Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nat Chem Biol ; 12(7): 511-5, 2016 07.
Article in English | MEDLINE | ID: mdl-27182663

ABSTRACT

CD437 is a retinoid-like small molecule that selectively induces apoptosis in cancer cells, but not in normal cells, through an unknown mechanism. We used a forward-genetic strategy to discover mutations in POLA1 that coincide with CD437 resistance (POLA1(R)). Introduction of one of these mutations into cancer cells by CRISPR-Cas9 genome editing conferred CD437 resistance, demonstrating causality. POLA1 encodes DNA polymerase α, the enzyme responsible for initiating DNA synthesis during the S phase of the cell cycle. CD437 inhibits DNA replication in cells and recombinant POLA1 activity in vitro. Both effects are abrogated by the identified POLA1 mutations, supporting POLA1 as the direct antitumor target of CD437. In addition, we detected an increase in the total fluorescence intensity and anisotropy of CD437 in the presence of increasing concentrations of POLA1 that is consistent with a direct binding interaction. The discovery of POLA1 as the direct anticancer target for CD437 has the potential to catalyze the development of CD437 into an anticancer therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Polymerase I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Retinoids/pharmacology , Antineoplastic Agents/chemistry , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , DNA Replication/drug effects , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Retinoids/chemistry
2.
bioRxiv ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36865268

ABSTRACT

Orphan cytotoxins are small molecules for which the mechanism of action (MoA) is either unknown or ambiguous. Unveiling the mechanism of these compounds may lead to useful tools for biological investigation and in some cases, new therapeutic leads. In select cases, the DNA mismatch repair-deficient colorectal cancer cell line, HCT116, has been used as a tool in forward genetic screens to identify compound-resistant mutations, which have ultimately led to target identification. To expand the utility of this approach, we engineered cancer cell lines with inducible mismatch repair deficits, thus providing temporal control over mutagenesis. By screening for compound resistance phenotypes in cells with low or high rates of mutagenesis, we increased both the specificity and sensitivity of identifying resistance mutations. Using this inducible mutagenesis system, we implicate targets for multiple orphan cytotoxins, including a natural product and compounds emerging from a high-throughput screen, thus providing a robust tool for future MoA studies.

3.
Cell Chem Biol ; 30(11): 1453-1467.e8, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37607550

ABSTRACT

Orphan cytotoxins are small molecules for which the mechanism of action (MoA) is either unknown or ambiguous. Unveiling the mechanism of these compounds may lead to useful tools for biological investigation and new therapeutic leads. In selected cases, the DNA mismatch repair-deficient colorectal cancer cell line, HCT116, has been used as a tool in forward genetic screens to identify compound-resistant mutations, which have ultimately led to target identification. To expand the utility of this approach, we engineered cancer cell lines with inducible mismatch repair deficits, thus providing temporal control over mutagenesis. By screening for compound resistance phenotypes in cells with low or high rates of mutagenesis, we increased both the specificity and sensitivity of identifying resistance mutations. Using this inducible mutagenesis system, we implicate targets for multiple orphan cytotoxins, including a natural product and compounds emerging from a high-throughput screen, thus providing a robust tool for future MoA studies.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Humans , DNA Mismatch Repair , Antineoplastic Agents/pharmacology , Mutagenesis , Cytotoxins
4.
Sci Adv ; 7(47): eabj5405, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34788094

ABSTRACT

Aberrant alternative pre-mRNA splicing plays a critical role in MYC-driven cancers and therefore may represent a therapeutic vulnerability. Here, we show that neuroblastoma, a MYC-driven cancer characterized by splicing dysregulation and spliceosomal dependency, requires the splicing factor RBM39 for survival. Indisulam, a "molecular glue" that selectively recruits RBM39 to the CRL4-DCAF15 E3 ubiquitin ligase for proteasomal degradation, is highly efficacious against neuroblastoma, leading to significant responses in multiple high-risk disease models, without overt toxicity. Genetic depletion or indisulam-mediated degradation of RBM39 induces significant genome-wide splicing anomalies and cell death. Mechanistically, the dependency on RBM39 and high-level expression of DCAF15 determine the exquisite sensitivity of neuroblastoma to indisulam. Our data indicate that targeting the dysregulated spliceosome by precisely inhibiting RBM39, a vulnerability in neuroblastoma, is a valid therapeutic strategy.

5.
Cell Rep ; 29(6): 1499-1510.e6, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693891

ABSTRACT

Indisulam and related sulfonamides recruit the splicing factor RBM39 to the CRL4-DCAF15 E3 ubiquitin ligase, resulting in RBM39 ubiquitination and degradation. Here, we used a combination of domain mapping and random mutagenesis to identify domains or residues that are necessary for indisulam-dependent RBM39 ubiquitination. DCAF15 mutations at Q232 or D475 prevent RBM39 recruitment by indisulam. RBM39 is recruited to DCAF15 by its RRM2 (RNA recognition motif 2) and is ubiquitinated on its N terminus. RBM23, which is an RBM39 paralog, is also recruited to the CRL4-DCAF15 ligase through its RRM2 domain and undergoes sulfonamide-dependent degradation. Indisulam alters the expression of more than 3,000 genes and causes widespread intron retention and exon skipping. All of these changes can be attributed to RBM39, and none are the consequence of RBM23 degradation. Our findings demonstrate that indisulam selectively degrades RBM23 and RBM39, the latter of which is critically important for splicing and gene expression.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , RNA Recognition Motif Proteins/metabolism , RNA Splicing/drug effects , RNA-Binding Proteins/metabolism , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lysine , Mutagenesis , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , RNA Recognition Motif Proteins/genetics , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
6.
Science ; 356(6336)2017 04 28.
Article in English | MEDLINE | ID: mdl-28302793

ABSTRACT

Indisulam is an aryl sulfonamide drug with selective anticancer activity. Its mechanism of action and the basis for its selectivity have so far been unknown. Here we show that indisulam promotes the recruitment of RBM39 (RNA binding motif protein 39) to the CUL4-DCAF15 E3 ubiquitin ligase, leading to RBM39 polyubiquitination and proteasomal degradation. Mutations in RBM39 that prevent its recruitment to CUL4-DCAF15 increase RBM39 stability and confer resistance to indisulam's cytotoxicity. RBM39 associates with precursor messenger RNA (pre-mRNA) splicing factors, and inactivation of RBM39 by indisulam causes aberrant pre-mRNA splicing. Many cancer cell lines derived from hematopoietic and lymphoid lineages are sensitive to indisulam, and their sensitivity correlates with DCAF15 expression levels. Two other clinically tested sulfonamides, tasisulam and chloroquinoxaline sulfonamide, share the same mechanism of action as indisulam. We propose that DCAF15 expression may be a useful biomarker to guide clinical trials of this class of drugs, which we refer to as SPLAMs (splicing inhibitor sulfonamides).


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Pharmacological/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Nuclear Proteins/metabolism , RNA Splicing/drug effects , RNA-Binding Proteins/metabolism , Sulfonamides/pharmacology , Ubiquitin-Protein Ligase Complexes/metabolism , Amino Acid Substitution , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Mice , Mice, Knockout , Mutation , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA-Binding Proteins/genetics , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Ubiquitination , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL