Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 23(7): 949-960, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38507740

RESUMEN

The activated B cell (ABC) subset of diffuse large B-cell lymphoma (DLBCL) is characterized by chronic B-cell receptor signaling and associated with poor outcomes when treated with standard therapy. In ABC-DLBCL, MALT1 is a core enzyme that is constitutively activated by stimulation of the B-cell receptor or gain-of-function mutations in upstream components of the signaling pathway, making it an attractive therapeutic target. We discovered a novel small-molecule inhibitor, ABBV-MALT1, that potently shuts down B-cell signaling selectively in ABC-DLBCL preclinical models leading to potent cell growth and xenograft inhibition. We also identified a rational combination partner for ABBV-MALT1 in the BCL2 inhibitor, venetoclax, which when combined significantly synergizes to elicit deep and durable responses in preclinical models. This work highlights the potential of ABBV-MALT1 monotherapy and combination with venetoclax as effective treatment options for patients with ABC-DLBCL.


Asunto(s)
Sinergismo Farmacológico , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas Proto-Oncogénicas c-bcl-2 , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/antagonistas & inhibidores , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Humanos , Animales , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Línea Celular Tumoral , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Proliferación Celular/efectos de los fármacos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad
2.
Blood Adv ; 7(18): 5294-5303, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37315225

RESUMEN

We evaluated immune cell subsets in patients with chronic lymphocytic leukemia (CLL) who received first-line therapy with 3 cycles of ibrutinib then 13 cycles of ibrutinib plus venetoclax in the minimal residual disease (MRD) cohort of the CAPTIVATE study (NCT02910583). Patients with Confirmed undetectable MRD (uMRD) were randomly assigned to placebo or ibrutinib groups; patients without Confirmed uMRD were randomly assigned to ibrutinib or ibrutinib plus venetoclax groups. We compared immune cell subsets in samples collected at 7 time points with age-matched healthy donors. CLL cells decreased within 3 cycles after venetoclax initiation; from cycle 16 onward, levels were similar to healthy donor levels (HDL; ≤0.8 cells per µL) in patients with Confirmed uMRD and slightly above HDL in patients without Confirmed uMRD. By 4 months after cycle 16, normal B cells had recovered to HDL in patients randomly assigned to placebo. Regardless of randomized treatment, abnormal counts of T cells, classical monocytes, and conventional dendritic cells recovered to HDL within 6 months (median change from baseline -49%, +101%, and +91%, respectively); plasmacytoid dendritic cells recovered by cycle 20 (+598%). Infections generally decreased over time regardless of randomized treatment and were numerically lowest in patients randomly assigned to placebo within 12 months after cycle 16. Sustained elimination of CLL cells and recovery of normal B cells were confirmed in samples from patients treated with fixed-duration ibrutinib plus venetoclax in the GLOW study (NCT03462719). These results demonstrate promising evidence of restoration of normal blood immune composition with ibrutinib plus venetoclax.


Asunto(s)
Reconstitución Inmune , Leucemia Linfocítica Crónica de Células B , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Piperidinas/uso terapéutico
3.
Am J Hematol ; 98(3): 449-463, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36594167

RESUMEN

The treatment of patients with relapsed or refractory lymphoid neoplasms represents a significant clinical challenge. Here, we identify the pro-survival BCL-2 protein family member MCL-1 as a resistance factor for the BCL-2 inhibitor venetoclax in non-Hodgkin lymphoma (NHL) cell lines and primary NHL samples. Mechanistically, we show that the antibody-drug conjugate polatuzumab vedotin promotes MCL-1 degradation via the ubiquitin/proteasome system. This targeted MCL-1 antagonism, when combined with venetoclax and the anti-CD20 antibodies obinutuzumab or rituximab, results in tumor regressions in preclinical NHL models, which are sustained even off-treatment. In a Phase Ib clinical trial (NCT02611323) of heavily pre-treated patients with relapsed or refractory NHL, 25/33 (76%) patients with follicular lymphoma and 5/17 (29%) patients with diffuse large B-cell lymphoma achieved complete or partial responses with an acceptable safety profile when treated with the recommended Phase II dose of polatuzumab vedotin in combination with venetoclax and an anti-CD20 antibody.


Asunto(s)
Inmunoconjugados , Linfoma no Hodgkin , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/patología , Rituximab/uso terapéutico , Inmunoconjugados/uso terapéutico
4.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36073548

RESUMEN

Heterozygous mutations in FLT3ITD, TET2, and DNMT3A are associated with hematologic malignancies in humans. In patients, cooccurrence of mutations in FLT3ITD combined with TET2 (TF) or FLT3ITD combined with DNMT3A (DF) are frequent. However, in some rare complex acute myeloid leukemia (AML), all 3 mutations cooccur - i.e., FLT3ITD, TET2, and DNMT3A (TFD). Whether the presence of these mutations in combination result in quantitative or qualitative differences in disease manifestation has not been investigated. We generated mice expressing heterozygous Flt3ITD and concomitant for either heterozygous loss of Tet2 (TF) or Dnmt3a (DF) or both (TFD). TF and DF mice did not induce disease early on, in spite of similar changes in gene expression; during the same time frame, an aggressive form of transplantable leukemia was observed in TFD mice, which was mostly associated with quantitative but not qualitative differences in gene expression relative to TF or DF mice. The gene expression signature of TFD mice showed remarkable similarity to the human TFD gene signature at the single-cell RNA level. Importantly, TFD-driven AML responded to a combination of drugs that target Flt3ITD, inflammation, and methylation in a mouse model, as well as in a PDX model of AML bearing 3 mutations.


Asunto(s)
ADN Metiltransferasa 3A , Proteínas de Unión al ADN , Dioxigenasas , Leucemia Mieloide Aguda , Tirosina Quinasa 3 Similar a fms , Animales , ADN Metiltransferasa 3A/genética , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Proteínas Proto-Oncogénicas/genética , Tirosina Quinasa 3 Similar a fms/genética
5.
Stem Cell Reports ; 15(1): 95-109, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32502465

RESUMEN

Studies of patients with acute myeloid leukemia (AML) have led to the identification of mutations that affect different cellular pathways. Some of these have been classified as preleukemic, and a stepwise evolution program whereby cells acquire additional mutations has been proposed in the development of AML. How the timing of acquisition of these mutations and their impact on transformation and the bone marrow (BM) microenvironment occurs has only recently begun to be investigated. We show that constitutive and early loss of the epigenetic regulator, TET2, when combined with constitutive activation of FLT3, results in transformation of chronic myelomonocytic leukemia-like or myeloproliferative neoplasm-like phenotype to AML, which is more pronounced in double-mutant mice relative to mice carrying mutations in single genes. Furthermore, we show that in preleukemic and leukemic mice there are alterations in the BM niche and secreted cytokines, which creates a permissive environment for the growth of mutation-bearing cells relative to normal cells.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación/genética , Animales , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Heterocigoto , Homocigoto , Humanos , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas/metabolismo , Índice de Severidad de la Enfermedad , Microambiente Tumoral , Tirosina Quinasa 3 Similar a fms/metabolismo
6.
Cell Stem Cell ; 23(6): 833-849.e5, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30526882

RESUMEN

Inflammation is a risk factor for cancer development. Individuals with preleukemic TET2 mutations manifest clonal hematopoiesis and are at a higher risk of developing leukemia. How inflammatory signals influence the survival of preleukemic hematopoietic stem and progenitor cells (HSPCs) is unclear. We show a rapid increase in the frequency and absolute number of Tet2-KO mature myeloid cells and HSPCs in response to inflammatory stress, which results in enhanced production of inflammatory cytokines, including interleukin-6 (IL-6), and resistance to apoptosis. IL-6 induces hyperactivation of the Shp2-Stat3 signaling axis, resulting in increased expression of a novel anti-apoptotic long non-coding RNA (lncRNAs), Morrbid, in Tet2-KO myeloid cells and HSPCs. Expression of activated Shp2 in HSPCs phenocopies Tet2 loss with regard to hyperactivation of Stat3 and Morrbid. In vivo, pharmacologic inhibition of Shp2 or Stat3 or genetic loss of Morrbid in Tet2 mutant mice rescues inflammatory-stress-induced abnormalities in HSPCs and mature myeloid cells, including clonal hematopoiesis.


Asunto(s)
Benzoquinonas/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Inflamación/tratamiento farmacológico , Células Mieloides/efectos de los fármacos , Piperidinas/farmacología , Propionatos/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Inflamación/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Células Mieloides/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
7.
JCI Insight ; 3(4)2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29467326

RESUMEN

Mutations in KIT and TET2 are associated with myeloid malignancies. We show that loss of TET2-induced PI3K activation and -increased proliferation is rescued by targeting the p110α/δ subunits of PI3K. RNA-Seq revealed a hyperactive c-Myc signature in Tet2-/- cells, which is normalized by inhibiting PI3K signaling. Loss of TET2 impairs the maturation of myeloid lineage-derived mast cells by dysregulating the expression of Mitf and Cebpa, which is restored by low-dose ascorbic acid and 5-azacytidine. Utilizing a mouse model in which the loss of TET2 precedes the expression of oncogenic Kit, similar to the human disease, results in the development of a non-mast cell lineage neoplasm (AHNMD), which is responsive to PI3K inhibition. Thus, therapeutic approaches involving hypomethylating agents, ascorbic acid, and isoform-specific PI3K inhibitors are likely to be useful for treating patients with TET2 and KIT mutations.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica/genética , Mastocitos/patología , Mastocitosis/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Dioxigenasas , Modelos Animales de Enfermedad , Mutación con Ganancia de Función , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Humanos , Mastocitosis/tratamiento farmacológico , Mastocitosis/patología , Ratones , Ratones Noqueados , Células Mieloides/efectos de los fármacos , Células Mieloides/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-kit/genética
8.
J Clin Invest ; 126(7): 2621-5, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27294524

RESUMEN

Hyperactivation of the mTOR pathway impairs hematopoietic stem cell (HSC) functions and promotes leukemogenesis. mTORC1 and mTORC2 differentially control normal and leukemic stem cell functions. mTORC1 regulates p70 ribosomal protein S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E-binding (eIF4E-binding) protein 1 (4E-BP1), and mTORC2 modulates AKT activation. Given the extensive crosstalk that occurs between mTORC1 and mTORC2 signaling pathways, we assessed the role of the mTORC1 substrate S6K1 in the regulation of both normal HSC functions and in leukemogenesis driven by the mixed lineage leukemia (MLL) fusion oncogene MLL-AF9. We demonstrated that S6K1 deficiency impairs self-renewal of murine HSCs by reducing p21 expression. Loss of S6K1 also improved survival in mice transplanted with MLL-AF9-positive leukemic stem cells by modulating AKT and 4E-BP1 phosphorylation. Taken together, these results suggest that S6K1 acts through multiple targets of the mTOR pathway to promote self-renewal and leukemia progression. Given the recent interest in S6K1 as a potential therapeutic target in cancer, our results further support targeting this molecule as a potential strategy for treatment of myeloid malignancies.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia/sangre , Complejos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Progresión de la Enfermedad , Factores Eucarióticos de Iniciación , Humanos , Leucemia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Células Madre/metabolismo
9.
Oncotarget ; 7(13): 16936-47, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26943578

RESUMEN

Understanding mast cell development is essential due to their critical role in regulating immunity and autoimmune diseases. Here, we show how Rho kinases (ROCK) regulate mast cell development and can function as therapeutic targets for treating allergic diseases. Rock1 deficiency results in delayed maturation of bone marrow derived mast cells (BMMCs) in response to IL-3 stimulation and reduced growth in response to stem cell factor (SCF) stimulation. Further, integrin-mediated adhesion and migration, and IgE-mediated degranulation are all impaired in Rock1-deficient BMMCs. To understand the mechanism behind altered mast cell development in Rock1-/- BMMCs, we analyzed the activation of ROCK and its downstream targets including LIM kinase (LIMK). We observed reduced activation of ROCK, LIMK, AKT and ERK1/2 in Rock1-deficient BMMCs in response to SCF stimulation. Further, loss of either Limk1 or Limk2 also demonstrated altered BMMC maturation and growth; combined deletion of both Limk1 and Limk2 resulted in further reduction in BMMC maturation and growth. In passive cutaneous anaphylaxis model, deficiency of Rock1 or treatment with ROCK inhibitor Fasudil protected mice against IgE-mediated challenge. Our results identify ROCK/LIMK pathway as a novel therapeutic target for treating allergic diseases involving mast cells.


Asunto(s)
Diferenciación Celular/inmunología , Quinasas Lim/metabolismo , Mastocitos/citología , Mastocitos/metabolismo , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/inmunología , Degranulación de la Célula/inmunología , Movimiento Celular/inmunología , Hipersensibilidad/inmunología , Ratones , Ratones Noqueados
10.
Cell Rep ; 9(4): 1333-48, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25456130

RESUMEN

Oncogenic mutations of FLT3 and KIT receptors are associated with poor survival in patients with acute myeloid leukemia (AML) and myeloproliferative neoplasms (MPNs), and currently available drugs are largely ineffective. Although Stat5 has been implicated in regulating several myeloid and lymphoid malignancies, how precisely Stat5 regulates leukemogenesis, including its nuclear translocation to induce gene transcription, is poorly understood. In leukemic cells, we show constitutive activation of focal adhesion kinase (FAK) whose inhibition represses leukemogenesis. Downstream of FAK, activation of Rac1 is regulated by RacGEF Tiam1, whose inhibition prolongs the survival of leukemic mice. Inhibition of the Rac1 effector PAK1 prolongs the survival of leukemic mice in part by inhibiting the nuclear translocation of Stat5. These results reveal a leukemic pathway involving FAK/Tiam1/Rac1/PAK1 and demonstrate an essential role for these signaling molecules in regulating the nuclear translocation of Stat5 in leukemogenesis.


Asunto(s)
Carcinogénesis/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Transcripción STAT5/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Benzotiazoles/farmacología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Mastocitosis Sistémica/patología , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Mutación/genética , Compuestos de Fenilurea/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Quinasas p21 Activadas/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo
11.
Curr Opin Hematol ; 21(4): 271-5, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24867289

RESUMEN

PURPOSE OF REVIEW: Rho kinases (ROCKs) are involved in regulating a variety of physiologic functions including cytoskeletal reorganization, migration, adhesion, survival and proliferation. They do so via activating several different downstream substrates such as myosin light chain phosphatase, LIM kinase and ezrin/radixin/moesin proteins. To date, most of the conclusions with regard to the function of ROCKs have involved the use of cell line models, pharmacologic inhibitors and dominant negative approaches. Importantly, the role of ROCK in hematopoiesis or leukemogenesis in the context of whole organism remains poorly understood. RECENT FINDINGS: Recent studies utilizing mice deficient in the expression of ROCK1 have begun to shed some light into the physiologic role(s) of ROCK in both normal and abnormal hematopoiesis. Findings, thus far, suggest that ROCK plays an essential role in regulating growth and survival in different hematopoietic lineages via distinct mechanisms, in part, by utilizing distinct downstream substrates including maintaining the activation of tumor-suppressor genes. SUMMARY: In blood cells, emerging data suggest that ROCK plays an essential role in negatively regulating inflammatory and erythropoietic stress and positively regulates the growth and survival of leukemic cells.


Asunto(s)
Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Hematopoyesis/fisiología , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Eritropoyesis , Humanos , Leucemia/genética , Leucemia/metabolismo , Células Mieloides/fisiología , Estrés Fisiológico
12.
J Clin Invest ; 123(10): 4449-63, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24091327

RESUMEN

An acquired somatic mutation at codon 816 in the KIT receptor tyrosine kinase is associated with poor prognosis in patients with systemic mastocytosis and acute myeloid leukemia (AML). Treatment of leukemic cells bearing this mutation with an allosteric inhibitor of p21-activated kinase (Pak) or its genetic inactivation results in growth repression due to enhanced apoptosis. Inhibition of the upstream effector Rac abrogates the oncogene-induced growth and activity of Pak. Although both Rac1 and Rac2 are constitutively activated via the guanine nucleotide exchange factor (GEF) Vav1, loss of Rac1 or Rac2 alone moderately corrected the growth of KIT-bearing leukemic cells, whereas the combined loss resulted in 75% growth repression. In vivo, the inhibition of Vav or Rac or Pak delayed the onset of myeloproliferative neoplasms (MPNs) and corrected the associated pathology in mice. To assess the role of Rac GEFs in oncogene-induced transformation, we used an inhibitor of Rac, EHop-016, which specifically targets Vav1 and found that EHop-016 was a potent inhibitor of human and murine leukemic cell growth. These studies identify Pak and Rac GTPases, including Vav1, as potential therapeutic targets in MPN and AML involving an oncogenic form of KIT.


Asunto(s)
Aminoquinolinas/farmacología , Antineoplásicos/farmacología , Carbazoles/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/farmacología , Quinasas p21 Activadas/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Activación Enzimática , Humanos , Mastocitosis/tratamiento farmacológico , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutación Missense , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/antagonistas & inhibidores , Proteínas de Unión al GTP rac/antagonistas & inhibidores
15.
Oncotarget ; 3(9): 909-10, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23006984

RESUMEN

If ROCKs are going to be targeted as anti-leukemic agents either as a monotherapy or in combination with existing drugs, a better characterization of the relative contribution of ROCK1 and ROCK2 in disease progression is essential.


Asunto(s)
Leucemia/tratamiento farmacológico , Leucemia/enzimología , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/enzimología , Quinasas Asociadas a rho/metabolismo , Humanos , Isoenzimas , Leucemia/genética , Terapia Molecular Dirigida/métodos , Trastornos Mieloproliferativos/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
16.
Blood ; 120(14): 2868-78, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-22889758

RESUMEN

Erythropoiesis is a dynamic, multistep process whereby hematopoietic stem cells differentiate toward a progressively committed erythroid lineage through intermediate progenitors. Although several downstream signaling molecules have been identified that regulate steady-state erythropoiesis, the major regulators under conditions of stress remain poorly defined. Rho kinases (ROCKs) belong to a family of serine/threonine kinases. Using gene-targeted ROCK1-deficient mice, we show that lack of ROCK1 in phenylhydrazine-induced oxidative stress model results in enhanced recovery from hemolytic anemia as well as enhanced splenic stress erythropoiesis compared with control mice. Deficiency of ROCK1 also results in enhanced survival, whereas wild-type mice die rapidly in response to stress. Enhanced survivability of ROCK1-deficient mice is associated with reduced level of reactive oxygen species. BM transplantation studies revealed that enhanced stress erythropoiesis in ROCK1-deficient mice is stem cell autonomous. We show that ROCK1 binds to p53 and regulates its stability and expression. In the absence of ROCK1, p53 phosphorylation and expression is significantly reduced. Our findings reveal that ROCK1 functions as a physiologic regulator of p53 under conditions of erythroid stress. These findings are expected to offer new perspectives on stress erythropoiesis and may provide a potential therapeutic target in human disease characterized by anemia.


Asunto(s)
Anemia Hemolítica/mortalidad , Anemia Hemolítica/prevención & control , Apoptosis , Eritropoyesis/fisiología , Estrés Oxidativo/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Quinasas Asociadas a rho/fisiología , Anemia Hemolítica/inducido químicamente , Animales , Antimetabolitos Antineoplásicos/toxicidad , Western Blotting , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/patología , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Eritropoyesis/efectos de los fármacos , Eritropoyetina/sangre , Femenino , Citometría de Flujo , Fluorouracilo/toxicidad , Inmunoprecipitación , Masculino , Ratones , Ratones Noqueados , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Fenilhidrazinas/toxicidad , Fosforilación , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/genética
17.
PLoS One ; 7(8): e43090, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22905207

RESUMEN

We hypothesized that analysis of single nucleotide polymorphism arrays (SNP-A) and new molecular defects may provide new insight in the pathogenesis of systemic mastocytosis (SM). SNP-A karyotyping was applied to identify recurrent areas of loss of heterozygosity and bidirectional sequencing was performed to evaluate the mutational status of TET2, DNMT3A, ASXL1, EZH2, IDH1/IDH2 and the CBL gene family. Overall survival (OS) was analyzed using the Kaplan-Meier method. We studied a total of 26 patients with SM. In 67% of SM patients, SNP-A karyotyping showed new chromosomal abnormalities including uniparental disomy of 4q and 2p spanning TET2/KIT and DNMT3A. Mutations in TET2, DNMT3A, ASXL1 and CBL were found in 23%, 12%, 12%, and 4% of SM patients, respectively. No mutations were observed in EZH2 and IDH1/IDH2. Significant differences in OS were observed for SM mutated patients grouped based on the presence of combined TET2/DNMT3A/ASXL1 mutations independent of KIT (P = 0.04) and sole TET2 mutations (P<0.001). In conclusion, TET2, DNMT3A and ASXL1 mutations are also present in mastocytosis and these mutations may affect prognosis, as demonstrated by worse OS in mutated patients.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Mastocitosis Sistémica/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Adolescente , Adulto , Anciano , ADN Metiltransferasa 3A , Análisis Mutacional de ADN , Cartilla de ADN/genética , Dioxigenasas , Femenino , Predisposición Genética a la Enfermedad , Humanos , Cariotipificación , Persona de Mediana Edad , Mutación , Pronóstico
18.
Blood ; 120(13): 2669-78, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22806893

RESUMEN

Intracellular mechanism(s) that contribute to promiscuous signaling via oncogenic KIT in systemic mastocytosis and acute myelogenous leukemia are poorly understood. We show that SHP2 phosphatase is essential for oncogenic KIT-induced growth and survival in vitro and myeloproliferative disease (MPD) in vivo. Genetic disruption of SHP2 or treatment of oncogene-bearing cells with a novel SHP2 inhibitor alone or in combination with the PI3K inhibitor corrects MPD by disrupting a protein complex involving p85α, SHP2, and Gab2. Importantly, a single tyrosine at position 719 in oncogenic KIT is sufficient to develop MPD by recruiting p85α, SHP2, and Gab2 complex to oncogenic KIT. Our results demonstrate that SHP2 phosphatase is a druggable target that cooperates with lipid kinases in inducing MPD.


Asunto(s)
Transformación Celular Neoplásica/patología , Proteína Adaptadora GRB2/fisiología , Mutación/genética , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/prevención & control , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Apoptosis , Western Blotting , Trasplante de Médula Ósea , Proliferación Celular , Transformación Celular Neoplásica/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunoprecipitación , Integrasas/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/mortalidad , Fosforilación/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia , Tirosina/metabolismo
19.
Mol Cell Biol ; 32(14): 2653-63, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22566685

RESUMEN

Mast cells require KIT receptor tyrosine kinase signaling for development and survival. Here, we report that SH2 domain-containing phosphatase 2 (SHP2) signaling downstream of KIT is essential for mast cell survival and homeostasis in mice. Using a novel mouse model with shp2 deletion within mature mast cells (MC-shp2 knockout [KO]), we find that SHP2 is required for the homeostasis of connective tissue mast cells. Consistently with the loss of skin mast cells, MC-shp2 KO mice fail to mount a passive late-phase cutaneous anaphylaxis response. To better define the phenotype of shp2-deficient mast cells, we used an inducible shp2 knockout approach in bone marrow-derived mast cells (BMMCs) or cultured peritoneal mast cells and found that SHP2 promotes mast cell survival. We show that SHP2 promotes KIT signaling to extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase and downregulation of the proapoptotic protein Bim in BMMCs. Also, SHP2-deficient BMMCs failed to repopulate mast cells in mast cell-deficient mice. Silencing of Bim partially rescued survival defects in shp2-deficient BMMCs, consistent with the importance of a KIT → SHP2 → Ras/ERK pathway in suppressing Bim and promoting mast cell survival. Thus, SHP2 is a key node in a mast cell survival pathway and a new potential therapeutic target in diseases involving mast cells.


Asunto(s)
Mastocitos/citología , Mastocitos/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Secuencia de Bases , Proteína 11 Similar a Bcl2 , Supervivencia Celular , Cartilla de ADN/genética , Silenciador del Gen , Homeostasis , Sistema de Señalización de MAP Quinasas , Mastocitos/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Anafilaxis Cutánea Pasiva/inmunología , Anafilaxis Cutánea Pasiva/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-kit/fisiología , ARN Interferente Pequeño/genética , Transducción de Señal , Piel/citología , Piel/enzimología
20.
J Immunol ; 188(8): 3839-50, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22430739

RESUMEN

Although SHIP is a well-established suppressor of IgE plus Ag-induced degranulation and cytokine production in bone marrow-derived mast cells (BMMCs), little is known about its role in connective tissue (CTMCs) or mucosal (MMCs) mast cells. In this study, we compared SHIP's role in the development as well as the IgE plus Ag and TLR-induced activation of CTMCs, MMCs, and BMMCs and found that SHIP delays the maturation of all three mast cell subsets and, surprisingly, that it is a positive regulator of IgE-induced BMMC survival. We also found that SHIP represses IgE plus Ag-induced degranulation of all three mast cell subsets and that TLR agonists do not trigger their degranulation, whether SHIP is present or not, nor do they enhance IgE plus Ag-induced degranulation. In terms of cytokine production, we found that in MMCs and BMMCs, which are poor producers of TLR-induced cytokines, SHIP is a potent negative regulator of IgE plus Ag-induced IL-6 and TNF-α production. Surprisingly, however, in splenic or peritoneal derived CTMCs, which are poor producers of IgE plus Ag-induced cytokines, SHIP is a potent positive regulator of TLR-induced cytokine production. Lastly, cell signaling and cytokine production studies with and without LY294002, wortmannin, and PI3Kα inhibitor-2, as well as with PI3K p85α(-/-) BMMCs and CTMCs, are consistent with SHIP positively regulating TLR-induced cytokine production via an adaptor-mediated pathway while negatively regulating IgE plus Ag-induced cytokine production by repressing the PI3K pathway.


Asunto(s)
Inmunoglobulina E/inmunología , Mastocitos/inmunología , Membrana Mucosa/inmunología , Monoéster Fosfórico Hidrolasas/inmunología , Animales , Antígenos/inmunología , Degranulación de la Célula/inmunología , Diferenciación Celular , Linaje de la Célula/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Regulación de la Expresión Génica , Inositol Polifosfato 5-Fosfatasas , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Mastocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/citología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...