Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Blood ; 119(21): 4898-907, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22490678

RESUMEN

Aging degrades hematopoietic stem cell (HSC) functions, including stress response; however, the involved molecular pathways are incompletely defined. Murine BM conditionally deleted for One-Twenty-Two-1 (Ott1), is able to maintain lifelong hematopoiesis and has preserved numbers of long-term HSCs, yet cannot repopulate nor sustain itself after transplantation against a competitor even when Ott1 is excised after engraftment. We show, specifically under replicative stress, that Ott1-deleted HSCs have a significant reduction of the G(0) cell-cycle fraction associated with self-renewal and undergo early failure. Therefore, Ott1 is required to preserve HSC quiescence during stress but not steady-state hematopoiesis. Reduced tolerance of replicative stress, increased myeloid potential, and greater absolute numbers are mutual characteristics of both Ott1-deleted and aged HSCs, and comparison of their gene expression profiles reveals a shared signature. Ott1-deleted HSCs share multiple aging-associated physiologic changes, including increases in NF-κB activation and DNA damage. Loss of Ott1 causes increased reactive oxygen species; however, antioxidant treatment does not rescue the competitive defect, indicating the existence of additional essential Ott1-dependent HSC pathways. In conclusion, our data establish a requirement for Ott1 in stress hematopoiesis and suggest that Ott1-dependent processes may converge with those affected by aging.


Asunto(s)
Proliferación Celular , Senescencia Celular/genética , Células Madre Hematopoyéticas/fisiología , Proteínas de Unión al ARN/fisiología , Fase de Descanso del Ciclo Celular/genética , Estrés Fisiológico , Animales , Células Cultivadas , Senescencia Celular/fisiología , Técnicas de Inactivación de Genes , Hematopoyesis/genética , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Fase de Descanso del Ciclo Celular/fisiología , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología
2.
Clin Cancer Res ; 29(5): 943-956, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36537918

RESUMEN

PURPOSE: The Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) polycythemia vera, essential thrombocythemia, and primary myelofibrosis are characterized by JAK/STAT pathway activation. JAK inhibitors are approved for MPN treatment, but persistence has been observed, due to JAK/STAT reactivation. EXPERIMENTAL DESIGN: Using MPN patient samples, JAK2-mutated cell lines, and MPN mouse models, we examined both the efficacy and mechanism by which crizotinib, the ALK/MET/RON/ROS1 inhibitor approved for the treatment of non-small cell lung cancer, alters MPN cell proliferation and JAK/STAT activation. RESULTS: We found that crizotinib suppresses proliferation and activation of JAK/STAT signaling, and decreases the disease burden in the JAK2V617F mouse model of MPN. Furthermore, we found that crizotinib could overcome JAK inhibitor persistence to ruxolitinib. Interestingly, phosphorylation of the crizotinib target RON kinase was enhanced in ruxolitinib-persistent cells. We show that phospho-JAK2 and phospho-RON can physically interact to sustain JAK/STAT signaling, and that the combination of crizotinib and ruxolitinib disrupts this interaction. Furthermore, RON knockdown suppresses proliferation and activation of JAK/STAT signaling in JAK2-mutated cells, and RON deletion in a JAK2V617F mouse MPN model decreases the disease burden. We also observed RON hyperactivation in MPN patient cells, suggesting that RON may be an important target of crizotinib in MPN. CONCLUSIONS: In summary, we demonstrate that crizotinib has preclinical efficacy in MPN patient cells, JAK2-mutated cell lines, and a JAK2-mutated mouse model, and that the combination of crizotinib with JAK inhibitors suppresses JAK inhibitor persistence. Our work suggests that crizotinib should be investigated for the treatment of patients with MPN.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Inhibidores de las Cinasas Janus , Neoplasias Pulmonares , Trastornos Mieloproliferativos , Animales , Ratones , Inhibidores de las Cinasas Janus/uso terapéutico , Crizotinib/farmacología , Crizotinib/uso terapéutico , Quinasas Janus/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Transducción de Señal , Factores de Transcripción STAT/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Janus Quinasa 2/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Mutación
3.
Blood ; 115(7): 1406-15, 2010 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-20008787

RESUMEN

Human cancers, including acute myeloid leukemia (AML), commonly display constitutive phosphoinositide 3-kinase (PI3K) AKT signaling. However, the exact role of AKT activation in leukemia and its effects on hematopoietic stem cells (HSCs) are poorly understood. Several members of the PI3K pathway, phosphatase and tensin homolog (Pten), the forkhead box, subgroup O (FOXO) transcription factors, and TSC1, have demonstrated functions in normal and leukemic stem cells but are rarely mutated in leukemia. We developed an activated allele of AKT1 that models increased signaling in normal and leukemic stem cells. In our murine bone marrow transplantation model using a myristoylated AKT1 (myr-AKT), recipients develop myeloproliferative disease, T-cell lymphoma, or AML. Analysis of the HSCs in myr-AKT mice reveals transient expansion and increased cycling, associated with impaired engraftment. myr-AKT-expressing bone marrow cells are unable to form cobblestones in long-term cocultures. Rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR) rescues cobblestone formation in myr-AKT-expressing bone marrow cells and increases the survival of myr-AKT mice. This study demonstrates that enhanced AKT activation is an important mechanism of transformation in AML and that HSCs are highly sensitive to excess AKT/mTOR signaling.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , División Celular/fisiología , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/citología , Leucemia Mieloide Aguda/tratamiento farmacológico , Linfoma de Células T/tratamiento farmacológico , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Ratones , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/farmacología , Bazo/citología , Serina-Treonina Quinasas TOR
4.
JCI Insight ; 52019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31120863

RESUMEN

Many cytokines and chemokines that are important for hematopoiesis activate the PI3K signaling pathway. Because this pathway is frequently mutated and activated in cancer, PI3K inhibitors have been developed for the treatment of several malignancies, and are now being tested in the clinic in combination with chemotherapy. However, the role of PI3K in adult hematopoietic stem cells (HSCs), particularly during hematopoietic stress, is still unclear. We previously showed that the individual PI3K catalytic isoforms P110α or P110ß have dispensable roles in HSC function, suggesting redundancy between PI3K isoforms in HSCs. We now demonstrate that simultaneous deletion of P110α and P110δ in double knockout (DKO) HSCs uncovers their redundant requirement in HSC cycling after 5-fluorouracil (5-FU) chemotherapy administration. In contrast, DKO HSCs are still able to exit quiescence in response to other stress stimuli, such as LPS. We found that DKO HSCs and progenitors have impaired sensing of inflammatory signals ex vivo, and that levels of IL1-ß and MIG are higher in the bone marrow after LPS than after 5-FU administration. Furthermore, exogenous in vivo administration of IL1-ß can induce cell cycle entry of DKO HSCs. Our findings have important clinical implications for the use of PI3K inhibitors in combination with chemotherapy.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Ciclo Celular , Fosfatidilinositol 3-Quinasa Clase I/genética , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Fluorouracilo/farmacología , Regulación Enzimológica de la Expresión Génica , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/efectos de los fármacos , Lipopolisacáridos/efectos adversos , Masculino , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Isoformas de Proteínas
5.
Oncogene ; 24(56): 8259-67, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16091734

RESUMEN

Reccurent chromosomal translocation t(4;14) (p16.3;q32.3) occurs in patients with multiple myeloma (MM) and is associated with ectopic overexpression of fibroblast growth factor receptor 3 (FGFR3) that sometimes may contain the activation mutations such as K650E thanatophoric dysplasia type II (TDII). Although there have been significant advances in therapy for MM including the use of proteasome inhibitors, t(4;14) MM has a particularly poor prognosis and most patients still die from complications related to their disease or therapy. One potential therapeutic strategy is to inhibit FGFR3 in those myeloma patients that overexpress the receptor tyrosine kinase due to chromosomal translocation. Here we evaluated PKC412, a small molecule tyrosine kinase inhibitor, for treatment of FGFR3-induced hematopoietic malignancies. PKC412 inhibited kinase activation and proliferation of hematopoietic Ba/F3 cells transformed by FGFR3 TDII or a TEL-FGFR3 fusion. Similar results were obtained in PKC412 inhibition of several different t(4;14)-positive human MM cell lines. Furthermore, treatment with PKC412 resulted in a statistically significant prolongation of survival in murine bone marrow transplant models of FGFR3 TDII-induced pre-B cell lymphoma, or a peripheral T-cell lymphoma associated TEL-FGFR3 fusion-induced myeloproliferative disease. These data indicate that PKC412 may be a useful molecularly targeted therapy for MM associated with overexpression of FGFR3, and perhaps other diseases associated with dysregulation of FGFR3 or related mutants.


Asunto(s)
Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/fisiología , Estaurosporina/análogos & derivados , Animales , Trasplante de Médula Ósea , Línea Celular Tumoral , Modelos Animales de Enfermedad , Inhibidores de Crecimiento/farmacología , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/metabolismo , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Estaurosporina/farmacología , Displasia Tanatofórica/tratamiento farmacológico , Displasia Tanatofórica/metabolismo , Transfección
6.
Nat Commun ; 7: 10739, 2016 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-26898884

RESUMEN

Myelodysplastic syndromes (MDS) are driven by complex genetic and epigenetic alterations. The MSI2 RNA-binding protein has been demonstrated to have a role in acute myeloid leukaemia and stem cell function, but its role in MDS is unknown. Here, we demonstrate that elevated MSI2 expression correlates with poor survival in MDS. Conditional deletion of Msi2 in a mouse model of MDS results in a rapid loss of MDS haematopoietic stem and progenitor cells (HSPCs) and reverses the clinical features of MDS. Inversely, inducible overexpression of MSI2 drives myeloid disease progression. The MDS HSPCs remain dependent on MSI2 expression after disease initiation. Furthermore, MSI2 expression expands and maintains a more activated (G1) MDS HSPC. Gene expression profiling of HSPCs from the MSI2 MDS mice identifies a signature that correlates with poor survival in MDS patients. Overall, we identify a role for MSI2 in MDS representing a therapeutic target in this disease.


Asunto(s)
Síndromes Mielodisplásicos/metabolismo , Proteínas de Unión al ARN/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/mortalidad
7.
J Clin Invest ; 124(4): 1794-809, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24569456

RESUMEN

The genes encoding RAS family members are frequently mutated in juvenile myelomonocytic leukemia (JMML) and acute myeloid leukemia (AML). RAS proteins are difficult to target pharmacologically; therefore, targeting the downstream PI3K and RAF/MEK/ERK pathways represents a promising approach to treat RAS-addicted tumors. The p110α isoform of PI3K (encoded by Pik3ca) is an essential effector of oncogenic KRAS in murine lung tumors, but it is unknown whether p110α contributes to leukemia. To specifically examine the role of p110α in murine hematopoiesis and in leukemia, we conditionally deleted p110α in HSCs using the Cre-loxP system. Postnatal deletion of p110α resulted in mild anemia without affecting HSC self-renewal; however, deletion of p110α in mice with KRASG12D-associated JMML markedly delayed their death. Furthermore, the p110α-selective inhibitor BYL719 inhibited growth factor-independent KRASG12D BM colony formation and sensitized cells to a low dose of the MEK inhibitor MEK162. Furthermore, combined inhibition of p110α and MEK effectively reduced proliferation of RAS-mutated AML cell lines and disease in an AML murine xenograft model. Together, our data indicate that RAS-mutated myeloid leukemias are dependent on the PI3K isoform p110α, and combined pharmacologic inhibition of p110α and MEK could be an effective therapeutic strategy for JMML and AML.


Asunto(s)
Genes ras , Hematopoyesis/genética , Hematopoyesis/fisiología , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Juvenil/enzimología , Leucemia Mielomonocítica Juvenil/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Eritropoyesis/genética , Eritropoyesis/fisiología , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/patología , Leucemia Mielomonocítica Juvenil/patología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/deficiencia , Transducción de Señal
8.
Cancer Res ; 73(1): 373-84, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23149921

RESUMEN

MOZ-TIF2 is a leukemogenic fusion oncoprotein that confers self-renewal capability to hematopoietic progenitor cells and induces acute myelogenous leukemia (AML) with long latency in bone marrow transplantation assays. Here, we report that FLT3-ITD transforms hematopoietic cells in cooperation with MOZ-TIF2 in vitro and in vivo. Coexpression of FLT3-ITD confers growth factor independent survival/proliferation, shortens disease latency, and results in an increase in the number of leukemic stem cells (LSC). We show that STAT5, a major effector of aberrant FLT3-ITD signal transduction, is both necessary and sufficient for this cooperative effect. In addition, STAT5 signaling is essential for MOZ-TIF2-induced leukemic transformation itself. Lack of STAT5 in fetal liver cells caused rapid differentiation and loss of replating capacity of MOZ-TIF2-transduced cells enriched for LSCs. Furthermore, mice serially transplanted with Stat5(-/-) MOZ-TIF2 leukemic cells develop AML with longer disease latency and finally incomplete penetrance when compared with mice transplanted with Stat5(+/+) MOZ-TIF2 leukemic cells. These data suggest that STAT5AB is required for the self-renewal of LSCs and represents a combined signaling node of FLT3-ITD and MOZ-TIF2 driven leukemogenesis. Therefore, targeting aberrantly activated STAT5 or rewired downstream signaling pathways may be a promising therapeutic option.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Southern Blotting , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología
9.
Cancer Res ; 71(12): 4117-29, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21505102

RESUMEN

Fusion oncogenes in acute myeloid leukemia (AML) promote self-renewal from committed progenitors, thereby linking transformation and self-renewal pathways. Like most cancers, AML is a genetically and biologically heterogeneous disease, but it is unclear whether transformation results from common or overlapping genetic programs acting downstream of multiple mutations or by the engagement of unique genetic programs acting cooperatively downstream of individual mutations. This distinction is important, because the involvement of common programs would imply the existence of common molecular targets to treat AML, no matter which oncogenes are involved. Here we show that the ability to promote self-renewal is a generalized property of leukemia-associated oncogenes. Disparate oncogenes initiated overlapping transformation and self-renewal gene expression programs, the common elements of which were defined in established leukemic stem cells from an animal model as well as from a large cohort of patients with differing AML subtypes, where they strongly predicted pathobiological character. Notably, individual genes commonly activated in these programs could partially phenocopy the self-renewal function of leukemia-associated oncogenes in committed murine progenitors. Furthermore, they could generate AML following expression in murine bone marrow. In summary, our findings reveal the operation of common programs of self-renewal and transformation downstream of leukemia-associated oncogenes, suggesting that mechanistically common therapeutic approaches to AML are likely to be possible, regardless of the identity of the driver oncogene involved.


Asunto(s)
Leucemia Mieloide Aguda/etiología , Oncogenes , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Perfilación de la Expresión Génica , Proteínas de Homeodominio/fisiología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Fusión Oncogénica/fisiología , Proteína 1 Compañera de Translocación de RUNX1
10.
Cancer Cell ; 17(6): 584-96, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20541703

RESUMEN

We report a Jak2V617F knockin mouse myeloproliferative neoplasm (MPN) model resembling human polycythemia vera (PV). The MPN is serially transplantable and we demonstrate that the hematopoietic stem cell (HSC) compartment has the unique capacity for disease initiation but does not have a significant selective competitive advantage over wild-type HSCs. In contrast, myeloid progenitor populations are expanded and skewed toward the erythroid lineage, but cannot transplant the disease. Treatment with a JAK2 kinase inhibitor ameliorated the MPN phenotype, but did not eliminate the disease-initiating population. These findings provide insights into the consequences of JAK2 activation on HSC differentiation and function and have the potential to inform therapeutic approaches to JAK2V617F-positive MPN.


Asunto(s)
Sustitución de Aminoácidos , Células Madre Hematopoyéticas/patología , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Animales , Antígenos CD/metabolismo , Médula Ósea/patología , Células de la Médula Ósea/efectos de los fármacos , Trasplante de Médula Ósea , Recuento de Células , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Eritropoyetina/farmacología , Expresión Génica/genética , Perfilación de la Expresión Génica , Hematócrito , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Heterocigoto , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Células Progenitoras de Megacariocitos/metabolismo , Células Progenitoras de Megacariocitos/patología , Células Progenitoras de Megacariocitos y Eritrocitos/efectos de los fármacos , Células Progenitoras de Megacariocitos y Eritrocitos/metabolismo , Células Progenitoras de Megacariocitos y Eritrocitos/patología , Células Progenitoras de Megacariocitos y Eritrocitos/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Trastornos Mieloproliferativos/tratamiento farmacológico , Policitemia Vera/genética , Policitemia Vera/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , Bazo/efectos de los fármacos , Bazo/patología , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Análisis de Supervivencia
11.
Nat Med ; 16(8): 903-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20616797

RESUMEN

RNA-binding proteins of the Musashi (Msi) family are expressed in stem cell compartments and in aggressive tumors, but they have not yet been widely explored in the blood. Here we demonstrate that Msi2 is the predominant form expressed in hematopoietic stem cells (HSCs), and its knockdown leads to reduced engraftment and depletion of HSCs in vivo. Overexpression of human MSI2 in a mouse model increases HSC cell cycle progression and cooperates with the chronic myeloid leukemia-associated BCR-ABL1 oncoprotein to induce an aggressive leukemia. MSI2 is overexpressed in human myeloid leukemia cell lines, and its depletion leads to decreased proliferation and increased apoptosis. Expression levels in human myeloid leukemia directly correlate with decreased survival in patients with the disease, thereby defining MSI2 expression as a new prognostic marker and as a new target for therapy in acute myeloid leukemia (AML).


Asunto(s)
Transformación Celular Neoplásica/genética , Hematopoyesis/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de Unión al ARN/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Células Cultivadas , Progresión de la Enfermedad , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/mortalidad , Ratones , Ratones Transgénicos , Modelos Biológicos , Invasividad Neoplásica , Pronóstico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba/genética
12.
Cell Stem Cell ; 4(6): 559-67, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19497284

RESUMEN

We report the unexpected finding that loss of Hh signaling through conditional deletion of Smoothened (Smo) in the adult hematopoietic compartment has no apparent effect on adult hematopoiesis, including peripheral blood count, number or cell-cycle status of stem or progenitor cells, hematopoietic colony-forming potential, long-term repopulating activity in competitive repopulation assays, or stress response to serial 5-fluorouracil treatment. Furthermore, pharmacologic inhibition of Hh signaling with a potent and selective small molecule antagonist has no substantive effect on hematopoiesis in the mouse. In addition, Hh signaling is not required for the development of MLL-AF9-mediated acute myeloid leukemia (AML). Taken together, these data demonstrate that Hh signaling is dispensable for normal hematopoietic development and hematopoietic stem cell function, indicating that targeting of Hh signaling in solid tumors is not likely to result in hematopoietic toxicity. Furthermore, the Hh pathway may not be a compelling target in certain hematopoietic malignancies.


Asunto(s)
Proteínas Hedgehog/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Transducción de Señal/fisiología , Células Madre Adultas/fisiología , Animales , Leucemia Mieloide Aguda/etiología , Ratones , Neoplasias Experimentales/etiología , Receptores Acoplados a Proteínas G/deficiencia , Receptor Smoothened
13.
Cancer Cell ; 13(4): 311-20, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18394554

RESUMEN

We report that TG101348, a selective small-molecule inhibitor of JAK2 with an in vitro IC50 of approximately 3 nM, shows therapeutic efficacy in a murine model of myeloproliferative disease induced by the JAK2V617F mutation. In treated animals, there was a statistically significant reduction in hematocrit and leukocyte count, a dose-dependent reduction/elimination of extramedullary hematopoiesis, and, at least in some instances, evidence for attenuation of myelofibrosis. There were no apparent toxicities and no effect on T cell number. In vivo responses were correlated with surrogate endpoints, including reduction/elimination of JAK2V617F disease burden assessed by quantitative genomic PCR, suppression of endogenous erythroid colony formation, and in vivo inhibition of JAK-STAT signal transduction as assessed by flow cytometric measurement of phosphorylated Stat5.


Asunto(s)
Sustitución de Aminoácidos , Modelos Animales de Enfermedad , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Policitemia Vera/tratamiento farmacológico , Policitemia Vera/enzimología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirrolidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Trasplante de Médula Ósea , Línea Celular Tumoral , Ensayo de Unidades Formadoras de Colonias , Determinación de Punto Final , Citometría de Flujo , Sistema Hematopoyético/citología , Sistema Hematopoyético/efectos de los fármacos , Humanos , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenilalanina/genética , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Pirrolidinas/farmacocinética , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacocinética , Tasa de Supervivencia , Resultado del Tratamiento , Valina/genética
14.
Blood ; 107(11): 4274-81, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16478879

RESUMEN

An acquired somatic mutation, Jak2V617F, was recently discovered in most patients with polycythemia vera (PV), chronic idiopathic myelofibrosis (CIMF), and essential thrombocythemia (ET). To investigate the role of this mutation in vivo, we transplanted bone marrow (BM) transduced with a retrovirus expressing either Jak2 wild-type (wt) or Jak2V617F into lethally irradiated syngeneic recipient mice. Expression of Jak2V617F, but not Jak2wt, resulted in clinicopathologic features that closely resembled PV in humans. These included striking elevation in hemoglobin level/hematocrit, leukocytosis, megakaryocyte hyperplasia, extramedullary hematopoiesis resulting in splenomegaly, and reticulin fibrosis in the bone marrow. Histopathologic and flow cytometric analyses showed an increase in maturing myeloid lineage progenitors, although megakaryocytes showed decreased polyploidization and staining for acetylcholinesterase. In vitro analysis of primary cells showed constitutive activation of Stat5 and cytokine-independent growth of erythroid colony-forming unit (CFU-E) and erythropoietin hypersensitivity, and Southern blot analysis for retroviral integration indicated that the disease was oligoclonal. Furthermore, we observed strain-specific differences in phenotype, with Balb/c mice demonstrating markedly elevated leukocyte counts, splenomegaly, and reticulin fibrosis compared with C57Bl/6 mice. We conclude that Jak2V617F expression in bone marrow progenitors results in a PV-like syndrome with myelofibrosis and that there are strain-specific modifiers that may in part explain phenotypic pleiotropy of Jak2V617F-associated myeloproliferative disease in humans.


Asunto(s)
Trasplante de Médula Ósea/métodos , Policitemia Vera/etiología , Mielofibrosis Primaria/etiología , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Sustitución de Aminoácidos , Animales , Linaje de la Célula , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/citología , Janus Quinasa 2 , Ratones , Ratones Endogámicos , Policitemia Vera/genética , Policitemia Vera/patología , Especificidad de la Especie , Transducción Genética , Trasplante Isogénico
15.
Blood ; 108(4): 1339-45, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16627759

RESUMEN

Acquired mutations in the FLT3 receptor tyrosine kinase are common in acute myeloid leukemia and result in constitutive activation. The most frequent mechanism of activation is disruption of the juxtamembrane autoregulatory domain by internal tandem duplications (ITDs). FLT3-ITDs confer factor-independent growth to hematopoietic cells and induce a myeloproliferative syndrome in murine bone marrow transplant models. We and others have observed that FLT3-ITD activates STAT5 and its downstream effectors, whereas ligand-stimulated wild-type FLT3 (FLT3WT) does not. In vitro mapping of tyrosine phosphorylation sites in FLT3-ITD identified 2 candidate STAT5 docking sites within the juxtamembrane domain that are disrupted by the ITD. Tyrosine to phenylalanine substitution of residues 589 and 591 in the context of the FLT3-ITD did not affect tyrosine kinase activity, but abrogated STAT5 activation. Furthermore, FLT3-ITD-Y589/591F was incapable of inducing a myeloproliferative phenotype when transduced into primary murine bone marrow cells, whereas FLT3-ITD induced myeloproliferative disease with a median latency of 50 days. Thus, the conformational change in the FLT3 juxtamembrane domain induced by the ITD activates the kinase through dysregulation of autoinhibition and results in qualitative differences in signal transduction through STAT5 that are essential for the transforming potential of FLT3-ITD in vivo.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Leucemia Mieloide Aguda/metabolismo , Mutación , Trastornos Mieloproliferativos/metabolismo , Factor de Transcripción STAT5/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Activación Enzimática/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Estructura Terciaria de Proteína/genética , Factor de Transcripción STAT5/genética , Transducción de Señal/genética , Tirosina/genética , Tirosina/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
16.
Biol Reprod ; 73(4): 695-702, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15958731

RESUMEN

Early placental insulin-like protein (INSL4 or EPIL) is a member of the insulin superfamily of hormones, which is highly expressed in the placenta. We have confirmed this at term and shown it to be expressed by the maternal decidua. Although an abundance of locally acting growth factors are produced within the uterus during pregnancy, we hypothesized that INSL4 plays an important role in fetal and placental growth. We have demonstrated with cell lines and primary cells that it has a growth-inhibitory effect by causing apoptosis and loss of cell viability. We used primary amniotic epithelial cells for flow cytometry to show that INSL4 caused apoptosis, which was dose-related and significant (P < 0.05) at 50 ng/ml. This was confirmed by measurement of the nuclear matrix protein in the media. In comparison, relaxin treatment (up to 200 ng/ml) had no effect on apoptosis. The addition of INSL4 (3-30 ng/ml) also caused a loss of cell viability, although it had no effect on the numbers of cells at different phases of the cell cycle. Placental apoptosis is an important process in both normal placental development and in fetal growth restriction. Therefore, an in vivo clinical correlate was sought in fraternal twins exhibiting discordant growth. Expression of the INSL4 gene was doubled in the placenta of the growth-restricted twin compared to the normally grown sibling, suggesting that it may be linked to a higher level of apoptosis and loss of cell viability and, therefore, that it may contribute to fetal growth restriction.


Asunto(s)
Membrana Celular/metabolismo , Membranas Extraembrionarias/crecimiento & desarrollo , Membranas Extraembrionarias/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Placenta/metabolismo , Amnios/citología , Amnios/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Cultivadas , Femenino , Retardo del Crecimiento Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Placenta/citología , Embarazo , Valores de Referencia , Relaxina/farmacología , Estaurosporina/farmacología , Gemelos/genética
17.
Blood ; 106(9): 3206-13, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16030188

RESUMEN

AMN107 is a small molecule tyrosine kinase inhibitor developed, in the first instance, as a potent inhibitor of breakpoint cluster region-abelson (BCR-ABL). We tested its effectiveness against fusion tyrosine kinases TEL-platelet-derived growth factor receptorbeta (TEL-PDGFRbeta) and FIP1-like-1 (FIP1L1)-PDGFRalpha, which cause chronic myelomonocytic leukemia and hypereosinophilic syndrome, respectively. In vitro, AMN107 inhibited proliferation of Ba/F3 cells transformed by both TEL-PDGFRbeta and FIP1L1-PDGFRalpha with IC50 (inhibitory concentration 50%) values less than 25 nM and inhibited phosphorylation of the fusion kinases and their downstream signaling targets. The imatinib mesylate-resistant mutant TEL-PDGFRbeta T681I was sensitive to AMN107, whereas the analogous mutation in FIP1L1-PDGFRalpha, T674I, was resistant. In an in vivo bone marrow transplantation assay, AMN107 effectively treated myeloproliferative disease induced by TEL-PDGFRbeta and FIP1L1-PDGFRalpha, significantly increasing survival and disease latency and reducing disease severity as assessed by histopathology and flow cytometry. In summary, AMN107 can inhibit myeloid proliferation driven by TEL-PDGFRbeta and FIP1L1-PDGFRalpha and may be a useful drug for treatment of patients with myeloproliferative disease who harbor these kinase fusions.


Asunto(s)
Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/antagonistas & inhibidores , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Animales , Trasplante de Médula Ósea , Línea Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Mutación/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Proteínas de Fusión Oncogénica/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Tasa de Supervivencia , Factores de Escisión y Poliadenilación de ARNm/genética
18.
J Immunol ; 173(6): 4147-53, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15356165

RESUMEN

NO synthase 2 (NOS2) plays an important role in endotoxemia through overproduction of NO. Distamycin A (Dist A) belongs to a class of drugs termed minor-groove DNA binders, which can inhibit transcription factor binding to AT-rich regions of DNA. We and others have previously shown that AT-rich regions of DNA surrounding transcription factor binding sites in the NOS2 promoter are critical for NOS2 induction by inflammatory stimuli in vitro. Therefore, we hypothesized that Dist A would attenuate NOS2 up-regulation in vivo during endotoxemia and improve animal survival. C57BL/6 wild-type (WT) mice treated with Dist A and LPS (endotoxin) showed significantly improved survival compared with animals treated with LPS alone. In contrast, LPS-treated C57BL/6 NOS2-deficient (NOS2-/-) mice did not benefit from the protective effect of Dist A on mortality from endotoxemia. Treatment with Dist A resulted in protection from hypotension in LPS-treated WT mice, but not in NOS2-/- mice. Furthermore, LPS-induced NOS2 expression was attenuated in vivo (WT murine tissues) and in vitro (primary peritoneal and RAW 264.7 murine macrophages) with addition of Dist A. Dist A selectively decreased IFN regulatory factor-1 DNA binding in the enhancer region of the NOS2 promoter, and this IFN regulatory factor-1 site is critical for the effect of Dist A in attenuating LPS induction of NOS2. Our data point to a novel approach in modulating NOS2 expression in vivo during endotoxemia and suggest the potential for alternative treatment approaches for critical illness.


Asunto(s)
Distamicinas/farmacología , Endotoxemia/mortalidad , Endotoxemia/prevención & control , Inhibidores Enzimáticos/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/biosíntesis , Animales , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Endotoxemia/enzimología , Endotoxemia/genética , Inducción Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Hipotensión/enzimología , Hipotensión/genética , Hipotensión/prevención & control , Factor 1 Regulador del Interferón , Interferón gamma/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa/deficiencia , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Unión Proteica/genética
19.
Proc Natl Acad Sci U S A ; 101(40): 14479-84, 2004 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-15448205

RESUMEN

Human stem cell leukemia-lymphoma syndrome usually presents itself as a myeloproliferative disorder (MPD) that evolves to acute myeloid leukemia and/or lymphoma. The syndrome associated with t(8;13)(p11;q12) results in expression of the ZNF198-fibroblast growth factor receptor (FGFR) 1 fusion tyrosine kinase. Current empirically derived cytotoxic chemotherapy is inadequate for treatment of this disease. We hypothesized that small-molecule inhibitors of the ZNF198-FGFR1 fusion would have therapeutic efficacy. We characterized the transforming activity of ZNF198-FGFR1 in hematopoietic cells in vitro and in vivo. Expression of ZNF198-FGFR1 in primary murine hematopoietic cells caused a myeloproliferative syndrome in mice that recapitulated the human MPD phenotype. Transformation in these assays, and activation of the downstream effector molecules PLC-gamma, STAT5, and phosphatidylinositol 3-kinase/AKT, required the proline-rich domains, but not the ZNF domains, of ZNF198. A small-molecule tyrosine kinase inhibitor, PKC412 (N-benzoyl-staurosporine) effectively inhibited ZNF198-FGFR1 tyrosine kinase activity and activation of downstream effector pathways, and inhibited proliferation of ZNF198-FGFR1 transformed Ba/F3 cells. Furthermore, treatment with PKC412 resulted in statistically significant prolongation of survival in the murine model of ZNF198-FGFR1-induced MPD. Based in part on these data, PKC412 was administered to a patient with t(8;13)(p11;q12) and was efficacious in treatment of progressive myeloproliferative disorder with organomegaly. Therefore, PKC412 may be a useful therapy for treatment of human stem cell leukemia-lymphoma syndrome.


Asunto(s)
Trastornos Mieloproliferativos/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Animales , Línea Celular , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 8 , Modelos Animales de Enfermedad , Femenino , Variación Genética , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/química , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transformación Genética , Translocación Genética , Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA