Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Biol Chem ; 288(47): 33542-33558, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24089526

RESUMEN

The development of strategies to eradicate primary human acute myelogenous leukemia (AML) cells is a major challenge to the leukemia research field. In particular, primitive leukemia cells, often termed leukemia stem cells, are typically refractory to many forms of therapy. To investigate improved strategies for targeting of human AML cells we compared the molecular mechanisms regulating oxidative state in primitive (CD34(+)) leukemic versus normal specimens. Our data indicate that CD34(+) AML cells have elevated expression of multiple glutathione pathway regulatory proteins, presumably as a mechanism to compensate for increased oxidative stress in leukemic cells. Consistent with this observation, CD34(+) AML cells have lower levels of reduced glutathione and increased levels of oxidized glutathione compared with normal CD34(+) cells. These findings led us to hypothesize that AML cells will be hypersensitive to inhibition of glutathione metabolism. To test this premise, we identified compounds such as parthenolide (PTL) or piperlongumine that induce almost complete glutathione depletion and severe cell death in CD34(+) AML cells. Importantly, these compounds only induce limited and transient glutathione depletion as well as significantly less toxicity in normal CD34(+) cells. We further determined that PTL perturbs glutathione homeostasis by a multifactorial mechanism, which includes inhibiting key glutathione metabolic enzymes (GCLC and GPX1), as well as direct depletion of glutathione. These findings demonstrate that primitive leukemia cells are uniquely sensitive to agents that target aberrant glutathione metabolism, an intrinsic property of primary human AML cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Dioxolanos/farmacología , Glutatión/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Sesquiterpenos/farmacología , Antígenos CD34 , Femenino , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/antagonistas & inhibidores , Glutatión Peroxidasa/antagonistas & inhibidores , Glutatión Peroxidasa/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Oxidación-Reducción/efectos de los fármacos , Células Tumorales Cultivadas , Glutatión Peroxidasa GPX1
2.
J Immunol ; 188(8): 3784-90, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22427635

RESUMEN

Negative regulation of innate immune responses is essential to prevent excess inflammation and tissue injury and promote homeostasis. Lysophosphatidic acid (LPA) is a pleiotropic lipid that regulates cell growth, migration, and activation and is constitutively produced at low levels in tissues and in serum. Extracellular LPA binds to specific G protein-coupled receptors, whose function in regulating innate or adaptive immune responses remains poorly understood. Of the classical LPA receptors belonging to the Edg family, lpa2 (edg4) is expressed by dendritic cells (DC) and other innate immune cells. In this article, we show that DC from lpa2(-/-) mice are hyperactive compared with their wild-type counterparts and are less susceptible to inhibition by different LPA species. In transient-transfection assays, we found that lpa2 overexpression inhibits NF-κB-driven gene transcription. Using an adoptive-transfer approach, we found that allergen-pulsed lpa2(-/-) DC induced substantially more lung inflammation than did wild-type DC after inhaled allergen challenge. Finally, lpa2(-/-) mice develop greater allergen-driven lung inflammation than do their wild-type counterparts in models of allergic asthma involving both systemic and mucosal sensitization. Taken together, these findings identify LPA acting via lpa2 as a novel negative regulatory pathway that inhibits DC activation and allergic airway inflammation.


Asunto(s)
Asma/inmunología , Células Dendríticas/inmunología , Pulmón/inmunología , Lisofosfolípidos/inmunología , FN-kappa B/inmunología , Receptores del Ácido Lisofosfatídico/inmunología , Administración por Inhalación , Traslado Adoptivo , Alérgenos/inmunología , Animales , Asma/patología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Inflamación/inmunología , Inflamación/patología , Pulmón/metabolismo , Pulmón/patología , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Receptores del Ácido Lisofosfatídico/deficiencia , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal , Transcripción Genética
3.
Sci Rep ; 14(1): 12868, 2024 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834690

RESUMEN

Acute myeloid leukemia (AML) is fatal in the majority of adults. Identification of new therapeutic targets and their pharmacologic modulators are needed to improve outcomes. Previous studies had shown that immunization of rabbits with normal peripheral WBCs that had been incubated with fluorodinitrobenzene elicited high titer antibodies that bound to a spectrum of human leukemias. We report that proteomic analyses of immunoaffinity-purified lysates of primary AML cells showed enrichment of scaffolding protein IQGAP1. Immunohistochemistry and gene-expression analyses confirmed IQGAP1 mRNA overexpression in various cytogenetic subtypes of primary human AML compared to normal hematopoietic cells. shRNA knockdown of IQGAP1 blocked proliferation and clonogenicity of human leukemia cell-lines. To develop small molecules targeting IQGAP1 we performed in-silico screening of 212,966 compounds, selected 4 hits targeting the IQGAP1-GRD domain, and conducted SAR of the 'fittest hit' to identify UR778Br, a prototypical agent targeting IQGAP1. UR778Br inhibited proliferation, induced apoptosis, resulted in G2/M arrest, and inhibited colony formation by leukemia cell-lines and primary-AML while sparing normal marrow cells. UR778Br exhibited favorable ADME/T profiles and drug-likeness to treat AML. In summary, AML shows response to IQGAP1 inhibition, and UR778Br, identified through in-silico studies, selectively targeted AML cells while sparing normal marrow.


Asunto(s)
Proliferación Celular , Leucemia Mieloide Aguda , Proteínas Activadoras de ras GTPasa , Humanos , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/antagonistas & inhibidores , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/genética , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Simulación por Computador , Antineoplásicos/farmacología , Dominios Proteicos , Animales , Proteómica/métodos
4.
Blood ; 116(26): 5983-90, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20889920

RESUMEN

We have previously shown that the plant-derived compound parthenolide (PTL) can impair the survival and leukemogenic activity of primary human acute myeloid leukemia (AML) stem cells. However, despite the activity of this agent, PTL also induces cellular protective responses that likely function to reduce its overall cytotoxicity. Thus, we sought to identify pharmacologic agents that enhance the antileukemic potential of PTL. Toward this goal, we used the gene expression signature of PTL to identify compounds that inhibit cytoprotective responses by performing chemical genomic screening of the Connectivity Map database. This screen identified compounds acting along the phosphatidylinositol 3-kinase and mammalian target of rapamycin pathways. Compared with single agent treatment, exposure of AML cells to the combination of PTL and phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitors significantly decreased viability of AML cells and reduced tumor burden in vitro and in murine xenotransplantation models. Taken together, our data show that rational drug combinations can be identified using chemical genomic screening strategies and that inhibition of cytoprotective functions can enhance the eradication of primary human AML cells.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Sesquiterpenos/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirolimus/análogos & derivados , Sirolimus/farmacología
5.
Blood Adv ; 1(8): 500-503, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29296967

RESUMEN

The persistence of a CEBPA mutation at the time of complete remission warrants germ line analysis.Not all patients harboring germ line CEBPA mutations have a family history of AML.

6.
Cell Stem Cell ; 19(1): 23-37, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27374788

RESUMEN

Adipose tissue (AT) has previously been identified as an extra-medullary reservoir for normal hematopoietic stem cells (HSCs) and may promote tumor development. Here, we show that a subpopulation of leukemic stem cells (LSCs) can utilize gonadal adipose tissue (GAT) as a niche to support their metabolism and evade chemotherapy. In a mouse model of blast crisis chronic myeloid leukemia (CML), adipose-resident LSCs exhibit a pro-inflammatory phenotype and induce lipolysis in GAT. GAT lipolysis fuels fatty acid oxidation in LSCs, especially within a subpopulation expressing the fatty acid transporter CD36. CD36(+) LSCs have unique metabolic properties, are strikingly enriched in AT, and are protected from chemotherapy by the GAT microenvironment. CD36 also marks a fraction of human blast crisis CML and acute myeloid leukemia (AML) cells with similar biological properties. These findings suggest striking interplay between leukemic cells and AT to create a unique microenvironment that supports the metabolic demands and survival of a distinct LSC subpopulation.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo/patología , Antineoplásicos/farmacología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Animales , Antineoplásicos/uso terapéutico , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/patología , Antígenos CD36/metabolismo , Citoprotección/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Gónadas/patología , Humanos , Inflamación/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Lipólisis/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
7.
Neurotoxicology ; 26(4): 675-90, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15970329

RESUMEN

This project was undertaken to more completely understand the consequences of lifetime exposure to methylmercury. A series of experiments examined how perinatal or lifetime exposure to methylmercury affected behavioral performances in the adult mouse at different ages. One hundred female B6C3F1/HSD mice were assigned to one of three dose groups, 0 ppm, 1 ppm, or 3 ppm methylmercury chloride administered in a 5 nM sodium carbonate drinking solution. Four weeks after initiating dosing, the females were bred with male CBA/J HSD mice to produce the trihybrid offspring B6C3F1/HSD x CBA/J HSD. The methylmercury-treated litters were split into two subgroups, one exposed throughout its lifetime to the original dose, the other exposed through postnatal day 13. Altogether, then, five groups were studied: Control, 1 ppm perinatal, 1 ppm lifetime, 3 ppm perinatal, and 3 ppm lifetime. Three neurobehavioral indices were evaluated: (1) delayed spatial alternation (a test of memory) and (2) running in a wheel to earn food pellets (schedule-controlled operant behavior) were assessed starting at 5 and 15 months of age; (3) hindlimb splay, a measure of motor function, was assessed at 5, 15, and 26 months of age. Subjects tested at one age were littermates of those tested at the other ages. MeHg altered the hindlimb splay distance; control mice differed from methylmercury-exposed mice, the 1 ppm lifetime and 3 ppm lifetime groups differed from each other, and the analysis yielded an age by dose interaction. MeHg exposure altered different measures of wheel running under the 3 ppm lifetime condition. In the delayed alternation procedure, the mouse was required to respond to one of two locations in a strictly alternating sequence. More mice from the treated groups, except for the 1 ppm perinatal group, failed to meet the criterion at longer delay values. Overall, the results show that exposure to low levels of methylmercury produces behavioral effects that depend on the test procedure, the dose, the duration of exposure, and the age. Lifetime evaluations of exposure to toxicants, beginning with early development, should be a component of the risk assessment process for neurotoxicity.


Asunto(s)
Conducta Animal/efectos de los fármacos , Compuestos de Metilmercurio/toxicidad , Envejecimiento/fisiología , Animales , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Femenino , Alimentos , Miembro Posterior/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos , Actividad Motora/efectos de los fármacos , Síndromes de Neurotoxicidad/psicología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Desempeño Psicomotor/efectos de los fármacos , Reflejo/efectos de los fármacos , Percepción Espacial/efectos de los fármacos
8.
PLoS One ; 10(7): e0132375, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26177509

RESUMEN

Cytogenetically normal acute myeloid leukemia (CN-AML) patients harboring RUNX1 mutations have a dismal prognosis with anthracycline/cytarabine-based chemotherapy. We aimed to develop an in vivo model of RUNX1-mutated, CN-AML in which the nature of residual disease in this molecular disease subset could be explored. We utilized a well-characterized patient-derived, RUNX1-mutated CN-AML line (CG-SH). Tail vein injection of CG-SH into NOD scid gamma mice led to leukemic engraftment in the bone marrow, spleen, and peripheral blood within 6 weeks. Treatment of leukemic mice with anthracycline/cytarabine-based chemotherapy resulted in clearance of disease from the spleen and peripheral blood, but persistence of disease in the bone marrow as assessed by flow cytometry and secondary transplantation. Whole exome sequencing of CG-SH revealed mutations in ASXL1, CEBPA, GATA2, and SETBP1, not previously reported. We conclude that CG-SH xenografts are a robust, reproducible in vivo model of CN-AML in which to explore mechanisms of chemotherapy resistance and novel therapeutic approaches.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Análisis Citogenético , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación/genética , Neoplasia Residual/genética , Neoplasia Residual/patología , Animales , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Médula Ósea/patología , Línea Celular Tumoral , Citarabina/farmacología , Citarabina/uso terapéutico , Análisis Mutacional de ADN , Exoma/genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones Endogámicos NOD , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Physiol Genomics ; 8(2): 107-14, 2002 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-11875188

RESUMEN

Parotid gland acinar cells undergo marked hypertrophy and hyperplasia upon systemic exposure to the beta-adrenergic agonist, isoproterenol. This glandular enlargement is accompanied by substantial cellular changes including DNA synthesis, an increase in glandular protein synthesis, and differential changes in RNA transcription. To gain a more detailed understanding of the underlying changes induced by isoproterenol, we have examined the parotid gland gene expression profile of mice up to 24 h post-isoproterenol injection using high-density oligonucleotide arrays. Depending upon the exposure time, between 22 and 48 of the approximately 6,500 mouse genes and expressed sequence tags (ESTs) analyzed displayed significant changes in expression patterns. Genes that were previously shown to be repressed (alpha-amylase) or activated (proline-rich proteins) following isoproterenol exposure were found to be similarly affected in this experiment, validating this technique. This study demonstrates that the oligonucleotide array technology is a useful tool for examining isoproterenol-induced salivary gland gene expression changes. Using this as a starting point, we can begin to dissect the specific pathways involved in mediating isoproterenol action within the parotid gland.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Isoproterenol/farmacología , Glándula Parótida/efectos de los fármacos , Glándula Parótida/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Perfilación de la Expresión Génica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Mol Cancer Ther ; 13(8): 1979-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24934933

RESUMEN

Most patients with acute myelogenous leukemia (AML) relapse and die of their disease. Increasing evidence indicates that AML relapse is driven by the inability to eradicate leukemia stem cells (LSC). Thus, it is imperative to identify novel therapies that can ablate LSCs. Using an in silico gene expression-based screen for compounds evoking transcriptional effects similar to the previously described anti-LSC agent parthenolide, we identified AR-42 (OSU-HDAC42), a novel histone deacetylase inhibitor that is structurally similar to phenylbutyrate, but with improved activity at submicromolar concentrations. Here, we report that AR-42 induces NF-κB inhibition, disrupts the ability of Hsp90 to stabilize its oncogenic clients, and causes potent and specific cell death of LSCs but not normal hematopoietic stem and progenitor cells. Unlike parthenolide, the caspase-dependent apoptosis caused by AR-42 occurs without activation of Nrf-2-driven cytoprotective pathways. As AR-42 is already being tested in early clinical trials, we expect that our results can be extended to the clinic.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Fenilbutiratos/farmacología , Animales , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Dosificación Letal Mediana , Leucemia Mieloide Aguda/patología , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/fisiología , Sesquiterpenos/farmacología , Transcriptoma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cell Stem Cell ; 11(3): 359-72, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22863534

RESUMEN

Leukemia stem cells (LSCs) represent a biologically distinct subpopulation of myeloid leukemias, with reduced cell cycle activity and increased resistance to therapeutic challenge. To better characterize key properties of LSCs, we employed a strategy based on identification of genes synergistically dysregulated by cooperating oncogenes. We hypothesized that such genes, termed "cooperation response genes" (CRGs), would represent regulators of LSC growth and survival. Using both a primary mouse model and human leukemia specimens, we show that CRGs comprise genes previously undescribed in leukemia pathogenesis in which multiple pathways modulate the biology of LSCs. In addition, our findings demonstrate that the CRG expression profile can be used as a drug discovery tool for identification of compounds that selectively target the LSC population. We conclude that CRG-based analyses provide a powerful means to characterize the basic biology of LSCs as well as to identify improved methods for therapeutic targeting.


Asunto(s)
Leucemia/genética , Leucemia/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Oncogenes/genética , Animales , Benzotiazoles/farmacología , Crisis Blástica/genética , Crisis Blástica/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/efectos de los fármacos , Serpinas/metabolismo , Tirfostinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA