Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 127(20): 2451-9, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-26980726

RESUMEN

Acute myeloid leukemia (AML) with t(8;21) or inv(16) have been recognized as unique entities within AML and are usually reported together as core binding factor AML (CBF-AML). However, there is considerable clinical and biological heterogeneity within this group of diseases, and relapse incidence reaches up to 40%. Moreover, translocations involving CBFs are not sufficient to induce AML on its own and the full spectrum of mutations coexisting with CBF translocations has not been elucidated. To address these issues, we performed extensive mutational analysis by high-throughput sequencing in 215 patients with CBF-AML enrolled in the Phase 3 Trial of Systematic Versus Response-adapted Timed-Sequential Induction in Patients With Core Binding Factor Acute Myeloid Leukemia and Treating Patients with Childhood Acute Myeloid Leukemia with Interleukin-2 trials (age, 1-60 years). Mutations in genes activating tyrosine kinase signaling (including KIT, N/KRAS, and FLT3) were frequent in both subtypes of CBF-AML. In contrast, mutations in genes that regulate chromatin conformation or encode members of the cohesin complex were observed with high frequencies in t(8;21) AML (42% and 18%, respectively), whereas they were nearly absent in inv(16) AML. High KIT mutant allele ratios defined a group of t(8;21) AML patients with poor prognosis, whereas high N/KRAS mutant allele ratios were associated with the lack of KIT or FLT3 mutations and a favorable outcome. In addition, mutations in epigenetic modifying or cohesin genes were associated with a poor prognosis in patients with tyrosine kinase pathway mutations, suggesting synergic cooperation between these events. These data suggest that diverse cooperating mutations may influence CBF-AML pathophysiology as well as clinical behavior and point to potential unique pathogenesis of t(8;21) vs inv(16) AML.


Asunto(s)
Inversión Cromosómica , Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Factores de Unión al Sitio Principal/genética , ADN de Neoplasias/genética , Leucemia Mieloide Aguda/genética , Mutación , Translocación Genética , Adolescente , Adulto , Alelos , Proteínas de Ciclo Celular/genética , Niño , Preescolar , Cromatina/genética , Cromatina/ultraestructura , Proteínas Cromosómicas no Histona/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Pronóstico , Proteína 1 Compañera de Translocación de RUNX1 , Adulto Joven , Cohesinas
2.
J Pediatr Hematol Oncol ; 40(1): 43-47, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29189507

RESUMEN

Central nervous system (CNS) involvement at diagnosis of pediatric acute myeloid leukemia (AML) is not considered as an independent prognostic factor. This study describes the prognostic value of pediatric AML with CNS involvement at diagnosis. Pediatric patients were treated for de novo AML in the French multicenter trial ELAM02. Lumbar puncture was carried out in the first week, and the treatment was adapted to the CNS status. No patient received CNS radiotherapy. The patients were classified into 2 groups: CNS+ and CNS-. Of the 438 patients, 16% (n=70) had CNS involvement at diagnosis, and 29% showed clinical signs. The patients with CNS disease were younger (40% were below 2 y old), had a higher white blood cell count (median of 45 vs. 13 G/L), and had M4 and M5 morphologies. The complete remission rate was similar at 92.8% for CNS+ and 88.5% for CNS-. There was no significant difference between the CNS+ and the CNS- group in overall survival (76% and 71%, respectively) and event-free survival (57% and 52%, respectively). Regarding the occurrence of first relapse, the CNS+ group had a higher combined relapse rate of 26.1% compared with 10% for the CNS- group. The results indicate that CNS involvement at diagnosis of pediatric AML is not an independent prognostic factor. Triple intrathecal chemotherapy combined with high-dose intravenous cytarabine should be the first-line treatment for CNS disease.


Asunto(s)
Neoplasias del Sistema Nervioso Central/diagnóstico , Leucemia Mieloide Aguda/diagnóstico , Pronóstico , Adolescente , Factores de Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Estudios de Casos y Controles , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/mortalidad , Niño , Preescolar , Citarabina/administración & dosificación , Francia , Humanos , Lactante , Leucemia Monocítica Aguda , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Leucemia Mielomonocítica Aguda , Recuento de Leucocitos , Recurrencia , Análisis de Supervivencia
3.
Pediatr Hematol Oncol ; 34(8): 425-427, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29303660

RESUMEN

We report the outcome of 27 children with de novo acute megakaryoblastic leukemia (AMKL) (excluding Down syndrome) enrolled in the French multicenter prospective study ELAM02 (2005-2011). There was no difference in gender, initial leukocyte count, CNS involvement, and complete remission rate (88.9%), as compared to other acute myeloid leukemia (AML) subtypes. AMKL patients had a significantly poorer outcome (5-year overall survival 54% [CI 95% 33%-71%] than children with other AML subtypes (5-year overall survival 73% [CI 95% 68%-77%] p = 0.02). Gender, age, CNS leukemia, hyperleukocytosis, complete remission or cytogenetic subgroups were not significant prognostic factors of disease-free survival. AMKL (excluding Down syndrom) remains an AML subgroup with inferior outcome.


Asunto(s)
Leucemia Megacarioblástica Aguda/mortalidad , Niño , Preescolar , Supervivencia sin Enfermedad , Síndrome de Down , Femenino , Francia/epidemiología , Humanos , Lactante , Leucemia Megacarioblástica Aguda/sangre , Leucemia Megacarioblástica Aguda/terapia , Masculino , Estudios Prospectivos , Tasa de Supervivencia
4.
Eur J Nucl Med Mol Imaging ; 43(7): 1220-30, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26660144

RESUMEN

PURPOSE: To evaluate, in children with Hodgkin lymphoma (HL), the frequency and intensity of visually diffuse FDG uptake by selected organs at baseline (bPET) and on interim PET/CT (iPET), and to evaluate the relation between FDG uptake, metabolic response and evolution of the disease with treatment. PATIENTS AND METHODS: Thirty children with HL had bPET and then iPET after two cycles of treatment, which were blind-read retrospectively. Excluding sites with focal uptake, diffuse FDG uptake by thymus, bone marrow at iliac crests, liver, spleen, and the spinal cord at the 12th thoracic vertebra (Th12) was evaluated visually using a three-point scoring method and semiquantitatively by measuring SUVmax. Visualisation of activated brown adipose tissue (BAT) was also quoted. Five children had refractory HL. Recurrence-free survival was determined for each patient. Nine patients relapsed; in 21 non-relapsing patients, the median follow-up period was 43 months (range: 28-61). RESULTS: On bPET, the rate of diffuse and intense (visual score = 3) FDG uptake was 48 % in the spleen, 43 % in the spinal cord at Th12, 37 % in bone marrow, 21 % in the thymus and 7 % in BAT. At least one of those sites showed diffuse and intense FDG uptake in 77 % of patients. On iPET, a significant decrease in SUVmax was observed in thymus, iliac crest bone marrow and spleen, but not in spinal cord. In contrast, the FDG uptake by the liver significantly increased. The absence of SUVmax increase in the liver between bPET and iPET was the best criterion to predict a refractory disease (PPV = 55 %, NPV = 100 %). Its area under ROC (AUC) was 0.9 vs. 0.73 for five-point Deauville criteria. For prediction of relapse, two criteria were derived from the evolution of diffuse uptake between bPET and iPET: no increase in liver uptake and an increase > 5 % in spinal cord uptake. As compared with 13 patients who matched none of those criteria, the hazard ratio (HR) for relapse was 2.1 in 13 patients who matched one criterion, and 10.3 in four patients who matched both (Kaplan-Meier analysis p = 0.005). CONCLUSION: Diffuse and intense FDG uptake by organs is frequent in children with HL on bPET. On iPET, it is frequently reduced in all sites except the liver, which may pose problems for visual quotation of the FDG intensity of HL foci. The variation of SUVmax between bPET and iPET permitted us to achieve a prediction of refractory or relapsing HL that was at least as effective as using criteria based on FDG uptake by the HL lesions. The results of this retrospective pilot study need further validation.


Asunto(s)
Fluorodesoxiglucosa F18/metabolismo , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Adolescente , Transporte Biológico , Niño , Preescolar , Difusión , Femenino , Enfermedad de Hodgkin/terapia , Humanos , Masculino , Proyectos Piloto
5.
Blood ; 118(10): 2849-56, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21765021

RESUMEN

Hematopoietic stem cells (HSCs) engage in complex bidirectional signals with the hematopoietic microenvironment (HM), and there is emerging evidence that leukemia stem cells (LSCs) may use similar interactions. Using a syngeneic retroviral model of MLL-AF9 induced acute myeloid leukemia (AML), we have identified 2 different stages of leukemia progression, propagated by "pre-LSCs" and established leukemia (LSCs) and compared the homing properties of these distinctive entities to that of normal HSCs. The homing and microlocalization of pre-LSCs was most similar to long-term HSCs and was dependent on cell-intrinsic Wnt signaling. In contrast, the homing of established LSCs was most similar to that of committed myeloid progenitors and distinct from HSCs. Although osteoblast-derived Dickkopf-1, a potent Wnt inhibitor known to impair HSC function, dramatically impaired normal HSC localization within the bone marrow, it did not affect pre-LSCs, LSC homing, or AML development. Mechanistically, cell-intrinsic Wnt activation was observed in human and murine AML samples, explaining the independence of MLL-AF9 LSCs from niche-derived Wnt signals. These data identify differential engagement of HM associated with leukemic progression and identify an LSC niche that is physically distinct and independent of the constraints of Wnt signaling that apply to normal HSCs.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Nicho de Células Madre , Proteínas Wnt/metabolismo , Animales , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Citometría de Flujo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Proteínas Wnt/genética
6.
Blood ; 116(22): 4464-73, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20709909

RESUMEN

Adhesion properties of hematopoietic stem cells (HSCs) in the bone marrow (BM) niches control their migration and affect their cell-cycle dynamics. The serum response factor (Srf) regulates growth factor-inducible genes and genes controlling cytoskeleton structures involved in cell spreading, adhesion, and migration. We identified a role for Srf in HSC adhesion and steady-state hematopoiesis. Conditional deletion of Srf in BM cells resulted in a 3-fold expansion of the long- and short-term HSCs and multipotent progenitors (MPPs), which occurs without long-term modification of cell-cycle dynamics. Early differentiation steps to myeloid and lymphoid lineages were normal, but Srf loss results in alterations in mature-cell production and severe thrombocytopenia. Srf-null BM cells also displayed compromised engraftment properties in transplantation assays. Gene expression analysis identified Srf target genes expressed in HSCs, including a network of genes associated with cell migration and adhesion. Srf-null stem cells and MPPs displayed impair expression of the integrin network and decreased adherence in vitro. In addition, Srf-null mice showed increase numbers of circulating stem and progenitor cells, which likely reflect their reduced retention in the BM. Altogether, our results demonstrate that Srf is an essential regulator of stem cells and MPP adhesion, and suggest that Srf acts mainly through cell-matrix interactions and integrin signaling.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/citología , Factor de Respuesta Sérica/metabolismo , Animales , Adhesión Celular , Ciclo Celular , Linaje de la Célula , Eliminación de Gen , Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Integrinas/metabolismo , Ratones , Factor de Respuesta Sérica/genética
7.
Haematologica ; 97(3): 379-87, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22058212

RESUMEN

BACKGROUND: The nucleoporin gene NUP98 is rearranged in more than 27 chromosomal abnormalities observed in childhood and adult, de novo and therapy-related acute leukemias of myeloid and T-lymphoid origins, resulting in the creation of fusion genes and the expression of chimeric proteins. We report here the functional analysis of the NUP98-coiled-coil domain-containing protein 28A (NUP98-CCDC28A) fusion protein, expressed as the consequence of a recurrent t(6;11)(q24.1;p15.5) translocation. DESIGN AND METHODS: To gain insight into the function of the native CCDC28A gene, we collected information on any differential expression of CCDC28A among normal hematologic cell types and within subgroups of acute leukemia. To assess the in vivo effects of the NUP98-CCDC28A fusion, NUP98-CCDC28A or full length CCDC28A were retrovirally transduced into primary murine bone marrow cells and transduced cells were next transplanted into sub-lethally irradiated recipient mice. RESULTS: Our in silico analyses supported a contribution of CCDC28A to discrete stages of murine hematopoietic development. They also suggested selective enrichment of CCDC28A in the French-American-British M6 class of human acute leukemia. Primary murine hematopoietic progenitor cells transduced with NUP98-CCDC28A generated a fully penetrant and transplantable myeloproliferative neoplasm-like myeloid leukemia and induced selective expansion of granulocyte/macrophage progenitors in the bone marrow of transplanted recipients, showing that NUP98-CCDC28A promotes the proliferative capacity and self-renewal potential of myeloid progenitors. In addition, the transformation mediated by NUP98-CCDC28A was not associated with deregulation of the Hoxa-Meis1 pathway, a feature shared by a diverse set of NUP98 fusions. CONCLUSIONS: Our results demonstrate that the recurrent NUP98-CCDC28A is an oncogene that induces a rapid and transplantable myeloid neoplasm in recipient mice. They also provide additional evidence for an alternative leukemogenic mechanism for NUP98 oncogenes.


Asunto(s)
Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Núcleo Celular/metabolismo , Proliferación Celular , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 6 , Expresión Génica , Células Progenitoras de Granulocitos y Macrófagos/patología , Proteínas de Homeodominio/metabolismo , Humanos , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/mortalidad , Proteínas de Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Isoformas de Proteínas/genética , Transporte de Proteínas , Proteínas/metabolismo , Alineación de Secuencia , Translocación Genética
8.
Blood ; 112(10): 4220-6, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18755984

RESUMEN

Oncogenic activation of tyrosine kinase signaling pathway is recurrent in human leukemia. To gain insight into the oncogenic process leading to acute megakaryoblastic leukemia (AMKL), we performed sequence analyses of a subset of oncogenes known to be activated in human myeloid and myeloproliferative disorders. In a series of human AMKL samples from both Down syndrome and non-Down syndrome patients, mutations were identified within KIT, FLT3, JAK2, JAK3, and MPL genes, with a higher frequency in DS than in non-DS patients. The novel mutations were analyzed using BaF3 cells, showing that JAK3 mutations were activating mutations. Finally, we report a novel constitutively active MPL mutant, MPLT487A, observed in a non-Down syndrome childhood AMKL that induces a myeloproliferative disease in mouse bone marrow transplantation assay.


Asunto(s)
Síndrome de Down/genética , Leucemia Megacarioblástica Aguda/genética , Mutación , Proteínas de Neoplasias/genética , Adulto , Anciano , Animales , Línea Celular Tumoral , Niño , Preescolar , Síndrome de Down/metabolismo , Femenino , Humanos , Lactante , Recién Nacido , Leucemia Megacarioblástica Aguda/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Trasplante de Neoplasias
9.
Hemasphere ; 2(6): e159, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31723797

RESUMEN

Despite significant progress in the treatment of pediatric acute myeloblastic leukemia (AML), relapse remains the commonest cause of death. Randomized ELAM02 trial questioned if maintenance therapy with interleukin-2 (IL2), for 1 year, improves disease-free survival (DFS). Patients aged 0 to 18 years, with newly diagnosed AML (excluding patients with acute promyelocytic leukemia or down syndrome AML) were enrolled. They received 1 course of induction treatment (cytarabine and mitoxantrone) and 3 courses of consolidation treatment (high-dose cytarabine in courses 1 and 3). According to the cytogenetics risk, patients not undergoing hematopoietic stem cell transplantation, still in complete remission (CR) after the third course of consolidation treatment, were eligible for randomization to 1 year of maintenance therapy with monthly courses of IL2 or no maintenance treatment. There were 438 evaluable patients, 154 of whom were randomized to the IL2/no maintenance groups. Relapse occurred in 28 patients from the IL2+ group and 29 patients in the IL2- group. Survival was similar in the 2 groups, with a 4-year DFS of 62% without IL2 and 66% with IL2 (P = 0.75). In the CBF population, 4-year DFS was 55% without IL2 and 78% with IL2 (P = 0.07). No deaths from toxicity or excess of serious adverse events related to IL2 treatment were recorded. Prolonged IL2 for maintenance therapy after intensive chemotherapy is feasible and safe in pediatric AML patients in their first CR. Such treatment did not improve DFS in this study, but a positive trend was observed in favor of IL2 maintenance therapy among core binding factor acute myeloblastic leukemia.

10.
J Clin Oncol ; 33(27): 2949-62, 2015 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-26304895

RESUMEN

Diagnosis, treatment, response monitoring, and outcome of pediatric acute myeloid leukemia (AML) have made enormous progress during the past decades. Because AML is a rare type of childhood cancer, with an incidence of approximately seven occurrences per 1 million children annually, national and international collaborative efforts have evolved. This overview describes these efforts and includes a summary of the history and contributions of each of the main collaborative pediatric AML groups worldwide. The focus is on translational and clinical research, which includes past, current, and future clinical trials. Separate sections concern acute promyelocytic leukemia, myeloid leukemia of Down syndrome, and relapsed AML. A plethora of novel antileukemic agents that have emerged, including new classes of drugs, are summarized as well. Finally, an important aspect of the treatment of pediatric AML--supportive care--and late effects are discussed. The future is bright, with a wide range of emerging innovative therapies and with more and more international collaboration that ultimately aim to cure all children with AML, with fewer adverse effects and without late effects.


Asunto(s)
Comunicación Interdisciplinaria , Cooperación Internacional , Leucemia Mieloide Aguda/terapia , Oncología Médica/tendencias , Pediatría/tendencias , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Conducta Cooperativa , Difusión de Innovaciones , Supervivencia sin Enfermedad , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Lactante , Recién Nacido , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/historia , Leucemia Mieloide Aguda/mortalidad , Oncología Médica/historia , Pediatría/historia , Factores de Riesgo , Sobrevivientes , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
12.
J Clin Invest ; 123(1): 299-314, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23202735

RESUMEN

Aberrant expression of the homeodomain transcription factor CDX2 occurs in most cases of acute myeloid leukemia (AML) and promotes leukemogenesis, making CDX2, in principle, an attractive therapeutic target. Conversely, CDX2 acts as a tumor suppressor in colonic epithelium. The effectors mediating the leukemogenic activity of CDX2 and the mechanism underlying its context-dependent properties are poorly characterized, and strategies for interfering with CDX2 function in AML remain elusive. We report data implicating repression of the transcription factor KLF4 as important for the oncogenic activity of CDX2, and demonstrate that CDX2 differentially regulates KLF4 in AML versus colon cancer cells through a mechanism that involves tissue-specific patterns of promoter binding and epigenetic modifications. Furthermore, we identified deregulation of the PPARγ signaling pathway as a feature of CDX2-associated AML and observed that PPARγ agonists derepressed KLF4 and were preferentially toxic to CDX2+ leukemic cells. These data delineate transcriptional programs associated with CDX2 expression in hematopoietic cells, provide insight into the antagonistic duality of CDX2 function in AML versus colon cancer, and suggest reactivation of KLF4 expression, through modulation of PPARγ signaling, as a therapeutic modality in a large proportion of AML patients.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/biosíntesis , Leucemia Mieloide Aguda/metabolismo , PPAR gamma/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Factor de Transcripción CDX2 , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células HL-60 , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Proteínas de Homeodominio/genética , Humanos , Células K562 , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Especificidad de Órganos/genética , PPAR gamma/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Células U937
13.
Cell Stem Cell ; 11(3): 429-39, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22958934

RESUMEN

The mechanistic target of rapamycin (mTOR) pathway serves as a key sensor of cellular-energetic state and functions to maintain tissue homeostasis. Hyperactivation of the mTOR pathway impairs hematopoietic stem cell (HSC) function and is associated with leukemogenesis. However, the roles of the unique mTOR complexes (mTORCs) in hematopoiesis and leukemogenesis have not been adequately elucidated. We deleted the mTORC1 component, regulatory-associated protein of mTOR (Raptor), in mouse HSCs and its loss causes a nonlethal phenotype characterized by pancytopenia, splenomegaly, and the accumulation of monocytoid cells. Furthermore, Raptor is required for HSC regeneration, and plays largely nonredundant roles with rapamycin-insensitive companion of mTOR (Rictor) in these processes. Ablation of Raptor also significantly extends survival of mice in models of leukemogenesis evoked by Pten deficiency. These data delineate critical roles for mTORC1 in hematopoietic function and leukemogenesis and inform clinical strategies based on chronic mTORC1 inhibition.


Asunto(s)
Transformación Celular Neoplásica/patología , Hematopoyesis , Leucemia/enzimología , Leucemia/patología , Complejos Multiproteicos/metabolismo , Fosfohidrolasa PTEN/deficiencia , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras , Ciclo Celular/genética , Diferenciación Celular , Linaje de la Célula , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica , Hematopoyesis/genética , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Homeostasis , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Fosfohidrolasa PTEN/metabolismo , Proteína Reguladora Asociada a mTOR , Análisis de Supervivencia
14.
Nat Rev Immunol ; 12(1): 49-60, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22193770

RESUMEN

Progenitor cells that are the basis for all blood cell production share the bone marrow with more mature elements of the adaptive immune system. Specialized niches within the bone marrow guide and, at times, constrain the development of haematopoietic stem and progenitor cells (HSPCs) and lineage-restricted immune progenitor cells. Specific niche components are organized into distinct domains to create a diversified landscape in which specialized cell differentiation or population expansion programmes proceed. Local cues that reflect the tissue and organismal state affect cellular interactions to alter the production of a range of cell types. Here, we review the organization of regulatory elements in the bone marrow and discuss how these elements provide a dynamic means for the host to modulate stem cell and adaptive immune cell responses to physiological challenges.


Asunto(s)
Médula Ósea/inmunología , Médula Ósea/metabolismo , Inmunidad , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Nicho de Células Madre/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA