Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nat Chem Biol ; 16(10): 1120-1128, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32690945

RESUMEN

The NUDIX hydrolase NUDT15 was originally implicated in sanitizing oxidized nucleotides, but was later shown to hydrolyze the active thiopurine metabolites, 6-thio-(d)GTP, thereby dictating the clinical response of this standard-of-care treatment for leukemia and inflammatory diseases. Nonetheless, its physiological roles remain elusive. Here, we sought to develop small-molecule NUDT15 inhibitors to elucidate its biological functions and potentially to improve NUDT15-dependent chemotherapeutics. Lead compound TH1760 demonstrated low-nanomolar biochemical potency through direct and specific binding into the NUDT15 catalytic pocket and engaged cellular NUDT15 in the low-micromolar range. We also employed thiopurine potentiation as a proxy functional readout and demonstrated that TH1760 sensitized cells to 6-thioguanine through enhanced accumulation of 6-thio-(d)GTP in nucleic acids. A biochemically validated, inactive structural analog, TH7285, confirmed that increased thiopurine toxicity takes place via direct NUDT15 inhibition. In conclusion, TH1760 represents the first chemical probe for interrogating NUDT15 biology and potential therapeutic avenues.


Asunto(s)
Pirofosfatasas/antagonistas & inhibidores , Pirofosfatasas/metabolismo , Sitios de Unión , Línea Celular , Diseño de Fármacos , Desarrollo de Medicamentos , Escherichia coli , Humanos , Pirofosfatasa Inorgánica/antagonistas & inhibidores , Pirofosfatasa Inorgánica/genética , Pirofosfatasa Inorgánica/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Pirofosfatasas/química , Pirofosfatasas/genética , Relación Estructura-Actividad
2.
EMBO Mol Med ; 12(3): e10419, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31950591

RESUMEN

The deoxycytidine analogue cytarabine (ara-C) remains the backbone treatment of acute myeloid leukaemia (AML) as well as other haematological and lymphoid malignancies, but must be combined with other chemotherapeutics to achieve cure. Yet, the underlying mechanism dictating synergistic efficacy of combination chemotherapy remains largely unknown. The dNTPase SAMHD1, which regulates dNTP homoeostasis antagonistically to ribonucleotide reductase (RNR), limits ara-C efficacy by hydrolysing the active triphosphate metabolite ara-CTP. Here, we report that clinically used inhibitors of RNR, such as gemcitabine and hydroxyurea, overcome the SAMHD1-mediated barrier to ara-C efficacy in primary blasts and mouse models of AML, displaying SAMHD1-dependent synergy with ara-C. We present evidence that this is mediated by dNTP pool imbalances leading to allosteric reduction of SAMHD1 ara-CTPase activity. Thus, SAMHD1 constitutes a novel biomarker for combination therapies of ara-C and RNR inhibitors with immediate consequences for clinical practice to improve treatment of AML.


Asunto(s)
Citarabina/farmacología , Leucemia Mieloide Aguda , Pirofosfatasas/metabolismo , Ribonucleótido Reductasas/antagonistas & inhibidores , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Animales , Trifosfato de Arabinofuranosil Citosina/metabolismo , Ratones
3.
Virology ; 531: 260-268, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959264

RESUMEN

SAMHD1 is a human restriction factor known to prevent infection of macrophages, resting CD4+ T cells, and dendritic cells by HIV-1. To test the contribution of MxB to the ability of SAMHD1 to block HIV-1 infection, we created human THP-1 cell lines that were knocked out for expression of MxB, SAMHD1, or both. Interestingly, MxB depletion renders SAMHD1 ineffective against HIV-1 but not SIVmac. We observed similar results in human primary macrophages that were knockdown for the expression of MxB. To understand how MxB assists SAMHD1 restriction of HIV-1, we examined direct interaction between SAMHD1 and MxB in pull-down experiments. In addition, we investigated several properties of SAMHD1 in the absence of MxB expression, including subcellular localization, phosphorylation of the SAMHD1 residue T592, and dNTPs levels. These experiments showed that SAMHD1 restriction of HIV-1 requires expression of MxB.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1/fisiología , Proteínas de Resistencia a Mixovirus/metabolismo , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Secuencias de Aminoácidos , Infecciones por VIH/genética , Infecciones por VIH/virología , Humanos , Macrófagos/metabolismo , Macrófagos/virología , Proteínas de Resistencia a Mixovirus/genética , Fosforilación , Unión Proteica , Proteína 1 que Contiene Dominios SAM y HD/química , Proteína 1 que Contiene Dominios SAM y HD/genética , Especificidad de la Especie
4.
J Gen Virol ; 100(4): 656-661, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30767852

RESUMEN

The release of porcine endogenous retrovirus (PERV) particles from pig cells is a potential risk factor during xenotransplantation by way of productively infecting the human transplant recipient. Potential countermeasures against PERV replication are restriction factors that block retroviral replication. SAMHD1 is a triphosphohydrolase that depletes the cellular pool of dNTPs in non-cycling cells starving retroviral reverse transcription. We investigated the antiviral activity of human SAMHD1 against PERV and found that SAMHD1 potently restricts its reverse transcription in human monocytes, monocyte-derived dendritic cells (MDDC), or macrophages (MDM) and in monocytic THP-1 cells. Degradation of SAMHD1 by SIVmac Vpx or CRISPR/Cas9 knock-out of SAMHD1 allowed for PERV reverse transcription. Addition of deoxynucleosides alleviated the SAMHD1-mediated restriction suggesting that SAMHD1-mediated degradation of dNTPs restricts PERV replication in these human immune cells. In conclusion, our findings highlight SAMHD1 as a potential barrier to PERV transmission from pig transplants to human recipients during xenotransplantation.


Asunto(s)
Retrovirus Endógenos/fisiología , Xenoinjertos/metabolismo , Xenoinjertos/virología , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Animales , Sistemas CRISPR-Cas/fisiología , Línea Celular , Células HEK293 , Humanos , Macrófagos/metabolismo , Macrófagos/virología , Monocitos/metabolismo , Monocitos/virología , Transcripción Reversa/fisiología , Porcinos , Células THP-1 , Trasplante Heterólogo/métodos , Replicación Viral/fisiología
5.
Exp Hematol ; 52: 32-39, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28502830

RESUMEN

Sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1) is a (deoxy)guanosine triphosphate (dGTP/GTP)-activated deoxyribonucleoside triphosphate (dNTP) triphosphohydrolase involved in cellular dNTP homoeostasis. Mutations in SAMHD1 have been associated with the hyperinflammatory disease Aicardi-Goutières syndrome (AGS). SAMHD1 also limits cells' permissiveness to infection with diverse viruses, including human immunodeficiency virus (HIV-1), and controls endogenous retroviruses. Increasing evidence supports the role of SAMHD1 as a tumor suppressor. However, SAMHD1 also can act as a resistance factor to nucleoside-based chemotherapies by hydrolyzing their active triphosphate metabolites, thereby reducing response of various malignancies to these anticancer drugs. Hence, informed cancer therapies must take into account the ambiguous properties of SAMHD1 as both an inhibitor of uncontrolled proliferation and a resistance factor limiting the efficacy of anticancer treatments. Here, we provide evidence that SAMHD1 is a double-edged sword for patients with acute myelogenous leukemia (AML). Our time-dependent analyses of The Cancer Genome Atlas (TCGA) AML cohort indicate that high expression of SAMHD1, even though it critically limits the efficacy of high-dose ara-C therapy, might be associated with more favorable disease progression.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/genética , Resistencia a Medicamentos/genética , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Malformaciones del Sistema Nervioso/genética , Proteínas Supresoras de Tumor/genética , Enfermedad Aguda , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Azacitidina/análogos & derivados , Azacitidina/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Citarabina/uso terapéutico , Decitabina , Humanos , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Análisis de Supervivencia , Resultado del Tratamiento , Proteínas Supresoras de Tumor/metabolismo
6.
Cell Cycle ; 16(11): 1029-1038, 2017 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-28436707

RESUMEN

Recently, we demonstrated that sterile α motif and HD domain containing protein 1 (SAMHD1) is a major barrier in acute myelogenous leukemia (AML) cells to the cytotoxicity of cytarabine (ara-C), the most important drug in AML treatment. Ara-C is intracellularly converted by the canonical dNTP synthesis pathway to ara-CTP, which serves as a substrate but not an allosteric activator of SAMHD1. Using an AML mouse model, we show here that wild type but not catalytically inactive SAMHD1 reduces ara-C treatment efficacy in vivo. Expanding the clinically relevant substrates of SAMHD1, we demonstrate that THP-1 CRISPR/Cas9 cells lacking a functional SAMHD1 gene showed increased sensitivity to the antimetabolites nelarabine, fludarabine, decitabine, vidarabine, clofarabine, and trifluridine. Within this Extra View, we discuss and build upon both these and our previously reported findings, and propose SAMHD1 is likely active against a variety of nucleoside analog antimetabolites present in anti-cancer chemotherapies. Thus, SAMHD1 may constitute a promising target to improve a wide range of therapies for both hematological and non-haematological malignancies.


Asunto(s)
Antimetabolitos/efectos adversos , Neoplasias/tratamiento farmacológico , Nucleósidos/efectos adversos , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Modelos Biológicos , Neoplasias/patología , Nucleósidos/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína 1 que Contiene Dominios SAM y HD/genética
7.
Nat Med ; 23(2): 256-263, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28067901

RESUMEN

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR-Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Citarabina/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Unión al GTP Monoméricas/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Trifosfato de Arabinofuranosil Citosina/metabolismo , Niño , Preescolar , Citarabina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Técnicas In Vitro , Lactante , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Terapia Molecular Dirigida , Proteínas de Unión al GTP Monoméricas/metabolismo , Pronóstico , Proteína 1 que Contiene Dominios SAM y HD
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA