Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nature ; 511(7507): 90-3, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24870236

RESUMO

Drug resistance is a major hurdle in oncology. Responses of acute myeloid leukaemia (AML) patients to cytarabine (Ara-C)-based therapies are often short lived with a median overall survival of months. Therapies are under development to improve outcomes and include targeting the eukaryotic translation initiation factor (eIF4E) with its inhibitor ribavirin. In a Phase II clinical trial in poor prognosis AML, ribavirin monotherapy yielded promising responses including remissions; however, all patients relapsed. Here we identify a novel form of drug resistance to ribavirin and Ara-C. We observe that the sonic hedgehog transcription factor glioma-associated protein 1 (GLI1) and the UDP glucuronosyltransferase (UGT1A) family of enzymes are elevated in resistant cells. UGT1As add glucuronic acid to many drugs, modifying their activity in diverse tissues. GLI1 alone is sufficient to drive UGT1A-dependent glucuronidation of ribavirin and Ara-C, and thus drug resistance. Resistance is overcome by genetic or pharmacological inhibition of GLI1, revealing a potential strategy to overcome drug resistance in some patients.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Ácido Glucurônico/metabolismo , Glucuronosiltransferase/metabolismo , Proteínas Hedgehog/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Citarabina/metabolismo , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Deleção de Genes , Glucuronosiltransferase/biossíntese , Humanos , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/patologia , Ribavirina/metabolismo , Ribavirina/farmacologia , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco
2.
Blood ; 127(7): 858-68, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26603836

RESUMO

Aggressive double- and triple-hit (DH/TH) diffuse large B-cell lymphomas (DLBCLs) feature activation of Hsp90 stress pathways. Herein, we show that Hsp90 controls posttranscriptional dynamics of key messenger RNA (mRNA) species including those encoding BCL6, MYC, and BCL2. Using a proteomics approach, we found that Hsp90 binds to and maintains activity of eIF4E. eIF4E drives nuclear export and translation of BCL6, MYC, and BCL2 mRNA. eIF4E RNA-immunoprecipitation sequencing in DLBCL suggests that nuclear eIF4E controls an extended program that includes B-cell receptor signaling, cellular metabolism, and epigenetic regulation. Accordingly, eIF4E was required for survival of DLBCL including the most aggressive subtypes, DH/TH lymphomas. Indeed, eIF4E inhibition induces tumor regression in cell line and patient-derived tumorgrafts of TH-DLBCL, even in the presence of elevated Hsp90 activity. Targeting Hsp90 is typically limited by counterregulatory elevation of Hsp70B, which induces resistance to Hsp90 inhibitors. Surprisingly, we identify Hsp70 mRNA as an eIF4E target. In this way, eIF4E inhibition can overcome drug resistance to Hsp90 inhibitors. Accordingly, rational combinatorial inhibition of eIF4E and Hsp90 inhibitors resulted in cooperative antilymphoma activity in DH/TH DLBCL in vitro and in vivo.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Núcleo Celular/metabolismo , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/patologia , Humanos , Linfoma de Células B/patologia , Proteínas de Neoplasias/metabolismo
3.
Biochem Biophys Res Commun ; 434(3): 614-9, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23583375

RESUMO

The eukaryotic translation initiation factor eIF4E is highly elevated in human cancers including acute myeloid leukemia (AML). A potential anticancer agent, ribavirin, targets eIF4E activity in AML patients corresponding to clinical responses. To date, ribavirin is the only direct inhibitor of eIF4E to reach clinical trials. We showed that ribavirin acts as a competitive inhibitor of the methyl 7-guanosine (m(7)G) cap, the natural ligand of eIF4E. Here we examine the conformational changes occurring in human eIF4E upon binding the active metabolite of ribavirin, ribavirin triphosphate (RTP). Our NMR data revealed an unexpected concentration dependence on RTP affinity for eIF4E. We observed NMR spectra characteristic of tight binding at low micromolar concentrations (2-5 µM eIF4E) but much weaker affinity at more typical NMR concentrations (50- ). Comparison of chemical shift perturbation and line broadening suggest that the two eIF4E-RTP complexes differ in the precise positioning of RTP within the cap binding pocket, with the high affinity complex showing more extensive changes to the central ß-sheet and dorsal surface of eIF4E, similar to m(7)G cap. The differences between high and low affinity complexes arise due to concentration dependent aggregation of eIF4E and RTP. Given the intracellular concentrations of eIF4E and RTP and the differential binding toward the W56A eIF4E mutant the high affinity complex is the most physiologically relevant. In summary, these findings demonstrate that RTP binds in the cap-binding site but also suggests new features of this pocket that should be considered in drug design efforts and reveal new insights into ligand eIF4E recognition.


Assuntos
Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Ribavirina/farmacologia , Fator de Iniciação 4E em Eucariotos/química , Humanos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica , Ribavirina/metabolismo
5.
ACS Chem Biol ; 14(3): 348-355, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30763062

RESUMO

Cancer therapies are plagued by resistance. Previously, we discovered a novel form of cancer drug resistance where the Glioma-associated protein 1 (GLI1) elevates UGT1A glucuronidation enzymes, thereby glucuronidating cytarabine and ribavirin, leading to resistance in leukemia patients. Here, we demonstrate that GLI1 imparts resistance to ∼40 compounds, including FDA-approved drugs with disparate chemotypes ( e.g., methotrexate and venetoclax). GLI1 indirectly elevates UGT1As via the chaperone calreticulin, which is required for resistance. Further, we demonstrate that resistant cells are more sensitive to ATP inhibitors, suggesting an Achilles' heel, which could be exploited in the future. In all, we identify GLI1-inducible glucuronidation as a broad-spectrum multidrug resistance pathway.


Assuntos
Antimetabólitos Antineoplásicos/química , Citarabina/química , Glucuronosiltransferase/metabolismo , Bibliotecas de Moléculas Pequenas/química , Proteína GLI1 em Dedos de Zinco/antagonistas & inibidores , Trifosfato de Adenosina/antagonistas & inibidores , Antimetabólitos Antineoplásicos/metabolismo , Calreticulina/metabolismo , Citarabina/metabolismo , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Bibliotecas de Moléculas Pequenas/metabolismo
6.
J Mol Biol ; 431(2): 258-272, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30428301

RESUMO

Drug resistance is a major cause of cancer-related mortality. Glucuronidation of drugs via elevation of UDP-glucuronosyltransferases (UGT1As) correlates with clinical resistance. The nine UGT1A family members have broad substrate specificities attributed to their variable N-terminal domains and share a common C-terminal domain. Development of UGT1As as pharmacological targets has been hampered by toxicity of pan-UGT inhibitors and by difficulty in isolating pure N-terminal domains or full-length proteins. Here, we developed a strategy to target selected UGT1As which exploited the biochemical tractability of the C-domain and its ability to allosterically communicate with the catalytic site. By combining NMR fragment screening with in vitro glucuronidation assays, we identified inhibitors selective for UGT1A4. Significantly, these compounds selectively restored sensitivity in resistant cancer cells only for substrates of the targeted UGT1A. This strategy represents a crucial first step toward developing compounds to overcome unwanted glucuronidation thereby reversing resistance in patients.


Assuntos
Resistência a Medicamentos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Domínio Catalítico/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Especificidade por Substrato
7.
Elife ; 62017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29111978

RESUMO

The microenvironment provides a functional substratum supporting tumour growth. Hyaluronan (HA) is a major component of this structure. While the role of HA in malignancy is well-defined, the mechanisms driving its biosynthesis in cancer are poorly understood. We show that the eukaryotic translation initiation factor eIF4E, an oncoprotein, drives HA biosynthesis. eIF4E stimulates production of enzymes that synthesize the building blocks of HA, UDP-Glucuronic acid and UDP-N-Acetyl-Glucosamine, as well as hyaluronic acid synthase which forms the disaccharide chain. Strikingly, eIF4E inhibition alone repressed HA levels as effectively as directly targeting HA with hyaluronidase. Unusually, HA was retained on the surface of high-eIF4E cells, rather than being extruded into the extracellular space. Surface-associated HA was required for eIF4E's oncogenic activities suggesting that eIF4E potentiates an oncogenic HA program. These studies provide unique insights into the mechanisms driving HA production and demonstrate that an oncoprotein can co-opt HA biosynthesis to drive malignancy.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Ácido Hialurônico/biossíntese , Biossíntese de Proteínas , Vias Biossintéticas/genética , Linhagem Celular , Humanos
8.
Clin Cancer Res ; 21(10): 2207-10, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25810373

RESUMO

Drug resistance remains a major impediment in the development of durable cancer therapies. Studies in acute myelogenous leukemia (AML) patients revealed a new form of multidrug resistance. Here, increased glioma-associated protein GLI1 leads to elevation of the UDP-glucuronosyl transferase (UGT) enzymes. UGTs add glucuronic acid to xenobiotics and metabolites. Traditionally, the loss of these enzymes is thought to contribute to cancer as a result of impaired clearance of environmental carcinogens. However, we demonstrate that overexpression of UGTs can contribute to oncogenesis by promoting drug resistance. Indeed, UGT levels in AML patients treated with ribavirin and/or cytarabine were elevated at relapse relative to diagnosis. This was reversed by GLI1 inhibition, suggesting a clinically relevant strategy to overcome drug resistance. Further, overexpression of UGTs can also lead to drug resistance in other cancers, such as certain Hsp90 inhibitors and vorinostat in colorectal and chronic lymphoblastic leukemia, respectively. Not all drugs are targets of glucuronidation, suggesting that UGT status could be relevant to treatment choice. Here, we describe several facets of UGT biology and how these could be exploited clinically. These studies demonstrate how drugs in cancer cells can be metabolized differentially than their normal counterparts. In summary, we describe a new form of drug resistance relevant to a variety of cancer contexts.


Assuntos
Antineoplásicos/metabolismo , Resistencia a Medicamentos Antineoplásicos , Fatores de Transcrição/fisiologia , Animais , Antineoplásicos/farmacologia , Ácido Glucurônico/metabolismo , Glucuronosiltransferase/metabolismo , Glicosilação , Humanos , Inativação Metabólica , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Proteína GLI1 em Dedos de Zinco
9.
Mol Cell Oncol ; 2(1): e961827, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27308395

RESUMO

Chemoresistance remains a major impediment in cancer therapy. Although major progress has been made in understanding the mechanisms underlying resistance in cancer, there is still more to learn. Our studies provide evidence that Gli1 drives a novel form of drug resistance involving Phase II drug metabolism enzymes, specifically the UGT1A family.

10.
Front Pharmacol ; 4: 28, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23504227

RESUMO

Cancer drug resistance continues to be a major impediment in medical oncology. Clinically, resistance can arise prior to or as a result of cancer therapy. In this review, we discuss different mechanisms adapted by cancerous cells to resist treatment, including alteration in drug transport and metabolism, mutation and amplification of drug targets, as well as genetic rewiring which can lead to impaired apoptosis. Tumor heterogeneity may also contribute to resistance, where small subpopulations of cells may acquire or stochastically already possess some of the features enabling them to emerge under selective drug pressure. Making the problem even more challenging, some of these resistance pathways lead to multidrug resistance, generating an even more difficult clinical problem to overcome. We provide examples of these mechanisms and some insights into how understanding these processes can influence the next generation of cancer therapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA