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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 108(5): 1272-1283, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36700399

RESUMEN

Hypodiploid acute lymphoblastic leukemia (ALL) is an aggressive blood cancer with a poor prognosis despite intensive chemotherapy or stem cell transplant. Children and adolescents with positive end-of-induction minimal residual disease have an overall survival lower than 30%. However, data regarding therapeutic alternatives for this disease is nearly nonexistent, emphasizing the critical need for new or adjunctive therapies that can improve outcomes. We previously reported on the therapeutic efficacy of venetoclax (ABT-199) in hypodiploid B-lineage ALL but with limitations as monotherapy. In this study, we set out to identify drugs enhancing the anti-leukemic effect of venetoclax in hypodiploid ALL. Using a highthroughput drug screen, we identified dinaciclib, a cyclin-dependent kinase inhibitor that worked synergistically with venetoclax to induce cell death in hypodiploid cell lines. This combination eradicated leukemic blasts within hypodiploid ALL patient-derived xenografts mice with low off-target toxicity. Our findings suggest that dual inhibition of BCL-2 (venetoclax) and CDK9/MCL-1 (dinaciclib) is a promising therapeutic approach in hypodiploid ALL, warranting further investigation to inform clinical trials in this high-risk patient population.


Asunto(s)
Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Animales , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Línea Celular Tumoral , Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2 , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Antineoplásicos/farmacología
2.
Br J Haematol ; 190(5): 718-722, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32542672

RESUMEN

Complementary tools are warranted to increase the sensitivity of the initial testing for COVID-19. We identified a specific 'sandglass' aspect on the white blood cell scattergram of COVID-19 patients reflecting the presence of circulating plasmacytoid lymphocytes. Patients were dichotomized as COVID-19-positive or -negative based on reverse transcriptase polymerase chain reaction (RT-PCR) and chest computed tomography (CT) scan results. Sensitivity and specificity of the 'sandglass' aspect were 85·9% and 83·5% respectively. The positive predictive value was 94·3%. Our findings provide a non-invasive and simple tool to quickly categorize symptomatic patients as either COVID-19-probable or -improbable especially when RT-PCR and/or chest CT are not rapidly available.


Asunto(s)
Betacoronavirus/metabolismo , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/diagnóstico , Linfocitos/metabolismo , Tamizaje Masivo , Neumonía Viral/sangre , Neumonía Viral/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , COVID-19 , Infecciones por Coronavirus/diagnóstico por imagen , Femenino , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/diagnóstico por imagen , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , SARS-CoV-2 , Tomografía Computarizada por Rayos X
3.
Blood ; 129(11): e26-e37, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28122742

RESUMEN

Drug sensitivity and resistance testing on diagnostic leukemia samples should provide important functional information to guide actionable target and biomarker discovery. We provide proof of concept data by profiling 60 drugs on 68 acute lymphoblastic leukemia (ALL) samples mostly from resistant disease in cocultures of bone marrow stromal cells. Patient-derived xenografts retained the original pattern of mutations found in the matched patient material. Stromal coculture did not prevent leukemia cell cycle activity, but a specific sensitivity profile to cell cycle-related drugs identified samples with higher cell proliferation both in vitro and in vivo as leukemia xenografts. In patients with refractory relapses, individual patterns of marked drug resistance and exceptional responses to new agents of immediate clinical relevance were detected. The BCL2-inhibitor venetoclax was highly active below 10 nM in B-cell precursor ALL (BCP-ALL) subsets, including MLL-AF4 and TCF3-HLF ALL, and in some T-cell ALLs (T-ALLs), predicting in vivo activity as a single agent and in combination with dexamethasone and vincristine. Unexpected sensitivity to dasatinib with half maximal inhibitory concentration values below 20 nM was detected in 2 independent T-ALL cohorts, which correlated with similar cytotoxic activity of the SRC inhibitor KX2-391 and inhibition of SRC phosphorylation. A patient with refractory T-ALL was treated with dasatinib on the basis of drug profiling information and achieved a 5-month remission. Thus, drug profiling captures disease-relevant features and unexpected sensitivity to relevant drugs, which warrants further exploration of this functional assay in the context of clinical trials to develop drug repurposing strategies for patients with urgent medical needs.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Células Cultivadas , Técnicas de Cocultivo , Xenoinjertos , Humanos , Células Madre Mesenquimatosas/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
4.
Genes Dev ; 24(13): 1389-402, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20595231

RESUMEN

The p53 tumor suppressor limits proliferation in response to cellular stress through several mechanisms. Here, we test whether the recently described ability of p53 to limit stem cell self-renewal suppresses tumorigenesis in acute myeloid leukemia (AML), an aggressive cancer in which p53 mutations are associated with drug resistance and adverse outcome. Our approach combined mosaic mouse models, Cre-lox technology, and in vivo RNAi to disable p53 and simultaneously activate endogenous Kras(G12D)-a common AML lesion that promotes proliferation but not self-renewal. We show that p53 inactivation strongly cooperates with oncogenic Kras(G12D) to induce aggressive AML, while both lesions on their own induce T-cell malignancies with long latency. This synergy is based on a pivotal role of p53 in limiting aberrant self-renewal of myeloid progenitor cells, such that loss of p53 counters the deleterious effects of oncogenic Kras on these cells and enables them to self-renew indefinitely. Consequently, myeloid progenitor cells expressing oncogenic Kras and lacking p53 become leukemia-initiating cells, resembling cancer stem cells capable of maintaining AML in vivo. Our results establish an efficient new strategy for interrogating oncogene cooperation, and provide strong evidence that the ability of p53 to limit aberrant self-renewal contributes to its tumor suppressor activity.


Asunto(s)
Leucemia Mieloide Aguda/fisiopatología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proliferación Celular , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN/genética , Eliminación de Secuencia/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
5.
Blood ; 124(22): 3274-83, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25316678

RESUMEN

Mutant RAS oncoproteins activate signaling molecules that drive oncogenesis in multiple human tumors including acute myelogenous leukemia (AML). However, the specific functions of these pathways in AML are unclear, thwarting the rational application of targeted therapeutics. To elucidate the downstream functions of activated NRAS in AML, we used a murine model that harbors Mll-AF9 and a tetracycline-repressible, activated NRAS (NRAS(G12V)). Using computational approaches to explore our gene-expression data sets, we found that NRAS(G12V) enforced the leukemia self-renewal gene-expression signature and was required to maintain an MLL-AF9- and Myb-dependent leukemia self-renewal gene-expression program. NRAS(G12V) was required for leukemia self-renewal independent of its effects on growth and survival. Analysis of the gene-expression patterns of leukemic subpopulations revealed that the NRAS(G12V)-mediated leukemia self-renewal signature is preferentially expressed in the leukemia stem cell-enriched subpopulation. In a multiplexed analysis of RAS-dependent signaling, Mac-1(Low) cells, which harbor leukemia stem cells, were preferentially sensitive to NRAS(G12V) withdrawal. NRAS(G12V) maintained leukemia self-renewal through mTOR and MEK pathway activation, implicating these pathways as potential targets for cancer stem cell-specific therapies. Together, these experimental results define a RAS oncogene-driven function that is critical for leukemia maintenance and represents a novel mechanism of oncogene addiction.


Asunto(s)
Proliferación Celular/genética , GTP Fosfohidrolasas/fisiología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de la Membrana/fisiología , Sustitución de Aminoácidos , Animales , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , GTP Fosfohidrolasas/genética , Regulación Leucémica de la Expresión Génica , Glicina/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones SCID , Oncogenes/fisiología , Transcriptoma , Células Tumorales Cultivadas , Valina/genética
6.
Haematologica ; 101(10): 1190-1199, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27418650

RESUMEN

Juvenile myelomonocytic leukemia is a rare myeloproliferative neoplasm characterized by hyperactive RAS signaling. Neurofibromin1 (encoded by the NF1 gene) is a negative regulator of RAS activation. Patients with neurofibromatosis type 1 harbor loss-of-function mutations in NF1 and have a 200- to 500-fold increased risk of juvenile myelomonocytic leukemia. Leukemia cells from patients with juvenile myelomonocytic leukemia display hypersensitivity to certain cytokines, such as granulocyte-macrophage colony-stimulating factor. The granulocyte-macrophage colony-stimulating factor receptor utilizes pre-associated JAK2 to initiate signals after ligand binding. JAK2 subsequently activates STAT5, among other downstream effectors. Although STAT5 is gaining recognition as an important mediator of growth factor signaling in myeloid leukemias, the contribution of STAT5 to the development of hyperactive RAS-initiated myeloproliferative disease has not been well described. In this study, we investigated the consequence of STAT5 attenuation via genetic and pharmacological approaches in Nf1-deficient murine models of juvenile myelomonocytic leukemia. We found that homozygous Stat5 deficiency extended the lifespan of Nf1-deficient mice and eliminated the development of myeloproliferative neoplasm associated with Nf1 gene loss. Likewise, we found that JAK inhibition with ruxolitinib attenuated myeloproliferative neoplasm in Nf1-deficient mice. Finally, we found that primary cells from a patient with KRAS-mutant juvenile myelomonocytic leukemia displayed reduced colony formation in response to JAK2 inhibition. Our findings establish a central role for STAT5 activation in the pathogenesis of juvenile myelomonocytic leukemia and suggest that targeting this pathway may be of clinical utility in these patients.


Asunto(s)
Janus Quinasa 2/metabolismo , Leucemia Mielomonocítica Juvenil/etiología , Trastornos Mieloproliferativos/etiología , Neurofibromina 1/deficiencia , Factor de Transcripción STAT5/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Leucemia Mieloide/etiología , Leucemia Mieloide/genética , Leucemia Mielomonocítica Juvenil/genética , Ratones , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
7.
Nature ; 461(7262): 411-4, 2009 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-19727076

RESUMEN

The cascade comprising Raf, mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase (ERK) is a therapeutic target in human cancers with deregulated Ras signalling, which includes tumours that have inactivated the Nf1 tumour suppressor. Nf1 encodes neurofibromin, a GTPase-activating protein that terminates Ras signalling by stimulating hydrolysis of Ras-GTP. We compared the effects of inhibitors of MEK in a myeloproliferative disorder (MPD) initiated by inactivating Nf1 in mouse bone marrow and in acute myeloid leukaemias (AMLs) in which cooperating mutations were induced by retroviral insertional mutagenesis. Here we show that MEK inhibitors are ineffective in MPD, but induce objective regression of many Nf1-deficient AMLs. Drug resistance developed because of outgrowth of AML clones that were present before treatment. We cloned clone-specific retroviral integrations to identify candidate resistance genes including Rasgrp1, Rasgrp4 and Mapk14, which encodes p38alpha. Functional analysis implicated increased RasGRP1 levels and reduced p38 kinase activity in resistance to MEK inhibitors. This approach represents a robust strategy for identifying genes and pathways that modulate how primary cancer cells respond to targeted therapeutics and for probing mechanisms of de novo and acquired resistance.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas ras/metabolismo , Animales , Benzamidas/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Genes ras , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Ratones , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas ras/genética
8.
Adv Biochem Eng Biotechnol ; 182: 23-60, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35262750

RESUMEN

Artificial intelligence represents a powerful computational tool to analyze large amounts of data with and without supervision. Such powerful techniques have significantly impacted the advancement of multiple sectors from social media to data security, the automotive industry, gaming, finances, and healthcare. Only recently, however, artificial intelligence has been making significant strides in healthcare and biomedical research. Despite such advancements and the expectation of potential breakthroughs arising from implementing artificial intelligence in these fields, there is limited knowledge transfer and training for most healthcare professionals on using these computational techniques. While there is a wide array of publications on artificial intelligence applied to scientific research, most are too technical (not aimed at the non-initiated), too broad (with an overwhelming amount of data), or too focused on a particular application. This chapter presents an overview of artificial intelligence and its derivatives, giving a historical perspective, a succinct technical explanation of the underlying basis, and some examples of its applications. It finishes with a brief discussion on the challenges for implementing AI to be fully accepted in the scientific community.


Asunto(s)
Inteligencia Artificial , Investigación Biomédica , Atención a la Salud , Humanos
9.
Cancer Res ; 79(9): 2339-2351, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30862722

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common cancer in children. The highest rates of treatment failure occur in specific genetic subsets of ALL, including hypodiploid B-cell ALL (B-ALL), for which effective alternative therapies to current intensive chemotherapy treatments have yet to be developed. Here, we integrated biochemical and genomic profiling with functional drug assays to select effective agents with therapeutic potential against hypodiploid B-ALL. ABT-199, a selective Bcl-2 inhibitor, was effective in reducing leukemic burden in vitro and in vivo in patient-derived xenograft models of hypodiploid B-ALL. Daily oral treatment with ABT-199 significantly increased survival in xenografted mice. The unexpected efficacy of ABT-199 observed in hypodiploid leukemias lacking BIM expression (the major reported mediator of ABT-199-induced apoptosis) led us to investigate the mechanism of action of ABT-199 in the absence of BIM. Treatment with ABT-199 elicited responses in a dose-dependent manner, from cell-cycle arrest at low nanomolar concentrations to cell death at concentrations above 100 nmol/L. Collectively, these results demonstrate the efficacy of Bcl-2 inhibition and potential therapeutic strategy in hypodiploid B-ALL. SIGNIFICANCE: These results demonstrate the efficacy of ABT-199 in vivo and provide encouraging preclinical data of Bcl-2 as a potential target for the treatment of hypodiploid B-ALL.


Asunto(s)
Antineoplásicos/farmacología , Diploidia , Leucemia Experimental/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Linaje de la Célula , Proliferación Celular , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Nat Genet ; 47(5): 539-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25822087

RESUMEN

RAS network activation is common in human cancers, and in acute myeloid leukemia (AML) this activation is achieved mainly through gain-of-function mutations in KRAS, NRAS or the receptor tyrosine kinase FLT3. We show that in mice, premalignant myeloid cells harboring a Kras(G12D) allele retained low levels of Ras signaling owing to negative feedback involving Spry4 that prevented transformation. In humans, SPRY4 is located on chromosome 5q, a region affected by large heterozygous deletions that are associated with aggressive disease in which gain-of-function mutations in the RAS pathway are rare. These 5q deletions often co-occur with chromosome 17 alterations involving the deletion of NF1 (another RAS negative regulator) and TP53. Accordingly, combined suppression of Spry4, Nf1 and p53 produces high levels of Ras signaling and drives AML in mice. Thus, SPRY4 is a tumor suppressor at 5q whose disruption contributes to a lethal AML subtype that appears to acquire RAS pathway activation through a loss of negative regulators.


Asunto(s)
Carcinogénesis/genética , Leucemia Mieloide Aguda/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Genes Supresores de Tumor , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Mutación Missense , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética
12.
Clin Cancer Res ; 20(2): 490-8, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24178622

RESUMEN

PURPOSE: The clinical relevance of targeting the RAS/RAF/MEK/ERK pathway, activated in 70% to 80% of patients with acute myelogenous leukemia (AML), is unknown. EXPERIMENTAL DESIGN: Selumetinib is an oral small-molecule inhibitor of MAP-ERK kinase (MEK)-1/2. Forty-seven patients with relapsed/refractory AML or 60 years old or more with untreated AML were enrolled on a phase II study. Patients were stratified by FLT3 ITD mutation status. The primary endpoint was response rate (complete, partial, and minor). Leukemia cells were analyzed for extracellular signal-regulated kinase (ERK) and mTOR phosphorylation. RESULTS: Common drug-related toxicities were grade 1-2 diarrhea, fatigue, nausea, vomiting, and skin rash. In the FLT3 wild-type cohort, six of 36 (17%) patients had a response [one partial response, three minor responses, two unconfirmed minor responses (uMR)]. No patient with FLT3 ITD responded. NRAS and KRAS mutations were detected in 7% and 2% of patients, respectively. The sole patient with KRAS mutation had uMR with hematologic improvement in platelets. Baseline p-ERK activation was observed in 85% of patients analyzed but did not correlate with a response. A single-nucleotide polymorphism (SNP) rs3733542 in exon 18 of the KIT gene was detected in significantly higher number of patients with response/stable disease compared with nonresponders (60% vs. 23%; P = 0.027). CONCLUSIONS: Selumetinib is associated with modest single-agent antileukemic activity in advanced AML. However, given its favorable toxicity profile, combination with drugs that target other signaling pathways in AML should be considered. The potential association of SNP rs3733542 in exon 18 of the KIT gene with antileukemic activity of selumetinib is intriguing, but will require validation in larger trials.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Femenino , Genes ras , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Mutación , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Proto-Oncogénicas c-kit/genética , Resultado del Tratamiento , Tirosina Quinasa 3 Similar a fms/genética
13.
Sci Signal ; 6(304): ra105, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24300897

RESUMEN

Oncogenic K-Ras proteins, such as K-Ras(G12D), accumulate in the active, guanosine triphosphate (GTP)-bound conformation and stimulate signaling through effector kinases. The presence of the K-Ras(G12D) oncoprotein at a similar abundance to that of endogenous wild-type K-Ras results in only minimal phosphorylation and activation of the canonical Raf-mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling cascades in primary hematopoietic cells, and these pathways remain dependent on growth factors for efficient activation. We showed that phospholipase C-γ (PLC-γ), PI3K, and their generated second messengers link activated cytokine receptors to Ras and ERK signaling in differentiated bone marrow cells and in a cell population enriched for leukemia stem cells. Cells expressing endogenous oncogenic K-Ras(G12D) remained dependent on the second messenger diacylglycerol for the efficient activation of Ras-ERK signaling. These data raise the unexpected possibility of therapeutically targeting proteins that function upstream of oncogenic Ras in cancer.


Asunto(s)
Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sustitución de Aminoácidos , Animales , Células Cultivadas , Citocinas/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Células Madre Hematopoyéticas/patología , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Mutación Missense , Células Madre Neoplásicas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfolipasa C gamma/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Sistemas de Mensajero Secundario/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
14.
Nat Genet ; 45(3): 242-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23334668

RESUMEN

The genetic basis of hypodiploid acute lymphoblastic leukemia (ALL), a subtype of ALL characterized by aneuploidy and poor outcome, is unknown. Genomic profiling of 124 hypodiploid ALL cases, including whole-genome and exome sequencing of 40 cases, identified two subtypes that differ in the severity of aneuploidy, transcriptional profiles and submicroscopic genetic alterations. Near-haploid ALL with 24-31 chromosomes harbor alterations targeting receptor tyrosine kinase signaling and Ras signaling (71%) and the lymphoid transcription factor gene IKZF3 (encoding AIOLOS; 13%). In contrast, low-hypodiploid ALL with 32-39 chromosomes are characterized by alterations in TP53 (91.2%) that are commonly present in nontumor cells, IKZF2 (encoding HELIOS; 53%) and RB1 (41%). Both near-haploid and low-hypodiploid leukemic cells show activation of Ras-signaling and phosphoinositide 3-kinase (PI3K)-signaling pathways and are sensitive to PI3K inhibitors, indicating that these drugs should be explored as a new therapeutic strategy for this aggressive form of leukemia.


Asunto(s)
Aneuploidia , Aberraciones Cromosómicas , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Haploidia , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Ratones , Datos de Secuencia Molecular , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Trasplante Heterólogo , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
15.
Cancer Cell ; 14(4): 335-43, 2008 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-18835035

RESUMEN

Progress in understanding the molecular pathogenesis of human myeloproliferative disorders (MPDs) has led to guidelines incorporating genetic assays with histopathology during diagnosis. Advances in flow cytometry have made it possible to simultaneously measure cell type and signaling abnormalities arising as a consequence of genetic pathologies. Using flow cytometry, we observed a specific evoked STAT5 signaling signature in a subset of samples from patients suspected of having juvenile myelomonocytic leukemia (JMML), an aggressive MPD with a challenging clinical presentation during active disease. This signature was a specific feature involving JAK-STAT signaling, suggesting a critical role of this pathway in the biological mechanism of this disorder and indicating potential targets for future therapies.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Citometría de Flujo , Leucemia Mielomonocítica Juvenil/metabolismo , Trastornos Mieloproliferativos/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Adulto , Proliferación Celular , Células Cultivadas , Niño , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Janus Quinasa 2/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patología , Leucemia Mielomonocítica Juvenil/terapia , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Trastornos Mieloproliferativos/terapia , Estadificación de Neoplasias , Fosforilación , Recurrencia , Transducción de Señal/genética , Resultado del Tratamiento
16.
Blood ; 109(9): 3945-52, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17192389

RESUMEN

Defining how cancer-associated mutations perturb signaling networks in stem/progenitor populations that are integral to tumor formation and maintenance is a fundamental problem with biologic and clinical implications. Point mutations in RAS genes contribute to many cancers, including myeloid malignancies. We investigated the effects of an oncogenic Kras(G12D) allele on phosphorylated signaling molecules in primary c-kit(+) lin(-/low) hematopoietic stem/progenitor cells. Comparison of wild-type and Kras(G12D) c-kit(+) lin(-/low) cells shows that K-Ras(G12D) expression causes hyperproliferation in vivo and results in abnormal levels of phosphorylated STAT5, ERK, and S6 under basal and stimulated conditions. Whereas Kras(G12D) cells demonstrate hyperactive signaling after exposure to granulocyte-macrophage colony-stimulating factor, we unexpectedly observe a paradoxical attenuation of ERK and S6 phosphorylation in response to stem cell factor. These studies provide direct biochemical evidence that cancer stem/progenitor cells remodel signaling networks in response to oncogenic stress and demonstrate that multi-parameter flow cytometry can be used to monitor the effects of targeted therapeutics in vivo. This strategy has broad implications for defining the architecture of signaling networks in primary cancer cells and for implementing stem cell-targeted interventions.


Asunto(s)
Proliferación Celular , Células Madre Hematopoyéticas/metabolismo , Trastornos Mieloproliferativos/metabolismo , Células Madre Neoplásicas/metabolismo , Mutación Puntual , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Transducción de Señal , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Madre Hematopoyéticas/patología , Ratones , Ratones Transgénicos , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/genética
17.
Blood ; 110(1): 116-24, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17374739

RESUMEN

Gab2 is an important adapter molecule for cytokine signaling. Despite its major role in signaling by receptors associated with hematopoiesis, the role of Gab2 in hematopoiesis has not been addressed. We report that despite normal numbers of peripheral blood cells, bone marrow cells, and c-Kit(+)Lin(-)Sca-1(+) (KLS) cells, Gab2-deficient hematopoietic cells are deficient in cytokine responsiveness. Significant reductions in the number of colony-forming units in culture (CFU-C) in the presence of limiting cytokine concentrations were observed, and these defects could be completely corrected by retroviral complementation. In earlier hematopoiesis, Gab2-deficient KLS cells isolated in vitro responded poorly to hematopoietic growth factors, resulting in an up to 11-fold reduction in response to a cocktail of stem cell factor, flt3 ligand, and thrombopoietin. Gab2-deficient c-Kit(+)Lin(-) cells also demonstrate impaired activation of extracellular signal-regulated kinase (ERK) and S6 in response to IL-3, which supports defects in activating the phosphatidylinositol-3 kinase (PI-3K) and mitogen-associated protein kinase (MAPK) signaling cascades. Associated with the early defects in cytokine response, competitive transplantation of Gab2(-/-) bone marrow cells resulted in defective long-term multilineage repopulation. Therefore, we demonstrate that Gab2 adapter function is intrinsically required for hematopoietic cell response to early-acting cytokines, resulting in defective hematopoiesis in Gab2-deficient mice.


Asunto(s)
Hematopoyesis , Fosfoproteínas/deficiencia , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Trasplante de Médula Ósea , Células Cultivadas , Citocinas/farmacología , Células Madre Hematopoyéticas/patología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Mutantes , Fosfoproteínas/genética , Células Madre/citología
18.
J Cell Sci ; 119(Pt 1): 141-52, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16352660

RESUMEN

The small GTPase Rac contributes to regulation of cytoskeletal rearrangement during chemokine-induced lymphocyte adhesion and migration in a multi-step process that is very precisely coordinated. Chimaerins are Rac1-specific GTPase-activating proteins of unknown biological function, which have a canonical diacylglycerol C1-binding domain. Here we demonstrate endogenous expression of beta2-chimaerin in T lymphocytes and study the functional role of this protein in phorbol ester and chemokine (CXCL12)-regulated T-cell responses. We used green fluorescent protein-tagged beta2-chimaerin and phorbol ester stimulation to investigate changes in protein localization in living lymphocytes. Our results demonstrate that active Rac cooperates with C1-dependent phorbol ester binding to induce sustained GFP-beta2-chimaerin localization to the membrane. Subcellular distribution of GFP beta2-chimaerin in living cells showed no major changes following CXCL12 stimulation. Nonetheless Rac1-GTP levels were severely inhibited in GFP-beta2-chimaerin-expressing cells, which displayed reduced CXCL12-induced integrin-dependent adhesion and spreading. This effect was dependent on chimaerin GTPase-activating protein function and required diacylglycerol generation. Whereas beta2-chimaerin overexpression decreased static adhesion, it enhanced CXCL12-dependent migration via receptor-dependent diacylglycerol production. These studies demonstrate that beta2-chimaerin provides a novel, diacylglycerol-dependent mechanism for Rac regulation in T cells and suggest a functional role for this protein in Rac-mediated cytoskeletal remodeling.


Asunto(s)
Adhesión Celular/fisiología , Quimiotaxis/fisiología , Diglicéridos/metabolismo , Proteínas de Neoplasias/metabolismo , Linfocitos T/fisiología , Animales , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Activación Enzimática , Humanos , Células Jurkat , Proteínas de Neoplasias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de Unión al GTP rac1/metabolismo
19.
J Biol Chem ; 278(31): 29208-15, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12738795

RESUMEN

Protein kinase C (PKC) is the only PKC isoform recruited to the immunological synapse after T cell receptor stimulation, suggesting that its activation mechanism differs from that of the other isoforms. Previous studies have suggested that this selective PKC recruitment may operate via a Vav-regulated, cytoskeletal-dependent mechanism, independent of the classical phospholipase C/diacylglycerol pathway. Here, we demonstrate that, together with tyrosine phosphorylation of PKC in the regulatory domain, binding of phospholipase C-dependent diacylglycerol is required for PKC recruitment to the T cell synapse. In addition, we demonstrate that diacylglycerol kinase alpha-dependent diacylglycerol phosphorylation provides the negative signal required for PKC inactivation, ensuring fine control of the T cell activation response.


Asunto(s)
Membrana Celular/enzimología , Diglicéridos/metabolismo , Isoenzimas/metabolismo , Activación de Linfocitos , Proteína Quinasa C/metabolismo , Linfocitos T/inmunología , Fosfolipasas de Tipo C/metabolismo , Diacilglicerol Quinasa/metabolismo , Humanos , Células Jurkat , Fosfolipasa C gamma , Fosforilación , Proteína Quinasa C-theta , Receptor Muscarínico M1 , Receptores de Antígenos de Linfocitos T/fisiología , Receptores Muscarínicos/genética , Transducción de Señal , Transfección , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA