Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Yi Chuan ; 41(4): 327-336, 2019 Apr 20.
Artículo en Zh | MEDLINE | ID: mdl-30992254

RESUMEN

There are two major pathways, homology-directed repair (HDR) and nonhomologous end joining (NHEJ), involved in double-strand break (DSB) repair. Single-stranded oligodeoxyribonucleotide (ssODN)-mediated homologous recombination repair is commonly used for animal site-directed genome editing, with great scientific and practical value. To improve ssODN-mediated HDR efficiency in the pig genome, we investigated the effect and molecular mechanism of mitogen-activated extracellular signal-regulated kinase (MEK) inhibitor PD0325901 on the HDR efficiency in porcine fetal fibroblasts (PFFs). The results showed that PD0325901 obviously increased the percentage of G2 and S phase cell populations and reduced the cell population ratio in the G1 phase of PFFs, and promoted the expression of HDR repair factor. At the optimal concentration of 250 nmol/L, PD0325901 increased the repair efficiency of ssODN-mediated GFP reporter vector by 58.8% and the directed editing efficiency of PFF DMD and ROSA26 locus by 48.16% and 17.64%, respectively. The results show that MEK inhibitor PD0325901 significantly promotes the efficiency of ssODN-mediated homologous-directed repair in the porcine genome, thus offering a new idea to generate genetically modified pigs more effectively.


Asunto(s)
Benzamidas/farmacología , Difenilamina/análogos & derivados , Edición Génica , Reparación del ADN por Recombinación , Animales , Roturas del ADN de Doble Cadena , ADN de Cadena Simple , Difenilamina/farmacología , Fibroblastos , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Oligodesoxirribonucleótidos , Porcinos
2.
J Proteome Res ; 16(10): 3585-3595, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28850235

RESUMEN

Mouse embryonic stem cells (mESCs) can be maintained in a pluripotent state when cultured with 2 inhibitors (2i) of extracellular signal-regulated kinase (MEK) and glycogen synthase kinase-3 (GSK3), and Royan 2 inhibitors (R2i) of FGF4 and TGFß. The molecular mechanisms that control ESC self-renewal and pluripotency are more important for translating stem cell technologies to clinical applications. In this study, we used the shotgun proteomics technique to compare the proteome of the ground state condition (R2i- and 2i-grown cells) to that of serum. Out of 1749 proteins identified, 171 proteins were differentially expressed (p < 0.05) in the 2i, R2i, and serum samples. Gene ontology (GO) analysis of differentially abundant proteins showed that the focal adhesion signaling pathway significantly down-regulated under ground state conditions. mESCs had highly adhesive attachment under the serum condition, whereas in the 2i and R2i culture conditions, a loss of adhesion was observed and the cells were rounded and grew in compact colonies on gelatin. Quantitative RT-PCR showed reduced expression of the integrins family in the 2i and R2i conditions. The serum culture had more prominent phosphorylation of focal adhesion kinase (FAK) compared to 2i and R2i cultures. Activity of the extracellular signal-regulated kinase (ERK)1/2 decreased in the 2i and R2i cultures compared to serum. Activation of integrins by Mn2+ in the 2i and R2i cultures resulted in reduced Nanog and increased the expression of lineage marker genes. In this study, we demonstrated that reduced focal adhesion enabled mESCs to be maintained in an undifferentiated and pluripotent state.


Asunto(s)
Adhesión Celular/genética , Diferenciación Celular/genética , Células Madre Embrionarias de Ratones/metabolismo , Proteoma/genética , Animales , Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor 4 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 4 de Crecimiento de Fibroblastos/genética , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/genética , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/genética , Ratones , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética
3.
Invest New Drugs ; 35(5): 616-626, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28194539

RESUMEN

Background Dual inhibition of activated MAPK and mTOR signaling pathways may enhance the antitumor efficacy of the MEK 1/2 inhibitor pimasertib and the mTOR inhibitor temsirolimus given in combination. Methods In this phase I study, patients with refractory advanced solid tumors (NCT01378377) received once-weekly temsirolimus plus once-daily oral pimasertib in 21-day cycles in a modified 3 + 3 dose-escalation design. The maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D) of pimasertib in combination with temsirolimus, safety and pharmacokinetics (PK) were investigated. Results Of 33 patients evaluated, all experienced ≥1 treatment-emergent adverse event (TEAE) and 31 had treatment-related TEAEs, most frequently stomatitis and thrombocytopenia. TEAEs were reversible. No deaths were attributed to treatment. Nine patients had dose-limiting toxicities (stomatitis, thrombocytopenia, serum creatinine phosphokinase increase, visual impairment) and the MTD was determined as 45 mg/day pimasertib plus 25 mg/week temsirolimus. However, due to overlapping toxicities no further investigations were performed and the RP2D was not defined. PK profiles of both agents were not adversely affected. Seventeen patients (17/26 patients) had a best response of stable disease; five had stable disease lasting >12 weeks. Conclusions The RP2D was not defined and the pimasertib plus temsirolimus combination investigated did not warrant further study.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 2/antagonistas & inhibidores , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/metabolismo , Niacinamida/administración & dosificación , Niacinamida/análogos & derivados , Inhibidores de Proteínas Quinasas/administración & dosificación , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Adulto Joven
4.
Eur J Clin Pharmacol ; 73(6): 717-726, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28283692

RESUMEN

PURPOSE: Emerging data on selumetinib, a MEK1/2 inhibitor in clinical development, suggest a possible difference in pharmacokinetics (PK) between Japanese and Western patients. This pooled analysis sought to assess the effect of ethnicity on selumetinib exposure in healthy Western and Asian subjects, and to identify any association between genetic variants in the UGT1A1, CYP2C19 and ABCG2 genes and observed differences in selumetinib PK. METHODS: A pooled analysis of data from ten Phase I studies, one in Asian subjects (encompassing Japanese, non-Japanese Asian and Indian Asian subjects) and nine in Western subjects, was conducted. Key findings were derived from the collective exposure data across doses of 25, 35, 50 and 75 mg selumetinib; primary variables were dose-normalized AUC and Cmax. RESULTS: PK data from 308 subjects (10 studies) were available for the pooled analysis; genetic data from 87 subjects (3 studies) were available for the pharmacogenetic analysis. Dose-normalized AUC and Cmax were 35% (95% CI: 25-47%) and 39% (95% CI: 24-56%) higher in the pooled Asian group, respectively, compared with Western subjects. PK exposure parameters were similar between the Japanese, non-Japanese Asian and Indian groups. There was no evidence that the polymorphisms assessed in the genes UGT1A1, CYP2C19 and ABCG2 account for observed PK differences. CONCLUSIONS: Selumetinib exposure was higher in healthy Asian subjects compared with Western subjects, and these data provide valuable insight for clinicians to consider when treating patients of Asian ethnicity with selumetinib.


Asunto(s)
Bencimidazoles/farmacocinética , Farmacogenética , Inhibidores de Proteínas Quinasas/farmacocinética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Área Bajo la Curva , Pueblo Asiatico , Población Negra , Ensayos Clínicos Fase I como Asunto , Citocromo P-450 CYP2C19/genética , Glucuronosiltransferasa/genética , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 2/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Población Blanca
5.
Eur J Clin Pharmacol ; 73(2): 175-184, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27889832

RESUMEN

PURPOSE: Two phase I, open-label trials in healthy subjects assessed whether co-administration with CYP3A4/CYP2C19 inhibitors, itraconazole/fluconazole (study A), or CYP3A4 inducer, rifampicin (study B), affects the exposure, safety/tolerability and pharmacokinetics of selumetinib and its metabolite N-desmethyl selumetinib. METHODS: In study A (n = 26), subjects received a single dose of selumetinib 25 mg and, after 4 days of washout, were randomized to treatment 1 (itraconazole 200 mg twice daily on days 1-11) or treatment 2 (fluconazole 400 mg on day 1 then 200 mg/day on days 2-11) plus co-administration of single-dose selumetinib 25 mg on day 8 (selumetinib staggered 4 h after itraconazole/fluconazole dose); Twenty-one days after discharge/washout, subjects received the alternate treatment. In study B (n = 22), subjects received a single dose of selumetinib 75 mg (day 1) then rifampicin 600 mg/day (days 4-14) plus a single dose of selumetinib 75 mg on day 12. Pharmacokinetic analysis and safety assessments were performed. RESULTS: Selumetinib co-administered with itraconazole, fluconazole (selumetinib staggered 4 h after itraconazole/fluconazole dose), or rifampicin was well tolerated. Selumetinib exposure was higher when co-administered with itraconazole or fluconazole (area under the plasma concentration-time curve (AUC) increased by 49 and 53%, respectively; maximum plasma concentration (C max) increased by 19 and 26%, respectively) but lower when co-dosed with rifampicin (AUC and C max decreased by 51 and 26%, respectively) versus selumetinib dosed alone. Co-administration with itraconazole or rifampicin decreased N-desmethyl selumetinib AUC(0-t) (11 and 55%, respectively), and C max (25 and 18%, respectively), with fluconazole, AUC(0-t) increased by 40%, but there was no effect on C max. CONCLUSIONS: Co-administration of CYP3A4/CYP2C19 inhibitors will likely increase exposure to selumetinib, while CYP3A4 inducers will likely reduce its exposure.


Asunto(s)
Bencimidazoles/farmacocinética , Inhibidores del Citocromo P-450 CYP2C19/farmacología , Inhibidores del Citocromo P-450 CYP3A/farmacología , Adolescente , Adulto , Bencimidazoles/sangre , Estudios Cruzados , Citocromo P-450 CYP2C19/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inductores de las Enzimas del Citocromo P-450/farmacología , Femenino , Fluconazol/farmacología , Voluntarios Sanos , Humanos , Itraconazol/farmacología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 2/antagonistas & inhibidores , Masculino , Rifampin/farmacología , Adulto Joven
6.
Proc Natl Acad Sci U S A ; 111(42): E4494-503, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288766

RESUMEN

LEOPARD syndrome (multiple Lentigines, Electrocardiographic conduction abnormalities, Ocular hypertelorism, Pulmonary stenosis, Abnormal genitalia, Retardation of growth, sensorineural Deafness; LS), also called Noonan syndrome with multiple lentigines (NSML), is a rare autosomal dominant disorder associating various developmental defects, notably cardiopathies, dysmorphism, and short stature. It is mainly caused by mutations of the PTPN11 gene that catalytically inactivate the tyrosine phosphatase SHP2 (Src-homology 2 domain-containing phosphatase 2). Besides its pleiotropic roles during development, SHP2 plays key functions in energetic metabolism regulation. However, the metabolic outcomes of LS mutations have never been examined. Therefore, we performed an extensive metabolic exploration of an original LS mouse model, expressing the T468M mutation of SHP2, frequently borne by LS patients. Our results reveal that, besides expected symptoms, LS animals display a strong reduction of adiposity and resistance to diet-induced obesity, associated with overall better metabolic profile. We provide evidence that LS mutant expression impairs adipogenesis, triggers energy expenditure, and enhances insulin signaling, three features that can contribute to the lean phenotype of LS mice. Interestingly, chronic treatment of LS mice with low doses of MEK inhibitor, but not rapamycin, resulted in weight and adiposity gains. Importantly, preliminary data in a French cohort of LS patients suggests that most of them have lower-than-average body mass index, associated, for tested patients, with reduced adiposity. Altogether, these findings unravel previously unidentified characteristics for LS, which could represent a metabolic benefit for patients, but may also participate to the development or worsening of some traits of the disease. Beyond LS, they also highlight a protective role of SHP2 global LS-mimicking modulation toward the development of obesity and associated disorders.


Asunto(s)
Dieta , Síndrome LEOPARD/genética , Obesidad/prevención & control , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Delgadez/genética , Adipocitos/citología , Tejido Adiposo/metabolismo , Adiposidad , Animales , Composición Corporal , Diferenciación Celular , Modelos Animales de Enfermedad , Metabolismo Energético , Insulina/metabolismo , Lentivirus/metabolismo , Lipólisis , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Masculino , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Recombinación Genética
7.
Hum Mol Genet ; 23(3): 682-92, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24057668

RESUMEN

RASopathies are syndromes caused by gain-of-function mutations in the Ras signaling pathway. One of these conditions, Costello syndrome (CS), is typically caused by an activating de novo germline mutation in HRAS and is characterized by a wide range of cardiac, musculoskeletal, dermatological and developmental abnormalities. We report that a majority of individuals with CS have hypo-mineralization of enamel, the outer covering of teeth, and that similar defects are present in a CS mouse model. Comprehensive analysis of the mouse model revealed that ameloblasts, the cells that generate enamel, lacked polarity, and the ameloblast progenitor cells were hyperproliferative. Ras signals through two main effector cascades, the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K) pathways. To determine through which pathway Ras affects enamel formation, inhibitors targeting either PI3K or MEK 1 and 2 (MEK 1/2), kinases in the MAPK pathway, were utilized. MEK1/2 inhibition rescued the hypo-mineralized enamel, normalized the ameloblast polarity defect and restored normal progenitor cell proliferation. In contrast, PI3K inhibition only corrected the progenitor cell proliferation phenotype. We demonstrate for the first time the central role of Ras signaling in enamel formation in CS individuals and present the mouse incisor as a model system to dissect the roles of the Ras effector pathways in vivo.


Asunto(s)
Síndrome de Costello/metabolismo , Esmalte Dental/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adolescente , Adulto , Ameloblastos/metabolismo , Ameloblastos/patología , Animales , Estudios de Casos y Controles , Polaridad Celular , Niño , Preescolar , Estudios de Cohortes , Síndrome de Costello/genética , Esmalte Dental/efectos de los fármacos , Esmalte Dental/metabolismo , Esmalte Dental/ultraestructura , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Lactante , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Masculino , Ratones , Ratones Mutantes , Microscopía Electrónica de Rastreo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal/genética , Adulto Joven
9.
Biochim Biophys Acta ; 1833(5): 1114-24, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23274856

RESUMEN

Connective tissue growth factor (CTGF; also known as CCN2) is an inflammatory mediator, and shows elevated levels in regions of severe injury and inflammatory diseases. CTGF is abundantly expressed in osteoarthritis (OA). Migration and infiltration of mononuclear cells to inflammatory sites are playing important roles during OA pathogenesis. Monocyte chemoattractant protein-1 (MCP-1/CCL2) is the key chemokine that regulates migration and infiltration of monocytes. However, the effect of CTGF on MCP-1 expression and monocyte migration is largely unknown. Our results showed that MCP-1 was highly expressed in OA synovial fibroblasts (OASFs) as compared with normal SFs. Directly applying OASFs with CTGF increased MCP-1 expression in a concentration- and a time-dependent manner. CTGF mediated MCP-1 production was attenuated by αvß5 integrin neutralized antibody. Pretreatment with focal adhesion kinase (FAK), MEK, AP-1, and NF-κB inhibitors also inhibited the potentiating action of CTGF. CTGF-mediated increase of NF-κB and AP-1 luciferase activity was inhibited by FAK, MEK, and ERK inhibitors or mutants. In vitro chemotaxis assay showed that OA synovial fluid and supernatants from CTGF treated OASFs increased migration of monocyte. In addition, CTGF-mediated migration was inhibited by the FAK and MEK inhibitors. Taken together, our results indicated that CTGF enhances the migration of monocyte cells by increasing MCP-1 expression through the αvß5 integrin, FAK, MEK, ERK, and NF-κB/AP-1 signal transduction pathway.


Asunto(s)
Quimiocina CCL2 , Factor de Crecimiento del Tejido Conjuntivo , Inflamación , Osteoartritis , Movimiento Celular/genética , Células Cultivadas , Quimiocina CCL2/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Monocitos , Osteoartritis/genética , Osteoartritis/metabolismo , Receptores de Vitronectina/metabolismo , Transducción de Señal , Líquido Sinovial/metabolismo
10.
J Neurophysiol ; 107(10): 2782-93, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357798

RESUMEN

Activation of nicotinic acetylcholine receptors (nAChRs) by systemic nicotine enhances sensory-cognitive function and sensory-evoked cortical responses. Although nAChRs mediate fast neurotransmission at many synapses in the nervous system, nicotinic regulation of cortical processing is neuromodulatory. To explore potential mechanisms of nicotinic neuromodulation, we examined whether intracellular signal transduction involving mitogen-activated protein kinase (MAPK) contributes to regulation of tone-evoked responses in primary auditory cortex (A1) in the mouse. Systemic nicotine enhanced characteristic frequency (CF) tone-evoked current-source density (CSD) profiles in A1, including the shortest-latency (presumed thalamocortical) current sink in layer 4 and longer-latency (presumed intracortical) sinks in layers 2-4, by increasing response amplitudes and decreasing response latencies. Microinjection of the MAPK kinase (MEK) inhibitor U0126 into the thalamus, targeting the auditory thalamocortical pathway, blocked the effect of nicotine on the initial (thalamocortical) CSD component but did not block enhancement of longer-latency (intracortical) responses. Conversely, microinjection of U0126 into supragranular layers of A1 blocked nicotine's effect on intracortical, but not thalamocortical, CSD components. Simultaneously with enhancement of CF-evoked responses, responses to spectrally distant (nonCF) stimuli were reduced, implying nicotinic "sharpening" of frequency receptive fields, an effect also blocked by MEK inhibition. Consistent with these physiological results, acoustic stimulation with nicotine produced immunolabel for activated MAPK in A1, primarily in layer 2/3 cell bodies. Immunolabel was blocked by intracortical microinjection of the nAChR antagonist dihydro-ß-erythroidine, but not methyllycaconitine, implicating α4ß2*, but not α7, nAChRs. Thus activation of MAPK in functionally distinct forebrain circuits--thalamocortical, local intracortical, and long-range intracortical--underlies nicotinic neuromodulation of A1.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Nicotina/farmacología , Receptores Nicotínicos/metabolismo , Tálamo/efectos de los fármacos , Estimulación Acústica , Animales , Corteza Auditiva/metabolismo , Butadienos/farmacología , Inhibidores Enzimáticos/farmacología , Potenciales Evocados Auditivos/efectos de los fármacos , Potenciales Evocados Auditivos/fisiología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Masculino , Ratones , Vías Nerviosas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nitrilos/farmacología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Tálamo/metabolismo
11.
Hepatol Commun ; 6(1): 133-160, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34558855

RESUMEN

Alcohol-associated liver disease (ALD) is a significant clinical problem for which the most effective therapy is alcohol abstinence. The two aims of this study were, first, to identify the liver transcriptome, fecal microbiome, and portal serum metabolome at peak injury and during early and late resolution from ALD; and second, to integrate their interactions and understand better the pathogenesis of ALD. To provoke alcohol-induced liver injury, female and male wild-type mice were fed the control or ethanol Lieber-DeCarli diets for 6 weeks. To study early and late resolution, alcohol was withdrawn from the diet and mice were sacrificed after 3 and 14 days, respectively. At peak injury, there was increased signal transducer and activator of transcription (Stat3), Rho-GTPases, Tec kinase and glycoprotein VI (Gp6), and decreased peroxisome proliferator-activated receptor signaling. During resolution from ALD, there was up-regulation of vitamin D receptor/retinoid X receptor, toll-like receptor, p38 and Stat3, and down-regulation of liver X receptor signaling. Females showed significant changes in catabolic pathways, whereas males increased cellular stress, injury, and immune-response pathways that decreased during resolution. The bacterial genus Alistipes and the metabolite dipeptide glycyl-L-leucine increased at peak but decreased during resolution from ALD in both genders. Hepatic induction of mitogen-activated protein kinase (Map3k1) correlated with changes in the microbiome and metabolome at peak but was restored during ALD resolution. Inhibition of MAP3K1 protected from ALD in mice. Conclusion: Alcohol abstinence restores the liver transcriptome, fecal microbiome, and portal serum metabolome in a gender-specific manner. Integration of multiomics data identified Map3k1 as a key gene driving pathogenesis and resolution from ALD.


Asunto(s)
Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Metaboloma , Microbiota , Transcriptoma , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Heces/microbiología , Femenino , Hepatocitos/metabolismo , Hepatopatías Alcohólicas/microbiología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Masculino , Ratones Endogámicos C57BL , Regulación hacia Arriba
12.
Neuropathology ; 31(3): 250-64, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21118399

RESUMEN

Hypotonicity following water intoxication and/or salt loss leads to mainly astrocytic brain swelling. Astrocytic swelling also occurs following brain trauma or ischemia, together with an increase in extracellular K(+) ([K(+)](o)), stimulating a bumetanide/furosemide/ethacrynic acid-inhibitable cotransporter, NKCC1, that accumulates Na(+) and K(+) together with 2 Cl(-) and osmotically obliged water. Either type of swelling may become fatal and is associated with phosphorylation of extracellular regulated kinases 1 and 2 (ERK(1/2)). Only the swelling associated with elevated [K(+)](o), leads to an increase in astrocytic proliferation and in expression of the astrocytic marker, glial fibrillary acidic protein. These differences prompted us to investigate key aspects of the molecular pathways between hypotonicity-induced and high-K(+)-mediated swelling in primary cultures of mouse astrocytes. In the latter Ca(2+)-mediated, AG1478-inhibitable transactivation of the epidermal growth factor (EGF) receptor leads, via bumetanide-inhibitable activation of the mitogen activated protein (MAP) kinase pathway to ERK phosphorylation and to NKCC1-mediated swelling. In the former, inhibition of the MAP kinase pathway, but not of EGF receptor activation, abolishes ERK phosphorylation, but has no effect on swelling, indicating that activation of ERK is a result, not a cause, of the swelling.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Edema Encefálico/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Intoxicación por Agua/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Astrocitos/efectos de los fármacos , Edema Encefálico/patología , Bumetanida/farmacología , División Celular/fisiología , Células Cultivadas , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Receptores ErbB/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina , Soluciones Hipotónicas/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Fosforilación/fisiología , Potasio/farmacología , ARN Interferente Pequeño , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Intoxicación por Agua/patología , Familia-src Quinasas/antagonistas & inhibidores
13.
J Pharmacol Exp Ther ; 335(1): 23-31, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20627998

RESUMEN

E6201 [(3S,4R,5Z,8S,9S,11E)-14-(ethylamino)-8,9,16-trihydroxy-3,4-dimethyl-3,4,9,10-tetrahydro-1H-2-benzoxacyclotetradecine-1,7(8H)-dione)] is a novel anti-inflammatory agent that has potent inhibitory effects on the production of proinflammatory cytokines from leukocytes and antiproliferative activity on keratinocytes. To characterize the in vivo pharmacological activity of E6201, topically administered E6201 was evaluated in several different animal models of dermatitis. E6201 formulated as an ointment or cream showed dose-dependent inhibition of croton oil-induced acute edema formation and neutrophil infiltration into mouse skin. In addition, E6201 cream inhibited the 1-fluoro-2,4-dinitrobenzene-induced contact hypersensitivity reaction mediated by T cells in mice. In this model, E6201 cream also suppressed the migration of neutrophils and lymphocytes into the inflammatory site. Pretreatment with E6201 cream attenuated phorbol-12 myristate 13-acetate-induced ornithine decarboxylase activity, a marker of proliferation in epidermis. Furthermore, E6201 ointment showed inhibitory effects on both mezerein-induced and interleukin (IL)-23-induced epidermal hyperplasia. E6201 also suppressed T cell receptor-stimulated IL-17 production from human T cells. These results indicate that topically administered E6201 may be a useful agent for the prevention and treatment of cutaneous inflammatory and hyperproliferative diseases such as psoriasis.


Asunto(s)
Erupciones por Medicamentos/patología , Lactonas/farmacología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Administración Tópica , Animales , Antineoplásicos Fitogénicos , Aceite de Crotón , Dinitrofluorobenceno , Diterpenos , Hiperplasia/inducido químicamente , Hiperplasia/patología , Indicadores y Reactivos , Interleucina-17/biosíntesis , Interleucina-23/toxicidad , Lactonas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ornitina Descarboxilasa/biosíntesis , Ornitina Descarboxilasa/metabolismo , Peroxidasa/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Psoriasis/inducido químicamente , Psoriasis/patología , Piel/patología , Linfocitos T/fisiología , Acetato de Tetradecanoilforbol/farmacología
14.
Expert Opin Pharmacother ; 21(16): 1943-1953, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32880495

RESUMEN

INTRODUCTION: RAS-RAF-MEK-ERK signaling is implicated in tumor development by promoting cell proliferation and other cancer hallmarks. MEK1/2 kinases are up-regulated in the majority of human cancers due to activation of tyrosine kinase receptors, RAS proteins, BRAF kinase, or some other members of the MAPK pathway. Targeting of MEK1/2 kinases may counterbalance cancer progression. AREAS COVERED: The authors analyze the scientific publications relevant to selumetinib (AZD6244, ARRY-142886) systematically and provide their expert opinion. EXPERT OPINION: Selumetinib is an oral selective allosteric inhibitor of MEK1 and MEK2 kinases. Single-agent selumetinib is usually administered in hydrogen sulfate capsules 75 mg twice a day; combination with other therapeutic compounds may or may not require reduced dosing of this drug. The established dose for pediatric patients is 25 mg per square meter twice a day. Selumetinib was extensively evaluated in non-small cell lung cancer (NSCLC) patients. Studies utilizing this drug as a monotherapy did not confirm its efficacy toward NSCLC. A phase II trial showed that the addition of selumetinib to docetaxel improved response rates and progression-free survival (PFS) in chemotherapy-pretreated KRAS-mutated NSCLC patients; however, a subsequent phase III study did not confirm these findings. There are several highly successful non-NSCLC selumetinib trials involving, e.g., patients with neurofibromatosis type 1 related tumors and children with low-grade BRAF-driven gliomas.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bencimidazoles/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Proliferación Celular/efectos de los fármacos , Niño , Ensayos Clínicos como Asunto , Esquema de Medicación , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 2/antagonistas & inhibidores , Supervivencia sin Progresión
15.
Biochim Biophys Acta Mol Basis Dis ; 1866(8): 165806, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32320827

RESUMEN

Diabetic cardiomyopathy (DCM) accounts for increasing deaths of diabetic patients, and effective therapeutic targets are urgently needed. Myocardial lipotoxicity, which is caused by cardiac non-oxidative metabolic fatty acids and cardiotoxic fatty acid metabolites accumulation, has gained more attention to explain the increasing prevalence of DCM. However, whether mammalian Ste20-like kinase 1 (Mst1) plays a role in lipotoxicity in type 2 diabetes-induced cardiomyopathy has not yet been illustrated. Here, we found that Mst1 expression was elevated transcriptionally in the hearts of type 2 diabetes mellitus mice and palmitic acid-treated neonatal rat ventricular myocytes. Adeno-associated virus 9 (AAV9)-mediated Mst1 silencing in db/db mouse hearts significantly alleviated cardiac dysfunction and fibrosis. Notably, Mst1 knockdown in db/db mouse hearts decreased lipotoxic apoptosis and inflammatory response. Mst1 knockdown exerted protective effects through inactivation of MAPK/ERK kinase kinase 1 (MEKK1)/c-Jun N-terminal kinase (JNK) signaling pathway. Moreover, lipotoxicity induced Mst1 expression through promoting the binding of forkhead box O3 (FoxO3) and Mst1 promoter. Conclusively, we elucidated for the first time that Mst1 expression is regulated by FOXO3 under lipotoxicity stimulation and downregulation of Mst1 protects db/db mice from lipotoxic cardiac injury through MEKK1/JNK signaling inhibition, indicating that Mst1 abrogation may be a potential treatment strategy for DCM in type 2 diabetic patients.


Asunto(s)
Diabetes Mellitus Experimental/genética , Cardiomiopatías Diabéticas/genética , Ácidos Grasos/toxicidad , Proteína Forkhead Box O3/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Animales Recién Nacidos , Apoptosis/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/terapia , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/prevención & control , Ácidos Grasos/metabolismo , Proteína Forkhead Box O3/agonistas , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica , Factor de Crecimiento de Hepatocito , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidación-Reducción , Cultivo Primario de Células , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal
16.
Cell Death Differ ; 15(6): 1030-41, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18309325

RESUMEN

DJ-1 is a multifunctional protein that performs functions in transcriptional regulation and oxidative stress, and the loss of its function is believed to result in the onset of Parkinson's disease (PD). In this study, we report that DJ-1 protects against UV-induced cell death through the suppression of the JNK1 signaling pathway. The results of both binding and kinase studies have revealed that MEKK1 is the direct target of DJ-1. The C-terminus of DJ-1 was crucial to the inhibition of the MEKK1 kinase activity. Wild-type DJ-1 sequesters MEKK1 within the cytoplasm and the L166P mutant facilitates the translocation of MEKK1 toward the nucleus without physical association. Both DJ-1 knockdown and pathogenic L166P mutant were determined to be highly susceptible to the UV-induced activation of the MEKK1-SEK1-JNK1 signaling cascade and cell death. Taken together, our findings show that missense mutation in DJ-1 sensitizes cells to stress-induced cell death through the MEKK1-SEK1-JNK1 signaling pathway, a process, which may trigger the early onset of PD.


Asunto(s)
Apoptosis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Proteínas Oncogénicas/metabolismo , Estrés Oxidativo , Línea Celular , Núcleo Celular/enzimología , Citoplasma/enzimología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Mutación , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/fisiología , Proteína Desglicasa DJ-1 , Rayos Ultravioleta
17.
Immunology ; 128(1 Suppl): e325-34, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19016906

RESUMEN

Using two-dimensional sodium dodecyl sulphate-polyacrylamide gel electrophoresis, we found that copper/zinc superoxide dismutase (Cu/Zn-SOD, SOD-1) was induced in constructed CCR5 stably transfected HEK 293 cells, but not in mock cells, treated with CCL5. CCL5-induced SOD-1 expression was also confirmed in HEK 293-CCR5 cells and CCR5-positive granulocyte-macrophage colony-stimulating factor-induced human macrophages and murine macrophage RAW264.7 cells. CCL5 and CCR5 interaction induced SOD-1 expression mainly via MEK-ERK activation. In addition, we provided evidence that upregulation of SOD-1 by CCL5/CCR5 activation occurred in parallel with the increased release of tumour necrosis factor-alpha and nitric oxide and production of intracellular reactive oxygen species as well as enhanced nuclear factor-kappaB transcriptional activity in CCR5-positive RAW264.7 cells. Conversely, the MEK1/2 inhibitor PD98059 significantly inhibited SOD-1 expression with the decrease of these biological responses. More importantly, inhibition of SOD-1 activity by disulfiram also strongly inhibited the CCL5-induced biological effects. These data suggest that SOD-1 mediates CCR5 activation by CCL5 and that pharmacological modulation of SOD-1 may be beneficial to CCR5-associated diseases.


Asunto(s)
Quimiocina CCL5/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/inmunología , MAP Quinasa Quinasa Quinasa 2/inmunología , Macrófagos/inmunología , Receptores CCR5/metabolismo , Superóxido Dismutasa/biosíntesis , Androstadienos/farmacología , Animales , Línea Celular , Quimiocina CCL5/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , MAP Quinasa Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 2/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/agonistas , Óxido Nítrico/inmunología , Óxido Nítrico/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR5/agonistas , Superóxido Dismutasa-1 , Transfección , Factor de Necrosis Tumoral alfa/agonistas , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Wortmanina , Quinasa de Factor Nuclear kappa B
18.
J Pharmacol Exp Ther ; 331(2): 485-95, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19684251

RESUMEN

The goal of this study is to identify a novel inhibitor with anti-inflammatory and antiproliferative properties for the treatment of psoriasis. Compound f152A1 [(3S,5Z,8S,11E)-8,9,16-trihydroxy-14-methoxy-3-methyl-3,4,9,10-tetrahydro-1H-benzo[c][1]oxacyclotetradecine1,7(8H)-dione] was identified as the main active metabolite with strong inhibitory activity against tumor necrosis factor-alpha (TNFalpha) transcription in a fraction originated from the fermentation broth of the fungus Curvularia verruculosa. Although active in cell-based assays, f152A1 was unstable in plasma and liver microsome preparations, thus limiting its pharmaceutical utilization. To improve the metabolic properties of f152A1, a medicinal chemistry program was undertaken, resulting in the generation of over 400 analogs of f152A1. Eventually, E6201 [(3S,4R,5Z,8S,9S,11E)-14-(ethylamino)-8,9,16-trihydroxy-3,4-dimethyl-3,4,9,19-tetrahydro-1H-2-benzoxacyclotetradecine-1,7(8H)-dione] was identified as a promising analog in this series. In the present study, we characterized the in vitro activities of E6201 and discovered that the compound inhibits lipopolysaccharide-activated TNFalpha reporter activity in THP-1-33 cells with an IC(50) value of 50 nM and selectively inhibits mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK)-1 and MEK kinase-1 in cell-free biochemical assays. In addition, E6201 showed inhibitory activity in several other cell-based systems: 1) phosphorylation of c-jun N-terminal kinase and p38 MAPKs; 2) nuclear factor-kappaB and activated protein-1 activation in various cell types; 3) interleukin (IL)-2 production from human lymphocytes; 4) hyperproliferation of human keratinocytes; 5) IL-8 production from human keratinocytes; and 6) proinflammatory cytokine production from human peripheral blood mononuclear cells. Based on the data presented here, E6201 may be beneficial for treatment of inflammatory and hyperproliferative diseases such as psoriasis through its anti-inflammatory activities on immune cells and antihyperproliferative activities on keratinocytes.


Asunto(s)
Antiinflamatorios/farmacología , Proliferación Celular/efectos de los fármacos , Lactonas/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Actinas/metabolismo , Línea Celular , Citocinas/biosíntesis , Humanos , Inmunidad Celular/efectos de los fármacos , Indicadores y Reactivos , Interleucina-2/biosíntesis , Interleucina-8/biosíntesis , Células Jurkat , Queratinocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Fosforilación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
19.
Mol Cancer Ther ; 18(3): 706-717, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30523048

RESUMEN

Tumor heterogeneity is a major challenge for cancer treatment, especially due to the presence of various subpopulations with stem cell or progenitor cell properties. In mouse melanomas, both CD34+p75- (CD34+) and CD34-p75- (CD34-) tumor subpopulations were characterized as melanoma-propagating cells (MPC) that exhibit some of those key features. However, these two subpopulations differ from each other in tumorigenic potential, ability to recapitulate heterogeneity, and chemoresistance. In this study, we demonstrate that CD34+ and CD34- subpopulations carrying the BRAFV600E mutation confer differential sensitivity to targeted BRAF inhibition. Through elevated KDM5B expression, melanoma cells shift toward a more drug-tolerant, CD34- state upon exposure to BRAF inhibitor or combined BRAF inhibitor and MEK inhibitor treatment. KDM5B loss or inhibition shifts melanoma cells to the more BRAF inhibitor-sensitive CD34+ state. These results support that KDM5B is a critical epigenetic regulator that governs the transition of key MPC subpopulations with distinct drug sensitivity. This study also emphasizes the importance of continuing to advance our understanding of intratumor heterogeneity and ultimately develop novel therapeutics by altering the heterogeneous characteristics of melanoma.


Asunto(s)
Antígenos CD34/genética , Proteínas de Unión al ADN/genética , Histona Demetilasas con Dominio de Jumonji/genética , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Línea Celular Tumoral , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/genética , Melanoma/genética , Melanoma/patología , Ratones , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/farmacología , Vemurafenib/farmacología
20.
Sci Rep ; 9(1): 18630, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31819100

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) is a key driver of acute myeloid leukemia (AML). Several tyrosine kinase inhibitors (TKIs) targeting FLT3 have been evaluated clinically, but their effects are limited when used in monotherapy due to the emergence of drug-resistance. Thus, a better understanding of drug-resistance pathways could be a good strategy to explore and evaluate new combinational therapies for AML. Here, we used phosphoproteomics to identify differentially-phosphorylated proteins in patients with AML and TKI resistance. We then studied resistance mechanisms in vitro and evaluated the efficacy and safety of rational combinational therapy in vitro, ex vivo and in vivo in mice. Proteomic and immunohistochemical studies showed the sustained activation of ERK1/2 in bone marrow samples of patients with AML after developing resistance to FLT3 inhibitors, which was identified as a common resistance pathway. We examined the concomitant inhibition of MEK-ERK1/2 and FLT3 as a strategy to overcome drug-resistance, finding that the MEK inhibitor trametinib remained potent in TKI-resistant cells and exerted strong synergy when combined with the TKI midostaurin in cells with mutated and wild-type FLT3. Importantly, this combination was not toxic to CD34+ cells from healthy donors, but produced survival improvements in vivo when compared with single therapy groups. Thus, our data point to trametinib plus midostaurin as a potentially beneficial therapy in patients with AML.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación , Estaurosporina/análogos & derivados , Tirosina Quinasa 3 Similar a fms/genética , Adulto , Anciano , Animales , Antígenos CD34/genética , Células de la Médula Ósea/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Persona de Mediana Edad , Estaurosporina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA