Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Blood ; 120(16): 3310-7, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-22942183

RESUMEN

Fms-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase with important roles in hematopoietic progenitor cell survival and proliferation. It is mutated in approximately one-third of AML patients, mostly by internal tandem duplications (ITDs). Adaptor protein Lnk is a negative regulator of hematopoietic cytokine signaling. In the present study, we show that Lnk interacts physically with both wild-type FLT3 (FLT3-WT) and FLT3-ITD through the SH2 domains. We have identified the tyrosine residues 572, 591, and 919 of FLT3 as phosphorylation sites involved in direct binding to Lnk. Lnk itself was tyrosine phosphorylated by both FLT3 ligand (FL)-activated FLT3-WT and constitutively activated FLT3-ITD. Both shRNA-mediated depletion and forced overexpression of Lnk demonstrated that activation signals emanating from both forms of FLT3 are under negative regulation by Lnk. Moreover, Lnk inhibited 32D cell proliferation driven by different FLT3 variants. Analysis of primary BM cells from Lnk-knockout mice showed that Lnk suppresses the expansion of FL-stimulated hematopoietic progenitors, including lymphoid-primed multipotent progenitors. The results of the present study show that through direct binding to FLT3, Lnk suppresses FLT3-WT/ITD-dependent signaling pathways involved in the proliferation of hematopoietic cells. Therefore, modulation of Lnk expression levels may provide a unique therapeutic approach for FLT3-ITD-associated hematopoietic disease.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Leucemia Mieloide Aguda/metabolismo , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Proliferación Celular , Transformación Celular Neoplásica/patología , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/patología , Proteínas de la Membrana , Ratones , Ratones Noqueados , Mutación/genética , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Secuencias Repetidas en Tándem/genética , Tirosina/metabolismo , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Dominios Homologos src
2.
Biol Blood Marrow Transplant ; 17(10): 1546-50, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21324375

RESUMEN

Seroconversion rates following influenza vaccination in patients with hematologic malignancies after hematopoietic stem cell transplantation (HSCT) are known to be lower compared to healthy adults. The aim of our diagnostic study was to determine the rate of seroconversion after 1 or 2 doses of a novel split virion, inactivated, AS03-adjuvanted pandemic H1N1 influenza vaccine (A/California/7/2009) in HSCT recipients (ClinicalTrials.gov Identifier: NCT01017172). Blood samples were taken before and 21 days after a first dose and 21 days after a second dose of the vaccine. Antibody (AB) titers were determined by hemagglutination inhibition assay. Seroconversion was defined by either an AB titer of ≤ 1:10 before and ≥ 1:40 after or ≥ 1:10 before and ≥ 4-fold increase in AB titer 21 days after vaccination. Seventeen patients (14 allogeneic, 3 autologous HSCT) received 1 dose and 11 of these patients 2 doses of the vaccine. The rate of seroconversion was 41.2% (95% confidence interval [CI] 18.4-67.1) after the first and 81.8% (95% CI 48.2-97.7) after the second dose. Patients who failed to seroconvert after 1 dose of the vaccine were more likely to receive any immunosuppressive agent (P = .003), but time elapsed after or type of HSCT, age, sex, or chronic graft-versus-host disease was not different when compared to patients with seroconversion. In patients with hematologic malignancies after HSCT the rate of seroconversion after a first dose of an adjuvanted H1N1 influenza A vaccine was poor, but increased after a second dose.


Asunto(s)
Anticuerpos Antivirales/sangre , Trasplante de Células Madre Hematopoyéticas , Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza/administración & dosificación , Pandemias , Adulto , Factores de Edad , Anciano , Anticuerpos Antivirales/inmunología , Enfermedad Crónica , Femenino , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/inmunología , Humanos , Vacunas contra la Influenza/inmunología , Masculino , Persona de Mediana Edad , Factores Sexuales , Trasplante Autólogo , Trasplante Homólogo , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
4.
Ann Hematol ; 88(12): 1177-83, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19424697

RESUMEN

RNA quality is critical to achieve valid results in microarray experiments and to save resources. The RNA integrity number (RIN) can be measured with minimal sample consumption by microfluidics-based capillary electrophoresis. To determine whether RIN can predict the qualitative outcome of microarray hybridization, we measured RIN in total RNA samples from 484 different experiments by the 2100 Bioanalyzer system and correlated with the percentage of present calls (%pc) of downstream oligonucleotide microarrays. The correlation coefficient for RNA and %pc in all 408 samples for which the bioanalyzer algorithm was able to produce an RIN was 0.475 (p < 0.05), ranging from 0.039 to 0.673 for different tissue- and assay-type subgroups. Multivariate analysis found RIN to be the best predictor of microarray quality as assessed by %pc, outperforming the 28S to 18S ratio. For a %pc threshold of 25% and 35%, we determined optimal cut points for RIN at 7.15 and 8.05, respectively. Using the suggested cut points, RIN can support the final decision whether a certain RNA sample is appropriate for successful microarray hybridization.


Asunto(s)
Electroforesis Capilar , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/análisis , Algoritmos , Animales , Electroforesis Capilar/instrumentación , Electroforesis Capilar/métodos , Perfilación de la Expresión Génica/instrumentación , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN/genética , Estabilidad del ARN , Curva ROC , Reproducibilidad de los Resultados
5.
Biochem J ; 415(2): 241-5, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18588518

RESUMEN

The adaptor protein Lnk is expressed in haemopoietic cells and plays a critical role in haemopoiesis. Animal model studies demonstrated that Lnk acts as a broad inhibitor of signalling pathways in haemopoietic lineages. Lnk belongs to a family of proteins sharing several structural motifs, including an SH2 (Src homology 2) domain which binds phosphotyrosine residues in various signal-transducing proteins. The SH2 domain is essential for Lnk-mediated negative regulation of several cytokine receptors [e.g. Mpl, EpoR (erythropoietin receptor), c-Kit]. Therefore inhibition of the binding of Lnk to cytokine receptors might lead to enhanced downstream signalling of the receptor and thereby to improved haemopoiesis in response to exposure to cytokines (e.g. erythropoietin in anaemic patients). This hypothesis led us to define the exact binding site of Lnk to the stem cell factor receptor c-Kit. Pull-down experiments using GST (glutathione transferase)-fusion proteins of the different domains of c-Kit showed that Lnk almost exclusively binds to the phosphorylated juxtamembrane domain. Binding of Lnk to the juxtamembrane domain was abolished by point mutation of Tyr(568) and was competed by peptides with a phosphotyrosine residue at position 568. Co-immunoprecipitation with full-length wild-type or Y568F mutant c-Kit and Lnk confirmed these results, thus showing the importance of this phosphorylated tyrosine residue. Lnk bound directly to c-Kit without requiring other interacting partners. The identification of the binding site of Lnk to c-Kit will be useful to discover inhibitory molecules that prevent the binding of these two proteins, thus making haemopoietic cells more sensitive to growth factors.


Asunto(s)
Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Tirosina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Línea Celular , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Mutación , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/química , Tirosina/genética , Dominios Homologos src/genética
6.
Leuk Res ; 32(9): 1366-73, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18405967

RESUMEN

Cucurbitacins have long been utilized for their abortifacient and anti-inflammatory effects; however, little is known about their mechanism of action. In this study, we have demonstrated robust antiproliferative effects of CuB on various leukemia and lymphoma cell lines, as well as on primary mononuclear bone marrow cells derived from patients with acute myeloid leukemia or myelodysplastic syndrome. Myeloid leukemic cells treated with CuB exhibit significant S-phase cell cycle arrest, enlarged cell size, multinucleation, and enhanced expression of a monocytic- and granulocytic-specific CD11b. Moreover, our data demonstrate that CuB prominently alters the cytoskeletal network of leukemic cells, inducing rapid and improper polymerization of the F-actin network. These encouraging results suggest the appropriateness of clinical trials of cucurbitacins for the treatment of hematopoietic malignancies.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Citoesqueleto/patología , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicos/patología , Fase S/efectos de los fármacos , Triterpenos/farmacología , Apoptosis/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/patología , Antígeno CD11b/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Granulocitos/metabolismo , Granulocitos/patología , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico
7.
Oncol Rep ; 20(5): 1237-42, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18949427

RESUMEN

LY573636-sodium is a promising anti-tumor agent, which causes growth arrest and apoptosis of a variety of human solid tumors in vitro and in vivo. Moreover, studies have shown that the compound is selectively toxic towards tumor cells over their normal counterparts. This targeted effect makes LY573636 a candidate for combined therapy regimens in patients with advanced or resistant cancers. We studied for the first time, the anti-tumor properties of LY573636 against a variety of human hematopoietic malignancies, including AML, B-ALL, large B-cell and mantle cell lymphoma cell lines. Cells were treated with the compound in vitro and its effect on cell proliferation, apoptosis and differentiation was determined. The cell lines underwent growth arrest in response to treatment with LY573636 in a dose-dependent manner. This antiproliferative activity was associated with the induction of apoptosis, loss of mitochondrial membrane potential and induction of reactive oxygen species. Furthermore, we showed that LY573636 was able to induce granulocytic/monocytic differentiation of HL60 and U937 cells. LY573636, as shown before in solid tumors, is effective in hematopoietic cell lines as well. These data suggest the use of LY573636 alone or in combination with conventional chemotherapeutic regimens in hematopoietic malignancies.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Sulfonamidas/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Especies Reactivas de Oxígeno/metabolismo
8.
Eur J Emerg Med ; 12(6): 306-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16276262

RESUMEN

We report the case of a 25-year-old, hepatitis C-infected man, who presented with severe rhabdomyolysis and acute renal failure, and later developed posterior encephalopathy with cortical blindness after the ingestion of magic mushrooms. Conventional respiratory and cardiovascular support including mechanical ventilation, continuous veno-venous hemodialysis and corticosteroids led to improvement and the patient recovered completely over the following months. Magic mushrooms are becoming increasingly fashionable among drug users, as they are believed to be more harmless than other hallucinogenic designer drugs. So far, little is known about their possible severe side effects.


Asunto(s)
Lesión Renal Aguda/complicaciones , Alucinógenos/envenenamiento , Intoxicación por Setas/fisiopatología , Psilocibina/envenenamiento , Rabdomiólisis/complicaciones , Lesión Renal Aguda/inducido químicamente , Adulto , Enfermedades del Sistema Nervioso Central/complicaciones , Humanos , Masculino , Intoxicación por Setas/terapia , Rabdomiólisis/etiología , Rabdomiólisis/fisiopatología , Trastornos Relacionados con Sustancias
9.
Exp Hematol ; 39(5): 591-600, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21310211

RESUMEN

OBJECTIVE: Platelet-derived growth factor receptors α and ß (PDGFRA, PDGFRB) are frequently expressed on hematopoietic cells and regulate cellular responses such as proliferation, differentiation, survival, and transformation. Stimulation by autocrine loops or activation by chromosomal translocation makes them important factors in development of hematopoietic disorders. Interaction with the ligand PDGF results in activation of the tyrosine kinase domain and phosphorylation of tyrosine residues, thereby creating binding sites for molecules containing Src homology 2 domains. We hypothesized that one such protein may be Lnk, a negative regulator of cytokine receptors, including Mpl, EpoR, c-Kit, and c-Fms. MATERIALS AND METHODS: Interaction of Lnk with PDGFRA, PDGFRB, or leukemogenic FIP1L1-PDGFRA or TEL-PDGFRB was studied in cotransfected 293T cells. Effects of Lnk on PDGFR signaling were shown in 293T and NIH3T3 cells, whereas its influence on either PDGF-dependent or factor-independent growth was investigated using Ba/F3 or 32D cells expressing wild-type PDGFR, FIP1L1-PDGFRA, or TEL-PDGFRB. RESULTS: We show that Lnk binds to PDGFR after exposure of cells to PDGF. Furthermore, Lnk can bind the FIP1L1-PDGFRA fusion protein. Mutation or deletion of the Lnk Src homology 2 domain completely abolished binding of Lnk to FIP1L1-PDGFRA, but just partly prevented binding to PDGFRA or PDGFRB. Expression of Lnk inhibited proliferation of PDGF-dependent Ba/F3 cells and diminished phosphorylation of Erk in PDGF-treated NIH3T3. 32D cells transformed by either FIP1L1-PDGFRA or TEL-PDGFRB stopped growing when Lnk was expressed. CONCLUSIONS: Lnk is a negative regulator of PDGFR signaling. Development of Lnk mimetic drugs might provide a novel therapeutic strategy for myeloproliferative disorders.


Asunto(s)
Proteínas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Células 3T3 NIH , Unión Proteica , Proteínas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química
10.
Exp Hematol ; 38(9): 718-32, 732.e1-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20621626

RESUMEN

OBJECTIVE: Development of myelodysplastic syndrome (MDS) is suggested to follow a multistep pathogenesis and is characterized by accumulation of molecular defects of the hematopoietic stem/progenitor cells, resulting in aberrant differentiation and proliferation. MATERIALS AND METHODS: To detect alterations within the transcriptional program in MDS-derived CD34(+) cells during lineage-specific differentiation, we performed serial gene expression analysis of in vitro differentiated erythro-, granulo-, and megakaryopoietic cells using oligonucleotide microarrays (HG-U133A, Affymetrix, Santa Clara, CA, USA). For selected genes, expression data were confirmed using real-time polymerase chain reaction. RESULTS: We identified genes with altered expression during lineage-specific differentiation in either low- or high-risk MDS cells compared to the expression patterns of continuously up- or downregulated genes from the normal transcriptional program of hematopoiesis. In cluster analyses, we could show that MDS samples have a distinct expression pattern of a set of selected genes compared to normal cells, which allows prediction of the affiliation of a sample to one group. Furthermore, this study gives an overview of genes that are differentially expressed in MDS cells compared to normal hematopoiesis. CONCLUSION: Our data provide the first comprehensive transcriptional analysis of differentiating human CD34(+) cells derived from MDS patients compared to normal individuals. It gives new insights into the alteration of differentiation and proliferation of MDS stem cells.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Síndromes Mielodisplásicos/metabolismo , Transcripción Genética , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/patología , Humanos , Separación Inmunomagnética , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Riesgo
11.
J Leukoc Biol ; 88(4): 699-706, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20571037

RESUMEN

The M-CSFR (c-Fms) participates in proliferation, differentiation, and survival of macrophages and is involved in the regulation of distinct macrophage functions. Interaction with the ligand M-CSF results in phosphorylation of tyrosine residues on c-Fms, thereby creating binding sites for molecules containing SH2 domains. Lnk is a SH2 domain adaptor protein that negatively regulates hematopoietic cytokine receptors. Here, we show that Lnk binds to c-Fms. Biological and functional effects of this interaction were examined in macrophages from Lnk-deficient (KO) and WT mice. Clonogenic assays demonstrated an elevated number of M-CFUs in the bone marrow of Lnk KO mice. Furthermore, the M-CSF-induced phosphorylation of Akt in Lnk KO macrophages was increased and prolonged, whereas phosphorylation of Erk was diminished. Zymosan-stimulated production of ROS was increased dramatically in a M-CSF-dependent manner in Lnk KO macrophages. Lastly, Lnk inhibited M-CSF-induced migration of macrophages. In summary, we show that Lnk binds to c-Fms and can blunt M-CSF stimulation. Modulation of levels of Lnk in macrophages may provide a unique therapeutic approach to increase innate host defenses.


Asunto(s)
Macrófagos/metabolismo , Proteínas/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Diferenciación Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Ensayo de Inmunoadsorción Enzimática , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Macrófagos/citología , Macrófagos/inmunología , Proteínas de la Membrana , Ratones , Ratones Noqueados , Fagocitosis/inmunología , Proteínas/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Leukoc Biol ; 85(6): 957-65, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19293402

RESUMEN

The JAK2 mutation JAK2V617F is found frequently in patients with myeloproliferative disorders (MPD) and transforms hematopoietic cells to cytokine-independent proliferation when expressed with specific cytokine receptors. The Src homology 2 (SH2) and pleckstrin homology (PH) domain-containing adaptor protein Lnk (SH2B3) is a negative regulator of hematopoietic cytokine signaling. Here, we show that Lnk is a potent inhibitor of JAK2V617F constitutive activity. Lnk down-regulates JAK2V617F-mediated signaling and transformation in hematopoietic Ba/F3-erythropoietin receptor cells. Furthermore, in CFU assays, Lnk-deficient murine bone marrow cells are significantly more sensitive to transformation by JAK2V617F than wild-type (WT) cells. Lnk, through its SH2 and PH domains, interacts with WT and mutant JAK2 and is phosphorylated by constitutively activated JAK2V617F. Finally, we found that Lnk levels are high in CD34(+) hematopoietic progenitors from MPD patients and that Lnk expression is induced following JAK2 activation. Our data suggest that JAK2V617F is susceptible to endogenous negative-feedback regulation, providing new insights into the molecular pathogenesis of MPD.


Asunto(s)
Sustitución de Aminoácidos/genética , Janus Quinasa 2/antagonistas & inhibidores , Proteínas Mutantes/antagonistas & inhibidores , Trastornos Mieloproliferativos/enzimología , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular Transformada , Proliferación Celular , Activación Enzimática , Sistema Hematopoyético/citología , Sistema Hematopoyético/enzimología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Janus Quinasa 2/metabolismo , Proteínas de la Membrana , Ratones , Células Mieloides/citología , Células Mieloides/enzimología , Fenilalanina/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas/química , Valina/genética
13.
Exp Hematol ; 37(5): 585-592.e2, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19375649

RESUMEN

OBJECTIVE: Tyrosine kinases are involved in cytokine signaling and are frequently aberrantly activated in hematological malignancies. Lnk, a negative regulator of cytokine signaling, plays critical nonredundant roles in hematopoiesis. By binding to phosphorylated tyrosine kinases, Lnk inhibits major cytokine receptor signaling, including c-KIT; erythropoietin receptor-Janus kinase 2 (JAK2); and MPL-JAK2. In the present study, we investigated Lnk expression and possible function in transformed hematopoietic cells. MATERIALS AND METHODS: Coimmunoprecipitations were performed to identify binding between Lnk and mutant tyrosine kinases. Proliferation assays were done to examine the affect of Lnk overexpression on cancer cell growth. Real-time polymerase chain reaction analysis was used to determine Lnk expression in patient samples. RESULTS: We show that, in parallel to binding wild-type JAK2 and c-KIT, Lnk associates with and is phosphorylated by mutant alleles of JAK2 and c-KIT. In contrast, Lnk does not bind to and is not phosphorylated by BCR-ABL fusion protein. Ectopic expression of Lnk strongly attenuates growth of some leukemia cell lines, while others as well as most solid tumor cancer cell lines are either moderately inhibited or completely insensitive to Lnk. Furthermore, Lnk-mediated growth inhibition is associated with differential downregulation of phosphatidylinositol 3 kinase/Akt/mammalian target of rapamycin and mitogen-activated protein kinase/extracellular signal-regulated kinase signaling in leukemia cell lines. Surprisingly, analysis of Lnk in a large panel of myelodysplastic syndrome and acute myeloid leukemia patient samples revealed high levels of Lnk in nearly half of the samples. CONCLUSION: Although how leukemic cells overcome the antiproliferative effects of Lnk is not yet clear, our data highlight the multifaceted role negative feedback mechanisms play in malignant transformation.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Janus Quinasa 2/metabolismo , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicos/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Alelos , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Janus Quinasa 2/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Serina-Treonina Quinasas TOR
14.
Exp Hematol ; 37(8): 937-46, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19477219

RESUMEN

OBJECTIVE: Mantle cell lymphoma (MCL) is a lymphoma characterized by aberrant activation of CCND1/cyclin D1 followed by sequential genetic abnormalities. Genomic abnormalities in MCL have been extensively examined by classical cytogenetics and microarray-based comparative genomic hybridization techniques, pointing out a number of alterations in genomic regions that correlate with the neoplastic phenotype and survival. Recently, single nucleotide polymorphism genomic microarrays (SNP-chip) have been developed and used for analysis of cancer genomics. This technique allows detection of genomic changes with higher resolution, including loss of heterozygosity without changes of gene dosage, so-called acquired uniparental disomy (aUPD). MATERIALS AND METHODS: We have examined 33 samples of MCL (28 primary MCL and 5 cell lines) using the 250,000 SNP-chip from Affymetrix. RESULTS: Known alterations were confirmed by SNP arrays, including deletion of INK4A/ARF, duplication/amplification of MYC, deletion of ATM, and deletion of TP53. We also identified a duplication/amplification that occurred at 13q involving oncogenic microRNA, miR17-92. We found other genomic abnormalities, including duplication/amplification of cyclin D1, del(1p), del(6q), dup(3q) and dup(18q). Our SNP-chip analysis detected these abnormalities at high resolution, allowing us to narrow the size of the commonly deleted regions, including 1p and 6q. Our SNP-chip analysis detected a number of aUPD sites, including whole chromosome 9 aUPD and 9p aUPD. We also found an MCL case with 19p, leading to homozygous deletion of TNFSF genes. CONCLUSION: SNP-chip analysis detected in MCL very small genomic gains/losses, as well as aUPDs, which could not be detected by more conventional methods.


Asunto(s)
Análisis Mutacional de ADN/métodos , Linfoma de Células del Manto/genética , Polimorfismo de Nucleótido Simple , Células Cultivadas , Análisis Mutacional de ADN/normas , Eliminación de Gen , Duplicación de Gen , Humanos , Pérdida de Heterocigocidad , Análisis de Secuencia por Matrices de Oligonucleótidos , Eliminación de Secuencia , Disomía Uniparental
15.
Blood ; 110(9): 3360-4, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17693582

RESUMEN

Recently, activating myeloproliferative leukemia virus oncogene (MPL) mutations, MPLW515L/K, were described in myeloproliferative disorder (MPD) patients. MPLW515L leads to activation of downstream signaling pathways and cytokine-independent proliferation in hematopoietic cells. The adaptor protein Lnk is a negative regulator of several cytokine receptors, including MPL. We show that overexpression of Lnk in Ba/F3-MPLW515L cells inhibits cytokine-independent growth, while suppression of Lnk in UT7-MPLW515L cells enhances proliferation. Lnk blocks the activation of Jak2, Stat3, Erk, and Akt in these cells. Furthermore, MPLW515L-expressing cells are more susceptible to Lnk inhibitory functions than their MPL wild-type (MPLWT)-expressing counterparts. Lnk associates with activated MPLWT and MPLW515L and colocalizes with the receptors at the plasma membrane. The SH2 domain of Lnk is essential for its binding and for its down-regulation of MPLWT and MPLW515L. Lnk itself is tyrosine-phosphorylated following thrombopoietin stimulation. Further elucidating the cellular pathways that attenuate MPLW515L will provide insight into the pathogenesis of MPD and could help develop specific therapeutic approaches.


Asunto(s)
Regulación hacia Abajo , Proteínas Mutantes/genética , Trastornos Mieloproliferativos/genética , Proteínas/fisiología , Receptores de Trombopoyetina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Predisposición Genética a la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones Noqueados , Proteínas Mutantes/metabolismo , Mutación Puntual/fisiología , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Receptores de Trombopoyetina/metabolismo , Trombopoyetina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA