Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 331: 54-61, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28501332

RESUMEN

Emerging evidence suggests that increased nicotinamide phosphoribosyltransferase (NAMPT) expression is associated with the development and prognosis of many cancers, but it remains unknown regarding its role in oral squamous cell carcinoma (OSCC). In the present study, the results from tissue microarray showed that NAMPT was overexpressed in OSCC patients and its expression level was directly correlated with differential grades of cancer. Interestingly, treatment of OSCC cells with chemotherapy agent arsenic trioxide (ATO) decreased the levels of NAMPT protein and increased cellular death in an ATO dose- and time-dependent manner. Most importantly, combination of low concentration ATO with FK866 (a NAMPT inhibitor) exerted enhanced inhibitive effect on NAMPT protein and mRNA expressions, leading to synergistic cytotoxicity on cancer cells through increasing cell apoptosis and depleting intracellular nicotinamide adenine dinucleotide levels. These findings demonstrate the crucial role of NAMPT in the prognosis of OSCC and reveal inhibition of NAMPT as a novel mechanism of ATO in suppressing cancer cell growth. Our results suggest that ATO can significantly enhance therapeutic efficacy of NAMPT inhibitor, and combined treatment may be a novel and effective therapeutic strategy for OSCC patients.


Asunto(s)
Arsenicales/farmacología , Carcinoma de Células Escamosas/metabolismo , Citocinas/antagonistas & inhibidores , Neoplasias de la Boca/metabolismo , NAD/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Óxidos/farmacología , Adulto , Trióxido de Arsénico , Arsenicales/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citocinas/biosíntesis , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/tratamiento farmacológico , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/biosíntesis , Óxidos/uso terapéutico
2.
Lipids Health Dis ; 16(1): 191, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974242

RESUMEN

BACKGROUND: Nicotinamide phosphoribosyltransferase (NAMPT) and nicotinamide adenine dinucleotide (NAD) levels are crucial for liver function. The saturated fatty acid palmitate and the unsaturated fatty acid oleate are the main free fatty acids in adipose tissue and human diet. We asked how these fatty acids affect cell survival, NAMPT and NAD levels in HepG2 cells and primary human hepatocytes. METHODS: HepG2 cells were stimulated with palmitate (0.5mM), oleate (1mM) or a combination of both (0.5mM/1mM) as well as nicotinamide mononucleotide (NMN) (0.5 mM) or the specific NAMPT inhibitor FK866 (10nM). Cell survival was measured by WST-1 assay and Annexin V/propidium iodide staining. NAD levels were determined by NAD/NADH Assay or HPLC. Protein and mRNA levels were analysed by Western blot analyses and qPCR, respectively. NAMPT enzyme activity was measured using radiolabelled 14C-nicotinamide. Lipids were stained by Oil red O staining. RESULTS: Palmitate significantly reduced cell survival and induced apoptosis at physiological doses. NAMPT activity and NAD levels significantly declined after 48h of palmitate. In addition, NAMPT mRNA expression was enhanced which was associated with increased NAMPT release into the supernatant, while intracellular NAMPT protein levels remained stable. Oleate alone did not influence cell viability and NAMPT activity but ameliorated the negative impact of palmitate on cell survival, NAMPT activity and NAD levels, as well as the increased NAMPT mRNA expression and secretion. NMN was able to normalize intracellular NAD levels but did not ameliorate cell viability after co-stimulation with palmitate. FK866, a specific NAMPT inhibitor did not influence lipid accumulation after oleate-treatment. CONCLUSIONS: Palmitate targets NAMPT activity with a consequent cellular depletion of NAD. Oleate protects from palmitate-induced apoptosis and variation of NAMPT and NAD levels. Palmitate-induced cell stress leads to an increase of NAMPT mRNA and accumulation in the supernatant. However, the proapoptotic action of palmitate seems not to be mediated by decreased NAD levels.


Asunto(s)
Citocinas/genética , Hepatocitos/efectos de los fármacos , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Ácido Oléico/farmacología , Ácido Palmítico/farmacología , Acrilamidas/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , Expresión Génica , Células Hep G2 , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , NAD/antagonistas & inhibidores , Mononucleótido de Nicotinamida/farmacología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/metabolismo , Ácido Palmítico/antagonistas & inhibidores , Piperidinas/farmacología , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Biochim Biophys Acta ; 1834(12): 2520-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24035885

RESUMEN

Schistosoma mansoni NAD(+) catabolizing enzyme (SmNACE), a distant homolog of mammalian CD38, shows significant structural and functional analogy to the members of the CD38/ADP-ribosyl cyclase family. The hallmark of SmNACE is the lack of ADP-ribosyl cyclase activity that might be ascribed to subtle changes in its active site. To better characterize the residues of the active site we determined the kinetic parameters of nine mutants encompassing three acidic residues: (i) the putative catalytic residue Glu202 and (ii) two acidic residues within the 'signature' region (the conserved Glu124 and the downstream Asp133), (iii) Ser169, a strictly conserved polar residue and (iv) two aromatic residues (His103 and Trp165). We established the very important role of Glu202 and of the hydrophobic domains overwhelmingly in the efficiency of the nicotinamide-ribosyl bond cleavage step. We also demonstrated that in sharp contrast with mammalian CD38, the 'signature' Glu124 is as critical as Glu202 for catalysis by the parasite enzyme. The different environments of the two Glu residues in the crystal structure of CD38 and in the homology model of SmNACE could explain such functional discrepancies. Mutagenesis data and 3D structures also indicated the importance of aromatic residues, especially His103, in the stabilization of the reaction intermediate as well as in the selection of its conformation suitable for cyclization to cyclic ADP-ribose. Finally, we showed that inhibition of SmNACE by the natural product cyanidin requires the integrity of Glu202 and Glu124, but not of His103 and Trp165, hence suggesting different recognition modes for substrate and inhibitor.


Asunto(s)
Proteínas del Helminto/química , Hidrolasas/química , Schistosoma mansoni/enzimología , ADP-Ribosil Ciclasa 1/química , ADP-Ribosil Ciclasa 1/genética , Aminoácidos , Animales , Antocianinas/química , Dominio Catalítico , Proteínas del Helminto/genética , Humanos , Hidrolasas/antagonistas & inhibidores , Hidrolasas/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , NAD/antagonistas & inhibidores , NAD/química , NAD/genética , Estructura Terciaria de Proteína , Schistosoma mansoni/genética , Homología Estructural de Proteína
4.
Apoptosis ; 19(4): 581-93, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24292505

RESUMEN

Regulation of neutrophil apoptosis plays a critical role in the inflammatory response. Inflammation has previously been shown to increase levels of extracellular ß-nicotinamide adenine dinucleotide (NAD(+)). The present study demonstrates that extracellular NAD(+) at concentrations found in the inflamed tissues profoundly delays spontaneous apoptosis of human neutrophils as was evidenced by inhibition of phosphatidylserine (PS) exposure, DNA fragmentation and caspase-3 activation. The effect was abrogated by NF157, an antagonist of P2Y11 receptor, and was pertussis toxin-insensitive. The NAD(+)-mediated delay of neutrophil apoptosis was reversed by 2',5'-dideoxyadenosine, an inhibitor of adenylyl cyclase, and Rp-8-Br-cAMPS, an inhibitor of type I cAMP-dependent protein kinase A (PKA). Blocking of NAD(+)-induced influx of extracellular Ca(2+) with EGTA did not abolish the pro-survival effect of NAD(+). Extracellular NAD(+) inhibited proteasome-dependent degradation of Mcl-1 upstream of caspase activation and, furthermore, suppressed Bax translocation to the mitochondria and attenuated both dissipation of mitochondrial transmembrane potential (ΔΨm) and cytochrome c release from the mitochondria into the cytosol. Finally, we found that extracellular NAD(+) inhibited spontaneous activation of caspase-9, but not caspase-8, and the pro-survival effect of extracellular NAD(+) was abrogated by the inhibitor of caspase-9, but not by the inhibitor of caspase-8. Together, these results demonstrate that extracellular NAD(+) inhibits neutrophil apoptosis via P2Y11 receptor and cAMP/PKA pathway by regulating Mcl-1 level, Bax targeting to the mitochondria and mitochondrial apoptotic pathway. Thus, extracellular NAD(+) acts as a neutrophil survival factor that can contribute to prolonged neutrophil lifespan in inflammatory response.


Asunto(s)
Apoptosis , NAD/metabolismo , Neutrófilos/citología , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Espacio Extracelular/enzimología , Humanos , Inflamación/patología , Mitocondrias/fisiología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , NAD/antagonistas & inhibidores , Neutrófilos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores Purinérgicos P2/metabolismo , Transducción de Señal
5.
Blood ; 120(17): 3519-29, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22955917

RESUMEN

Malignant cells have a higher nicotinamide adenine dinucleotide (NAD(+)) turnover rate than normal cells, making this biosynthetic pathway an attractive target for cancer treatment. Here we investigated the biologic role of a rate-limiting enzyme involved in NAD(+) synthesis, Nampt, in multiple myeloma (MM). Nampt-specific chemical inhibitor FK866 triggered cytotoxicity in MM cell lines and patient MM cells, but not normal donor as well as MM patients PBMCs. Importantly, FK866 in a dose-dependent fashion triggered cytotoxicity in MM cells resistant to conventional and novel anti-MM therapies and overcomes the protective effects of cytokines (IL-6, IGF-1) and bone marrow stromal cells. Nampt knockdown by RNAi confirmed its pivotal role in maintenance of both MM cell viability and intracellular NAD(+) stores. Interestingly, cytotoxicity of FK866 triggered autophagy, but not apoptosis. A transcriptional-dependent (TFEB) and independent (PI3K/mTORC1) activation of autophagy mediated FK866 MM cytotoxicity. Finally, FK866 demonstrated significant anti-MM activity in a xenograft-murine MM model, associated with down-regulation of ERK1/2 phosphorylation and proteolytic cleavage of LC3 in tumor cells. Our data therefore define a key role of Nampt in MM biology, providing the basis for a novel targeted therapeutic approach.


Asunto(s)
Acrilamidas/farmacología , Antineoplásicos/farmacología , Citocinas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Piperidinas/farmacología , Proteínas/antagonistas & inhibidores , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Línea Celular Tumoral , Supervivencia Celular , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Terapia Molecular Dirigida , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Complejos Multiproteicos , NAD/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Especificidad de Órganos , Proteínas/genética , Proteínas/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioorg Med Chem Lett ; 24(2): 630-5, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24360556

RESUMEN

The structure-activity relationship (SAR) study of two chemotypes identified as inhibitors of the human NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (HPGD, 15-PGDH) was conducted. Top compounds from both series displayed potent inhibition (IC50 <50 nM), demonstrate excellent selectivity towards HPGD and potently induce PGE2 production in A549 lung cancer and LNCaP prostate cancer cells.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , NAD/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , NAD/metabolismo , Relación Estructura-Actividad
7.
Chem Commun (Camb) ; 57(34): 4162-4165, 2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33908442

RESUMEN

Two mitochondria-localized Ru(ii) complexes with photo-labile ligands were reported to exert one- and two-photon activatable anticancer activity through a dual-function mechanism, i.e. mitochondrial DNA covalent binding after photo-induced ligand dissociation and photo-catalyzed NADH depletion, thus displaying good activity towards cisplatin-resistant cancer cells under both normoxic and hypoxic conditions.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , ADN Mitocondrial/efectos de los fármacos , NAD/antagonistas & inhibidores , Dióxido de Nitrógeno/metabolismo , Rutenio/farmacología , Células A549 , Antineoplásicos/química , Antineoplásicos/metabolismo , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Daño del ADN , ADN Mitocondrial/metabolismo , Humanos , Ligandos , Estructura Molecular , NAD/análisis , NAD/metabolismo , Procesos Fotoquímicos , Fotones , Rutenio/química , Rutenio/metabolismo
8.
Int J Cancer ; 126(12): 2773-89, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20112275

RESUMEN

High-risk neuroblastoma is a rapidly growing tumor with a survival rate below 50%. A new treatment strategy is to administer chemotherapeutic drugs metronomically, i.e., at lower doses and frequent intervals. The aim of the study was to investigate the effects of GMX1777, a chemotherapeutic drug affecting cellular energy metabolism, in a mouse model for high-risk neuroblastoma. Female SCID mice were injected s.c. with MYCN-amplified human neuroblastoma cells and randomized to either treatment with GMX1777 or vehicle. In some animals, treatment was discontinued allowing tumor relapse. Treatment response was evaluated using the pediatric preclinical testing program (PPTP). Immunohistochemistry and qRT-PCR was performed on tumor cryosections to investigate the microscopic and molecular changes in tumors in response to GMX1777. Despite an increase in vessel density, tumor regression and a high group response score according to PPTP criteria was induced by GMX1777 without inducing drug resistance. Treatment resulted in inhibition of tumor cell proliferation, vessel maturation, reduced hypoxia, increased infiltration of MHC class II negative macrophages and expansion of the nonvascular stromal compartment. Decreased stromal VEGF-A and PDGF-B mRNA in response to treatment together with the structural data suggest a "deactivation" or "silencing" of the tumor stroma as a paracrine entity. In conclusion, GMX1777 was highly efficient against high-risk neuroblastoma xenografts through modulation of both the tumor cell and stromal compartment.


Asunto(s)
Resistencia a Antineoplásicos , Guanidinas/administración & dosificación , NAD/antagonistas & inhibidores , Neovascularización Patológica , Neuroblastoma/patología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Immunoblotting , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/irrigación sanguínea , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Trends Cancer ; 6(1): 9-12, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31952784

RESUMEN

Antibodies targeting CD38, a NAD+-degrading enzyme, have emerged as a promising immunotherapy against multiple myeloma (MM). Currently, the mechanisms by which anti-CD38 antibodies establish their therapeutic effects are poorly understood. Here, we advocate for the depletion of NAD+ to enhance the efficacy of anti-CD38-based immunotherapies in MM.


Asunto(s)
ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Citocinas/antagonistas & inhibidores , Glicoproteínas de Membrana/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , NAD/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , ADP-Ribosil Ciclasa 1/metabolismo , Acrilamidas/farmacología , Acrilamidas/uso terapéutico , Adenosina/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Citocinas/metabolismo , Sinergismo Farmacológico , Humanos , Glicoproteínas de Membrana/metabolismo , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , NAD/metabolismo , Niacinamida/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Piperidinas/farmacología , Piperidinas/uso terapéutico , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Escape del Tumor/efectos de los fármacos , Efecto Warburg en Oncología/efectos de los fármacos
10.
Biochemistry ; 48(50): 12005-13, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19899808

RESUMEN

NADH:ubiquinone oxidoreductase (complex I) is a complicated respiratory chain enzyme that conserves the energy from NADH oxidation, coupled to ubiquinone reduction, as a proton motive force across the mitochondrial inner membrane. Alternatively, NADH oxidation, by the flavin mononucleotide in complex I, can be coupled to the reduction of hydrophilic electron acceptors, in non-energy-transducing reactions. The reduction of molecular oxygen and hydrophilic quinones leads to the production of reactive oxygen species, the reduction of nicotinamide nucleotides leads to transhydrogenation, and "artificial" electron acceptors are widely used to study the mechanism of NADH oxidation. Here, we use a combined modeling strategy to accurately describe data from three flavin-linked electron acceptors (molecular oxygen, APAD(+), and ferricyanide), in the presence and absence of a competitive inhibitor, ADP-ribose. Our combined ping-pong (or ping-pong-pong) mechanism comprises the Michaelis-Menten equation for the reactions of NADH and APAD(+), simple dissociation constants for nonproductive nucleotide-enzyme complexes (defined for specific flavin oxidation states), and second-order rate constants for the reactions of ferricyanide and oxygen. The NADH-dependent parameters are independent of the identity of the electron acceptor. In contrast, a further flavin-linked acceptor, hexaammineruthenium(III), does not obey ping-pong-pong kinetics, and alternative sites for its reaction are discussed. Our analysis provides kinetic and thermodynamic information about the reactions of the flavin active site in complex I that is relevant to understanding the physiologically relevant mechanisms of NADH oxidation and superoxide formation.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Ferricianuros/metabolismo , Mononucleótido de Flavina/química , Mononucleótido de Flavina/metabolismo , NAD/análogos & derivados , NAD/metabolismo , Oxígeno/metabolismo , Animales , Unión Competitiva , Catálisis , Bovinos , Ferricianuros/antagonistas & inhibidores , Cinética , NAD/antagonistas & inhibidores , NAD/química , Oxidación-Reducción , Oxígeno/antagonistas & inhibidores , Unión Proteica , Compuestos de Rutenio/química
11.
J Cell Biol ; 51(3): 664-73, 1971 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-4331501

RESUMEN

The sensitivity of nicotinamide adenine dinucleotide (NADH) oxidase and succinoxidase to metal chelators, the generation of an electron paramagnetic resonance (EPR) signal upon addition of these substrates, and the rate of formation of the EPR signal relative to the rate of the cytochrome reduction suggest the participation of nonheme iron proteins in the respiratory process of Escherichia coli. The most inhibitory metal chelator, thenoyltrifluoro acetone, inhibited the reduction of nonheme iron and cytochromes but did not prevent the reoxidation of the reduced forms. The EPR signal, dehydrogenase, and oxidase activities evoked by NADH are considerably greater than the corresponding activities evoked by succinate. Because both substrates can reduce almost all of the cytochromes, a model in which fewer succinate dehydrogenase-nonheme iron protein complexes are linked to a common cytochrome chain than NADH dehydrogenase-nonheme iron protein complexes is considered likely.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Membrana Celular/metabolismo , Citocromos/metabolismo , Hierro/metabolismo , NAD/metabolismo , Succinatos/metabolismo , Acetona/farmacología , Butanonas/farmacología , Quelantes/metabolismo , Quelantes/farmacología , Citocromos/análisis , Citocromos/antagonistas & inhibidores , Espectroscopía de Resonancia por Spin del Electrón , Escherichia coli , Flúor/farmacología , Hierro/antagonistas & inhibidores , NAD/antagonistas & inhibidores , Oxidación-Reducción , Oxidorreductasas/metabolismo , Consumo de Oxígeno , Albúmina Sérica Bovina/farmacología , Espectrofotometría , Succinato Deshidrogenasa/antagonistas & inhibidores , Succinato Deshidrogenasa/metabolismo , Tiofenos/farmacología
12.
ACS Chem Biol ; 14(5): 949-958, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30969758

RESUMEN

Conventional treatments to combat the tuberculosis (TB) epidemic are falling short, thus encouraging the search for novel antitubercular drugs acting on unexplored molecular targets. Several whole-cell phenotypic screenings have delivered bioactive compounds with potent antitubercular activity. However, their cellular target and mechanism of action remain largely unknown. Further evaluation of these compounds may include their screening in search for known antitubercular drug targets hits. Here, a collection of nearly 1400 mycobactericidal compounds was screened against Mycobacterium tuberculosis NaMN adenylyltransferase ( MtNadD), a key enzyme in the biogenesis of NAD cofactor that was recently validated as a new drug target for dormant and active tuberculosis. We found three chemotypes that efficiently inhibit MtNadD in the low micromolar range in vitro. SAR and cheminformatics studies of commercially available analogues point to a series of benzimidazolium derivatives, here named N2, with bactericidal activity on different mycobacteria, including M. abscessus, multidrug-resistant M. tuberculosis, and dormant M. smegmatis. The on-target activity was supported by the increased resistance of an M. smegmatis strain overexpressing the target and by a rapid decline in NAD(H) levels. A cocrystal structure of MtNadD with N2-8 inhibitor reveals that the binding of the inhibitor induced the formation of a new quaternary structure, a dimer-of-dimers where two copies of the inhibitor occupy symmetrical positions in the dimer interface, thus paving the way for the development of a new generation of selective MtNadD bioactive inhibitors. All these results strongly suggest that pharmacological inhibition of MtNadD is an effective strategy to combat dormant and resistant Mtb strains.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , NAD/antagonistas & inhibidores , Nicotinamida-Nucleótido Adenililtransferasa/antagonistas & inhibidores , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , NAD/biosíntesis , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Relación Estructura-Actividad
13.
Antioxid Redox Signal ; 30(2): 251-294, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29634344

RESUMEN

Significance: Nicotinamide adenine dinucleotide (NAD+) is an essential pyridine nucleotide that serves as an essential cofactor and substrate for a number of critical cellular processes involved in oxidative phosphorylation and ATP production, DNA repair, epigenetically modulated gene expression, intracellular calcium signaling, and immunological functions. NAD+ depletion may occur in response to either excessive DNA damage due to free radical or ultraviolet attack, resulting in significant poly(ADP-ribose) polymerase (PARP) activation and a high turnover and subsequent depletion of NAD+, and/or chronic immune activation and inflammatory cytokine production resulting in accelerated CD38 activity and decline in NAD+ levels. Recent studies have shown that enhancing NAD+ levels can profoundly reduce oxidative cell damage in catabolic tissue, including the brain. Therefore, promotion of intracellular NAD+ anabolism represents a promising therapeutic strategy for age-associated degenerative diseases in general, and is essential to the effective realization of multiple benefits of healthy sirtuin activity. The kynurenine pathway represents the de novo NAD+ synthesis pathway in mammalian cells. NAD+ can also be produced by the NAD+ salvage pathway. Recent Advances: In this review, we describe and discuss recent insights regarding the efficacy and benefits of the NAD+ precursors, nicotinamide (NAM), nicotinic acid (NA), nicotinamide riboside (NR), and nicotinamide mononucleotide (NMN), in attenuating NAD+ decline in degenerative disease states and physiological aging. Critical Issues: Results obtained in recent years have shown that NAD+ precursors can play important protective roles in several diseases. However, in some cases, these precursors may vary in their ability to enhance NAD+ synthesis via their location in the NAD+ anabolic pathway. Increased synthesis of NAD+ promotes protective cell responses, further demonstrating that NAD+ is a regulatory molecule associated with several biochemical pathways. Future Directions: In the next few years, the refinement of personalized therapy for the use of NAD+ precursors and improved detection methodologies allowing the administration of specific NAD+ precursors in the context of patients' NAD+ levels will lead to a better understanding of the therapeutic role of NAD+ precursors in human diseases.


Asunto(s)
Envejecimiento/metabolismo , Biomarcadores , NAD/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Animales , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Terapia Molecular Dirigida , NAD/antagonistas & inhibidores , Enfermedades Neurodegenerativas/tratamiento farmacológico , Oxidación-Reducción , Transducción de Señal
14.
Nat Chem ; 11(11): 1041-1048, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31548671

RESUMEN

Hypoxic tumours are a major problem for cancer photodynamic therapy. Here, we show that photoredox catalysis can provide an oxygen-independent mechanism of action to combat this problem. We have designed a highly oxidative Ir(III) photocatalyst, [Ir(ttpy)(pq)Cl]PF6 ([1]PF6, where 'ttpy' represents 4'-(p-tolyl)-2,2':6',2''-terpyridine and 'pq' represents 3-phenylisoquinoline), which is phototoxic towards both normoxic and hypoxic cancer cells. Complex 1 photocatalytically oxidizes 1,4-dihydronicotinamide adenine dinucleotide (NADH)-an important coenzyme in living cells-generating NAD• radicals with a high turnover frequency in biological media. Moreover, complex 1 and NADH synergistically photoreduce cytochrome c under hypoxia. Density functional theory calculations reveal π stacking in adducts of complex 1 and NADH, facilitating photoinduced single-electron transfer. In cancer cells, complex 1 localizes in mitochondria and disrupts electron transport via NADH photocatalysis. On light irradiation, complex 1 induces NADH depletion, intracellular redox imbalance and immunogenic apoptotic cancer cell death. This photocatalytic redox imbalance strategy offers a new approach for efficient cancer phototherapy.


Asunto(s)
Citocromos c/metabolismo , Neoplasias/metabolismo , Hipoxia Tumoral , Antineoplásicos/química , Antineoplásicos/farmacología , Biocatálisis , Muerte Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Teoría Funcional de la Densidad , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Moleculares , Conformación Molecular , NAD/antagonistas & inhibidores , NAD/metabolismo , Neoplasias/patología , Oxidación-Reducción , Procesos Fotoquímicos , Hipoxia Tumoral/efectos de los fármacos
15.
Mol Med Rep ; 20(6): 5163-5171, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31702813

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease of the central nervous system that is characterized by progressive cognitive dysfunction and which ultimately leads to dementia. Studies have shown that energy dysmetabolism contributes significantly to the pathogenesis of a variety of aging­associated diseases and degenerative diseases of the nervous system, including AD. One focus of research thus has been how to regulate the expression of nicotinamide phosphoribosyltransferase (NAMPT) to prevent against neurodegenerative diseases. Therefore, the present study used 6­month­old APPswe/PS1ΔE9 (APP/PS1) transgenic mice as early AD mouse models and sought to evaluate nicotinamide adenine dinucleotide (NAD+) and FK866 (a NAMPT inhibitor) treatment in APP/PS1 mice to study NAMPT dysmetabolism in the process of AD and elucidate the underlying mechanisms. As a result of this treatment, the expression of NAMPT decreased, the synthesis of ATP and NAD+ became insufficient and the NAD+/NADH ratio was reduced. The administration of NAD+ alleviated the spatial learning and memory of APP/PS1 mice and reduced senile plaques. Administration of NAD+ may also increase the expression of the key protein NAMPT and its related protein sirtuin 1 as well as the synthesis of NAD+. Therefore, increasing NAMPT expression levels may promote NAD+ production. Their regulation could form the basis for a new therapeutic strategy.


Asunto(s)
Acrilamidas/antagonistas & inhibidores , Enfermedad de Alzheimer/metabolismo , Citocinas/efectos de los fármacos , Citocinas/metabolismo , NAD/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/efectos de los fármacos , Nicotinamida Fosforribosiltransferasa/metabolismo , Piperidinas/antagonistas & inhibidores , Transducción de Señal/fisiología , Acrilamidas/farmacología , Amiloide/metabolismo , Animales , Conducta Animal , Citocinas/genética , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NAD/metabolismo , NAD/farmacología , Nicotinamida Fosforribosiltransferasa/genética , Piperidinas/farmacología , Sirtuina 1/metabolismo
16.
Biol Res Nurs ; 9(4): 311-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18398226

RESUMEN

Treatment advances, including central nervous system (CNS) treatment with methotrexate, have led to significant gains in disease-free survival from childhood acute lymphoblastic leukemia (ALL). However, methotrexate has been associated with neurological problems such as declines in cognitive and academic abilities. The purpose of this study was to investigate methotrexate-induced changes in beta-oxidation in children with ALL receiving methotrexate for CNS treatment. Specific aims were to investigate effects of methotrexate on beta-oxidation of the two most prevalent fatty acids (palmitic acid and stearic acid) in cerebrospinal fluid (CSF) samples and correlate the ratio of monounsaturation to saturation of these fatty acids with cognitive and academic abilities. The sample included 12 females and 14 males with low-risk (n = 7), standard-risk ( n = 13), or high-risk (n = 6) ALL. Mean age at diagnosis was 94.1 months (SD = 34.4). CSF samples were obtained in conjunction with diagnostic lumbar punctures; subsequent samples were obtained prior to intrathecal methotrexate administration during the induction, consolidation, and continuation phases of treatment. Fatty acids were analyzed by gas chromatography. Results showed a significant increase in the ratio of monounsaturation to saturation of both fatty acids, which was greatest during the most intensive phase of treatment. Ratios of monounsaturated to saturated fatty acids were negatively correlated with full-scale IQ, verbal IQ, and math calculations. Findings suggest that methotrexate alters beta-oxidation and that the resulting increase in fatty acid monounsaturation is related to declines in some domains of cognitive ability.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Trastornos del Conocimiento/inducido químicamente , Metotrexato/efectos adversos , Ácido Palmítico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Ácidos Esteáricos , Análisis de Varianza , Niño , Cromatografía de Gases , Trastornos del Conocimiento/líquido cefalorraquídeo , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/metabolismo , Femenino , Humanos , Pruebas de Inteligencia , Masculino , NAD/antagonistas & inhibidores , NAD/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Ácido Palmítico/líquido cefalorraquídeo , Ácido Palmítico/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Desempeño Psicomotor , Factores de Riesgo , Estadísticas no Paramétricas , Ácidos Esteáricos/líquido cefalorraquídeo , Ácidos Esteáricos/metabolismo
17.
Org Lett ; 20(23): 7650-7655, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30479128

RESUMEN

We report the chemical synthesis of a set of nicotinamide adenine dinucleotide (NAD) cap analogues containing chemical modifications that reduce their susceptibility to NAD-RNA-degrading enzymes. These analogues can be incorporated into transcripts in a similar way as NAD. Biochemical characterization of RNAs carrying these caps with DXO, NudC, and Nudt12 enzymes led to the identification of compounds that can be instrumental in unraveling so far unaddressed biological aspects of NAD-RNAs.


Asunto(s)
Adenina/farmacología , NAD/antagonistas & inhibidores , Niacinamida/farmacología , Caperuzas de ARN/antagonistas & inhibidores , Adenina/análogos & derivados , Adenina/química , Conformación Molecular , NAD/metabolismo , Niacinamida/análogos & derivados , Niacinamida/química , Caperuzas de ARN/metabolismo
18.
Trends Pharmacol Sci ; 39(4): 424-436, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29482842

RESUMEN

Recent reports indicate that intracellular NAD levels decline in tissues during chronological aging, and that therapies aimed at increasing cellular NAD levels could have beneficial effects in many age-related diseases. The protein CD38 (cluster of differentiation 38) is a multifunctional enzyme that degrades NAD and modulates cellular NAD homeostasis. At the physiological level, CD38 has been implicated in the regulation of metabolism and in the pathogenesis of multiple conditions including aging, obesity, diabetes, heart disease, asthma, and inflammation. Interestingly, many of these functions are mediated by CD38 enzymatic activity. In addition, CD38 has also been identified as a cell-surface marker in hematologic cancers such as multiple myeloma, and a cytotoxic anti-CD38 antibody has been approved by the FDA for use in this disease. Although this is a remarkable development, killing CD38-positive tumor cells with cytotoxic anti-CD38 antibodies is only one of the potential pharmacological uses of targeting CD38. The present review discusses the biology of the CD38 enzyme and the current state of development of pharmacological tools aimed at CD38, and explores how these agents may represent a novel approach for treating human conditions including cancer, metabolic disease, and diseases of aging.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , NAD+ Nucleosidasa/metabolismo , Neoplasias/terapia , ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , ADP-Ribosil Ciclasa 1/inmunología , Envejecimiento/fisiología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Humanos , Terapia Molecular Dirigida , NAD/antagonistas & inhibidores , NAD/metabolismo , NAD+ Nucleosidasa/antagonistas & inhibidores , Neoplasias/enzimología , Neoplasias/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología
19.
Mol Cancer Res ; 15(12): 1714-1721, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28860121

RESUMEN

Nicotinamide phosphoribosyltransferase (NAMPT) and nicotinic acid phosphoribosyltransferase (NAPRT) are rate-limiting enzymes in the NAD+ synthesis pathway. Chondrosarcoma is a malignant cartilage forming bone tumor, in which mutations altering isocitrate dehydrogenase-1 and -2 (IDH1 and IDH2) activity have been identified as potential driver mutations. Vulnerability for NAD+ depletion has been reported for IDH1/2-mutant cells. Here, the potency of NAMPT inhibitors as a treatment of chondrosarcoma was explored. Eleven chondrosarcoma cell lines were treated with NAMPT inhibitors, in which the effect on cell viability, colony formation, and 3D collagen invasion was assessed. The expression level of NAMPT and NAPRT transcripts in chondrosarcoma cells was determined by qRT-PCR. Methylation of the NAPRT promoter was evaluated using a previously published dataset of genome-wide methylation. In addition, a methylation dataset was used to determine methylation of the NAPRT promoter in 20 IDH1/2-mutated cartilage tumors. Chondrosarcoma cells showed a dose-dependent decrease in cell viability, 3D collagen invasion, and colony formation upon treatment with NAMPT inhibitors, in which nearly half of the cell lines demonstrated absolute IC50s in the low nanomolar range. Increasing IC50s correlated to increasing NAPRT expression levels and decreasing NAPRT promoter methylation. No correlation between IDH1/2 mutation status and sensitivity for NAMPT inhibitors was observed. Strikingly, higher methylation of the NAPRT promoter was observed in high-grade versus low-grade chondrosarcomas. In conclusion, this study identified NAMPT as a potential target for treatment of chondrosarcoma.Implications: Chondrosarcoma patients, especially those of high histologic grade with lower expression and hypermethylation of NAPRT, may benefit from inhibition of the NAD synthesis pathway. Mol Cancer Res; 15(12); 1714-21. ©2017 AACR.


Asunto(s)
Condrosarcoma/genética , Citocinas/genética , Isocitrato Deshidrogenasa/genética , Neoplasias de Tejido Óseo/genética , Nicotinamida Fosforribosiltransferasa/genética , Pentosiltransferasa/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Condrosarcoma/tratamiento farmacológico , Condrosarcoma/patología , Citocinas/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mutación , NAD/antagonistas & inhibidores , NAD/biosíntesis , NAD/genética , Invasividad Neoplásica/genética , Neoplasias de Tejido Óseo/tratamiento farmacológico , Neoplasias de Tejido Óseo/patología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Pentosiltransferasa/antagonistas & inhibidores , Regiones Promotoras Genéticas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
20.
Eur J Med Res ; 11(8): 313-21, 2006 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-17052966

RESUMEN

FK866 is a novel anticancer agent that was previously shown to interfere with NAD superset+ biosynthesis by inhibition of nicotinamide phosphoribosyltransferase and to initiate apoptosis in cancer cells. As NAD superset+ is involved in cellular DNA repair processes, the present in vitro study on THP-1 and K562 leukemia cells was conducted to investigate the cytotoxicity of FK866 combination treatment with various cytotoxic agents: the antimetabolite Ara-C, the DNA-intercalating agent daunorubicin and the alkylating compounds 1-methyl-3-nitro-1-nitrosoguanidinium (MNNG) and melphalan. Cell viability after drug exposure was assessed by propidium iodide (PI) staining. Non-cytotoxic concentrations of FK866 (10 superset-9 M or less), applied simultaneously or 24 hours before adding cytotoxic agents, caused a depletion in the intracellular NAD superset+ and--to a lesser extent-- NADH levels in THP-1 cells. After 48 and 72 hours treatment with daunorubicin and Ara-C, respectively, increased cell death was observed in THP-1 cells that were pretreated with FK866, as compared to cells exposed to antineoplastic drugs alone. However, this effect was transient, and there was no difference in cell survival after 72 hours incubation with daunorubicin or 96 hours with Ara-C. - Non-toxic concentrations of FK866 added 8, 16, or 24 hours before starting treatment with the PARP-activating agent MNNG synergistically decreased intracellular NAD superset+ contents, and increased MNNG-induced cytotoxicity both in THP-1 and K562 cells for at least 72 hours. This effect was less pronounced when FK866 was used in combination with another alkylating agent, melphalan. The PARP inhibitor 3-aminobenzamide delayed MNNG-induced cytotoxicity by 24 hours both in cells that were pretreated with FK866 and in non-pretreated cells. 48 hours later, the protective effect of 3-aminobenzamide could no longer be observed, but FK866-pretreated cells retained increased sensitivity to MNNG. - In conclusion, the chemosensitizing effect of FK866 on cell death induced by antineoplastic drugs was particularly obvious in combination with substances like MNNG that cause NAD superset+ depletion per se. It was less pronounced and only transiently measurable in combination with daunorubicin, Ara-C, and melphalan, respectively. These results may indicate different levels of DNA damage implicated in the action of the cytotoxic agents used.


Asunto(s)
Acrilamidas/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Monocitos/efectos de los fármacos , NAD/antagonistas & inhibidores , Piperidinas/farmacología , Antibióticos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Antineoplásicos Alquilantes/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citarabina/farmacología , Daunorrubicina/farmacología , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Melfalán/farmacología , Metilnitronitrosoguanidina/farmacología , Monocitos/citología , NAD/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA