Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Blood ; 115(7): 1406-15, 2010 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-20008787

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Divisão Celular/fisiologia , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Rim/citologia , Leucemia Mieloide Aguda/tratamento farmacológico , Linfoma de Células T/tratamento farmacológico , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Baço/citologia , Serina-Treonina Quinases TOR
2.
Blood ; 115(17): 3489-97, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20197553

RESUMO

Apc, a negative regulator of the canonical Wnt signaling pathway, is a bona-fide tumor suppressor whose loss of function results in intestinal polyposis. APC is located in a commonly deleted region on human chromosome 5q, associated with myelodysplastic syndrome (MDS), suggesting that haploinsufficiency of APC contributes to the MDS phenotype. Analysis of the hematopoietic system of mice with the Apc(min) allele that results in a premature stop codon and loss of function showed no abnormality in steady state hematopoiesis. Bone marrow derived from Apc(min) mice showed enhanced repopulation potential, indicating a cell intrinsic gain of function in the long-term hematopoietic stem cell (HSC) population. However, Apc(min) bone marrow was unable to repopulate secondary recipients because of loss of the quiescent HSC population. Apc(min) mice developed a MDS/myeloproliferative phenotype. Our data indicate that Wnt activation through haploinsufficiency of Apc causes insidious loss of HSC function that is only evident in serial transplantation strategies. These data provide a cautionary note for HSC-expansion strategies through Wnt pathway activation, provide evidence that cell extrinsic factors can contribute to the development of myeloid disease, and indicate that loss of function of APC may contribute to the phenotype observed in patients with MDS and del(5q).


Assuntos
Proteína da Polipose Adenomatosa do Colo , Medula Óssea/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Síndromes Mielodisplásicas/metabolismo , Alelos , Animais , Sequência de Bases , Medula Óssea/patologia , Cromossomos Humanos Par 5/genética , Cromossomos Humanos Par 5/metabolismo , Códon de Terminação/genética , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Mutantes , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Fenótipo , Deleção de Sequência , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
3.
J Clin Invest ; 124(4): 1794-809, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24569456

RESUMO

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.


Assuntos
Genes ras , Hematopoese/genética , Hematopoese/fisiologia , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Eritropoese/genética , Eritropoese/fisiologia , Xenoenxertos , Humanos , Leucemia Mieloide Aguda/patologia , Leucemia Mielomonocítica Juvenil/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Transdução de Sinais
4.
Cancer Discov ; 3(8): 922-35, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23689072

RESUMO

The causes for malignant progression of disseminated tumors and the reasons recurrence rates differ in women with different breast cancer subtypes are unknown. Here, we report novel mechanisms of tumor plasticity that are mandated by microenvironmental factors and show that recurrence rates are not strictly due to cell-intrinsic properties. Specifically, outgrowth of the same population of incipient tumors is accelerated in mice with triple-negative breast cancer (TNBC) relative to those with luminal breast cancer. Systemic signals provided by overt TNBCs cause the formation of a tumor-supportive microenvironment enriched for EGF and insulin-like growth factor-I (IGF-I) at distant indolent tumor sites. Bioavailability of EGF and IGF-I enhances the expression of transcription factors associated with pluripotency, proliferation, and epithelial-mesenchymal transition. Combinatorial therapy with EGF receptor and IGF-I receptor inhibitors prevents malignant progression. These results suggest that plasticity and recurrence rates can be dictated by host systemic factors and offer novel therapeutic potential for patients with TNBC.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Fator de Crescimento Epidérmico/genética , Transição Epitelial-Mesenquimal , Receptores ErbB/antagonistas & inibidores , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/genética , Camundongos , Camundongos Nus , Metástase Neoplásica , Recidiva Local de Neoplasia , Transplante de Neoplasias , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Células Estromais/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Microambiente Tumoral/fisiologia
5.
Cancer Cell ; 17(6): 584-96, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20541703

RESUMO

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.


Assuntos
Substituição de Aminoácidos , Células-Tronco Hematopoéticas/patologia , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Animais , Antígenos CD/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Contagem de Células , Diferenciação Celular/genética , Modelos Animais de Doenças , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Eritropoetina/farmacologia , Expressão Gênica/genética , Perfilação da Expressão Gênica , Hematócrito , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Heterozigoto , Humanos , Janus Quinase 2/antagonistas & inibidores , Células Progenitoras de Megacariócitos/metabolismo , Células Progenitoras de Megacariócitos/patologia , Células Progenitoras de Megacariócitos e Eritrócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Células Progenitoras de Megacariócitos e Eritrócitos/patologia , Células Progenitoras de Megacariócitos e Eritrócitos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Transtornos Mieloproliferativos/tratamento farmacológico , Policitemia Vera/genética , Policitemia Vera/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Pirrolidinas/farmacologia , Pirrolidinas/uso terapêutico , Baço/efeitos dos fármacos , Baço/patologia , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Análise de Sobrevida
6.
Nat Med ; 16(8): 903-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20616797

RESUMO

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).


Assuntos
Transformação Celular Neoplásica/genética , Hematopoese/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Proteínas de Ligação a RNA/fisiologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Células Cultivadas , Progressão da Doença , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/mortalidade , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Invasividade Neoplásica , Prognóstico , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA