Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
JAMA ; 313(8): 815-23, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25710658

RESUMEN

IMPORTANCE: With cure rates of childhood acute lymphoblastic leukemia (ALL) exceeding 85%, there is a need to mitigate treatment toxicities that can compromise quality of life, including peripheral neuropathy from vincristine treatment. OBJECTIVE: To identify genetic germline variants associated with the occurrence or severity of vincristine-induced peripheral neuropathy in children with ALL. DESIGN, SETTING, AND PARTICIPANTS: Genome-wide association study of patients in 1 of 2 prospective clinical trials for childhood ALL that included treatment with 36 to 39 doses of vincristine. Genome-wide single-nucleotide polymorphism (SNP) analysis and vincristine-induced peripheral neuropathy were assessed in 321 patients from whom DNA was available: 222 patients (median age, 6.0 years; range, 0.1-18.8 years) enrolled in 1994-1998 in the St Jude Children's Research Hospital protocol Total XIIIB with toxic effects follow-up through January 2001, and 99 patients (median age, 11.4 years; range, 3.0-23.8 years) enrolled in 2007-2010 in the Children's Oncology Group (COG) protocol AALL0433 with toxic effects follow-up through May 2011. Human leukemia cells and induced pluripotent stem cell neurons were used to assess the effects of lower CEP72 expression on vincristine sensitivity. EXPOSURE: Treatment with vincristine at a dose of 1.5 or 2.0 mg/m2. MAIN OUTCOMES AND MEASURES: Vincristine-induced peripheral neuropathy was assessed at clinic visits using National Cancer Institute criteria and prospectively graded as mild (grade 1), moderate (grade 2), serious/disabling (grade 3), or life threatening (grade 4). RESULTS: Grade 2 to 4 vincristine-induced neuropathy during continuation therapy occurred in 28.8% of patients (64/222) in the St Jude cohort and in 22.2% (22/99) in the COG cohort. A SNP in the promoter region of the CEP72 gene, which encodes a centrosomal protein involved in microtubule formation, had a significant association with vincristine neuropathy (meta-analysis P = 6.3×10(-9)). This SNP had a minor allele frequency of 37% (235/642), with 50 of 321 patients (16%; 95% CI, 11.6%-19.5%) homozygous for the risk allele (TT at rs924607). Among patients with the high-risk CEP72 genotype (TT at rs924607), 28 of 50 (56%; 95% CI, 41.2%-70.0%) developed at least 1 episode of grade 2 to 4 neuropathy, a higher rate than in patients with the CEP72 CC or CT genotypes (58/271 patients [21.4%; 95% CI, 16.9%-26.7%]; P = 2.4×10(-6)). The severity of neuropathy was greater in patients homozygous for the TT genotype compared with patients with the CC or CT genotype (2.4-fold by Poisson regression [P<.0001] and 2.7-fold based on mean grade of neuropathy: 1.23 [95% CI, 0.74-1.72] vs 0.45 [95% CI, 0.3-0.6]; P = .004 by t test). Reducing CEP72 expression in human neurons and leukemia cells increased their sensitivity to vincristine. CONCLUSIONS AND RELEVANCE: In this preliminary study of children with ALL, an inherited polymorphism in the promoter region of CEP72 was associated with increased risk and severity of vincristine-related peripheral neuropathy. If replicated in additional populations, this finding may provide a basis for safer dosing of this widely prescribed anticancer agent.


Asunto(s)
Antineoplásicos Fitogénicos/efectos adversos , Proteínas Asociadas a Microtúbulos/genética , Enfermedades del Sistema Nervioso Periférico/genética , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Vincristina/efectos adversos , Adolescente , Antineoplásicos Fitogénicos/administración & dosificación , Niño , Preescolar , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Vincristina/administración & dosificación , Adulto Joven
2.
Hum Mol Genet ; 21(21): 4793-804, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22846425

RESUMEN

Treatment-related toxicity can be life-threatening and is the primary cause of interruption or discontinuation of chemotherapy for acute lymphoblastic leukemia (ALL), leading to an increased risk of relapse. Mercaptopurine is an essential component of continuation therapy in all ALL treatment protocols worldwide. Genetic polymorphisms in thiopurine S-methyltransferase (TPMT) are known to have a marked effect on mercaptopurine metabolism and toxicity; however, some patients with wild-type TPMT develop toxicity during mercaptopurine treatment for reasons that are not well understood. To identify additional genetic determinants of mercaptopurine toxicity, a genome-wide analysis was performed in a panel of human HapMap cell lines to identify trans-acting genes whose expression and/or single-nucleotide polymorphisms (SNPs) are related to TPMT activity, then validated in patients with ALL. The highest ranking gene with both mRNA expression and SNPs associated with TPMT activity in HapMap cell lines was protein kinase C and casein kinase substrate in neurons 2 (PACSIN2). The association of a PACSIN2 SNP (rs2413739) with TPMT activity was confirmed in patients and knock-down of PACSIN2 mRNA in human leukemia cells (NALM6) resulted in significantly lower TPMT activity. Moreover, this PACSIN2 SNP was significantly associated with the incidence of severe gastrointestinal (GI) toxicity during consolidation therapy containing mercaptopurine, and remained significant in a multivariate analysis including TPMT and SLCO1B1 as covariates, consistent with its influence on TPMT activity. The association with GI toxicity was also validated in a separate cohort of pediatric patients with ALL. These data indicate that polymorphism in PACSIN2 significantly modulates TPMT activity and influences the risk of GI toxicity associated with mercaptopurine therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Tracto Gastrointestinal , Mercaptopurina/efectos adversos , Metiltransferasas/genética , Línea Celular Tumoral , Niño , Preescolar , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/fisiopatología , Femenino , Tracto Gastrointestinal/fisiopatología , Expresión Génica , Estudio de Asociación del Genoma Completo , Genotipo , Proyecto Mapa de Haplotipos , Humanos , Masculino , Mercaptopurina/uso terapéutico , Metiltransferasas/metabolismo , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
4.
Cancer Epidemiol Biomarkers Prev ; 30(12): 2256-2267, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34583966

RESUMEN

BACKGROUND: Childhood cancer survivors are at elevated risk for motor and/or sensory neuropathy. The study aims to evaluate the concordance between self-report peripheral neuropathy compared with clinically ascertained peripheral neuropathy, and to identify factors associated with misclassification of peripheral neuropathy among survivors. METHODS: The concordance between self-report and clinically ascertained peripheral neuropathy was evaluated among 2,933 5+ years old childhood cancer survivors (mean age 33.3, SD = 8.9). The sensitivity, specificity, and accuracy of self-report peripheral motor neuropathy (PMN) and peripheral sensory neuropathy (PSN) were calculated with reference to clinically assessed peripheral neuropathy. RESULTS: Female survivors were more likely than male survivors to have clinically ascertained PMN (8.4% vs. 5.6%, P = 0.004). For females, having either PSN or PMN the most sensitive, specific, and accurate self-reported symptom was endorsing ≥2 symptoms on the self-report questionnaire (43.2%, 90.3%, and 85.2%, respectively), with kappa of 0.304. For males, having either PSN or PMN the most sensitive, specific, and accurate self-reported symptom was endorsing ≥2 symptoms on the self-report questionnaire (38.8%, 90.5%, and 86.3%, respectively) with kappa of 0.242. Age at diagnosis, emotional distress, and reporting pain in legs in the past 4 weeks were associated with an increased risk for false-positive reporting of peripheral neuropathy. Race (White), age at assessment, and emotional distress were associated with increased risk for false-negative reporting of peripheral neuropathy. CONCLUSIONS: Agreement between self-report and clinically ascertained peripheral neuropathy was poor in survivors. Choosing self-report versus clinical ascertained peripheral neuropathy should be carefully considered. IMPACT: The current study identifies the need for a self-report questionnaire that accurately assesses symptoms of peripheral neuropathy among cancer survivors.


Asunto(s)
Supervivientes de Cáncer/psicología , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Trastornos Somatomorfos/diagnóstico , Adulto , Supervivientes de Cáncer/estadística & datos numéricos , Femenino , Humanos , Masculino , Enfermedades del Sistema Nervioso Periférico/epidemiología , Enfermedades del Sistema Nervioso Periférico/psicología , Calidad de Vida , Autoinforme , Distribución por Sexo , Trastornos Somatomorfos/epidemiología , Trastornos Somatomorfos/psicología
5.
Clin Transl Sci ; 14(4): 1490-1504, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33742760

RESUMEN

Vincristine (VCR) is one of the most widely prescribed medications for treating solid tumors and acute lymphoblastic leukemia (ALL) in children and adults. However, its major dose-limiting toxicity is peripheral neuropathy that can disrupt curative therapy. Peripheral neuropathy can also persist into adulthood, compromising quality of life of childhood cancer survivors. Reducing VCR-induced neurotoxicity without compromising its anticancer effects would be ideal. Here, we show that low expression of NHP2L1 is associated with increased sensitivity of primary leukemia cells to VCR, and that concomitant administration of VCR with inhibitors of NHP2L1 increases VCR cytotoxicity in leukemia cells, prolongs survival of ALL xenograft mice, but decreases VCR effects on human-induced pluripotent stem cell-derived neurons and mitigates neurotoxicity in mice. These findings offer a strategy for increasing VCR's antileukemic effects while reducing peripheral neuropathy in patients treated with this widely prescribed medication.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Enfermedades del Sistema Nervioso Periférico/prevención & control , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Ribonucleoproteínas Nucleares Pequeñas/antagonistas & inhibidores , Vincristina/efectos adversos , Adolescente , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Células Cultivadas , Niño , Resistencia a Antineoplásicos/genética , Femenino , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Ratones , Neuronas , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Cultivo Primario de Células , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Vincristina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
6.
J Clin Invest ; 130(12): 6600-6615, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33164984

RESUMEN

BACKGROUNDInterpatient differences in the accumulation of methotrexate's active polyglutamylated metabolites (MTXPGs) in leukemia cells influence its antileukemic effects.METHODSTo identify genomic and epigenomic and patient variables determining the intracellular accumulation of MTXPGs, we measured intracellular MTXPG levels in acute lymphoblastic leukemia (ALL) cells from 388 newly diagnosed patients after in vivo high-dose methotrexate (HDMTX) (1 g/m2) treatment, defined ALL subtypes, and assessed genomic and epigenomic variants influencing folate pathway genes (mRNA, miRNA, copy number alterations [CNAs], SNPs, single nucleotide variants [SNVs], CpG methylation).RESULTSWe documented greater than 100-fold differences in MTXPG levels, which influenced its antileukemic effects (P = 4 × 10-5). Three ALL subtypes had lower MTXPG levels (T cell ALL [T-ALL] and B cell ALL [B-ALL] with the TCF3-PBX1 or ETV6-RUNX1 fusions), and 2 subtypes had higher MTXPG levels (hyperdiploid and BCR-ABL like). The folate pathway genes SLC19A1, ABCC1, ABCC4, FPGS, and MTHFD1 significantly influenced intracellular MTXPG levels (P = 2.9 × 10-3 to 3.7 × 10-8). A multivariable model including the ALL subtype (P = 1.1 × 10-14), the SLC19A1/(ABCC1 + ABCC4) transporter ratio (P = 3.6 × 10-4), the MTX infusion time (P = 1.5 × 10-3), FPGS mRNA expression (P = 2.1 × 10-3), and MTX systemic clearance (P = 4.4 × 10-2) explained 42% of the variation in MTXPG accumulation (P = 1.1 × 10-38). Model simulations indicated that a longer infusion time (24 h vs. 4 h) was superior in achieving higher intracellular MTXPG levels across all subtypes if ALL.CONCLUSIONSThese findings provide insights into mechanisms underlying interpatient differences in intracellular accumulation of MTXPG in leukemia cells and its antileukemic effectsFUNDINGTHE National Cancer Institute (NCI) and the Institute of General Medical Sciences of the NIH, the Basque Government Programa Posdoctoral de Perfeccionamiento de Personal Investigador doctor, and the American Lebanese Syrian Associated Charities (ALSAC).


Asunto(s)
Metotrexato/análogos & derivados , Proteínas de Neoplasias , Ácido Poliglutámico/análogos & derivados , Leucemia-Linfoma Linfoblástico de Células Precursoras , Adolescente , Línea Celular Tumoral , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Metotrexato/farmacocinética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Pruebas de Farmacogenómica , Ácido Poliglutámico/farmacocinética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
7.
Nat Cancer ; 1(3): 329-344, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32885175

RESUMEN

Identification of genomic and epigenomic determinants of drug resistance provides important insights for improving cancer treatment. Using agnostic genome-wide interrogation of mRNA and miRNA expression, DNA methylation, SNPs, CNAs and SNVs/Indels in primary human acute lymphoblastic leukemia cells, we identified 463 genomic features associated with glucocorticoid resistance. Gene-level aggregation identified 118 overlapping genes, 15 of which were confirmed by genome-wide CRISPR screen. Collectively, this identified 30 of 38 (79%) known glucocorticoid-resistance genes/miRNAs and all 38 known resistance pathways, while revealing 14 genes not previously associated with glucocorticoid-resistance. Single cell RNAseq and network-based transcriptomic modelling corroborated the top previously undiscovered gene, CELSR2. Manipulation of CELSR2 recapitulated glucocorticoid resistance in human leukemia cell lines and revealed a synergistic drug combination (prednisolone and venetoclax) that mitigated resistance in mouse xenograft models. These findings illustrate the power of an integrative genomic strategy for elucidating genes and pathways conferring drug resistance in cancer cells.


Asunto(s)
MicroARNs , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Resistencia a Antineoplásicos/genética , Genómica , Glucocorticoides/farmacología , Humanos , Ratones , MicroARNs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
8.
Mol Cell Biol ; 26(6): 2247-61, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16508001

RESUMEN

In pituitary GH3B6 cells, signaling involving the protein kinase C (PKC) multigene family can self-organize into a spatiotemporally coordinated cascade of isoform activation. Indeed, thyrotropin-releasing hormone (TRH) receptor activation sequentially activated green fluorescent protein (GFP)-tagged or endogenous PKCbeta1, PKCalpha, PKCepsilon, and PKCdelta, resulting in their accumulation at the entire plasma membrane (PKCbeta and -delta) or selectively at the cell-cell contacts (PKCalpha and -epsilon). The duration of activation ranged from 20 s for PKCalpha to 20 min for PKCepsilon. PKCalpha and -epsilon selective localization was lost in the presence of Gö6976, suggesting that accumulation at cell-cell contacts is dependent on the activity of a conventional PKC. Constitutively active, dominant-negative PKCs and small interfering RNAs showed that PKCalpha localization is controlled by PKCbeta1 activity and is calcium independent, while PKCepsilon localization is dependent on PKCalpha activity. PKCdelta was independent of the cascade linking PKCbeta1, -alpha, and -epsilon. Furthermore, PKCalpha, but not PKCepsilon, is involved in the TRH-induced beta-catenin relocation at cell-cell contacts, suggesting that PKCepsilon is not the unique functional effector of the cascade. Thus, TRH receptor activation results in PKCbeta1 activation, which in turn initiates a calcium-independent but PKCbeta1 activity-dependent sequential translocation of PKCalpha and -epsilon. These results challenge the current understanding of PKC signaling and raise the question of a functional dependence between isoforms.


Asunto(s)
Proteína Quinasa C/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Carbazoles/farmacología , Comunicación Celular , Línea Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Ionomicina/farmacología , Isoenzimas/genética , Isoenzimas/metabolismo , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/genética , Proteína Quinasa C beta , Transporte de Proteínas , Ratas , Receptores de Hormona Liberadora de Tirotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Tirotropina/metabolismo , Hormona Liberadora de Tirotropina/metabolismo , Hormona Liberadora de Tirotropina/farmacología , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo
9.
Lancet Child Adolesc Health ; 2(10): 744-754, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30236383

RESUMEN

Peripheral neuropathy is a well recognised treatment-related toxicity in children with cancer, associated with exposure to neurotoxic chemotherapy agents. Acute damage can occur in sensory, motor, or autonomic neurons, with symptoms that are rarely life threatening, but often severe enough to interfere with function during therapy and after treatment ends. The type of neuropathy and specific symptoms are associated with multiple factors including age at time of therapy, genetic predisposition, chemotherapy type and cumulative dose, and exposure to other agents during therapy. In this Review, we describe the peripheral neuropathy phenotype in children during cancer therapy and among survivors who have completed therapy, to summarise genetic and treatment-related risk factors for neuropathy, and to outline strategies to monitor and detect neuropathy during and after therapy. Additionally, we outline strategies for medical management of neuropathy during treatment and potential rehabilitation interventions to prevent or remediate functional loss.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Adolescente , Niño , Humanos
10.
SLAS Discov ; 23(2): 164-173, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28985478

RESUMEN

Splicing is an important eukaryotic mechanism for expanding the transcriptome and proteome, influencing a number of biological processes. Understanding its regulation and identifying small molecules that modulate this process remain a challenge. We developed an assay based on time-resolved fluorescence resonance energy transfer (TR-FRET) to detect the interaction between the protein NHP2L1 and U4 RNA, which are two key components of the spliceosome. We used this assay to identify small molecules that interfere with this interaction in a high-throughput screening (HTS) campaign. Topotecan and other camptothecin derivatives were among the top hits. We confirmed that topotecan disrupts the interaction between NHP2L1 and U4 by binding to U4 and inhibits RNA splicing. Our data reveal new functions of known drugs that could facilitate the development of therapeutic strategies to modify splicing and alter gene function.


Asunto(s)
Empalme del ARN/efectos de los fármacos , ARN Nuclear Pequeño/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Topotecan/farmacología , Transferencia Resonante de Energía de Fluorescencia/métodos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Empalmosomas/efectos de los fármacos
12.
Sci Rep ; 6: 30757, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27476972

RESUMEN

A feature in patients with constitutional DNA-mismatch repair deficiency is agenesis of the corpus callosum, the cause of which has not been established. Here we report a previously unrecognized consequence of deficiency in MSH2, a protein known primarily for its function in correcting nucleotide mismatches or insertions and deletions in duplex DNA caused by errors in DNA replication or recombination. We documented that Msh2 deficiency causes dysmyelination of the axonal projections in the corpus callosum. Evoked action potentials in the myelinated corpus callosum projections of Msh2-null mice were smaller than wild-type mice, whereas unmyelinated axons showed no difference. Msh2-null mice were also impaired in locomotive activity and had an abnormal response to heat. These findings reveal a novel pathogenic consequence of MSH2 deficiency, providing a new mechanistic hint to previously recognized neurological disorders in patients with inherited DNA-mismatch repair deficiency.


Asunto(s)
Cuerpo Calloso , Reparación de la Incompatibilidad de ADN , Enfermedades Desmielinizantes , Potenciales Evocados , Locomoción , Proteína 2 Homóloga a MutS/deficiencia , Animales , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Cuerpo Calloso/fisiopatología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/fisiopatología , Ratones , Ratones Noqueados , Proteína 2 Homóloga a MutS/metabolismo
13.
Nat Genet ; 47(6): 607-14, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25938942

RESUMEN

Glucocorticoids are universally used in the treatment of acute lymphoblastic leukemia (ALL), and resistance to glucocorticoids in leukemia cells confers poor prognosis. To elucidate mechanisms of glucocorticoid resistance, we determined the prednisolone sensitivity of primary leukemia cells from 444 patients newly diagnosed with ALL and found significantly higher expression of CASP1 (encoding caspase 1) and its activator NLRP3 in glucocorticoid-resistant leukemia cells, resulting from significantly lower somatic methylation of the CASP1 and NLRP3 promoters. Overexpression of CASP1 resulted in cleavage of the glucocorticoid receptor, diminished the glucocorticoid-induced transcriptional response and increased glucocorticoid resistance. Knockdown or inhibition of CASP1 significantly increased glucocorticoid receptor levels and mitigated glucocorticoid resistance in CASP1-overexpressing ALL. Our findings establish a new mechanism by which the NLRP3-CASP1 inflammasome modulates cellular levels of the glucocorticoid receptor and diminishes cell sensitivity to glucocorticoids. The broad impact on the glucocorticoid transcriptional response suggests that this mechanism could also modify glucocorticoid effects in other diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Receptores de Glucocorticoides/metabolismo , Adolescente , Antineoplásicos Hormonales/farmacología , Secuencia de Bases , Niño , Preescolar , Metilación de ADN , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Lactante , Recién Nacido , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Recurrencia Local de Neoplasia/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Prednisolona/farmacología , Proteolisis , Transcripción Genética , Células Tumorales Cultivadas , Regulación hacia Arriba
15.
Nat Med ; 17(10): 1298-303, 2011 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-21946537

RESUMEN

DNA mismatch repair enzymes (for example, MSH2) maintain genomic integrity, and their deficiency predisposes to several human cancers and to drug resistance. We found that leukemia cells from a substantial proportion of children (∼11%) with newly diagnosed acute lymphoblastic leukemia have low or undetectable MSH2 protein levels, despite abundant wild-type MSH2 mRNA. Leukemia cells with low levels of MSH2 contained partial or complete somatic deletions of one to four genes that regulate MSH2 degradation (FRAP1 (also known as MTOR), HERC1, PRKCZ and PIK3C2B); we also found these deletions in individuals with adult acute lymphoblastic leukemia (16%) and sporadic colorectal cancer (13.5%). Knockdown of these genes in human leukemia cells recapitulated the MSH2 protein deficiency by enhancing MSH2 degradation, leading to substantial reduction in DNA mismatch repair and increased resistance to thiopurines. These findings reveal a previously unrecognized mechanism whereby somatic deletions of genes regulating MSH2 degradation result in undetectable levels of MSH2 protein in leukemia cells, DNA mismatch repair deficiency and drug resistance.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Resistencia a Antineoplásicos/genética , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Adulto , Western Blotting , Línea Celular Tumoral , Niño , Fosfatidilinositol 3-Quinasas Clase II , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/deficiencia , Humanos , Fosfatidilinositol 3-Quinasas/deficiencia , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales , Proteína Quinasa C/deficiencia , Serina-Treonina Quinasas TOR/deficiencia , Tioguanina , Ubiquitina-Proteína Ligasas
16.
J Biol Chem ; 284(28): 18808-15, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19429675

RESUMEN

In the pituitary gland, activated protein kinase C (PKC) isoforms accumulate either selectively at the cell-cell contact (alpha and epsilon) or at the entire plasma membrane (beta1 and delta). The molecular mechanisms underlying these various subcellular locations are not known. Here, we demonstrate the existence within PKCepsilon of a cell-cell contact targeting sequence (3CTS) that, upon stimulation, is capable of targeting PKCdelta, chimerin-alpha1, and the PKCepsilon C1 domain to the cell-cell contact. We show that this selective targeting of PKCepsilon is lost upon overexpression of 3CTS fused to a (R-Ahx-R)(4) (where Ahx is 6-aminohexanoic acid) vectorization peptide, reflecting a dominant-negative effect of the overexpressed 3CTS on targeting selectivity. 3CTS contains a putative amphipathic alpha-helix, a 14-3-3-binding site, and the Glu-374 amino acid, involved in targeting selectivity. We show that the integrity of the alpha-helix is important for translocation but that 14-3-3 is not involved in targeting selectivity. However, PKCepsilon translocation is increased when PKCepsilon/14-3-3 interaction is abolished, suggesting that phorbol 12-myristate 13-acetate activation may initiate two sets of PKCepsilon functions, those depending on 14-3-3 and those depending on translocation to cell-cell contacts. Thus, 3CTS is involved in the modulation of translocation via its 14-3-3-binding site, in cytoplasmic desequestration via the alpha-helix, and in selective PKCepsilon targeting at the cell-cell contact via Glu-374.


Asunto(s)
Proteína Quinasa C-epsilon/fisiología , Proteínas 14-3-3/química , Secuencia de Aminoácidos , Aminoácidos/química , Sitios de Unión , Comunicación Celular , Ácido Glutámico/química , Humanos , Datos de Secuencia Molecular , Hipófisis/metabolismo , Conformación Proteica , Proteína Quinasa C-epsilon/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Acetato de Tetradecanoilforbol/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA