Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Biol Chem ; 295(11): 3431-3446, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32005668

ABSTRACT

Cytotoxic molecules can kill cancer cells by disrupting critical cellular processes or by inducing novel activities. 6-(4-(Diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one (DNMDP) is a small molecule that kills cancer cells by generation of novel activity. DNMDP induces complex formation between phosphodiesterase 3A (PDE3A) and schlafen family member 12 (SLFN12) and specifically kills cancer cells expressing elevated levels of these two proteins. Here, we examined the characteristics and covariates of the cancer cell response to DNMDP. On average, the sensitivity of human cancer cell lines to DNMDP is correlated with PDE3A expression levels. However, DNMDP could also bind the related protein, PDE3B, and PDE3B supported DNMDP sensitivity in the absence of PDE3A expression. Although inhibition of PDE3A catalytic activity did not account for DNMDP sensitivity, we found that expression of the catalytic domain of PDE3A in cancer cells lacking PDE3A is sufficient to confer sensitivity to DNMDP, and substitutions in the PDE3A active site abolish compound binding. Moreover, a genome-wide CRISPR screen identified the aryl hydrocarbon receptor-interacting protein (AIP), a co-chaperone protein, as required for response to DNMDP. We determined that AIP is also required for PDE3A-SLFN12 complex formation. Our results provide mechanistic insights into how DNMDP induces PDE3A-SLFN12 complex formation, thereby killing cancer cells with high levels of PDE3A and SLFN12 expression.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/pathology , Base Sequence , Biomarkers, Tumor/metabolism , CRISPR-Cas Systems/genetics , Catalytic Domain , Cell Death/drug effects , Cell Line, Tumor , Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry , Frameshift Mutation/genetics , Genome , Heterozygote , Humans , Protein Binding/drug effects , Pyridazines/pharmacology
2.
Nature ; 506(7488): 371-5, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24390348

ABSTRACT

Cervical cancer is responsible for 10-15% of cancer-related deaths in women worldwide. The aetiological role of infection with high-risk human papilloma viruses (HPVs) in cervical carcinomas is well established. Previous studies have also implicated somatic mutations in PIK3CA, PTEN, TP53, STK11 and KRAS as well as several copy-number alterations in the pathogenesis of cervical carcinomas. Here we report whole-exome sequencing analysis of 115 cervical carcinoma-normal paired samples, transcriptome sequencing of 79 cases and whole-genome sequencing of 14 tumour-normal pairs. Previously unknown somatic mutations in 79 primary squamous cell carcinomas include recurrent E322K substitutions in the MAPK1 gene (8%), inactivating mutations in the HLA-B gene (9%), and mutations in EP300 (16%), FBXW7 (15%), NFE2L2 (4%), TP53 (5%) and ERBB2 (6%). We also observe somatic ELF3 (13%) and CBFB (8%) mutations in 24 adenocarcinomas. Squamous cell carcinomas have higher frequencies of somatic nucleotide substitutions occurring at cytosines preceded by thymines (Tp*C sites) than adenocarcinomas. Gene expression levels at HPV integration sites were statistically significantly higher in tumours with HPV integration compared with expression of the same genes in tumours without viral integration at the same site. These data demonstrate several recurrent genomic alterations in cervical carcinomas that suggest new strategies to combat this disease.


Subject(s)
Genome, Human/genetics , Mutation/genetics , Uterine Cervical Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/virology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Case-Control Studies , Cell Cycle Proteins/genetics , Core Binding Factor beta Subunit/genetics , DNA Copy Number Variations/genetics , DNA Mutational Analysis , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein/genetics , Exome/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Gene Expression Regulation, Neoplastic/genetics , Genomics , HLA-B Antigens/genetics , Humans , Mitogen-Activated Protein Kinase 1/genetics , NF-E2-Related Factor 2/genetics , Papillomaviridae/genetics , Papillomaviridae/physiology , Papillomavirus Infections/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , Receptor, ErbB-2/genetics , Transcription Factors/genetics , Transcriptome/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Uterine Cervical Neoplasms/virology , Virus Integration/genetics
4.
Proc Natl Acad Sci U S A ; 109(36): 14476-81, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22908275

ABSTRACT

We assessed somatic alleles of six receptor tyrosine kinase genes mutated in lung adenocarcinoma for oncogenic activity. Five of these genes failed to score in transformation assays; however, novel recurring extracellular domain mutations of the receptor tyrosine kinase gene ERBB2 were potently oncogenic. These ERBB2 extracellular domain mutants were activated by two distinct mechanisms, characterized by elevated C-terminal tail phosphorylation or by covalent dimerization mediated by intermolecular disulfide bond formation. These distinct mechanisms of receptor activation converged upon tyrosine phosphorylation of cellular proteins, impacting cell motility. Survival of Ba/F3 cells transformed to IL-3 independence by the ERBB2 extracellular domain mutants was abrogated by treatment with small-molecule inhibitors of ERBB2, raising the possibility that patients harboring such mutations could benefit from ERBB2-directed therapy.


Subject(s)
Adenocarcinoma/enzymology , Lung Neoplasms/enzymology , Mutation/genetics , Receptor, ErbB-2/genetics , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Alleles , Animals , Cell Movement/physiology , Cloning, Molecular , DNA Primers/genetics , Dimerization , Immunoblotting , Lung Neoplasms/genetics , Mice , NIH 3T3 Cells , Phosphorylation , Protein Structure, Tertiary/genetics , Retroviridae , Tandem Mass Spectrometry
5.
J Med Chem ; 67(11): 8962-8987, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38748070

ABSTRACT

Dysregulation of histone methyl transferase nuclear receptor-binding SET domain 2 (NSD2) has been implicated in several hematological and solid malignancies. NSD2 is a large multidomain protein that carries histone writing and histone reading functions. To date, identifying inhibitors of the enzymatic activity of NSD2 has proven challenging in terms of potency and SET domain selectivity. Inhibition of the NSD2-PWWP1 domain using small molecules has been considered as an alternative approach to reduce NSD2-unregulated activity. In this article, we present novel computational chemistry approaches, encompassing free energy perturbation coupled to machine learning (FEP/ML) models as well as virtual screening (VS) activities, to identify high-affinity NSD2 PWWP1 binders. Through these activities, we have identified the most potent NSD2-PWWP1 binder reported so far in the literature: compound 34 (pIC50 = 8.2). The compounds identified herein represent useful tools for studying the role of PWWP1 domains for inhibition of human NSD2.


Subject(s)
Drug Design , Histone-Lysine N-Methyltransferase , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Ligands , Humans , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Structure-Activity Relationship , Machine Learning , Models, Molecular , Protein Domains
6.
Nat Commun ; 12(1): 4375, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34272366

ABSTRACT

DNMDP and related compounds, or velcrins, induce complex formation between the phosphodiesterase PDE3A and the SLFN12 protein, leading to a cytotoxic response in cancer cells that express elevated levels of both proteins. The mechanisms by which velcrins induce complex formation, and how the PDE3A-SLFN12 complex causes cancer cell death, are not fully understood. Here, we show that PDE3A and SLFN12 form a heterotetramer stabilized by binding of DNMDP. Interactions between the C-terminal alpha helix of SLFN12 and residues near the active site of PDE3A are required for complex formation, and are further stabilized by interactions between SLFN12 and DNMDP. Moreover, we demonstrate that SLFN12 is an RNase, that PDE3A binding increases SLFN12 RNase activity, and that SLFN12 RNase activity is required for DNMDP response. This new mechanistic understanding will facilitate development of velcrin compounds into new cancer therapies.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Pyridazines/chemistry , Adenosine Monophosphate/chemistry , Calorimetry, Differential Scanning , Catalytic Domain , Cell Survival/drug effects , Cell Survival/genetics , Cryoelectron Microscopy , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Endoribonucleases/chemistry , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kinetics , Mass Spectrometry , Multienzyme Complexes/ultrastructure , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Multimerization , Pyridazines/pharmacology , Recombinant Proteins , Tetrahydroisoquinolines/chemistry
7.
J Clin Invest ; 124(4): 1582-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569458

ABSTRACT

Targeted cancer therapies often induce "outlier" responses in molecularly defined patient subsets. One patient with advanced-stage lung adenocarcinoma, who was treated with oral sorafenib, demonstrated a near-complete clinical and radiographic remission for 5 years. Whole-genome sequencing and RNA sequencing of primary tumor and normal samples from this patient identified a somatic mutation, ARAF S214C, present in the cancer genome and expressed at high levels. Additional mutations affecting this residue of ARAF and a nearby residue in the related kinase RAF1 were demonstrated across 1% of an independent cohort of lung adenocarcinoma cases. The ARAF mutations were shown to transform immortalized human airway epithelial cells in a sorafenib-sensitive manner. These results suggest that mutant ARAF is an oncogenic driver in lung adenocarcinoma and an indicator of sorafenib response.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation, Missense , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Proto-Oncogene Proteins A-raf/genetics , Adenocarcinoma/enzymology , Adenocarcinoma of Lung , Aged , Amino Acid Substitution , Cell Transformation, Neoplastic/genetics , DNA, Neoplasm/genetics , Female , Humans , Lung Neoplasms/enzymology , Molecular Targeted Therapy , Niacinamide/therapeutic use , Oncogenes , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/genetics , Sorafenib
SELECTION OF CITATIONS
SEARCH DETAIL