Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(26): e2122897119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35700355

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolves rapidly under the pressure of host immunity, as evidenced by waves of emerging variants despite effective vaccinations, highlighting the need for complementing antivirals. We report that targeting a pyrimidine synthesis enzyme restores inflammatory response and depletes the nucleotide pool to impede SARS-CoV-2 infection. SARS-CoV-2 deploys Nsp9 to activate carbamoyl-phosphate synthetase, aspartate transcarbamoylase, and dihydroorotase (CAD) that catalyzes the rate-limiting steps of the de novo pyrimidine synthesis. Activated CAD not only fuels de novo nucleotide synthesis but also deamidates RelA. While RelA deamidation shuts down NF-κB activation and subsequent inflammatory response, it up-regulates key glycolytic enzymes to promote aerobic glycolysis that provides metabolites for de novo nucleotide synthesis. A newly synthesized small-molecule inhibitor of CAD restores antiviral inflammatory response and depletes the pyrimidine pool, thus effectively impeding SARS-CoV-2 replication. Targeting an essential cellular metabolic enzyme thus offers an antiviral strategy that would be more refractory to SARS-CoV-2 genetic changes.


Asunto(s)
Antivirales , Aspartato Carbamoiltransferasa , Tratamiento Farmacológico de COVID-19 , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante) , Dihidroorotasa , Inhibidores Enzimáticos , Pirimidinas , SARS-CoV-2 , Replicación Viral , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Aspartato Carbamoiltransferasa/antagonistas & inhibidores , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/antagonistas & inhibidores , Dihidroorotasa/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Ratones , Pirimidinas/antagonistas & inhibidores , Pirimidinas/biosíntesis , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Factor de Transcripción ReIA/metabolismo , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
2.
Invest New Drugs ; 39(2): 348-361, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32978717

RESUMEN

Multiple myeloma (MM) is a devastating disease with low survival rates worldwide. The mean lifetime of patients may be extendable with new drug alternatives. Aurora A kinase (AURKA) is crucial in oncogenesis, because its overexpression or amplification may incline the development of various types of cancer, including MM. Therefore, inhibitors of AURKA are innovative and promising targets. Natural compounds always represented a valuable resource for anticancer drug development. In the present study, based on virtual drug screening of more than 48,000 natural compounds, the antibiotic deschloro-chlorotricin (DCCT) has been identified to bind to AURKA with even higher binding affinity (free bindung energy: -12.25 kcal/mol) than the known AURKA inhibitor, alisertib (free binding energy: -11.25 kcal/mol). The in silico studies have been verified in vitro by using microscale thermophoresis. DCCT inhibited MM cell lines (KMS-11, L-363, RPMI-8226, MOLP-8, OPM-2, NCI-H929) with IC50 values in a range from 0.01 to 0.12 µM. Furthermore, DCCT downregulated AURKA protein expression, induced G2/M cell cycle arrest and disturbed the cellular microtubule network as determined by Western blotting, flow cytometry, and fluorescence microscopy. Thus, DCCT may be a promising lead structure for further derivatization and the development of specific AURKA inhibitors in MM therapy.


Asunto(s)
Aminoglicósidos/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Azepinas/antagonistas & inhibidores , Azepinas/farmacología , Ciclo Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Unión Proteica/fisiología , Pirimidinas/antagonistas & inhibidores , Pirimidinas/farmacología
3.
Nat Chem Biol ; 15(3): 250-258, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30643284

RESUMEN

Irreversible inhibition of disease-associated proteins with small molecules is a powerful approach for achieving increased and sustained pharmacological potency. Here, we introduce α-chlorofluoroacetamide (CFA) as a novel warhead of targeted covalent inhibitor (TCI). Despite weak intrinsic reactivity, CFA-appended quinazoline showed high reactivity toward Cys797 of epidermal growth factor receptor (EGFR). In cells, CFA-quinazoline showed higher target specificity for EGFR than the corresponding Michael acceptors in a wide concentration range (0.1-10 µM). The cysteine adduct of the CFA derivative was susceptible to hydrolysis and reversibly yielded intact thiol but was stable in solvent-sequestered ATP-binding pocket of EGFR. This environment-dependent hydrolysis can potentially reduce off-target protein modification by CFA-based drugs. Oral administration of CFA quinazoline NS-062 significantly suppressed tumor growth in a mouse xenograft model. Further, CFA-appended pyrazolopyrimidine irreversibly inhibited Bruton's tyrosine kinase with higher target specificity. These results demonstrate the utility of CFA as a new class warheads for TCI.


Asunto(s)
Acetamidas/síntesis química , Cisteína/metabolismo , Quinazolinas/síntesis química , Acetamidas/química , Acetamidas/farmacología , Animales , Antineoplásicos , Línea Celular , Receptores ErbB , Humanos , Ratones , Ratones Desnudos , Neoplasias , Fosfotransferasas/fisiología , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/antagonistas & inhibidores , Quinazolinas/química , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Recept Signal Transduct Res ; 39(2): 167-174, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31354087

RESUMEN

2,4-Disubstituted pyridopyrimidine derivatives were studied against ABCG2 enzyme. The modeling of pyridopyrimidine derivatives were done using two methods of multiple linear regression and support vector regression and four molecular descriptors of BIC4, log p, VRA2, and binding energy were selected for modeling. The statistical results were satisfactory. The interactions of ABCG2 enzyme with pyridopyrimidine derivatives were investigated using molecular docking method. Based on the results, increasing of binding energy and hydrophobicity of the compounds increase their inhibitory activity. Protein stability in complex with pharmaceutical derivatives was discussed using molecular dynamics simulation method.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Estabilidad Proteica/efectos de los fármacos , Pirimidinas/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Sitios de Unión/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Pirimidinas/antagonistas & inhibidores , Pirimidinas/farmacología , Relación Estructura-Actividad Cuantitativa
5.
Int J Mol Sci ; 20(14)2019 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-31340581

RESUMEN

This study was designed to investigate the protective effect of resveratrol (RES) on premature ovarian failure (POF) and the proliferation of female germline stem cells (FGSCs) at the tissue and cell levels. POF mice were lavaged with RES, and POF ovaries were co-cultured with RES and/or GANT61 in vitro. FGSCs were pretreated with Busulfan and RES and/or GANT61 and co-cultured with M1 macrophages, which were pretreated with RES. The weights of mice and their ovaries, as well as their follicle number, were measured. Ovarian function, antioxidative stress, inflammation, and FGSCs survival were evaluated. RES significantly increased the weights of POF mice and their ovaries as well as the number of follicles, while it decreased the atresia rate of follicles. Higher levels of Mvh, Oct4, SOD2, GPx, and CAT were detected after treatment with RES in vivo and in vitro. RES treatment resulted in significantly lower TNF-α and IL-6 concentrations and an obviously higher IL-10 concentration in the ovaries. In FGSCs, higher Mvh, Oct4, and SOD2 concentrations and lower TNF-α, IL-6, and MDA concentrations were measured in the RES group. Blockage of the Hh signaling pathway reversed the protective effect of RES on FGSCs. In conclusion, RES effectively improved the ovarian function of the POF model and the productive capacity of FGSCs via relieving oxidative stress and inflammation and a mechanism involving the Hh signaling pathway, suggesting that RES is a potential agent against POF and can aid in the survival of FGSCs.


Asunto(s)
Antioxidantes/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Oogoniales/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Insuficiencia Ovárica Primaria/tratamiento farmacológico , Resveratrol/farmacología , Animales , Peso Corporal/efectos de los fármacos , Busulfano/toxicidad , Catalasa/genética , Catalasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Femenino , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Oogoniales/metabolismo , Células Madre Oogoniales/patología , Tamaño de los Órganos/efectos de los fármacos , Folículo Ovárico/metabolismo , Folículo Ovárico/patología , Cultivo Primario de Células , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/patología , Piridinas/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/antagonistas & inhibidores , Pirimidinas/farmacología , Transducción de Señal , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Glutatión Peroxidasa GPX1
6.
J Periodontal Res ; 51(5): 661-8, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26740292

RESUMEN

BACKGROUND AND OBJECTIVE: LP533401 is an inhibitor of tryptophan hydroxylase 1, which regulates serotonin production in the gut. Previous work indicates that LP533401 has an anabolic effect in bone. Thus, we hypothesized that inhibition of gut serotonin production may modulate the host response in periodontal disease. In this study, we aimed to analyze the effects of LP533401 in a rat periodontitis model to evaluate the role of gut serotonin in periodontitis pathophysiology. MATERIAL AND METHODS: Twenty-four rats were divided into three groups: treated group (T: ligature-induced periodontal disease and LP533401, 25 mg/kg/d) by gavage; ligature group (L: ligature-induced periodontal disease only); and control group (C: without ligature-induced periodontal disease). After 28 d, radiographic alveolar bone support was measured on digital radiographs, and alveolar bone volume fraction, tissue mineral density and trabeculae characteristics were quantified by microcomputed tomography in the right hemi-mandible. Left hemi-mandibles were decalcified and alveolar bone loss, attachment loss and area of collagen in the gingiva were histologically analyzed. RESULTS: Significant difference between the L and C groups was found, confirming that periodontal disease was induced. We observed no difference between the T and L groups regarding alveolar bone destruction and area of collagen. CONCLUSION: LP533401 (25 mg/kg/d) for 28 d does not prevent bone loss and does not modulate host response in a rat model of induced periodontal disease.


Asunto(s)
Enfermedades Periodontales/tratamiento farmacológico , Enfermedades Periodontales/patología , Pirimidinas/antagonistas & inhibidores , Serotonina/metabolismo , Pérdida de Hueso Alveolar/diagnóstico por imagen , Pérdida de Hueso Alveolar/tratamiento farmacológico , Pérdida de Hueso Alveolar/patología , Pérdida de Hueso Alveolar/prevención & control , Animales , Colágeno , Modelos Animales de Enfermedad , Encía/patología , Ligadura/efectos adversos , Masculino , Mandíbula/patología , Pérdida de la Inserción Periodontal/diagnóstico por imagen , Pérdida de la Inserción Periodontal/prevención & control , Periodontitis/tratamiento farmacológico , Periodontitis/patología , Ratas , Ratas Wistar , Serotonina/fisiología , Microtomografía por Rayos X/métodos
7.
Retrovirology ; 12: 65, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26215564

RESUMEN

BACKGROUND: Hijacking of the cullin-RING E3 ubiquitin ligase (CRL) machinery is a common mechanism employed by diverse groups of viruses for the efficient counteraction and degradation of host proteins. In particular, HIV-1 Vpu usurps the SCF(ß-TrCP) E3 ubiquitin ligase complex to mark CD4 for degradation by the 26S proteasome. Vpu also interacts with and downmodulates a number of other host proteins, including the restriction factor BST-2. However, whether Vpu primarily relies on a cullin-dependent or -independent mechanism to antagonize its cellular targets has not been fully elucidated. RESULTS: We utilized a sulphamate AMP analog, MLN4924, to effectively block the activation of CRLs within infected primary CD4(+) T cells. MLN4924 treatment, in a dose dependent manner, efficiently relieved surface downmodulation and degradation of CD4 by NL4-3 Vpu. MLN4924 inhibition was highly specific, as this inhibitor had no effect on Nef's ability to downregulate CD4, which is accomplished by a CRL-independent mechanism. In contrast, NL4-3 Vpu's capacity to downregulate BST-2, NTB-A and CCR7 was not inhibited by the drug. Vpu's from both a transmitted founder (T/F) and chronic carrier (CC) virus preserved the ability to downregulate BST-2 in the presence of MLN4924. Finally, depletion of cellular pools of cullin 1 attenuated Vpu's ability to decrease CD4 but not BST-2 surface levels. CONCLUSIONS: We conclude that Vpu employs both CRL-dependent and CRL-independent modes of action against host proteins. Notably, we also establish that Vpu-mediated reduction of BST-2 from the cell surface is independent of ß-TrCP and the CRL- machinery and this function is conserved by Vpu's from primary isolates. Therefore, potential therapies aimed at antagonizing the activities of Vpu may need to address these distinct mechanisms of action in order to achieve a maximal effect.


Asunto(s)
Proteínas Cullin/metabolismo , Regulación hacia Abajo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Ciclopentanos/antagonistas & inhibidores , Ciclopentanos/farmacología , VIH-1/genética , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Pirimidinas/antagonistas & inhibidores , Pirimidinas/farmacología , Receptores CCR7/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/metabolismo
8.
J Virol ; 88(2): 878-89, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24198417

RESUMEN

Arenaviruses merit significant interest as important human pathogens, since several of them cause severe hemorrhagic fever disease that is associated with high morbidity and significant mortality. Currently, there are no FDA-licensed arenavirus vaccines available, and current antiarenaviral therapy is limited to an off-labeled use of the nucleoside analog ribavirin, which has limited prophylactic efficacy. The pyrimidine biosynthesis inhibitor A3, which was identified in a high-throughput screen for compounds that blocked influenza virus replication, exhibits a broad-spectrum antiviral activity against negative- and positive-sense RNA viruses, retroviruses, and DNA viruses. In this study, we evaluated the antiviral activity of A3 against representative Old World (lymphocytic choriomeningitis virus) and New World (Junin virus) arenaviruses in rodent, monkey, and human cell lines. We show that A3 is significantly more efficient than ribavirin in controlling arenavirus multiplication and that the A3 inhibitory effect is in part due to its ability to interfere with viral RNA replication and transcription. We document an additive antiarenavirus effect of A3 and ribavirin, supporting the potential combination therapy of ribavirin and pyrimidine biosynthesis inhibitors for the treatment of arenavirus infections.


Asunto(s)
Antivirales/farmacología , Infecciones por Arenaviridae/virología , Arenavirus/efectos de los fármacos , Pirimidinas/antagonistas & inhibidores , Animales , Infecciones por Arenaviridae/metabolismo , Arenavirus/genética , Arenavirus/fisiología , Línea Celular , Evaluación Preclínica de Medicamentos , Humanos , Pirimidinas/biosíntesis , Replicación Viral/efectos de los fármacos
9.
Cardiovasc Drugs Ther ; 28(5): 447-57, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25106875

RESUMEN

PURPOSE: Statins increase the incidence of new onset diabetes. Prolonged statin therapy upregulates PTEN expression. PTEN levels are also elevated in diabetic animals. Activation of protein kinase A by cAMP decreases PTEN expression. We assessed whether prolonged treatment with rosuvastatin (ROS) induces glucose intolerance by upregulating Phosphatase and Tensin Homologue on Chromosome 10 (PTEN) in mice receiving normal (ND) or Western Diet (WD) and whether concomitant treatment with cilostazol (CIL, a phosphodiesterase-3 inhibitor) attenuates the effects. METHODS: PTEN(loxp/cre) or PTEN(+/-) mice received ND or WD without or with ROS (10 mg/kg/day). Wild-type mice received ND or WD without or with ROS, CIL (10 mg/kg/day), or ROS+CIL for 30 days. Fasting insulin and glucose tolerance test were measured as well as PTEN and P-AKT levels in skeletal muscle. RESULTS: Serum glucose after intraperitoneal injection of glucose was higher in PTEN(loxp/cre) mice receiving WD or ROS and especially WD+ROS. Levels were lower in PTEN(+/-) mice compared to PTEN(loxp/cre) in each treatment group. CIL decreased glucose levels in mice receiving WD, ROS and their combination. Insulin levels were higher in the WD+ROS group. CIL decreased insulin in mice receiving WD+ROS. WD, ROS and especially their combination increased PTEN and decreased P-AKT levels. CIL attenuated the effect of WD, ROS and their combination. CONCLUSIONS: Long-term ROS can induce diabetes by upregulating PTEN. CIL attenuates these changes. Partial knockdown of PTEN also ameliorates ROS-induced insulin resistance. Further studies are needed to assess the effects of increasing cAMP levels to prevent the induction of diabetes by statins.


Asunto(s)
Diabetes Mellitus Tipo 2/inducido químicamente , Fluorobencenos/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Resistencia a la Insulina/genética , Fosfohidrolasa PTEN/biosíntesis , Pirimidinas/efectos adversos , Sulfonamidas/efectos adversos , Regulación hacia Arriba/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Glucemia/genética , Cilostazol , Diabetes Mellitus Tipo 2/sangre , Dieta Occidental , Fluorobencenos/administración & dosificación , Fluorobencenos/antagonistas & inhibidores , Fluorobencenos/farmacología , Técnicas de Silenciamiento del Gen , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Insulina/sangre , Ratones , Músculo Esquelético/metabolismo , Inhibidores de Fosfodiesterasa 3/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/administración & dosificación , Pirimidinas/antagonistas & inhibidores , Pirimidinas/farmacología , Rosuvastatina Cálcica , Sulfonamidas/administración & dosificación , Sulfonamidas/antagonistas & inhibidores , Sulfonamidas/farmacología , Tetrazoles/farmacología
10.
Toxicol Appl Pharmacol ; 267(1): 113-24, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23274516

RESUMEN

Compound C, a well-known inhibitor of the intracellular energy sensor AMP-activated protein kinase (AMPK), has been reported to cause apoptotic cell death in myeloma, breast cancer cells and glioma cells. In this study, we have demonstrated that compound C not only induced autophagy in all tested skin cancer cell lines but also caused more apoptosis in p53 wildtype skin cancer cells than in p53-mutant skin cancer cells. Compound C can induce upregulation, phosphorylation and nuclear translocalization of the p53 protein and upregulate expression of p53 target genes in wildtype p53-expressing skin basal cell carcinoma (BCC) cells. The changes of p53 status were dependent on DNA damage which was caused by compound C induced reactive oxygen species (ROS) generation and associated with activated ataxia-telangiectasia mutated (ATM) protein. Using the wildtype p53-expressing BCC cells versus stable p53-knockdown BCC sublines, we present evidence that p53-knockdown cancer cells were much less sensitive to compound C treatment with significant G2/M cell cycle arrest and attenuated the compound C-induced apoptosis but not autophagy. The compound C induced G2/M arrest in p53-knockdown BCC cells was associated with the sustained inactive Tyr15 phosphor-Cdc2 expression. Overall, our results established that compound C-induced apoptosis in skin cancer cells was dependent on the cell's p53 status.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Pirazoles/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Apoptosis/fisiología , Autofagia/efectos de los fármacos , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Pirazoles/farmacología , Pirimidinas/farmacología , Neoplasias Cutáneas/enzimología
11.
Glia ; 60(12): 2050-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23002008

RESUMEN

In neurons, small-conductance calcium-activated potassium (KCNN/SK/K(Ca)2) channels maintain calcium homeostasis after N-methyl-D-aspartate (NMDA) receptor activation, thereby preventing excitotoxic neuronal death. So far, little is known about the function of KCNN/SK/K(Ca)2 channels in non-neuronal cells, such as microglial cells. In this study, we addressed the question whether KCNN/SK/K(Ca)2 channels activation affected inflammatory responses of primary mouse microglial cells upon lipopolysaccharide (LPS) stimulation. We found that N-cyclohexyl-N-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-4-pyrimidinamine (CyPPA), a positive pharmacological activator of KCNN/SK/K(Ca)2 channels, significantly reduced LPS-stimulated activation of microglia in a concentration-dependent manner. The general KCNN/SK/K(Ca)2 channel blocker apamin reverted these effects of CyPPA on microglial proliferation. Since calcium plays a central role in microglial activation, we further addressed whether KCNN/SK/K(Ca)2 channel activation affected the changes of intracellular calcium levels, [Ca(2+)](i), in microglial cells. Our data show that LPS-induced elevation of [Ca(2+)](i) was attenuated following activation of KCNN2/3/K(Ca)2.2/K(Ca)2.3 channels by CyPPA. Furthermore, CyPPA reduced downstream events including tumor necrosis factor alpha and interleukin 6 cytokine production and nitric oxide release in activated microglia. Further, we applied specific peptide inhibitors of the KCNN/SK/K(Ca)2 channel subtypes to identify which particular channel subtype mediated the observed anti-inflammatory effects. Only inhibitory peptides targeting KCNN3/SK3/K(Ca)2.3 channels, but not KCNN2/SK2/K(Ca)2.2 channel inhibition, reversed the CyPPA-effects on LPS-induced microglial proliferation. These findings revealed that KCNN3/SK3/K(Ca)2.3 channels can modulate the LPS-induced inflammatory responses in microglial cells. Thus, KCNN3/SK3/K(Ca)2.3 channels may serve as a therapeutic target for reducing microglial activity and related inflammatory responses in the central nervous system.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Citocinas/biosíntesis , Mediadores de Inflamación/fisiología , Microglía/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Animales , Animales Recién Nacidos , Apamina/farmacología , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/fisiología , Regulación hacia Abajo/efectos de los fármacos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/toxicidad , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Pirazoles/antagonistas & inhibidores , Pirazoles/toxicidad , Pirimidinas/antagonistas & inhibidores , Pirimidinas/toxicidad , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
12.
J Virol ; 85(13): 6548-56, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21507975

RESUMEN

Viral replication relies on the host to supply nucleosides. Host enzymes involved in nucleoside biosynthesis are potential targets for antiviral development. Ribavirin (a known antiviral drug) is such an inhibitor that suppresses guanine biosynthesis; depletion of the intracellular GTP pool was shown to be the major mechanism to inhibit flavivirus. Along similar lines, inhibitors of the pyrimidine biosynthesis pathway could be targeted for potential antiviral development. Here we report on a novel antiviral compound (NITD-982) that inhibits host dihydroorotate dehydrogenase (DHODH), an enzyme required for pyrimidine biosynthesis. The inhibitor was identified through screening 1.8 million compounds using a dengue virus (DENV) infection assay. The compound contains an isoxazole-pyrazole core structure, and it inhibited DENV with a 50% effective concentration (EC(50)) of 2.4 nM and a 50% cytotoxic concentration (CC(50)) of >5 µM. NITD-982 has a broad antiviral spectrum, inhibiting both flaviviruses and nonflaviviruses with nanomolar EC(90)s. We also show that (i) the compound inhibited the enzymatic activity of recombinant DHODH, (ii) an NITD-982 analogue directly bound to the DHODH protein, (iii) supplementing the culture medium with uridine reversed the compound-mediated antiviral activity, and (iv) DENV type 2 (DENV-2) variants resistant to brequinar (a known DHODH inhibitor) were cross resistant to NITD-982. Collectively, the results demonstrate that the compound inhibits DENV through depleting the intracellular pyrimidine pool. In contrast to the in vitro potency, the compound did not show any efficacy in the DENV-AG129 mouse model. The lack of in vivo efficacy is likely due to the exogenous uptake of pyrimidine from the diet or to a high plasma protein-binding activity of the current compound.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Virus del Dengue/efectos de los fármacos , Dengue/tratamiento farmacológico , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Animales , Antivirales/química , Antivirales/farmacocinética , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Dengue/virología , Virus del Dengue/enzimología , Virus del Dengue/patogenicidad , Virus del Dengue/fisiología , Dihidroorotato Deshidrogenasa , Modelos Animales de Enfermedad , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Pirimidinas/biosíntesis , Sigmodontinae , Resultado del Tratamiento , Células Vero , Replicación Viral/efectos de los fármacos
13.
Can J Physiol Pharmacol ; 90(8): 1135-44, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22809038

RESUMEN

Peroxisome proliferator-activated receptors (PPAR) regulate the expression of genes involved in lipid metabolism, energy production, and inflammation. Their role in ischaemia-reperfusion (I/R) is less clear, although research indicates involvement of PPARs in some forms of preconditioning. This study aimed to explore the effects of PPAR-α activation on the I/R injury and potential cardioprotective downstream mechanisms involved. Langendorff-perfused hearts of rats pretreated with the selective PPAR-α agonist WY-14643 (WY, pirinixic acid; 3 mg·(kg body mass)·day(-1); 5 days) were subjected to 30 min ischaemia - 2 h reperfusion with or without the phosphatidylinositol 3-kinase (PI3K)-Akt inhibitor wortmannin for the evaluation of functional (left ventricular developed pressure, LVDP) recovery, infarct size (IS), and reperfusion-induced arrhythmias. A 2-fold increase in baseline PPAR-α mRNA levels (qPCR) in the WY-treated group and higher post-I/R PPAR-α levels compared with those in untreated controls were accompanied by similar changes in the expression of PPAR-α target genes PDK4 and mCPT-1, regulating glucose and fatty acid metabolism, and by enhanced Akt phosphorylation. Post-ischaemic LVDP restoration in WY-treated hearts reached 60% ± 9% of the pre-ischaemic values compared with 24% ± 3% in the control hearts (P < 0.05), coupled with reduced IS and incidence of ventricular fibrillation that was blunted by wortmannin. Results indicate that PPAR-α up-regulation may confer preconditioning-like protection via metabolic effects. Downstream mechanisms of PPAR-α-mediated cardioprotection may involve PI3K-Akt activation.


Asunto(s)
Daño por Reperfusión Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/fisiopatología , PPAR alfa/fisiología , Fosfatidilinositol 3-Quinasa/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Androstadienos/farmacología , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/prevención & control , Quimasas/biosíntesis , Modelos Animales de Enfermedad , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/metabolismo , PPAR alfa/biosíntesis , Proliferadores de Peroxisomas/antagonistas & inhibidores , Proliferadores de Peroxisomas/farmacología , Proliferadores de Peroxisomas/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/antagonistas & inhibidores , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Wortmanina
14.
Biochem Genet ; 50(9-10): 684-93, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22580794

RESUMEN

Purine and pyrimidine antimetabolites are used to treat leukemias, autoimmune diseases, and solid tumors. Detection of slow metabolizers before administration of the drugs is necessary to prevent any subsequent drug toxicity. With this aim, we determined the frequencies of normal and slow alleles in our population. Polymorphisms in genes encoding cytidine deaminase (CDA), dihydropyrimidine dehydrogenase (DPYD), and thiopurine-S-methyltransferase (TPMT) were documented in 225 healthy volunteers. The polymorphisms typed included CDA*3, DPYD*2A, TPMT*2A, TPMT*3B, and TPMT*3C. Methods used for genotyping included standard PCR-RFLP and allele-specific PCR reactions. The frequencies were 0.44 % for DPYD*2A, 0.67 % for TPMT*3B, and 0.89 % for TPMT*3C. The CDA*3 and TPMT*2A alleles were not detected. Although these polymorphisms have been demonstrated to be associated with drug toxicity in other populations, they appear to be very rare in the adult Indian population.


Asunto(s)
Citidina Desaminasa/genética , Dihidrouracilo Deshidrogenasa (NADP)/genética , Frecuencia de los Genes , Metiltransferasas/genética , Purinas/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Adolescente , Adulto , Alelos , Pueblo Asiatico/genética , Activación Enzimática , Femenino , Genética de Población/métodos , Genotipo , Técnicas de Genotipaje , Humanos , India , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Polimorfismo de Longitud del Fragmento de Restricción , Prevalencia , Adulto Joven
15.
Behav Brain Res ; 417: 113585, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34536428

RESUMEN

Tremor is one of the motor symptoms of Parkinson's disease (PD), present also in neuroleptic-induced parkinsonism. Tremulous Jaw Movements (TJMs) are suggested to be a well-validated rodent model of PD resting tremor. TJMs can be induced by typical antipsychotics and are known to be reduced by different drugs, including adenosine A2A receptor antagonists. The aim of the present study was to search for brain structures involved in the tremorolytic action of SCH58261, a selective A2A receptor antagonist, in TJMs induced by subchronic pimozide. Besides TJMs, we evaluated in the same animals the expression of zif-268 mRNA (neuronal responsiveness marker), and mRNA levels for glutamic acid decarboxylase 65-kDa isoform (GAD65) and vesicular glutamate transporters 1 and 2 (vGluT1/2) in selected brain structures, as markers of GABAergic and glutamatergic neurons, respectively. We found that SCH58261 reduced the pimozide-induced TJMs. Pimozide increased the zif-268 mRNA level in the striatum, nucleus accumbens (NAc) core, and substantia nigra pars reticulata (SNr). Additionally, it increased GAD65 mRNA in the striatum and SNr, and vGluT2 mRNA levels in the subthalamic nucleus (STN). A positive correlation between zif-268, GAD65 and vGluT2 mRNAs and TJMs was found. SCH58261 reversed the pimozide-increased zif-268 mRNA in the striatum and NAc core and GAD65 mRNA in the striatum and SNr. In contrast, SCH58261 did not influence vGluT2 mRNA in STN. The present study suggests an importance of the striato-subthalamo-nigro-thalamic circuit in neuroleptic-induced TJMs. The tremorolytic effect of A2A receptor blockade seems to involve this circuit bypassing, however, STN.


Asunto(s)
Antagonistas de Dopamina/efectos adversos , Maxilares/efectos de los fármacos , Movimiento/efectos de los fármacos , Pimozida/efectos adversos , Pirimidinas/antagonistas & inhibidores , Receptor de Adenosina A2A/efectos de los fármacos , Triazoles/antagonistas & inhibidores , Animales , Antipsicóticos/farmacología , Encéfalo/metabolismo , Cuerpo Estriado/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Glutamato Descarboxilasa/metabolismo , Masculino , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Enfermedad de Parkinson Secundaria/fisiopatología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Subtalámico/metabolismo , Temblor/inducido químicamente
16.
Lipids Health Dis ; 10: 165, 2011 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-21936891

RESUMEN

BACKGROUND: This study was an investigation of the effects of ingesting a daily dose of isolated glycinin soy protein (11S globulin), in association with rosuvastatin, on the control of hypercholesterolemia in experimental animals. METHODS: Male Wistar rats were kept in individual cages under appropriate controlled conditions of temperature, light and humidity. The animals were divided into five groups (n = 9): 1) standard (STD): fed on casein as protein source; 2) hypercholesterolemic (HC): STD plus 1% cholesterol and 0.5% cholic acid; 3) HC+11S: hypercholesterolemic + glycinin (300 mg/kg/day); 4) HC+ROS: hypercholesterolemic + rosuvastatin (10 mg/kg/day); 5) HC+11S+ROS: HC diet, the 11S protein and the drug in the doses given in (3) and (4). The protein and the drug were administered by gavage for 28 days. The results indicated that the addition of 1% cholesterol and 0.5% cholic acid induced hypercholesterolemia in the animals without interfering with their weight gain. RESULTS: A single daily dose of glycinin contributed an additional 2.8% of dietary protein intake and demonstrated its functional role, particularly in raising HDL-C, decreasing triglycerides in the liver and improving the atherogenic index in animals exposed to a hypercholesterolemic diet. CONCLUSION: Most of the beneficial effects of the isolated treatments disappeared when the drug (rosuvastatin) and the protein (glycinin) were taken simultaneously. The association was shown not to interact additively, as noted in the plasma levels of total cholesterol and non-HDL cholesterol, and in the significant increase of cholesterol in the liver. Studies are in progress to identify the effects of peptides derived from the 11S globulin and their role in cholesterol metabolism.


Asunto(s)
HDL-Colesterol/sangre , Suplementos Dietéticos , Fluorobencenos/antagonistas & inhibidores , Interacciones Alimento-Droga , Globulinas/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/química , Hipercolesterolemia/dietoterapia , Pirimidinas/antagonistas & inhibidores , Proteínas de Soja/uso terapéutico , Sulfonamidas/antagonistas & inhibidores , Animales , Aterosclerosis/prevención & control , Colesterol/sangre , Colesterol/metabolismo , Colesterol en la Dieta/efectos adversos , Ácido Cólico/efectos adversos , Terapia Combinada , Fluorobencenos/uso terapéutico , Globulinas/aislamiento & purificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipercolesterolemia/sangre , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/metabolismo , Lipoproteínas/metabolismo , Hígado/metabolismo , Masculino , Pirimidinas/uso terapéutico , Ratas , Ratas Wistar , Factores de Riesgo , Rosuvastatina Cálcica , Proteínas de Soja/aislamiento & purificación , Sulfonamidas/uso terapéutico , Triglicéridos/metabolismo
17.
Mol Cancer Ther ; 20(6): 975-985, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33722854

RESUMEN

KRASG12C inhibitors, including MRTX849, are promising treatment options for KRAS-mutant non-small cell lung cancer (NSCLC). PD-1 inhibitors are approved in NSCLC; however, strategies to enhance checkpoint inhibitor therapy (CIT) are needed. KRASG12C mutations are smoking-associated transversion mutations associated with high tumor mutation burden, PD-L1 positivity, and an immunosuppressive tumor microenvironment. To evaluate the potential of MRTX849 to augment CIT, its impact on immune signaling and response to CIT was evaluated. In human tumor xenograft models, MRTX849 increased MHC class I protein expression and decreased RNA and/or plasma protein levels of immunosuppressive factors. In a KrasG12C -mutant CT26 syngeneic mouse model, MRTX849 decreased intratumoral myeloid-derived suppressor cells and increased M1-polarized macrophages, dendritic cells, CD4+, and CD8+ T cells. Similar results were observed in lung KrasG12C -mutant syngeneic and a genetically engineered mouse (GEM) model. In the CT26 KrasG12C model, MRTX849 demonstrated marked tumor regression when tumors were established in immune-competent BALB/c mice; however, the effect was diminished when tumors were grown in T-cell-deficient nu/nu mice. Tumors progressed following anti-PD-1 or MRTX849 single-agent treatment in immune-competent mice; however, combination treatment demonstrated durable, complete responses (CRs). Tumors did not reestablish in the same mice that exhibited durable CRs when rechallenged with tumor cell inoculum, demonstrating these mice developed adaptive antitumor immunity. In a GEM model, treatment with MRTX849 plus anti-PD-1 led to increased progression-free survival compared with either single agent alone. These data demonstrate KRAS inhibition reverses an immunosuppressive tumor microenvironment and sensitizes tumors to CIT through multiple mechanisms.


Asunto(s)
Acetonitrilos/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Piperazinas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Microambiente Tumoral/efectos de los fármacos
18.
Eur J Med Chem ; 186: 111855, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31740051

RESUMEN

There is an increasing interest in the field of cancer therapy for small compounds targeting pyrimidine biosynthesis, and in particular dihydroorotate dehydrogenase (DHODH), the fourth enzyme of this metabolic pathway. Three available DHODH structures, featuring three different known inhibitors, were used as templates to screen in silico an original chemical library from Erevan University. This process led to the identification of P1788, a compound chemically related to the alkaloid cerpegin, as a new class of pyrimidine biosynthesis inhibitors. In line with previous reports, we investigated the effect of P1788 on the cellular innate immune response. Here we show that pyrimidine depletion by P1788 amplifies cellular response to both type-I and type II interferons, but also induces DNA damage as assessed by γH2AX staining. Moreover, the addition of inhibitors of the DNA damage response led to the suppression of the P1788 stimulatory effects on the interferon pathway. This demonstrates that components of the DNA damage response are bridging the inhibition of pyrimidine biosynthesis by P1788 to the interferon signaling pathway. Altogether, these results provide new insights on the mode of action of novel pyrimidine biosynthesis inhibitors and their development for cancer therapies.


Asunto(s)
Furanos/farmacología , Piridinas/farmacología , Piridonas/farmacología , Pirimidinas/antagonistas & inhibidores , Células Cultivadas , Daño del ADN , Relación Dosis-Respuesta a Droga , Furanos/síntesis química , Furanos/química , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Piridonas/química , Pirimidinas/biosíntesis , Relación Estructura-Actividad
19.
Sci Rep ; 10(1): 10100, 2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32572069

RESUMEN

RNA viruses are responsible for a large variety of animal infections. Equine Arteritis Virus (EAV) is a positive single-stranded RNA virus member of the family Arteriviridae from the order Nidovirales like the Coronaviridae. EAV causes respiratory and reproductive diseases in equids. Although two vaccines are available, the vaccination coverage of the equine population is largely insufficient to prevent new EAV outbreaks around the world. In this study, we present a high-throughput in vitro assay suitable for testing candidate antiviral molecules on equine dermal cells infected by EAV. Using this assay, we identified three molecules that impair EAV infection in equine cells: the broad-spectrum antiviral and nucleoside analog ribavirin, and two compounds previously described as inhibitors of dihydroorotate dehydrogenase (DHODH), the fourth enzyme of the pyrimidine biosynthesis pathway. These molecules effectively suppressed cytopathic effects associated to EAV infection, and strongly inhibited viral replication and production of infectious particles. Since ribavirin is already approved in human and small animal, and that several DHODH inhibitors are in advanced clinical trials, our results open new perspectives for the management of EAV outbreaks.


Asunto(s)
Infecciones por Arterivirus/tratamiento farmacológico , Equartevirus/metabolismo , Ribavirina/farmacología , Animales , Antivirales/farmacología , Infecciones por Arterivirus/veterinaria , Línea Celular , Efecto Citopatogénico Viral/efectos de los fármacos , Dihidroorotato Deshidrogenasa , Enfermedades de los Caballos/virología , Caballos/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Purinas/antagonistas & inhibidores , Purinas/biosíntesis , Purinas/farmacología , Pirimidinas/antagonistas & inhibidores , Pirimidinas/biosíntesis , Pirimidinas/farmacología , ARN/farmacología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
20.
Nat Commun ; 11(1): 577, 2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31996669

RESUMEN

The Bruton tyrosine kinase (BTK) inhibitor ibrutinib provides effective treatment for patients with chronic lymphocytic leukemia (CLL), despite extensive heterogeneity in this disease. To define the underlining regulatory dynamics, we analyze high-resolution time courses of ibrutinib treatment in patients with CLL, combining immune-phenotyping, single-cell transcriptome profiling, and chromatin mapping. We identify a consistent regulatory program starting with a sharp decrease of NF-κB binding in CLL cells, which is followed by reduced activity of lineage-defining transcription factors, erosion of CLL cell identity, and acquisition of a quiescence-like gene signature. We observe patient-to-patient variation in the speed of execution of this program, which we exploit to predict patient-specific dynamics in the response to ibrutinib based on the pre-treatment patient samples. In aggregate, our study describes time-dependent cellular, molecular, and regulatory effects for therapeutic inhibition of B cell receptor signaling in CLL, and it establishes a broadly applicable method for epigenome/transcriptome-based treatment monitoring.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/efectos de los fármacos , Cromatina/genética , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Pirazoles/antagonistas & inhibidores , Pirazoles/metabolismo , Pirazoles/uso terapéutico , Pirimidinas/antagonistas & inhibidores , Pirimidinas/metabolismo , Pirimidinas/uso terapéutico , Adenina/análogos & derivados , Epigenoma , Epigenómica , Perfilación de la Expresión Génica , Heterogeneidad Genética/efectos de los fármacos , Humanos , Leucemia Linfocítica Crónica de Células B/inmunología , Aprendizaje Automático , Piperidinas , Receptores de Antígenos de Linfocitos B/efectos de los fármacos , Análisis de Secuencia de ARN , Factores de Transcripción , Transcriptoma
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda