDTS) [originally called Su(DTS)], a missense mutation in the gene encoding the 20S proteasome alpha2 subunit. Pros25(SuDTS) acts in a dominant manner to rescue both Pros26(1) and Prosbeta2(1) from their DTS lethal phenotypes. Using an in vivo protein degradation assay it was shown that this suppression occurs by counteracting the dominant-negative effect of the DTS mutant on proteasome activity. Pros25(SuDTS) is a recessive polyphasic lethal at ambient temperatures. The effects of these mutants on larval neuroblast mitosis were also examined. While Prosbeta2(1) shows a modest increase in the number of defective mitotic figures, there were no defects seen with the other two mutants, other than slightly reduced mitotic indexes.


Subject(s)
Drosophila melanogaster/genetics , Endopeptidases/genetics , Genes, Lethal , Genes, Suppressor , Proteasome Endopeptidase Complex/genetics , Suppression, Genetic , Alleles , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Female , Male , Models, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Temperature
18.
Proc Natl Acad Sci U S A ; 101(14): 4930-5, 2004 Apr 06.
Article in English | MEDLINE | ID: mdl-15037740

ABSTRACT

The transcription factor and tumor suppressor protein p53 is frequently inactivated in human cancers. In many cases, p53 gene mutations result in high levels of inactive, full-length p53 protein with one amino acid change in the core domain that recognizes p53 DNA-binding sites. The ability to endow function to mutated p53 proteins would dramatically improve cancer therapy, because it would reactivate a central apoptotic pathway. By using genetic strategies and p53 assays in yeast and mammalian cells, we identified a global suppressor motif involving codons 235, 239, and 240. These intragenic suppressor mutations, either alone or in combination, restored function to 16 of 30 of the most common p53 cancer mutants tested. The 235-239-240 suppressor motif establishes that manipulation of a small region of the core domain is sufficient to activate a large number of p53 cancer mutants. Understanding the structural basis of the rescue mechanism will allow the pursuit of small compounds able to achieve a similar stabilization of p53 cancer mutants.


Subject(s)
Genes, Suppressor , Genes, p53 , Neoplasms/genetics , Animals , Cell Line , Cricetinae , Humans , Mutagenesis , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL