Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
2.
Front Oncol ; 13: 1096499, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969004

RESUMEN

Patients with pancreatic ductal adenocarcinoma (PDAC) have a dismal 5-year survival rate of less than 10%, predominantly due to delayed diagnosis and a lack of effective treatment options. In the PDAC tumor microenvironment (TME), neutrophils are among the immune cell types that are most prevalent and are linked to a poor clinical prognosis. However, treatments that target tumor-associated neutrophils are limited despite recent developments in our understanding of neutrophil function in cancer. The feline sarcoma oncogene (FES) is a nonreceptor tyrosine kinase previously associated with leukemia and hematopoietic homeostasis. Here we describe a newly derived FES null mouse with no distinct phenotype and no defects in hematopoietic homeostasis including neutrophil viability. The immune cell composition and neutrophil population were analyzed with flow cytometry, colony-forming unit (CFU) assay, and a neutrophil viability assay, while the response to PDAC was examined with an in vivo cancer model. In an experimental metastasis model, the FES null model displayed a reduced PDAC hepatic metastatic burden and a reduction in neutrophils granulocytes. Accordingly, our results indicate FES as a potential target for PDAC TME modulation.

3.
Bull Cancer ; 110(3): 331-335, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36775700

RESUMEN

This article highlights the presentations from the 2021 scientific meeting of the Club Hematopoiesis and Oncogenesis. This annual meeting focuses on hematopoiesis and oncogenic mechanisms. Various topics were presented: expansion of hematopoietic stem cells with in vivo and ex vivo strategies, the role of the hematopoietic stem cell niches in aging and leukemic resistance, the crossroad between hematology and immunology, the importance of the metabolism in normal hematopoiesis and hematopoietic defects, solid tumors and oncogenesis, the noncoding genome, inflammation in monocyte differentiation and leukemia, and importantly, the recent advances in myeloid malignancies, lymphoid leukemia and lymphoma.


Asunto(s)
Leucemia , Linfoma , Humanos , Hematopoyesis/genética , Células Madre Hematopoyéticas , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología
4.
JCI Insight ; 7(7)2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35393954

RESUMEN

Mutation of the TET2 DNA-hydroxymethylase has been associated with a number of immune pathologies. The disparity in phenotype and clinical presentation among these pathologies leads to questions regarding the role of TET2 mutation in promoting disease evolution in different immune cell types. Here we show that, in primary mast cells, Tet2 expression is induced in response to chronic and acute activation signals. In TET2-deficient mast cells, chronic activation via the oncogenic KITD816V allele associated with mastocytosis, selects for a specific epigenetic signature characterized by hypermethylated DNA regions (HMR) at immune response genes. H3K27ac and transcription factor binding is consistent with priming or more open chromatin at both HMR and non-HMR in proximity to immune genes in these cells, and this signature coincides with increased pathological inflammation signals. HMR are also associated with a subset of immune genes that are direct targets of TET2 and repressed in TET2-deficient cells. Repression of these genes results in immune tolerance to acute stimulation that can be rescued with vitamin C treatment or reiterated with a Tet inhibitor. Overall, our data support a model where TET2 plays a direct role in preventing immune tolerance in chronically activated mast cells, supporting TET2 as a viable target to reprogram the innate immune response for innovative therapies.


Asunto(s)
Proteínas de Unión al ADN , Dioxigenasas , Tolerancia Inmunológica , Mastocitos , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Mastocitos/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
5.
PLoS One ; 16(12): e0260852, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855882

RESUMEN

Establishing a universally applicable protocol to assess the impact of BRCA1 variants of uncertain significance (VUS) expression is a problem which has yet to be resolved despite major progresses have been made. The numerous difficulties which must be overcome include the choices of cellular models and functional assays. We hypothesised that the use of induced pluripotent stem (iPS) cells might facilitate the standardisation of protocols for classification, and could better model the disease process. We generated eight iPS cell lines from patient samples expressing either BRCA1 pathogenic variants, non-pathogenic variants, or BRCA1 VUSs. The impact of these variants on DNA damage repair was examined using a ɣH2AX foci formation assay, a Homologous Repair (HR) reporter assay, and a chromosome abnormality assay. Finally, all lines were tested for their ability to differentiate into mammary lineages in vitro. While the results obtained from the two BRCA1 pathogenic variants were consistent with published data, some other variants exhibited differences. The most striking of these was the BRCA1 variant Y856H (classified as benign), which was unexpectedly found to present a faulty HR repair pathway, a finding linked to the presence of an additional variant in the ATM gene. Finally, all lines were able to differentiate first into mammospheres, and then into more advanced mammary lineages expressing luminal- or basal-specific markers. This study stresses that BRCA1 genetic analysis alone is insufficient to establish a reliable and functional classification for assessment of clinical risk, and that it cannot be performed without considering the other genetic aberrations which may be present in patients. The study also provides promising opportunities for elucidating the physiopathology and clinical evolution of breast cancer, by using iPS cells.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/patología , Daño del ADN , Reparación del ADN , Predisposición Genética a la Enfermedad , Células Madre Pluripotentes Inducidas/patología , Mutación , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/genética , Femenino , Pruebas Genéticas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
6.
Cancers (Basel) ; 13(11)2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-34206000

RESUMEN

Protein kinases (PK) make up around 2% of the human genome and their expression profile varies depending on the organ and tissue [...].

7.
Blood ; 138(17): 1590-1602, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33974006

RESUMEN

Systemic mastocytosis (SM) is a KIT-driven hematopoietic neoplasm characterized by the excessive accumulation of neoplastic mast cells (MCs) in various organs and, mainly, the bone marrow (BM). Multiple genetic and epigenetic mechanisms contribute to the onset and severity of SM. However, little is known to date about the metabolic underpinnings underlying SM aggressiveness, which has thus far impeded the development of strategies to leverage metabolic dependencies when existing KIT-targeted treatments fail. Here, we show that plasma metabolomic profiles were able to discriminate indolent from advanced forms of the disease. We identified N-acetyl-d-glucosamine (GlcNAc) as the most predictive metabolite of SM severity. High plasma levels of GlcNAc in patients with advanced SM correlated with the activation of the GlcNAc-fed hexosamine biosynthesis pathway in patients BM aspirates and purified BM MCs. At the functional level, GlcNAc enhanced human neoplastic MCs proliferation and promoted rapid health deterioration in a humanized mouse model of SM. In addition, in the presence of GlcNAc, immunoglobulin E-stimulated MCs triggered enhanced release of proinflammatory cytokines and a stronger acute response in a mouse model of passive cutaneous anaphylaxis. Mechanistically, elevated GlcNAc levels promoted the transcriptional accessibility of chromatin regions that contain genes encoding mediators of receptor tyrosine kinases cascades and inflammatory responses, thus leading to a more aggressive phenotype. Therefore, GlcNAc is an oncometabolite driver of SM aggressiveness. This study suggests the therapeutic potential for targeting metabolic pathways in MC-related diseases to manipulate MCs effector functions.


Asunto(s)
Acetilglucosamina/análisis , Ensamble y Desensamble de Cromatina , Mastocitos/patología , Mastocitosis Sistémica/patología , Acetilglucosamina/metabolismo , Adulto , Animales , Progresión de la Enfermedad , Humanos , Mastocitos/metabolismo , Mastocitosis Sistémica/genética , Mastocitosis Sistémica/metabolismo , Metaboloma , Ratones SCID , Estudios Prospectivos
8.
Cancers (Basel) ; 12(7)2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32708273

RESUMEN

Protein tyrosine kinases have been recognized as important actors of cell transformation and cancer progression, since their discovery as products of viral oncogenes. SRC-family kinases (SFKs) play crucial roles in normal hematopoiesis. Not surprisingly, they are hyperactivated and are essential for membrane receptor downstream signaling in hematological malignancies such as acute myeloid leukemia (AML) and mastocytosis. The precise roles of SFKs are difficult to delineate due to the number of substrates, the functional redundancy among members, and the use of tools that are not selective. Yet, a large num ber of studies have accumulated evidence to support that SFKs are rational therapeutic targets in AML and mastocytosis. These two pathologies are regulated by two related receptor tyrosine kinases, which are well known in the field of hematology: FLT3 and KIT. FLT3 is one of the most frequently mutated genes in AML, while KIT oncogenic mutations occur in 80-90% of mastocytosis. Studies on oncogenic FLT3 and KIT signaling have shed light on specific roles for members of the SFK family. This review highlights the central roles of SFKs in AML and mastocytosis, and their interconnection with FLT3 and KIT oncoproteins.

9.
Nat Commun ; 8(1): 1420, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29127277

RESUMEN

Masitinib, a highly selective protein kinase inhibitor, can sensitise gemcitabine-refractory cancer cell lines when used in combination with gemcitabine. Here we report a reverse proteomic approach that identifies the target responsible for this sensitisation: the deoxycytidine kinase (dCK). Masitinib, as well as other protein kinase inhibitors, such as imatinib, interact with dCK and provoke an unforeseen conformational-dependent activation of this nucleoside kinase, modulating phosphorylation of nucleoside analogue drugs. This phenomenon leads to an increase of prodrug phosphorylation of most of the chemotherapeutic drugs activated by this nucleoside kinase. The unforeseen dual activity of protein kinase inhibition/nucleoside kinase activation could be of great therapeutic benefit, through either reducing toxicity of therapeutic agents by maintaining effectiveness at lower doses or by counteracting drug resistance initiated via down modulation of dCK target.


Asunto(s)
Desoxicitidina Quinasa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Tiazoles/farmacología , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Cristalografía por Rayos X , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina Quinasa/química , Diseño de Fármacos , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/farmacología , Modelos Biológicos , Modelos Moleculares , Fosforilación , Piperidinas , Polifarmacología , Inhibidores de Proteínas Quinasas/química , Proteómica , Piridinas , Tiazoles/química , Gemcitabina
10.
J Clin Invest ; 127(6): 2310-2325, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28463229

RESUMEN

Identification and functional validation of oncogenic drivers are essential steps toward advancing cancer precision medicine. Here, we have presented a comprehensive analysis of the somatic genomic landscape of the widely used BRAFV600E- and NRASQ61K-driven mouse models of melanoma. By integrating the data with publically available genomic, epigenomic, and transcriptomic information from human clinical samples, we confirmed the importance of several genes and pathways previously implicated in human melanoma, including the tumor-suppressor genes phosphatase and tensin homolog (PTEN), cyclin dependent kinase inhibitor 2A (CDKN2A), LKB1, and others. Importantly, this approach also identified additional putative melanoma drivers with prognostic and therapeutic relevance. Surprisingly, one of these genes encodes the tyrosine kinase FES. Whereas FES is highly expressed in normal human melanocytes, FES expression is strongly decreased in over 30% of human melanomas. This downregulation correlates with poor overall survival. Correspondingly, engineered deletion of Fes accelerated tumor progression in a BRAFV600E-driven mouse model of melanoma. Together, these data implicate FES as a driver of melanoma progression and demonstrate the potential of cross-species oncogenomic approaches combined with mouse modeling to uncover impactful mutations and oncogenic driver alleles with clinical importance in the treatment of human cancer.


Asunto(s)
Melanoma/genética , Proteínas Proto-Oncogénicas c-fes/genética , Neoplasias Cutáneas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Variaciones en el Número de Copia de ADN , Genes Supresores de Tumor , Genómica , Humanos , Melanoma/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Oncogenes , Proteínas Proto-Oncogénicas c-fes/metabolismo , Neoplasias Cutáneas/metabolismo , Vía de Señalización Wnt
11.
PLoS One ; 11(7): e0160165, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467080

RESUMEN

The receptors tyrosine kinases (RTKs) for the colony stimulating factor-1, CSF-1R, and for the stem cell factor, SCFR or KIT, are important mediators of signal transduction. The abnormal function of these receptors, promoted by gain-of-function mutations, leads to their constitutive activation, associated with cancer or other proliferative diseases. A secondary effect of the mutations is the alteration of receptors' sensitivity to tyrosine kinase inhibitors, compromising effectiveness of these molecules in clinical treatment. In particular, the mutation V560G in KIT increases its sensitivity to Imatinib, while the D816V in KIT, and D802V in CSF-1R, triggers resistance to the drug. We analyzed the Imatinib binding affinity to the native and mutated KIT (mutations V560G, S628N and D816V) and CSF-1R (mutation D802V) by using molecular dynamics simulations and energy calculations of Imatinib•target complexes. Further, we evaluated the sensitivity of the studied KIT receptors to Imatinib by measuring the inhibition of KIT phosphorylation. Our study showed that (i) the binding free energy of Imatinib to the targets is highly correlated with their experimentally measured sensitivity; (ii) the electrostatic interactions are a decisive factor affecting the binding energy; (iii) the most deleterious impact to the Imatinib sensitivity is promoted by D802V (CSF-1R) and D816V (KIT) mutations; (iv) the role of the juxtamembrane region, JMR, in the imatinib binding is accessory. These findings contribute to a better description of the mutation-induced effects alternating the targets sensitivity to Imatinib.


Asunto(s)
Mutación , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor de Células Madre/metabolismo , Animales , Células COS , Chlorocebus aethiops , Enlace de Hidrógeno , Mesilato de Imatinib/química , Mesilato de Imatinib/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Receptor de Factor Estimulante de Colonias de Macrófagos/química , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Factor de Células Madre/química , Factor de Células Madre/genética
12.
Oncotarget ; 7(32): 51163-51173, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27323399

RESUMEN

CDK4/CDK6 and RB proteins drive the progression through the G1 phase of the cell cycle. In acute myeloid leukemia (AML), the activity of the CDK/Cyclin D complex is increased. The mechanism involved is unknown, as are the respective roles played by CDK4 or CDK6 in this process. Here, we report that AML cells carrying FLT3-ITD mutations are dependent on CDK6 for cell proliferation while CDK4 is not essential. We showed that FLT3-ITD signaling is responsible for CDK6 overexpression, through a pathway involving the SRC-family kinase HCK. Accordingly, FLT3-ITD failed to transform primary hematopoietic progenitor cells from Cdk6-/- mice. Our results demonstrate that CDK6 is the primary target of CDK4/CDK6 inhibitors in FLT3-ITD positive AML. Furthermore, we delineate an essential protein kinase pathway -FLT3/HCK/CDK6- in the context of AML with FLT3-ITD mutations.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/genética , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas c-hck/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Línea Celular Tumoral , Quinasa 6 Dependiente de la Ciclina/metabolismo , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas Proto-Oncogénicas c-hck/metabolismo , Transducción de Señal/genética , Secuencias Repetidas en Tándem , Tirosina Quinasa 3 Similar a fms/metabolismo
13.
JAMA Dermatol ; 150(12): 1345-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25317746

RESUMEN

IMPORTANCE The KIT receptor is mutated in approximately 15%of acral, mucosal, and chronic, sun-damaged melanomas. The status of KIT mutations is of interest because they usually are mutually exclusive with N-RAS and B-RAF mutations and because of the availability of KIT kinase inhibitors in the clinic. Some recurrent KIT mutations are well characterized; others are poorly described.OBSERVATIONS We describe a novel KIT mutation in a patient with metastatic melanoma. The mutation, located in exon 13, resulted in S628N substitution in the KIT receptor. Using all-atom molecular dynamics simulations, biochemical assays, and cell-based assays, we showed that the mutation is a bona fide gain-of-function oncogenic mutation. Furthermore,we evaluated the sensitivity of the mutant to imatinib and dasatinib.CONCLUSIONS AND RELEVANCE We report a novel KIT gain-of-function mutation with S628N substitution (exon 13) and show that it is sensitive to imatinib in vitro. Therefore, patients with this mutation may be eligible for KIT kinase inhibitor­based therapy. Further studies are needed to evaluate the clinical benefit of such therapy.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Pulmonares/genética , Melanoma/genética , Proteínas Proto-Oncogénicas c-kit/genética , Neoplasias Cutáneas/genética , Neoplasias de la Columna Vertebral/genética , Anciano de 80 o más Años , Animales , Benzamidas/farmacología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Dasatinib , Resultado Fatal , Femenino , Fibroblastos/citología , Humanos , Mesilato de Imatinib , Neoplasias Pulmonares/secundario , Melanoma/secundario , Mutación Missense , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Mutación Puntual , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/farmacología , Ratas , Neoplasias Cutáneas/patología , Neoplasias de la Columna Vertebral/secundario , Tiazoles/farmacología
14.
Haematologica ; 99(5): 830-5, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24389310

RESUMEN

Mastocytosis is a rare and chronic disease with phenotypes ranging from indolent to severe. Prognosis for this disease is variable and very few biomarkers to predict disease evolution or outcome are currently known. We have performed comprehensive screening in our large cohort of mastocytosis patients for mutations previously found in other myeloid diseases and that could serve as prognostic indicators. KIT, SRSF2-P95 and TET2 mutations were by far the most frequent, detected in 81%, 24% and 21% of patients, respectively. Where TET2 and SRSF2-P95 mutation both correlated with advanced disease phenotypes, SRSF2-P95 hotspot mutation was found almost exclusively in patients diagnosed with associated clonal hematologic non-mast cell disease. Statistically, TET2 and SRSF2-P95 mutations were highly associated, suggesting a mechanistic link between these two factors. Finally, analysis of both clonal and sorted cell populations from patients confirms the presence of these mutations in the mast cell component of the disease, suggests an ontological mutation hierarchy and provides evidence for the expansion of multiple clones. This highlights the prognostic potential of such approaches, if applied systematically, for delineating the roles of specific mutations in predisposing and/or driving distinct disease phenotypes.


Asunto(s)
Epigénesis Genética , Mastocitosis/genética , Mastocitosis/patología , Mutación , Proteínas Nucleares/genética , Ribonucleoproteínas/genética , Evolución Clonal/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Femenino , Humanos , Masculino , Mastocitos/metabolismo , Mastocitos/patología , Mastocitosis/mortalidad , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-kit/genética , Factores de Empalme Serina-Arginina
15.
Blood ; 120(24): 4846-9, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-23074272

RESUMEN

Although a role for oncogenic KIT in driving mast cell disease is clear, the mechanisms driving the multiple phenotypic and clinical manifestations of this disorder are not well elucidated. We now show, using a large cohort of mastocytosis patients, including an almost equal number of aggressive and nonaggressive cases of systemic mastocytosis, that in contrast to the oncogenic KITD816V, TET2 mutation statistically associates with aggressive forms of the disease. By infecting primary murine bone marrow-derived mast cells with KITD816V, we also observe a significant and competitive growth advantage for KITD816V in Tet2-nullizygous compared with wild-type cells. TET2-deficient cells display increased proliferation and can survive in the absence of cytokines. Taken together, these data demonstrate a oncogenic cooperation in mast cells and reveal TET2 mutation as a potential marker to diagnose and predict severe forms of mastocytosis.


Asunto(s)
Proteínas de Unión al ADN/genética , Mastocitos/metabolismo , Mastocitosis/genética , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas/genética , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Animales , Células de la Médula Ósea/metabolismo , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Células Cultivadas , Estudios de Cohortes , Dioxigenasas , Femenino , Humanos , Masculino , Mastocitos/patología , Mastocitosis/patología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Factores de Tiempo , Transfección
16.
J Biol Chem ; 286(8): 5956-66, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21135090

RESUMEN

Mutations in the c-kit gene occur in the vast majority of mastocytosis. In adult patients as well as in the cell line derived from mast cell neoplasms, the mutations occur almost exclusively at amino acid 816 within the kinase domain of KIT. Among the downstream effectors of KIT signaling, STAT3 and STAT5 have been shown to be critical for cell proliferation elicited by the KIT-Asp(816) mutant protein. However, little is known about the mechanisms of activation of STAT proteins. In this study, we identify and clarify the contribution of various STAT kinases in two widely used neoplastic mast cell lines, P815 and HMC-1. We show that STAT1, -3, and -5 proteins are activated downstream of the KIT-Asp(816) mutant. All three STAT proteins are located in the nucleus and are phosphorylated on serine residues. KIT-Asp(816) mutant can directly phosphorylate STATs on the activation-specific tyrosine residues in vitro. However, within cells, SRC family kinases and JAKs diversely contribute to tyrosine phosphorylation of STAT proteins downstream of the KIT mutant. Using a panel of inhibitors, we provide evidence for the implication or exclusion of serine/threonine kinases as responsible for serine phosphorylation of STAT1, -3, and -5 in the two cell lines. Finally, we show that only STAT5 is transcriptionally active in these cells. This suggests that the contribution of STAT1 and STAT3 downstream of KIT mutant is independent of their transcription factor function.


Asunto(s)
Proliferación Celular , Mastocitos/metabolismo , Mastocitosis/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Adulto , Animales , Células COS , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Chlorocebus aethiops , Humanos , Mastocitos/patología , Mastocitosis/genética , Mastocitosis/patología , Ratones , Mutación , Fosforilación/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/genética , Factores de Transcripción STAT/genética , Transcripción Genética/genética
17.
BMC Biochem ; 11: 48, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21122136

RESUMEN

BACKGROUND: RhoGDI proteins are important regulators of the small GTPase Rac, because they shuttle Rac from the cytoplasm to membranes and also protect Rac from activation, deactivation and degradation. How the binding and release of Rac from RhoGDI is regulated is not precisely understood. RESULTS: We report that the non-receptor tyrosine kinase Fer is able to phosphorylate RhoGDIα and form a direct protein complex with it. This interaction is mediated by the C-terminal end of RhoGDIα. Activation of Fer by reactive oxygen species caused increased phosphorylation of RhoGDIα and pervanadate treatment further augmented this. Tyrosine phosphorylation of RhoGDIα by Fer prevented subsequent binding of Rac to RhoGDIα, but once a RhoGDIα-Rac complex was formed, the Fer kinase was not able to cause Rac release through tyrosine phosphorylation of preformed RhoGDIα-Rac complexes. CONCLUSIONS: These results identify tyrosine phosphorylation of RhoGDIα by Fer as a mechanism to regulate binding of RhoGDIα to Rac.


Asunto(s)
Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Inhibidores de Disociación de Guanina Nucleótido/química , Humanos , Fosforilación , Unión Proteica , Ratas , Especificidad por Sustrato , Tirosina/metabolismo , Inhibidor alfa de Disociación del Nucleótido Guanina rho , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
18.
Blood ; 116(7): 1114-23, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20484085

RESUMEN

Compared with adults, pediatric mastocytosis has a relatively favorable prognosis. Interestingly, a difference was also observed in the status of c-kit mutations according to the age of onset. Although most adult patients have a D(816)V mutation in phosphotransferase domain (PTD), we have described that half of the children carry mutations in extracellular domain (ECD). KIT-ECD versus KIT-PTD mutants were introduced into rodent Ba/F3, EML, Rat2, and human TF1 cells to investigate their biologic effect. Both ECD and PTD mutations induced constitutive receptor autophosphorylation and ligand-independent proliferation of the 3 hematopoietic cells. Unlike ECD mutants, PTD mutants enhanced cluster formation and up-regulated several mast cell-related antigens in Ba/F3 cells. PTD mutants failed to support colony formation and erythropoietin-mediated erythroid differentiation. ECD and PTD mutants also displayed distinct whole-genome transcriptional profiles in EML cells. We observed differences in their signaling properties: they both activated STAT, whereas AKT was only activated by ECD mutants. Consistently, AKT inhibitor suppressed ECD mutant-dependent proliferation, clonogenicity, and erythroid differentiation. Expression of myristoylated AKT restored erythroid differentiation in EML-PTD cells, suggesting the differential role of AKT in those mutants. Overall, our study implied different pathogenesis of pediatric versus adult mastocytosis, which might explain their diverse phenotypes.


Asunto(s)
Mastocitosis/genética , Mutación/genética , Fosfotransferasas/genética , Proteínas Proto-Oncogénicas c-kit/genética , Adulto , Animales , Apoptosis , Western Blotting , Células Cultivadas , Niño , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Linfocitos/metabolismo , Mastocitos/metabolismo , Mastocitosis/metabolismo , Mastocitosis/patología , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Biochem Biophys Res Commun ; 393(1): 174-8, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20117079

RESUMEN

FES is a cytoplasmic tyrosine kinase activated by several membrane receptors, originally identified as a viral oncogene product. We have recently identified FES as a crucial effector of oncogenic KIT mutant receptor. However, FES implication in wild-type KIT receptor function was not addressed. We report here that FES interacts with KIT and is phosphorylated following activation by its ligand SCF. Unlike in the context of oncogenic KIT mutant, FES is not involved in wild-type KIT proliferation signal, or in cell adhesion. Instead, FES is required for SCF-induced chemotaxis. In conclusion, FES kinase is a mediator of wild-type KIT signalling implicated in cell migration.


Asunto(s)
Quimiotaxis , Proteínas Proto-Oncogénicas c-fes/metabolismo , Factor de Células Madre/metabolismo , Dominios Homologos src , Adhesión Celular , Línea Celular Tumoral , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-fes/genética , Técnicas del Sistema de Dos Híbridos , Tirosina
20.
Blood ; 112(10): 4039-47, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18753636

RESUMEN

Stem cell factor (SCF) plays critical roles in proliferation, survival, migration, and function of hematopoietic progenitor and mast cells through binding to Kit receptor. Previous studies have implicated the adaptor protein Lnk as an important negative regulator of SCF signaling. However, the molecular mechanism underlying this regulation is unclear. Here, we showed that the Src homology 2 domain (SH2) of Lnk binds directly and preferentially to phosphorylated tyrosine 567 in Kit juxtamembrane domain. Using Lnk(-/-) bone marrow mast cells (BMMCs) transduced with different Lnk proteins, we demonstrated that Lnk down-regulates SCF-induced proliferation with attenuation of mitogen-activated protein kinase (MAPK) and c-jun N-terminal kinase signaling. Furthermore, we showed that Lnk(-/-) BMMCs displayed increased SCF-dependent migration compared with wild-type cells, revealing a novel Lnk-mediated inhibitory function. This correlated with enhanced Rac and p38 MAPK activation. Finally, we found that Lnk domains and carboxy-terminal tyrosine contribute differently to inhibition of in vitro expansion of hematopoietic progenitors. Altogether, our results demonstrate that Lnk, through its binding to Kit tyrosine 567, negatively modulates specific SCF-dependent signaling pathways involved in the proliferation and migration of primary hematopoietic cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Mastocitos/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células de la Médula Ósea/citología , Movimiento Celular/fisiología , Proliferación Celular , Supervivencia Celular/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Activación Enzimática/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Mastocitos/citología , Proteínas de la Membrana , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Factor de Células Madre/genética , Transducción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...