Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Mol Cell ; 82(16): 3030-3044.e8, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35764091

RESUMO

Characterized by intracellular lipid droplet accumulation, clear cell renal cell carcinoma (ccRCC) is resistant to cytotoxic chemotherapy and is a lethal disease. Through an unbiased siRNA screen of 2-oxoglutarate (2-OG)-dependent enzymes, which play a critical role in tumorigenesis, we identified Jumonji domain-containing 6 (JMJD6) as an essential gene for ccRCC tumor development. The downregulation of JMJD6 abolished ccRCC colony formation in vitro and inhibited orthotopic tumor growth in vivo. Integrated ChIP-seq and RNA-seq analyses uncovered diacylglycerol O-acyltransferase 1 (DGAT1) as a critical JMJD6 effector. Mechanistically, JMJD6 interacted with RBM39 and co-occupied DGAT1 gene promoter with H3K4me3 to induce DGAT1 expression. JMJD6 silencing reduced DGAT1, leading to decreased lipid droplet formation and tumorigenesis. The pharmacological inhibition (or depletion) of DGAT1 inhibited lipid droplet formation in vitro and ccRCC tumorigenesis in vivo. Thus, the JMJD6-DGAT1 axis represents a potential new therapeutic target for ccRCC.


Assuntos
Carcinoma de Células Renais , Diacilglicerol O-Aciltransferase , Histona Desmetilases com o Domínio Jumonji , Neoplasias Renais , Carcinogênese/genética , Carcinoma de Células Renais/genética , Diacilglicerol O-Aciltransferase/genética , Diacilglicerol O-Aciltransferase/metabolismo , Epigênese Genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias Renais/genética , Gotículas Lipídicas/metabolismo
2.
Cell ; 158(6): 1324-1334, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25215490

RESUMO

The P7C3 class of aminopropyl carbazole chemicals fosters the survival of neurons in a variety of rodent models of neurodegeneration or nerve cell injury. To uncover its mechanism of action, an active derivative of P7C3 was modified to contain both a benzophenone for photocrosslinking and an alkyne for CLICK chemistry. This derivative was found to bind nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme involved in the conversion of nicotinamide into nicotinamide adenine dinucleotide (NAD). Administration of active P7C3 chemicals to cells treated with doxorubicin, which induces NAD depletion, led to a rebound in intracellular levels of NAD and concomitant protection from doxorubicin-mediated toxicity. Active P7C3 variants likewise enhanced the activity of the purified NAMPT enzyme, providing further evidence that they act by increasing NAD levels through its NAMPT-mediated salvage.


Assuntos
NAD/metabolismo , Fármacos Neuroprotetores/farmacologia , Animais , Carbazóis/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/agonistas , Citocinas/genética , Citocinas/metabolismo , Doxorrubicina/farmacologia , Humanos , Redes e Vias Metabólicas , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo
3.
Cell ; 150(4): 842-54, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22901813

RESUMO

Due to genome instability, most cancers exhibit loss of regions containing tumor suppressor genes and collateral loss of other genes. To identify cancer-specific vulnerabilities that are the result of copy number losses, we performed integrated analyses of genome-wide copy number and RNAi profiles and identified 56 genes for which gene suppression specifically inhibited the proliferation of cells harboring partial copy number loss of that gene. These CYCLOPS (copy number alterations yielding cancer liabilities owing to partial loss) genes are enriched for spliceosome, proteasome, and ribosome components. One CYCLOPS gene, PSMC2, encodes an essential member of the 19S proteasome. Normal cells express excess PSMC2, which resides in a complex with PSMC1, PSMD2, and PSMD5 and acts as a reservoir protecting cells from PSMC2 suppression. Cells harboring partial PSMC2 copy number loss lack this complex and die after PSMC2 suppression. These observations define a distinct class of cancer-specific liabilities resulting from genome instability.


Assuntos
Genes Essenciais , Instabilidade Genômica , Neoplasias/genética , ATPases Associadas a Diversas Atividades Celulares , Animais , Linhagem Celular Tumoral , Deleção Cromossômica , Dosagem de Genes , Genes Supressores de Tumor , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Transplante Heterólogo
4.
Cell ; 151(7): 1457-73, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23245941

RESUMO

Wnt/ß-catenin signaling plays a key role in the pathogenesis of colon and other cancers; emerging evidence indicates that oncogenic ß-catenin regulates several biological processes essential for cancer initiation and progression. To decipher the role of ß-catenin in transformation, we classified ß-catenin activity in 85 cancer cell lines in which we performed genome-scale loss-of-function screens and found that ß-catenin active cancers are dependent on a signaling pathway involving the transcriptional regulator YAP1. Specifically, we found that YAP1 and the transcription factor TBX5 form a complex with ß-catenin. Phosphorylation of YAP1 by the tyrosine kinase YES1 leads to localization of this complex to the promoters of antiapoptotic genes, including BCL2L1 and BIRC5. A small-molecule inhibitor of YES1 impeded the proliferation of ß-catenin-dependent cancers in both cell lines and animal models. These observations define a ß-catenin-YAP1-TBX5 complex essential to the transformation and survival of ß-catenin-driven cancers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transformação Celular Neoplásica , Neoplasias do Colo/metabolismo , Fosfoproteínas/metabolismo , Proteínas com Domínio T/metabolismo , beta Catenina/metabolismo , Animais , Linhagem Celular Tumoral , Colo/embriologia , Colo/metabolismo , Neoplasias do Colo/patologia , Humanos , Proteínas Inibidoras de Apoptose/genética , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-yes/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-yes/metabolismo , Survivina , Fatores de Transcrição , Transcrição Gênica , Proteínas de Sinalização YAP , Peixe-Zebra/embriologia , Proteína bcl-X/genética , Quinases da Família src/antagonistas & inibidores
5.
J Am Chem Soc ; 142(13): 6128-6138, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32163279

RESUMO

TASIN (Truncated APC-Selective Inhibitors) compounds are selectively toxic to colorectal cancer cells with APC mutations, although their mechanism of action remains unknown. Here, we found that TASINs inhibit three enzymes in the postsqualene cholesterol biosynthetic pathway including EBP, DHCR7, and DHCR24. Even though all three of these enzymes are required for cholesterol biosynthesis, only inhibition of the most upstream enzyme, EBP, led to cancer cell death via depletion of downstream sterols, an observation that was confirmed by genetic silencing of EBP. Pharmacologic inhibition or genetic silencing of either DHCR7 or DHCR24 had no impact on cell viability. By using photoaffinity probes to generate a relationship between chemical structure and probe competition, we identified compounds that selectively inhibit either EBP or DHCR7. These studies identify EBP, but not downstream enzymes in the cholesterol biosynthetic pathway, as a target in APC mutant colorectal cancer and also have implications for the clinical development of highly selective EBP inhibitors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Esteroide Isomerases/antagonistas & inibidores , Proteína da Polipose Adenomatosa do Colo/genética , Antineoplásicos/química , Vias Biossintéticas/efeitos dos fármacos , Colesterol/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/química , Células HCT116 , Humanos , Mutação , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Esteroide Isomerases/metabolismo
6.
Nat Chem Biol ; 12(7): 511-5, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27182663

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , DNA Polimerase I/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Retinoides/farmacologia , Antineoplásicos/química , DNA Polimerase I/genética , DNA Polimerase I/metabolismo , Replicação do DNA/efeitos dos fármacos , Inibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estrutura Molecular , Retinoides/química
7.
Nat Chem Biol ; 12(4): 218-25, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26829472

RESUMO

A hallmark of targeted cancer therapies is selective toxicity among cancer cell lines. We evaluated results from a viability screen of over 200,000 small molecules to identify two chemical series, oxalamides and benzothiazoles, that were selectively toxic at low nanomolar concentrations to the same 4 of 12 human lung cancer cell lines. Sensitive cell lines expressed cytochrome P450 (CYP) 4F11, which metabolized the compounds into irreversible inhibitors of stearoyl CoA desaturase (SCD). SCD is recognized as a promising biological target in cancer and metabolic disease. However, SCD is essential to sebocytes, and accordingly SCD inhibitors cause skin toxicity. Mouse sebocytes did not activate the benzothiazoles or oxalamides into SCD inhibitors, providing a therapeutic window for inhibiting SCD in vivo. We thus offer a strategy to target SCD in cancer by taking advantage of high CYP expression in a subset of tumors.


Assuntos
Antineoplásicos/farmacologia , Benzotiazóis/farmacologia , Descoberta de Drogas/métodos , Neoplasias Pulmonares/enzimologia , Ácido Oxâmico/análogos & derivados , Estearoil-CoA Dessaturase/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Benzotiazóis/farmacocinética , Benzotiazóis/uso terapêutico , Benzotiazóis/toxicidade , Linhagem Celular Tumoral , Sistema Enzimático do Citocromo P-450/metabolismo , Família 4 do Citocromo P450 , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos SCID , Estrutura Molecular , Terapia de Alvo Molecular , Ácido Oxâmico/farmacocinética , Ácido Oxâmico/farmacologia , Ácido Oxâmico/uso terapêutico , Ácido Oxâmico/toxicidade , Ligação Proteica , Glândulas Sebáceas/efeitos dos fármacos , Glândulas Sebáceas/enzimologia , Glândulas Sebáceas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Am Chem Soc ; 138(33): 10561-70, 2016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27459345

RESUMO

Poylcyclic tetrahydroxanthones comprise a large class of cytototoxic natural products. No mechanism of action has been described for any member of the family. We report the synthesis of kibdelone C and several simplified analogs. Both enantiomers of kibdeleone C show low nanomolar cytotoxicity toward multiple human cancer cell lines. Moreover, several simplified derivatives with improved chemical stability display higher activity than the natural product itself. In vitro studies rule out interaction with DNA or inhibition of topoisomerase, both of which are common modes of action for polycyclic aromatic compounds. However, celluar studies reveal that kibdelone C and its simplified derivatives disrupt the actin cytoseketon without directly binding actin or affecting its polymerization in vitro.


Assuntos
Xantonas/síntese química , Xantonas/farmacologia , Linhagem Celular Tumoral , Técnicas de Química Sintética , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Humanos , Xantonas/química
9.
Cell Chem Biol ; 31(1): 139-149.e14, 2024 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-37967558

RESUMO

A novel class of benzoxaboroles was reported to induce cancer cell death but the mechanism was unknown. Using a forward genetics platform, we discovered mutations in cleavage and polyadenylation specific factor 3 (CPSF3) that reduce benzoxaborole binding and confer resistance. CPSF3 is the endonuclease responsible for pre-mRNA 3'-end processing, which is also important for RNA polymerase II transcription termination. Benzoxaboroles inhibit this endonuclease activity of CPSF3 in vitro and also curb transcriptional termination in cells, which results in the downregulation of numerous constitutively expressed genes. Furthermore, we used X-ray crystallography to demonstrate that benzoxaboroles bind to the active site of CPSF3 in a manner distinct from the other known inhibitors of CPSF3. The benzoxaborole compound impeded the growth of cancer cell lines derived from different lineages. Our results suggest benzoxaboroles may represent a promising lead as CPSF3 inhibitors for clinical development.


Assuntos
Antineoplásicos , Compostos de Boro , Fator de Especificidade de Clivagem e Poliadenilação , Endonucleases , Precursores de RNA , Processamento Pós-Transcricional do RNA , Fator de Especificidade de Clivagem e Poliadenilação/antagonistas & inibidores , Fator de Especificidade de Clivagem e Poliadenilação/química , Endonucleases/antagonistas & inibidores , Precursores de RNA/genética , Precursores de RNA/metabolismo , Compostos de Boro/química , Compostos de Boro/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Humanos , Linhagem Celular Tumoral
10.
ACS Chem Biol ; 19(6): 1339-1350, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38829020

RESUMO

N-Pyridinylthiophene carboxamide (compound 21) displays activity against peripheral nerve sheath cancer cells and mouse xenografts by an unknown mechanism. Through medicinal chemistry, we identified a more active derivative, compound 9, and found that only analogues with structures similar to nicotinamide retained activity. Genetic screens using compound 9 found that both NAMPT and NMNAT1, enzymes in the NAD salvage pathway, are necessary for activity. Compound 9 is metabolized by NAMPT and NMNAT1 into an adenine dinucleotide (AD) derivative in a cell-free system, cultured cells, and mice, and inhibition of this metabolism blocked compound activity. AD analogues derived from compound 9 inhibit IMPDH in vitro and cause cell death by inhibiting IMPDH in cells. These findings nominate these compounds as preclinical candidates for the development of tumor-activated IMPDH inhibitors to treat neuronal cancers.


Assuntos
NAD , Niacinamida , Tiofenos , Animais , NAD/metabolismo , Humanos , Camundongos , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Niacinamida/farmacologia , Niacinamida/química , Tiofenos/farmacologia , Tiofenos/química , Tiofenos/metabolismo , Linhagem Celular Tumoral , IMP Desidrogenase/antagonistas & inibidores , IMP Desidrogenase/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Nicotinamida Fosforribosiltransferase/metabolismo , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Neoplasias de Bainha Neural/tratamento farmacológico , Neoplasias de Bainha Neural/metabolismo , Neoplasias de Bainha Neural/patologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Nicotinamida-Nucleotídeo Adenililtransferase/antagonistas & inibidores
11.
J Med Chem ; 67(11): 9277-9301, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38804887

RESUMO

We recently discovered a novel N-aryl tetracyclic dicarboximide MM0299 (1) with robust activity against glioma stem-like cells that potently and selectively inhibits lanosterol synthase leading to the accumulation of the toxic shunt metabolite 24(S),25-epoxycholesterol. Herein, we delineate a systematic and comprehensive SAR study that explores the structural space surrounding the N-aryl tetracyclic dicarboximide scaffold. A series of 100 analogs were synthesized and evaluated for activity against the murine glioma stem-like cell line Mut6 and for metabolic stability in mouse liver S9 fractions. This study led to several analogs with single-digit nanomolar activity in Mut6 glioblastoma cells that were metabolically stable in S9 fractions. In vivo pharmacokinetic analysis of selected analogs identified compound 52a (IC50 = 63 nM; S9 T1/2 > 240 min) which was orally available (39% plasma; 58% brain) and displayed excellent brain exposure. Chronic oral dosing of 52a during a 2-week tolerability study indicated no adverse effect on body weight nor signs of hematologic, liver, or kidney toxicity.


Assuntos
Glioma , Células-Tronco Neoplásicas , Animais , Camundongos , Relação Estrutura-Atividade , Glioma/tratamento farmacológico , Glioma/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Humanos , Descoberta de Drogas , Masculino , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia
12.
Cell Chem Biol ; 30(2): 214-229.e18, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36758549

RESUMO

Glioblastoma (GBM) is an aggressive adult brain cancer with few treatment options due in part to the challenges of identifying brain-penetrant drugs. Here, we investigated the mechanism of MM0299, a tetracyclic dicarboximide with anti-glioblastoma activity. MM0299 inhibits lanosterol synthase (LSS) and diverts sterol flux away from cholesterol into a "shunt" pathway that culminates in 24(S),25-epoxycholesterol (EPC). EPC synthesis following MM0299 treatment is both necessary and sufficient to block the growth of mouse and human glioma stem-like cells by depleting cellular cholesterol. MM0299 exhibits superior selectivity for LSS over other sterol biosynthetic enzymes. Critical for its application in the brain, we report an MM0299 derivative that is orally bioavailable, brain-penetrant, and induces the production of EPC in orthotopic GBM tumors but not normal mouse brain. These studies have implications for the development of an LSS inhibitor to treat GBM or other neurologic indications.


Assuntos
Glioblastoma , Glioma , Adulto , Humanos , Lanosterol/farmacologia , Lanosterol/metabolismo , Encéfalo/metabolismo , Glioma/tratamento farmacológico , Glioma/metabolismo , Colesterol , Glioblastoma/tratamento farmacológico
13.
bioRxiv ; 2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36865268

RESUMO

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.

14.
Cell Chem Biol ; 30(11): 1453-1467.e8, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37607550

RESUMO

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.


Assuntos
Antineoplásicos , Neoplasias do Colo , Humanos , Reparo de Erro de Pareamento de DNA , Antineoplásicos/farmacologia , Mutagênese , Citotoxinas
15.
Nat Commun ; 14(1): 3762, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37353483

RESUMO

Colorectal cancers (CRCs) are prevalent worldwide, yet current treatments remain inadequate. Using chemical genetic screens, we identify that co-inhibition of topoisomerase I (TOP1) and NEDD8 is synergistically cytotoxic in human CRC cells. Combination of the TOP1 inhibitor irinotecan or its bioactive metabolite SN38 with the NEDD8-activating enzyme inhibitor pevonedistat exhibits synergy in CRC patient-derived organoids and xenografts. Mechanistically, we show that pevonedistat blocks the ubiquitin/proteasome-dependent repair of TOP1 DNA-protein crosslinks (TOP1-DPCs) induced by TOP1 inhibitors and that the CUL4-RBX1 complex (CRL4) is a prominent ubiquitin ligase acting on TOP1-DPCs for proteasomal degradation upon auto-NEDD8 modification during replication. We identify DCAF13, a DDB1 and Cullin Associated Factor, as the receptor of TOP1-DPCs for CRL4. Our study not only uncovers a replication-coupled ubiquitin-proteasome pathway for the repair of TOP1-DPCs but also provides molecular and translational rationale for combining TOP1 inhibitors and pevonedistat for CRC and other types of cancers.


Assuntos
Neoplasias Colorretais , Inibidores da Topoisomerase I , Humanos , Inibidores da Topoisomerase I/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Ligases/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Ligação a RNA
16.
Nat Commun ; 12(1): 2954, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34012010

RESUMO

How cancer cells cope with high levels of replication stress during rapid proliferation is currently unclear. Here, we show that macrophage migration inhibitory factor (MIF) is a 3' flap nuclease that translocates to the nucleus in S phase. Poly(ADP-ribose) polymerase 1 co-localizes with MIF to the DNA replication fork, where MIF nuclease activity is required to resolve replication stress and facilitates tumor growth. MIF loss in cancer cells leads to mutation frequency increases, cell cycle delays and DNA synthesis and cell growth inhibition, which can be rescued by restoring MIF, but not nuclease-deficient MIF mutant. MIF is significantly upregulated in breast tumors and correlates with poor overall survival in patients. We propose that MIF is a unique 3' nuclease, excises flaps at the immediate 3' end during DNA synthesis and favors cancer cells evading replication stress-induced threat for their growth.


Assuntos
Neoplasias da Mama/metabolismo , Replicação do DNA/fisiologia , Endonucleases Flap/metabolismo , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , DNA/química , DNA/metabolismo , Dano ao DNA , DNA Polimerase III/genética , DNA Polimerase III/metabolismo , Replicação do DNA/genética , Feminino , Endonucleases Flap/deficiência , Endonucleases Flap/genética , Técnicas de Inativação de Genes , Instabilidade Genômica , Células HCT116 , Humanos , Oxirredutases Intramoleculares/deficiência , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/deficiência , Fatores Inibidores da Migração de Macrófagos/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformação de Ácido Nucleico , Poli(ADP-Ribose) Polimerase-1/metabolismo , Fase S , Especificidade por Substrato
17.
Sci Adv ; 7(47): eabj5405, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34788094

RESUMO

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.

18.
ACS Chem Biol ; 15(1): 158-170, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31874028

RESUMO

Selective toxicity among cancer cells of the same lineage is a hallmark of targeted therapies. As such, identifying compounds that impair proliferation of a subset of non-small-cell lung cancer (NSCLC) cell lines represents one strategy to discover new drugs for lung cancer. Previously, phenotypic screens of 202 103 compounds led to the identification of 208 selective NSCLC toxins ( McMillan , E. A. , Cell , 2018 , 173 , 864 ). The mechanism of action for the majority of these compounds remains unknown. Here, we discovered the target for a series of quinazoline diones (QDC) that demonstrate selective toxicity among 96 NSCLC lines. Using photoreactive probes, we found that the QDC binds to both mitochondrial complex I of the electron transport chain and hydroxyacyl CoA dehydrogenase subunit alpha (HADHA), which catalyzes long-chain fatty acid oxidation. Inhibition of complex I is the on-target activity for QDC, while binding to HADHA is off-target. The sensitivity profile of the QDC across NSCLC lines correlated with the sensitivity profiles of six additional structurally distinct compounds. The antiproliferative activity of these compounds is also the consequence of binding to mitochondrial complex I, reflecting significant structural diversity among complex I inhibitors. Small molecules targeting complex I are currently in clinical development for the treatment of cancer. Our results highlight complex I as a target in NSCLC and report structurally diverse scaffolds that inhibit complex I.


Assuntos
Antineoplásicos/química , Carcinoma Pulmonar de Células não Pequenas/dietoterapia , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Inibidores Enzimáticos/química , Neoplasias Pulmonares/dietoterapia , Quinazolinonas/química , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/farmacologia , Ácidos Graxos/metabolismo , Técnicas de Inativação de Genes , Humanos , Subunidade alfa da Proteína Mitocondrial Trifuncional/genética , Subunidade alfa da Proteína Mitocondrial Trifuncional/metabolismo , Estrutura Molecular , Terapia de Alvo Molecular , Oxirredução , Consumo de Oxigênio , Ligação Proteica , Conformação Proteica , Proteômica , Quinazolinonas/farmacologia , Relação Estrutura-Atividade , Especificidade por Substrato
19.
J Med Chem ; 63(17): 9773-9786, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787093

RESUMO

A series of N-acyl benzothiazoles shows selective and potent cytotoxicity against cancer cell lines expressing cytochrome P450 4F11. A prodrug form is metabolized by cancer cells into an active inhibitor of stearoyl-CoA desaturase (SCD). Substantial variation on the acyl portion of the inhibitors allowed the identification of (R)-27, which balanced potency, solubility, and lipophilicity to allow proof-of-concept studies in mice. The prodrugs were activated inside the tumor, where they can arrest tumor growth. Together, these observations offer promise that a tumor-activated prodrug strategy might exploit the essentiality of SCD for tumor growth, while avoiding toxicity associated with systemic SCD inhibition.


Assuntos
Benzotiazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Estearoil-CoA Dessaturase/antagonistas & inibidores , Animais , Benzotiazóis/farmacocinética , Linhagem Celular Tumoral , Família 4 do Citocromo P450/metabolismo , Feminino , Humanos , Camundongos , Pró-Fármacos/metabolismo , Distribuição Tecidual
20.
Methods Mol Biol ; 1888: 175-187, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30519947

RESUMO

Many small molecule compounds with anticancer activities are discovered through phenotype-based screens. However, discovering the targets of these small molecules has been challenging. The gold standard for target identification requires the discovery of mutations in the target protein that block the effects of small molecules in vitro as well as in vivo. Here we describe the procedures for isolating drug resistant clones using the colorectal cancer cell line HCT-116 followed by whole-exome sequencing to identify recurrent mutations associated with compound resistance. Together with downstream in vitro and in vivo validation experiments, this strategy enables rapid target discovery for cytotoxic compounds.


Assuntos
Descoberta de Drogas/métodos , Resistência a Medicamentos/genética , Sequenciamento do Exoma , Exoma , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Biologia Computacional/métodos , Células HCT116 , Humanos , Concentração Inibidora 50 , Bibliotecas de Moléculas Pequenas , Software
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa