Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 33(3): 4525-4537, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30702927

RESUMEN

It has been shown that 5-amino-4-imidazolecarboxamide riboside (AICAr) can inhibit cell proliferation and induce apoptosis in childhood acute lymphoblastic leukemia (ALL) cells. Although AICAr could regulate cellular energy metabolism by activating AMPK, the cytotoxic mechanisms of AICAr are still unclear. Here, we knocked out TP53 or PRKAA1 gene (encoding AMPKα1) in NALM-6 and Reh cells by using the clustered regularly interspaced short palindromic repeats/Cas9 system and found that AICAr-induced proliferation inhibition was independent of AMPK activation but dependent on p53. Liquid chromatography-mass spectrometry analysis of nucleotide metabolites indicated that AICAr caused an increase in adenosine triphosphate, deoxyadenosine triphosphate, and deoxyguanosine triphosphate levels by up-regulating purine biosynthesis, while AICAr led to a decrease in cytidine triphosphate, uridine triphosphate, deoxycytidine triphosphate, and deoxythymidine triphosphate levels because of reduced phosphoribosyl pyrophosphate production, which consequently impaired the pyrimidine biosynthesis. Ribonucleoside triphosphate (NTP) pool imbalances suppressed the rRNA transcription efficiency. Furthermore, deoxy-ribonucleoside triphosphate (dNTP) pool imbalances induced DNA replication stress and DNA double-strand breaks, followed by cell cycle arrest and apoptosis in ALL cells. Exogenous uridine could rebalance the NTP and dNTP pools by supplementing pyrimidine and then attenuate AICAr-induced cytotoxicity. Our data indicate that RNA transcription inhibition and DNA replication stress induced by NTP and dNTP pool imbalances might play a key role in AICAr-mediated cytotoxic effects on ALL cells, suggesting a potential clinical application of AICAr in future ALL therapy.-Du, L., Yang, F., Fang, H., Sun, H., Chen, Y., Xu, Y., Li, H., Zheng, L., Zhou, B.-B. S. AICAr suppresses cell proliferation by inducing NTP and dNTP pool imbalances in acute lymphoblastic leukemia cells.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Nucleótidos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Ribonucleótidos/farmacología , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/fisiología , Aminoimidazol Carboxamida/antagonistas & inhibidores , Aminoimidazol Carboxamida/farmacología , Aminoimidazol Carboxamida/toxicidad , Apoptosis/efectos de los fármacos , Sistemas CRISPR-Cas , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Desoxirribonucleótidos/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Técnicas de Inactivación de Genes , Genes p53 , Genes de ARNr , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , ARN Ribosómico/biosíntesis , Ribonucleótidos/antagonistas & inhibidores , Ribonucleótidos/metabolismo , Ribonucleótidos/toxicidad , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/fisiología , Uridina/farmacología
2.
Sci Transl Med ; 9(396)2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659436

RESUMEN

Emerging viral infections are difficult to control because heterogeneous members periodically cycle in and out of humans and zoonotic hosts, complicating the development of specific antiviral therapies and vaccines. Coronaviruses (CoVs) have a proclivity to spread rapidly into new host species causing severe disease. Severe acute respiratory syndrome CoV (SARS-CoV) and Middle East respiratory syndrome CoV (MERS-CoV) successively emerged, causing severe epidemic respiratory disease in immunologically naïve human populations throughout the globe. Broad-spectrum therapies capable of inhibiting CoV infections would address an immediate unmet medical need and could be invaluable in the treatment of emerging and endemic CoV infections. We show that a nucleotide prodrug, GS-5734, currently in clinical development for treatment of Ebola virus disease, can inhibit SARS-CoV and MERS-CoV replication in multiple in vitro systems, including primary human airway epithelial cell cultures with submicromolar IC50 values. GS-5734 was also effective against bat CoVs, prepandemic bat CoVs, and circulating contemporary human CoV in primary human lung cells, thus demonstrating broad-spectrum anti-CoV activity. In a mouse model of SARS-CoV pathogenesis, prophylactic and early therapeutic administration of GS-5734 significantly reduced lung viral load and improved clinical signs of disease as well as respiratory function. These data provide substantive evidence that GS-5734 may prove effective against endemic MERS-CoV in the Middle East, circulating human CoV, and, possibly most importantly, emerging CoV of the future.


Asunto(s)
Alanina/análogos & derivados , Antivirales/farmacología , Coronavirus/efectos de los fármacos , Epidemias , Ribonucleótidos/farmacología , Zoonosis/epidemiología , Zoonosis/virología , Adenosina Monofosfato/análogos & derivados , Alanina/metabolismo , Alanina/farmacocinética , Alanina/farmacología , Alanina/toxicidad , Animales , Antivirales/metabolismo , Antivirales/farmacocinética , Antivirales/toxicidad , Callithrix , Línea Celular , Células Epiteliales/virología , Humanos , Pulmón/patología , Ratones , Ribonucleótidos/metabolismo , Ribonucleótidos/farmacocinética , Ribonucleótidos/toxicidad , Replicación Viral/efectos de los fármacos , Zoonosis/prevención & control
3.
Diabetologia ; 55(1): 245-54, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21993711

RESUMEN

AIMS/HYPOTHESIS: Neural tube defects (NTDs) are a common malformation associated with diabetic embryopathy. Maternal hyperglycaemia-induced oxidative stress inhibits the expression of Pax3, a gene that is essential for neural tube closure, and increases the incidence of NTDs. Because oxidative stress can stimulate AMP-activated kinase (AMPK) activity, and AMPK can regulate gene transcription, we hypothesised that increased AMPK activity would mediate the adverse effects of maternal hyperglycaemia-induced oxidative stress on Pax3 expression and NTDs. METHODS: Pregnant mice were made transiently hyperglycaemic by glucose injection, or hypoxic by housing in a hypoxic chamber, or were treated with antimycin A to induce oxidative stress, and AMPK activity in the embryos was assayed. The effects of stimulating AMPK activity with 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR) on Pax3 expression and NTDs were determined. Vitamin E or glutathione ethyl ester was used to reduce oxidative stress, and compound C was used to inhibit AMPK activation. Murine embryonic stem cells were employed as an in vitro model to study the effects of oxidative stress on AMPK activity and the effects of AMPK stimulation on Pax3 expression. RESULTS: Maternal hyperglycaemia stimulated AMPK activity, and stimulation of AMPK with AICAR inhibited Pax3 expression (in vivo and in vitro) and increased NTDs (in vivo). Stimulation of AMPK by hyperglycaemia, hypoxia or antimycin A was inhibited by antioxidants. The AMPK inhibitor compound C blocked the effects of hyperglycaemia or AA on Pax3 expression and NTDs. CONCLUSIONS/INTERPRETATION: Stimulation of AMPK in embryos during a diabetic pregnancy mediates the effects of hyperglycaemia-induced oxidative stress to disturb the expression of the critical Pax3 gene, thereby causing NTDs.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Estrés Oxidativo , Embarazo en Diabéticas/fisiopatología , Proteínas Quinasas Activadas por AMP/química , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/toxicidad , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Línea Celular , Embrión de Mamíferos/enzimología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hiperglucemia/fisiopatología , Hipoxia/fisiopatología , Ratones , Ratones Endogámicos ICR , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/prevención & control , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Embarazo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , ARN Mensajero/metabolismo , Ribonucleótidos/toxicidad
4.
Med Hypotheses ; 71(5): 657-63, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18710792

RESUMEN

The genetic deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT), located on the X chromosome, causes a severe neurological disorder in man, known as Lesch-Nyhan disease (LND). The enzyme HPRT is part of the savage pathway of purine biosynthesis and catalyzes the conversion of hypoxanthine and guanine to their respective nucleotides, IMP and GMP. HPRT deficiency is associated with a relatively selective dysfunction of brain dopamine systems. Several metabolites that accumulate in the patients (phosphoribosylpyrophosphate (PRPP), hypoxanthine, guanine, xanthine, and Z-nucleotides) have been proposed as toxic agents in LND. Some authors have pointed that Z-riboside, derived from the accumulation of ZMP, could be the toxic metabolite in LND. However, the available experimental data support a better hypothesis. I suggest that ZMP (and not Z-riboside) is the key toxic metabolite in LND. ZMP is an inhibitor of the bifunctional enzyme adenylosuccinate lyase, and a deficiency of this enzyme causes psychomotor and mental retardation in humans. Moreover, it has been reported that ZMP inhibits mitochondrial oxidative phosphorylation and induces apoptosis in certain cell types. ZMP is also an activator of the AMP-activated protein kinase (AMPK), a homeostatic regulator of energy levels in the cell. The AMPK has been implicated in the regulation of cell viability, catecholamine biosynthesis and cell structure. I propose that accumulation of ZMP will induce a pleiotropic effect in the brain by (1) a direct inhibition of mitochondrial respiration and the bifunctional enzyme adenylosuccinate lyase, and (2) a sustained activation of the AMPK which in turns would reduce cell viability, decrease dopamine synthesis, and alters cell morphology. In addition, a mechanism to explain the accumulation of ZMP in LND is presented. The knowledge of the toxic metabolite, and the way it acts, would help to design a better therapy.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Síndrome de Lesch-Nyhan/etiología , Ribonucleótidos/toxicidad , Aminoimidazol Carboxamida/química , Aminoimidazol Carboxamida/toxicidad , Línea Celular Tumoral , Humanos , Hipoxantina Fosforribosiltransferasa/metabolismo , Inosina Monofosfato/química , Síndrome de Lesch-Nyhan/metabolismo , Modelos Biológicos , Modelos Químicos , Modelos Teóricos , Fosforilación Oxidativa , Purinas/química , Ribonucleósidos/química , Ribonucleótidos/química
5.
Genetics ; 170(1): 61-70, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15744050

RESUMEN

Because some metabolic intermediates are involved in more than one pathway, crosstalk between pathways is crucial to maintaining homeostasis. AMP and histidine biosynthesis pathways are coregulated at the transcriptional level in response to adenine availability. 5'-Phosphoribosyl-4-carboxamide-5-aminoimidazole (AICAR), a metabolic intermediate at the crossroads between these two pathways, is shown here to be critical for activation of the transcriptional response in the absence of adenine. In this study, we show that both AMP and histidine pathways significantly contribute to AICAR synthesis. Furthermore, we show that upregulation of the histidine pathway clearly interferes with regulation of the AMP pathway, thus providing an explanation for the regulatory crosstalk between these pathways. Finally, we revisit the histidine auxotrophy of ade3 or ade16 ade17 mutants. Interestingly, overexpression of PMU1, encoding a potential phosphomutase, partially suppresses the histidine requirement of an ade3 ade16 ade17 triple mutant, most probably by reducing the level of AICAR in this mutant. Together our data clearly establish that AICAR is not just a metabolic intermediate but also acts as a true regulatory molecule.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Histidina/metabolismo , Purinas/metabolismo , Ribonucleótidos/fisiología , Saccharomyces cerevisiae/metabolismo , Adenosina Monofosfato/biosíntesis , Aminoimidazol Carboxamida/toxicidad , Ácido Fólico/metabolismo , Regulación Fúngica de la Expresión Génica/fisiología , Inosina Monofosfato/metabolismo , Ribonucleótidos/toxicidad , Saccharomyces cerevisiae/genética
6.
Nucleic Acids Res Suppl ; (1): 255-6, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-12836361

RESUMEN

The bicyclic pyrimidine analogue, 3,4-dihydro-6H,8H-pyrimido[4,5-c][1,2]oxazin-7-one (P) can base pair with both A and G. The riboside 5'-triphsophate of P (PTP) efficiently induces mutation during in vitro transcription and reverse transcription cycles using a phage promoter. In the present study, we have constructed an in vitro transcription system promoted by the human immunodeficiency virus type 1 (HIV-1) 5'-long terminal repeat (LTR) using HeLa nuclear extract supplemented with HIV-1 Tat protein. Using this system, the effects of mutagenic ribonucleotide analogues were studied.


Asunto(s)
Fármacos Anti-VIH/toxicidad , VIH-1/genética , Mutágenos/toxicidad , Ribonucleótidos/toxicidad , Fármacos Anti-VIH/química , Fármacos Anti-VIH/uso terapéutico , Duplicado del Terminal Largo de VIH , VIH-1/efectos de los fármacos , Células HeLa , Humanos , Mutagénesis , Mutágenos/química , Mutágenos/uso terapéutico , ADN Polimerasa Dirigida por ARN/metabolismo , Ribonucleótidos/química , Ribonucleótidos/uso terapéutico , Transcripción Genética
7.
J Med Chem ; 27(10): 1295-9, 1984 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6592359

RESUMEN

L-Alanosine [3-(hydroxynitrosoamino)-L-alanine] is an antitumor antibiotic that at the present is undergoing phase II clinical trials. Its mode of action as well as its metabolism has been extensively studied, and the metabolite N-[(5-amino-1-beta-D-ribofuranosyl-1H-imidazol-4-yl)carbonyl]-3- (hydroxynitrosoamino)-L-alanine ribonucleotide (L-alanosine AICOR) proved to be an extremely potent inhibitor of de novo purine biosynthesis and is thus primarily responsible for the antitumor activity of the drug. The synthesis of the corresponding ribonucleoside, i.e., N-[(5-amino-1-beta-D-ribofuranosyl-1H-imidazol-4-yl)carbonyl]-3- (hydroxynitrosamino)-L-alanine ribonucleoside (L-alanosine AICO ribonucleoside), was accomplished by condensation of a suitably protected derivative of L-alanosine with N-succinimidyl-5-amino-1-(2,3,5-tri-O-acetyl-beta-D-ribofuranosyl)-1H-im idazole-4-carboxylate followed by the removal of the protective groups. The biological activity of L-alanosine AICO ribonucleoside was tested in vitro on whole tumor cells and on the isolated enzyme adenylosuccinate synthetase and in vivo on murine experimental leukemia. The compound was found to be inactive in these tests.


Asunto(s)
Antibióticos Antineoplásicos/síntesis química , Ribonucleótidos/síntesis química , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Humanos , Indicadores y Reactivos , Leucemia Mieloide Aguda , Espectroscopía de Resonancia Magnética , Ribonucleótidos/toxicidad , Espectrofotometría Infrarroja
8.
Cancer Res ; 44(8): 3608-12, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6744283

RESUMEN

A Phase I trial of tricyclic nucleoside phosphate (1,4,5,6,8-pentaazaacenaphthylene-3-amino-1, 5-dihydro-5-methyl-1-beta-D-ribofuranosyl 5'-phosphate ester; NSC 280594) was conducted using a 5-day continuous infusion schedule. Thirty-seven patients with advanced cancer were entered on the study, of whom 33 patients were evaluable for response and toxicity. Dose levels ranged from 10 mg/sq m/day X 5 days to 40 mg/sq m/day X 5 days. Initially, courses were repeated every 3 to 4 weeks. As cumulative toxicity became manifested, the interval between courses was changed to every 6 weeks. Major toxicities included hyperglycemia, hepatotoxicity, and thrombocytopenia. Patients with a prior history of diabetes mellitus, extensive radiation therapy, or significant liver metastases were prone to severe toxicity. Other toxicities noted were nausea and vomiting, abdominal discomfort, anemia, and reduction in serum calcium, phosphorus, and albumin levels. Rare side effects included hypertriglyceridemia, hyperamylasemia, diarrhea, and stomatitis. Antitumor activity observed include improvement in s.c. metastases in a patient with papillary thyroid carcinoma, stabilization of disease in a patient with mesothelioma, and mixed responses in three patients (colon cancer, sarcoma, and tonsillar squamous cell cancer). Recommended schedule for Phase II studies is 20 mg/sq m/day for 5 days every 6 weeks.


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias/tratamiento farmacológico , Acenaftenos , Adulto , Anciano , Anemia/inducido químicamente , Plaquetas/efectos de los fármacos , Evaluación de Medicamentos , Femenino , Granulocitos/efectos de los fármacos , Humanos , Hiperglucemia/inducido químicamente , Infusiones Parenterales , Leucocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Ribonucleótidos/administración & dosificación , Ribonucleótidos/toxicidad
9.
J Cell Biol ; 98(4): 1296-303, 1984 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6715406

RESUMEN

Transcription of the Balbiani ring (BR) genes of the dipteran Chironomus tentans was inhibited by teh nucleoside analogue DRB (5,6-dichloro-1-beta-D-ribofuranosyl benzimidazole). The BR genes were emptied of RNA polymerases and the subsequent packing of the genes was monitored by transmission electron microscopy. The thin chromatin axis of the transcriptionally active genes condensed into a thick (20-25 nm) chromatin fiber, which was recorded as a linear structure, an open loop or a supercoiled loop. The compacted genes were finally packed into dense clumps of chromatin. It was proposed that upon repression of RNA synthesis the BR gene template attains the following consecutive stages with increasing compaction: transcription loop----linear thick fiber----open thick fiber loop----supercoiled thick fiber loop----dense chromatin. Within the chromatin blocks structures that resembled the supercoiled loops were discerned, suggesting that the final packing of the template might be accomplished by a close alignment of supercoiled loops.


Asunto(s)
Cromatina/fisiología , Genes/efectos de los fármacos , Ribonucleótidos/toxicidad , Transcripción Genética/efectos de los fármacos , Animales , Chironomidae , Cromatina/efectos de los fármacos , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Microscopía Electrónica , Glándulas Salivales/fisiología , Glándulas Salivales/ultraestructura , Moldes Genéticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...