Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38934082

RESUMEN

The treatment of blast phase chronic myeloid leukemia (bpCML) remains a challenge due at least in part to drug resistance of leukemia stem cells (LSCs). Recent clinical evidence suggests that the BCL-2 inhibitor venetoclax in combination with ABL-targeting tyrosine kinase inhibitors (TKIs) can eradicate bpCML LSCs. In this report, we employed preclinical models of bpCML to investigate the efficacy and underlying mechanism of LSC-targeting with venetoclax/TKI combinations. Transcriptional analysis of LSCs exposed to venetoclax and dasatinib revealed upregulation of genes involved in lysosomal biology, in particular lysosomal acid lipase A (LIPA), a regulator of free fatty acids. Metabolomic analysis confirmed increased levels of free fatty acids in response to venetoclax/dasatinib. Pre-treatment of leukemia cells with bafilomycin, a specific lysosome inhibitor, or genetic perturbation of LIPA, resulted in increased sensitivity of leukemia cells toward venetoclax/dasatinib, implicating LIPA in treatment resistance. Importantly, venetoclax/dasatinib treatment does not affect normal stem cell function, suggestive of a leukemia-specific response. These results demonstrate that venetoclax/dasatinib is an LSCselective regimen in bpCML and that disrupting LIPA and fatty acid transport enhances venetoclax/dasatinib response in targeting LSCs, providing a rationale for exploring lysosomal disruption as an adjunct therapeutic strategy to prolong disease remission.

2.
Br J Cancer ; 117(10): 1537-1543, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-28881356

RESUMEN

BACKGROUND: Aldehyde dehydrogenase (ALDH) 1A1 is an immunohistological biomarker of various solid tumours, but has not been successfully proved as a colorectal cancer (CRC) marker. We recently reported that ALDH1B1, which has functional roles in tumourigenesis, may be a better CRC marker than ALDH1A1. METHODS: Human CRC explants and cell lines were analysed to identify candidate CRC markers from eight ALDH isozymes including ALDH1A1 and ALDH1B1. A tissue microarray, including paired specimens of normal and tumour tissues, was subsequently analysed to determine if candidate ALDHs could distinguish CRC from normal tissue. RESULTS: Based on mRNA analysis, ALDH1B1 and ALDH2 were selected as suitable candidates. These were strongly and regularly expressed in tumour tissue and cell lines, including highly tumourigenic cell populations (ALDH+CD44+ cells), while other ALDHs, including ALDH1A1, showed differential or low expression. No genetic alteration of ALDH1B1 in CRC was suggested by the relationships between mRNA and protein levels/enzymatic activities, and cDNA sequences of CRC cell lines. Tissue microarray findings showed that ALDH1B1, but not ALDH2, could distinguish CRC from normal tissue. Furthermore, ratios of ALDH1B1 to ALDH1A1 or ALDH2 were found to be powerful CRC indicators. CONCLUSIONS: These results suggest that ALDH1B1 is a novel human CRC biomarker.


Asunto(s)
Aldehído Deshidrogenasa/análisis , Biomarcadores de Tumor/análisis , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/enzimología , Aldehído Deshidrogenasa/biosíntesis , Familia de Aldehído Deshidrogenasa 1 , Aldehído Deshidrogenasa Mitocondrial , Humanos , Inmunohistoquímica
3.
Haematologica ; 102(6): 1054-1065, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28280079

RESUMEN

Aldehyde dehydrogenase 1A1 (ALDH1A1) activity is high in hematopoietic stem cells and functions in part to protect stem cells from reactive aldehydes and other toxic compounds. In contrast, we found that approximately 25% of all acute myeloid leukemias expressed low or undetectable levels of ALDH1A1 and that this ALDH1A1- subset of leukemias correlates with good prognosis cytogenetics. ALDH1A1- cell lines as well as primary leukemia cells were found to be sensitive to treatment with compounds that directly and indirectly generate toxic ALDH substrates including 4-hydroxynonenal and the clinically relevant compounds arsenic trioxide and 4-hydroperoxycyclophosphamide. In contrast, normal hematopoietic stem cells were relatively resistant to these compounds. Using a murine xenotransplant model to emulate a clinical treatment strategy, established ALDH1A1- leukemias were also sensitive to in vivo treatment with cyclophosphamide combined with arsenic trioxide. These results demonstrate that targeting ALDH1A1- leukemic cells with toxic ALDH1A1 substrates such as arsenic and cyclophosphamide may be a novel targeted therapeutic strategy for this subset of acute myeloid leukemias.


Asunto(s)
Aldehído Deshidrogenasa/deficiencia , Quimioterapia Combinada/métodos , Leucemia Mieloide Aguda/tratamiento farmacológico , Familia de Aldehído Deshidrogenasa 1 , Animales , Trióxido de Arsénico , Arsenicales/uso terapéutico , Células Cultivadas , Ciclofosfamida/uso terapéutico , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/enzimología , Ratones , Terapia Molecular Dirigida , Óxidos/uso terapéutico , Retinal-Deshidrogenasa
4.
Blood ; 124(24): 3608-12, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25339361

RESUMEN

Leukemic transformation of human cells is a complex process. Here we show that forced expression of MN1 in primitive human cord blood cells maintained on stromal cells in vitro induces a transient, but not serially transplantable, myeloproliferation in engrafted mice. However, cotransduction of an activated HOX gene (NUP98HOXD13) with MN1 induces a serially transplantable acute myeloid leukemia (AML). Further characterization of the leukemic cells generated from the dually transduced cells showed the activation of stem cell gene expression signatures also found in primary human AML. These findings show a new forward genetic model of human leukemogenesis and further highlight the relevance of homeobox transcription factors in the transformation process.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Sangre Fetal/metabolismo , Leucemia Mieloide Aguda/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas de Fusión Oncogénica/genética , Transactivadores , Proteínas Supresoras de Tumor/genética
5.
Adv Exp Med Biol ; 815: 349-59, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25427917

RESUMEN

Hematopoiesis involves the orderly production of millions of blood cells per second from a small number of essential bone marrow cells termed hematopoietic stem cells (HSCs). Ethanol suppresses normal hematopoiesis resulting in leukopenia, anemia, and thrombocytopenia and may also predispose to the development of diseases such as myelodysplasia (MDS) and acute leukemia. Currently the exact mechanisms by which ethanol perturbs hematopoiesis are unclear. The aldehyde dehydrogenase (ALDH) gene family plays a major role in the metabolism of reactive aldehydes derived from ethanol in the liver and other organs. At least one of the ALDH isoforms, ALDH1A1, is expressed at high levels in HSCs in humans, mice, and other organisms. Recent data indicate that ALDH1A1 and possibly other ALDH isoforms may metabolize reactive aldehydes in HSCs and other hematopoietic cells as they do in the liver and elsewhere. In addition, loss of these ALDHs leads to perturbation of a variety of cell processes that may predispose HSCs to disorders in growth and leukemic transformation. From these findings, we suggest a hypothesis that the cytopenias and possible increased risk of MDS and acute leukemia in heavy alcohol users is due to polymorphisms in genes responsible for metabolism of alcohol derived reactive aldehydes and repair of their DNA adducts in HSCs and other hematopoietic cells. In the article, we will summarize the biological properties of hematopoietic cells and diseases related to ethanol consumption, discuss molecular characteristics of ethanol metabolism, and describe a model to explain how ethanol derived reactive aldehydes may promote HSC damage.


Asunto(s)
Aldehído Deshidrogenasa/fisiología , Etanol/toxicidad , Hematopoyesis/efectos de los fármacos , Leucemia Mieloide Aguda/inducido químicamente , Síndromes Mielodisplásicos/inducido químicamente , Animales , Humanos
6.
Int J Cancer ; 132(8): 1741-50, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22987615

RESUMEN

Patients with chronic myelogenous leukemia (CML) respond well to tyrosine kinase inhibitors (TKIs) of the Bcr-Abl oncoprotein. However, intolerance and resistance to these agents remains a challenge, and TKIs are unable to eradicate rare leukemia-initiating cells. Leukemia treatment would benefit from a better understanding of molecular signals that are necessary for the survival of leukemia-initiating cells but dispensable for normal hematopoietic stem cells. Leukemia-initiating cells in CML can arise from myeloid progenitor cells, a population that we have reported in normal hematopoiesis to depend on the RNA-editing enzyme adenosine deaminase acting on RNA-1 (ADAR1). We now report that Bcr-Abl transformed leukemic cells were ADAR1-dependent in a conditional ADAR1 knockout mouse model. ADAR1 deletion reversed leukocytosis and splenomegaly, and preferentially depleted primitive Lin-Sca+Kit+ (LSK) leukemic cells but not LSK cells lacking the leukemic oncoprotein. ADAR1 deletion ultimately normalized the peripheral white blood count, eliminating leukemic cells as assessed by PCR. These results uncover a novel requirement for ADAR1 in myeloid leukemic cells and indicate that ADAR1 may comprise a new molecular target for CML-directed therapeutics.


Asunto(s)
Adenosina Desaminasa/genética , Eliminación de Gen , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Inhibidores de la Adenosina Desaminasa/farmacología , Animales , Secuencia de Bases , Cartilla de ADN , Citometría de Flujo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamoxifeno/farmacología
7.
Blood ; 118(7): 1766-73, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21730353

RESUMEN

Strategies for expanding hematopoietic stem cells (HSCs) could have significant utility for transplantation-based therapies. However, deleterious consequences of such manipulations remain unknown. Here we examined the impact of HSC self-renewal divisions in vitro and in vivo on their subsequent regenerative and continuing ability to sustain blood cell production in the absence of telomerase. HSC expansion in vitro was obtained using a NUP98-HOXA10hd transduction strategy and, in vivo, using a serial transplant protocol. We observed ~ 10kb telomere loss in leukocytes produced in secondary mice transplanted with HSCs regenerated in primary recipients of NUP98-HOXA10hd-transduced and in vitro-expanded Tert(-/-) HSCs 6 months before. The second generation leukocytes also showed elevated expression of γH2AX (relative to control) indicative of greater accumulating DNA damage. In contrast, significant telomere shortening was not detected in leukocytes produced from freshly isolated, serially transplanted wild-type (WT) or Tert(-/-) HSCs, suggesting that HSC replication posttransplant is not limited by telomere shortening in the mouse. These findings document a role of telomerase in telomere homeostasis, and in preserving HSC functional integrity on prolonged self-renewal stimulation.


Asunto(s)
Daño del ADN , Células Madre Hematopoyéticas/enzimología , Telomerasa/metabolismo , Telómero , Animales , Proliferación Celular , Eliminación de Gen , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histonas/genética , Ratones , Ratones Endogámicos C57BL , Telomerasa/genética
8.
Blood ; 118(16): 4366-76, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-21865344

RESUMEN

Achieving high-level expansion of hematopoietic stem cells (HSCs) in vitro will have an important clinical impact in addition to enabling elucidation of their regulation. Here, we couple the ability of engineered NUP98-HOXA10hd expression to stimulate > 1000-fold net expansions of murine HSCs in 10-day cultures initiated with bulk lin(-)Sca-1(+)c-kit(+) cells, with strategies to purify fetal and adult HSCs and analyze their expansion clonally. We find that NUP98-HOXA10hd stimulates comparable expansions of HSCs from both sources at ∼ 60% to 90% unit efficiency in cultures initiated with single cells. Clonally expanded HSCs consistently show balanced long-term contributions to the lymphoid and myeloid lineages without evidence of leukemogenic activity. Although effects on fetal and adult HSCs were indistinguishable, NUP98-HOXA10hd-transduced adult HSCs did not thereby gain a competitive advantage in vivo over freshly isolated fetal HSCs. Live-cell image tracking of single transduced HSCs cultured in a microfluidic device indicates that NUP98-HOXA10hd does not affect their proliferation kinetics, and flow cytometry confirmed the phenotype of normal proliferating HSCs and allowed reisolation of large numbers of expanded HSCs at a purity of 25%. These findings point to the effects of NUP98-HOXA10hd on HSCs in vitro being mediated by promoting self-renewal and set the stage for further dissection of this process.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción/genética , Animales , Proliferación Celular , Separación Celular , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ingeniería de Proteínas , Transducción Genética
9.
Cancer Discov ; 13(9): 2032-2049, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37358260

RESUMEN

The BCL2 inhibitor venetoclax has recently emerged as an important component of acute myeloid leukemia (AML) therapy. Notably, use of this agent has revealed a previously unrecognized form of pathogenesis characterized by monocytic disease progression. We demonstrate that this form of disease arises from a fundamentally different type of leukemia stem cell (LSC), which we designate as monocytic LSC (m-LSC), that is developmentally and clinically distinct from the more well-described primitive LSC (p-LSC). The m-LSC is distinguished by a unique immunophenotype (CD34-, CD4+, CD11b-, CD14-, CD36-), unique transcriptional state, reliance on purine metabolism, and selective sensitivity to cladribine. Critically, in some instances, m-LSC and p-LSC subtypes can co-reside in the same patient with AML and simultaneously contribute to overall tumor biology. Thus, our findings demonstrate that LSC heterogeneity has direct clinical significance and highlight the need to distinguish and target m-LSCs as a means to improve clinical outcomes with venetoclax-based regimens. SIGNIFICANCE: These studies identify and characterize a new type of human acute myeloid LSC that is responsible for monocytic disease progression in patients with AML treated with venetoclax-based regimens. Our studies describe the phenotype, molecular properties, and drug sensitivities of this unique LSC subclass. This article is featured in Selected Articles from This Issue, p. 1949.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Antígenos CD34/metabolismo , Antígenos CD34/uso terapéutico , Leucemia Mieloide Aguda/genética , Células Madre Neoplásicas/metabolismo , Progresión de la Enfermedad
10.
PLoS Genet ; 5(9): e1000640, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19749980

RESUMEN

The phenotypic heterogeneity that characterizes human cancers reflects the enormous genetic complexity of the oncogenic process. This complexity can also be seen in mouse models where it is frequently observed that in addition to the initiating genetic alteration, the resulting tumor harbors additional, somatically acquired mutations that affect the tumor phenotype. To investigate the role of genetic interactions in the development of tumors, we have made use of the Emu-myc model of pre-B and B cell lymphoma. Since various studies point to a functional interaction between Myc and the Rb/E2F pathway, we have investigated the role of E2F activities in the process of Myc-induced lymphomagenesis. Whereas the absence of E2F1 and E2F3 function has no impact on Myc-mediated tumor development, the absence of E2F2 substantially accelerates the time of tumor onset. Conversely, tumor development is delayed by the absence of E2F4. The enhanced early onset of tumors seen in the absence of E2F2 coincides with an expansion of immature B lineage cells that are likely to be the target for Myc oncogenesis. In contrast, the absence of E2F4 mutes the response of the lineage to Myc and there is no expansion of immature B lineage cells. We also find that distinct types of tumors emerge from the Emu-myc mice, distinguished by different patterns of gene expression, and that the relative proportions of these tumor types are affected by the absence of either E2F2 or E2F4. From these results, we conclude that there are several populations of tumors that arise from the Emu-myc model, reflecting distinct populations of cells that are susceptible to Myc-mediated oncogenesis and that the proportion of these cell populations is affected by the presence or absence of E2F activities.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F2/metabolismo , Factor de Transcripción E2F3/metabolismo , Factor de Transcripción E2F4/metabolismo , Linfoma de Células B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F2/genética , Factor de Transcripción E2F3/genética , Factor de Transcripción E2F4/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma de Células B/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/genética
11.
JCO Clin Cancer Inform ; 6: e2200030, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36194842

RESUMEN

PURPOSE: There are currently limited objective criteria to help assist physicians in determining whether an individual patient with acute myeloid leukemia (AML) is likely to do better with induction with either standard 7 + 3 chemotherapy or targeted therapy with venetoclax plus azacitidine. The study goal was to address this need by developing exploratory clinical decision support methods. PATIENTS AND METHODS: Univariable and multivariable analysis as well as comparison of a range of machine learning (ML) predictors were performed using cohorts of 120 newly diagnosed 7 + 3-treated AML patients compared with 101 venetoclax plus azacitidine-treated patients. RESULTS: A variety of features in the two patient cohorts were identified that may potentially correlate with short- and long-term outcomes, toxicities, and other considerations. A subset of these diagnostic features was then used to develop ML-based predictors with relatively high areas under the curve of short- and long-term outcomes, hospital stays, transfusion requirements, and toxicities for individual patients treated with either venetoclax/azacitidine or 7 + 3. CONCLUSION: Potential ML-based approaches to clinical decision support to help guide individual patients with newly diagnosed AML to either 7 + 3 or venetoclax plus azacitidine induction therapy were identified. Larger cohorts with separate test and validation studies are necessary to confirm these initial findings.


Asunto(s)
Sistemas de Apoyo a Decisiones Clínicas , Leucemia Mieloide Aguda , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Azacitidina/efectos adversos , Azacitidina/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/etiología , Aprendizaje Automático , Sulfonamidas , Resultado del Tratamiento
12.
Blood ; 113(25): 6342-50, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19377048

RESUMEN

Hematopoietic stem cells (HSCs) are generally defined by their dual properties of pluripotency and extensive self-renewal capacity. However, a lack of experimental clarity as to what constitutes extensive self-renewal capacity coupled with an absence of methods to prospectively isolate long-term repopulating cells with defined self-renewal activities has made it difficult to identify the essential components of the self-renewal machinery and investigate their regulation. We now show that cells capable of repopulating irradiated congenic hosts for 4 months and producing clones of cells that can be serially transplanted are selectively and highly enriched in the CD150(+) subset of the EPCR(+)CD48(-)CD45(+) fraction of mouse fetal liver and adult bone marrow cells. In contrast, cells that repopulate primary hosts for the same period but show more limited self-renewal activity are enriched in the CD150(-) subset. Comparative transcriptome analyses of these 2 subsets with each other and with HSCs whose self-renewal activity has been rapidly extinguished in vitro revealed 3 new genes (VWF, Rhob, Pld3) whose elevated expression is a consistent and selective feature of the long-term repopulating cells with durable self-renewal capacity. These findings establish the identity of a phenotypically and molecularly distinct class of pluripotent hematopoietic cells with lifelong self-renewal capacity.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/citología , Animales , Animales Congénicos , Antígenos CD/análisis , Antígenos de Diferenciación/análisis , Células de la Médula Ósea/citología , División Celular , Células Cultivadas/trasplante , Perfilación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Antígenos Comunes de Leucocito/análisis , Hígado/citología , Hígado/embriología , Ratones , Ratones Endogámicos C57BL , Fosfolipasa D/análisis , Quimera por Radiación , Receptores de Superficie Celular/análisis , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Proteína de Unión al GTP rhoB/análisis , Proteína de Unión al GTP rhoB/genética , Factor de von Willebrand/análisis , Factor de von Willebrand/genética
13.
BMC Bioinformatics ; 10: 184, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19531228

RESUMEN

BACKGROUND: Flow cytometry technology is widely used in both health care and research. The rapid expansion of flow cytometry applications has outpaced the development of data storage and analysis tools. Collaborative efforts being taken to eliminate this gap include building common vocabularies and ontologies, designing generic data models, and defining data exchange formats. The Minimum Information about a Flow Cytometry Experiment (MIFlowCyt) standard was recently adopted by the International Society for Advancement of Cytometry. This standard guides researchers on the information that should be included in peer reviewed publications, but it is insufficient for data exchange and integration between computational systems. The Functional Genomics Experiment (FuGE) formalizes common aspects of comprehensive and high throughput experiments across different biological technologies. We have extended FuGE object model to accommodate flow cytometry data and metadata. METHODS: We used the MagicDraw modelling tool to design a UML model (Flow-OM) according to the FuGE extension guidelines and the AndroMDA toolkit to transform the model to a markup language (Flow-ML). We mapped each MIFlowCyt term to either an existing FuGE class or to a new FuGEFlow class. The development environment was validated by comparing the official FuGE XSD to the schema we generated from the FuGE object model using our configuration. After the Flow-OM model was completed, the final version of the Flow-ML was generated and validated against an example MIFlowCyt compliant experiment description. RESULTS: The extension of FuGE for flow cytometry has resulted in a generic FuGE-compliant data model (FuGEFlow), which accommodates and links together all information required by MIFlowCyt. The FuGEFlow model can be used to build software and databases using FuGE software toolkits to facilitate automated exchange and manipulation of potentially large flow cytometry experimental data sets. Additional project documentation, including reusable design patterns and a guide for setting up a development environment, was contributed back to the FuGE project. CONCLUSION: We have shown that an extension of FuGE can be used to transform minimum information requirements in natural language to markup language in XML. Extending FuGE required significant effort, but in our experiences the benefits outweighed the costs. The FuGEFlow is expected to play a central role in describing flow cytometry experiments and ultimately facilitating data exchange including public flow cytometry repositories currently under development.


Asunto(s)
Biología Computacional/métodos , Citometría de Flujo , Almacenamiento y Recuperación de la Información/métodos , Lenguajes de Programación , Bases de Datos Factuales
14.
Leuk Res ; 80: 1-10, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30852438

RESUMEN

Iron metabolism is altered in a variety of cancers; however, little is known about the role of iron metabolism in the biology and response to therapy of acute myeloid leukemia (AML). Here we show that SLC40A1, the gene encoding the iron exporter ferroportin (FPN), is variably expressed among primary AMLs and that low levels are associated with good prognosis and improved outcomes. In particular, core binding factor (CBF) AMLs, which are associated with good outcomes with chemotherapy, consistently have low level of SLC40A1 expression. AML cell lines that expressed relatively low levels of FPN endogenously, or were engineered via gene knockdown, had an increased sensitivity to chemotherapy relative to controls expressing high levels of FPN. Primary FPNlow AML bulk cells also had increased sensitivity to Ara-C treatment, iron treatment and the combination of Ara-C and iron relative to FPNhigh cells. FPNlow leukemic stem cells (LSCs) had decreased viability following addition of iron alone and in combination with Ara-C treatment relative to FPNhigh LSCs. Together these observations suggest a model where FPN mediated iron metabolism may play a role in chemosensitivity and outcome to therapy in AML.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proliferación Celular/efectos de los fármacos , Citarabina/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Biomarcadores de Tumor/genética , Proteínas de Transporte de Catión/genética , Análisis Citogenético , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
15.
Cell Rep ; 27(1): 238-254.e6, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30943405

RESUMEN

The NADPH-dependent oxidase NOX2 is an important effector of immune cell function, and its activity has been linked to oncogenic signaling. Here, we describe a role for NOX2 in leukemia-initiating stem cell populations (LSCs). In a murine model of leukemia, suppression of NOX2 impaired core metabolism, attenuated disease development, and depleted functionally defined LSCs. Transcriptional analysis of purified LSCs revealed that deficiency of NOX2 collapses the self-renewal program and activates inflammatory and myeloid-differentiation-associated programs. Downstream of NOX2, we identified the forkhead transcription factor FOXC1 as a mediator of the phenotype. Notably, suppression of NOX2 or FOXC1 led to marked differentiation of leukemic blasts. In xenotransplantation models of primary human myeloid leukemia, suppression of either NOX2 or FOXC1 significantly attenuated disease development. Collectively, these findings position NOX2 as a critical regulator of malignant hematopoiesis and highlight the clinical potential of inhibiting NOX2 as a means to target LSCs.


Asunto(s)
Autorrenovación de las Células , Leucemia/sangre , Leucopoyesis , Células Progenitoras Mieloides/metabolismo , NADPH Oxidasa 2/metabolismo , Animales , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Leucemia/genética , Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/patología , NADPH Oxidasa 2/genética
16.
Cytometry A ; 73(10): 926-30, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18752282

RESUMEN

A fundamental tenet of scientific research is that published results are open to independent validation and refutation. Minimum data standards aid data providers, users, and publishers by providing a specification of what is required to unambiguously interpret experimental findings. Here, we present the Minimum Information about a Flow Cytometry Experiment (MIFlowCyt) standard, stating the minimum information required to report flow cytometry (FCM) experiments. We brought together a cross-disciplinary international collaborative group of bioinformaticians, computational statisticians, software developers, instrument manufacturers, and clinical and basic research scientists to develop the standard. The standard was subsequently vetted by the International Society for Advancement of Cytometry (ISAC) Data Standards Task Force, Standards Committee, membership, and Council. The MIFlowCyt standard includes recommendations about descriptions of the specimens and reagents included in the FCM experiment, the configuration of the instrument used to perform the assays, and the data processing approaches used to interpret the primary output data. MIFlowCyt has been adopted as a standard by ISAC, representing the FCM scientific community including scientists as well as software and hardware manufacturers. Adoptionof MIFlowCyt by the scientific and publishing communities will facilitate third-party understanding and reuse of FCM data.


Asunto(s)
Sistemas de Administración de Bases de Datos , Citometría de Flujo/normas , Guías como Asunto , Separación Celular/instrumentación , Separación Celular/métodos , Separación Celular/normas , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Indicadores y Reactivos/normas , Manejo de Especímenes/métodos , Manejo de Especímenes/normas
17.
Exp Hematol ; 35(5): 817-30, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17577930

RESUMEN

OBJECTIVE: Strategies to expand hematopoietic stem cells (HSCs) ex vivo are of key interest. The objective of this study was to resolve if ability of HOXB4, previously documented to induce a significant expansion of HSCs in culture, may extend to other HOX genes and also to further analyze the HOX sequence requirements to achieve this effect. METHODS: To investigate the ability of Nucleoporin98-Homeobox fusion genes to stimulate HSC self-renewal, we evaluated their presence in 10- to 20-day cultures of transduced mouse bone marrow cells. Stem cell recovery was measured by limiting-dilution assay for long-term competitive repopulating cells (CRU Assay). RESULTS: These experiments revealed remarkable expansions of Nucleoporin98-Homeobox-transduced HSCs (1000-fold to 10,000-fold over input) in contrast to the expected decline of HSCs in control cultures. Nevertheless, the Nucleoporin98-Homeobox-expanded HSCs displayed no proliferative senescence and retained normal lympho-myeloid differentiation activity and a controlled pool size in vivo. Analysis of proviral integration patterns showed the cells regenerated in vivo were highly polyclonal, indicating they had derived from a large proportion of the initially targeted HSCs. Importantly, these effects were preserved when all HOX sequences flanking the homeodomain were removed, thus defining the homeodomain as a key and independent element in the fusion. CONCLUSION: These findings create new possibilities for investigating HSCs biochemically and genetically and for achieving clinically significant expansion of human HSCs.


Asunto(s)
Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas Recombinantes de Fusión/fisiología , Factores de Transcripción/genética , Animales , Técnicas de Cultivo de Célula/métodos , Trasplante de Células Madre Hematopoyéticas , Proteínas Homeobox A10 , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética
18.
Chem Biol Interact ; 276: 46-51, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28645468

RESUMEN

Multiple studies have demonstrated that ALDH1A1 is elevated in hematopoietic stem cells (HSCs). As a means to better characterize such cells, we previously developed the fluorescent ALDH1A1 substrate Aldefluor to facilitate HSC identification and isolation. This has proven useful for counting and isolating HSCs from human bone marrow, peripheral blood and cord blood as well as stem cells in other tissues and organisms. Given the high level expression of ALDH1A1, we explored its biology and that of other ALDHs in HSCs and found that ALDH1A1 and ALDH3A1 were important in metabolizing reactive aldehydes (RAlds) and reactive oxygen species (ROS). In murine models, loss of these two isoforms resulted in a variety of effects on HSC biology, increased DNA damage and predisposition to leukemia formation when combined with a genetic driver of HSC proliferation and self-renewal. Loss of ALDH activity may also predispose to marrow failure and AML in Fanconi's anemia (FA). ALDHs also have importance in mediating drug resistance in AML, may be useful in the identification of leukemia stem cells (LSCs) and ALDH activity levels may have prognostic significance. Together these findings suggest that further studying ALDH biology in AML and other blood cancers may provide important insights into malignant transformation and may point the way to the development of novel diagnostics and therapies.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Neoplasias Hematológicas/terapia , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/terapia , Aldehído Deshidrogenasa/genética , Animales , Antineoplásicos/uso terapéutico , Células Sanguíneas/citología , Células de la Médula Ósea/citología , Proliferación Celular , Autorrenovación de las Células , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Sangre Fetal/citología , Neoplasias Hematológicas/diagnóstico , Células Madre Hematopoyéticas/citología , Humanos , Isoenzimas/metabolismo , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Pronóstico , Especies Reactivas de Oxígeno/metabolismo
19.
Cancer Res ; 63(16): 5126-35, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941844

RESUMEN

Treatment with LAQ824 (Novartis Pharmaceutical, Inc.), a cinnamyl hydroxamic acid analogue inhibitor of histone deacetylases, depleted the mRNA and protein expression of Bcr-Abl in human chronic myeloid leukemia blast crisis (CML-BC) cells. Exposure to LAQ824 induced the expression of the cell cycle-dependent kinase inhibitors p21 and p27 and caused cell cycle G(1)-phase accumulation and apoptosis of CML-BC cells. LAQ824 also induced acetylation of heat shock protein 90. This inhibited the chaperone association of Bcr-Abl with heat shock protein 90, thereby promoting the proteasomal degradation of Bcr-Abl. Cotreatment with LAQ824 increased imatinib mesylate-induced apoptosis of CML-BC cells. Additionally, LAQ824 down-regulated the levels of mutant Bcr-Abl possessing the T315I point mutation, as well as induced apoptosis of imatinib-refractory primary CML-BC cells. Therefore, LAQ824 may be a promising therapeutic agent in the treatment of imatinib-sensitive or -refractory human leukemia.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Crisis Blástica/tratamiento farmacológico , Cisteína Endopeptidasas/fisiología , Inhibidores Enzimáticos/farmacología , Proteínas de Fusión bcr-abl/metabolismo , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Complejos Multienzimáticos/fisiología , Proteínas Musculares , Piperazinas/farmacología , Pirimidinas/farmacología , Acetilación , Benzamidas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/genética , Fase G1/efectos de los fármacos , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de Microfilamentos/biosíntesis , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal , Piridonas/farmacología , Células Tumorales Cultivadas
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA