Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Immunology ; 164(4): 722-736, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34534359

RESUMEN

Bruton's tyrosine kinase (BTK) is a TEC kinase with a multifaceted role in B-cell biology and function, highlighted by its position as a critical component of the B-cell receptor signalling pathway. Due to its role as a therapeutic target in several haematological malignancies including chronic lymphocytic leukaemia, BTK has been gaining tremendous momentum in recent years. Within the immune system, BTK plays a part in numerous pathways and cells beyond B cells (i.e. T cells, macrophages). Not surprisingly, BTK has been elucidated to be a driving factor not only in lymphoproliferative disorders but also in autoimmune diseases and response to infection. To extort this role, BTK inhibitors such as ibrutinib have been developed to target BTK in other diseases. However, due to rising levels of resistance, the urgency to develop new inhibitors with alternative modes of targeting BTK is high. To meet this demand, an expanding list of BTK inhibitors is currently being trialled. In this review, we synopsize recent discoveries regarding BTK and its role within different immune cells and pathways. Additionally, we discuss the broad significance and relevance of BTK for various diseases ranging from haematology and rheumatology to the COVID-19 pandemic. Overall, BTK signalling and its targetable nature have emerged as immensely important for a wide range of clinical applications. The development of novel, more specific and less toxic BTK inhibitors could be revolutionary for a significant number of diseases with yet unmet treatment needs.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Linfocitos B/enzimología , Sistema Inmunológico/enzimología , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , COVID-19/enzimología , COVID-19/inmunología , Humanos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/inmunología , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/inmunología , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Quimiocina/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Tratamiento Farmacológico de COVID-19
3.
Hum Pathol ; 68: 175-183, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28899738

RESUMEN

Angiomyomatous hamartoma of lymph nodes (AMH-LN) is an uncommon benign proliferation of smooth muscle, blood vessels, collagenous stroma, and adipocytes, most commonly affecting inguinal LN. A similar constellation of cell types constitutes various members of the perivascular epithelioid cell tumor (PEComa) family, including lymphangioleiomyomatosis (LAM), which can involve LN in women. Because some LN-LAM patients have tuberous sclerosis complex and/or other PEComa family lesions, it is clinically relevant to distinguish LN-LAM from AMH-LN. Given their similar features, however, the possibility that AMH-LN is a morphologic variant of LN-LAM merits inquiry. The dual melanocytic and myoid immunophenotype distinguishes the PEComa family from its mimics. Cathepsin K has recently emerged as a more sensitive marker for the PEComa family than HMB-45, which can be weak and focal, but cathepsin K has not been studied in AMH-LN. This study evaluated 21 AMH-LNs for clinical, morphologic, and immunophenotypic features of LN-LAM. None (0/21) had tuberous sclerosis complex or PEComas. Thirteen (62%) were male, unlike LN-LAM, which is restricted to women. All cases exhibited intraparenchymal proliferation of variable-sized, thick-walled blood vessels within collagenous stroma containing a sparse to focally cellular population of haphazardly distributed smooth muscle cells. Admixed adipocytes were commonly present. None exhibited classical features of LN-LAM such as subcapsular localization, extranodal extension, intralymphatic growth, compact nests, branching lymphatic channels, plump cell shape, or foamy/clear cytoplasm. None exhibited any staining for cathepsin K, HMB-45, or microphthalmia transcription factor. There is no clinical, morphologic, or immunohistochemical evidence to suggest that AMH-LN is a variant of LN-LAM.


Asunto(s)
Hamartoma/patología , Ganglios Linfáticos/patología , Linfangioleiomiomatosis/patología , Trastornos Linfoproliferativos/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Biopsia , Catepsina K/análisis , Diagnóstico Diferencial , Femenino , Hamartoma/enzimología , Humanos , Inmunohistoquímica , Ganglios Linfáticos/enzimología , Trastornos Linfoproliferativos/enzimología , Masculino , Antígenos Específicos del Melanoma/análisis , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Adulto Joven , Antígeno gp100 del Melanoma
4.
Dis Model Mech ; 9(12): 1483-1495, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27799148

RESUMEN

Histone deacetylase 9 (HDAC9) is expressed in B cells, and its overexpression has been observed in B-lymphoproliferative disorders, including B-cell non-Hodgkin lymphoma (B-NHL). We examined HDAC9 protein expression and copy number alterations in primary B-NHL samples, identifying high HDAC9 expression among various lymphoma entities and HDAC9 copy number gains in 50% of diffuse large B-cell lymphoma (DLBCL). To study the role of HDAC9 in lymphomagenesis, we generated a genetically engineered mouse (GEM) model that constitutively expressed an HDAC9 transgene throughout B-cell development under the control of the immunoglobulin heavy chain (IgH) enhancer (Eµ). Here, we report that the Eµ-HDAC9 GEM model develops splenic marginal zone lymphoma and lymphoproliferative disease (LPD) with progression towards aggressive DLBCL, with gene expression profiling supporting a germinal center cell origin, as is also seen in human B-NHL tumors. Analysis of Eµ-HDAC9 tumors suggested that HDAC9 might contribute to lymphomagenesis by altering pathways involved in growth and survival, as well as modulating BCL6 activity and p53 tumor suppressor function. Epigenetic modifications play an important role in the germinal center response, and deregulation of the B-cell epigenome as a consequence of mutations and other genomic aberrations are being increasingly recognized as important steps in the pathogenesis of a variety of B-cell lymphomas. A thorough mechanistic understanding of these alterations will inform the use of targeted therapies for these malignancies. These findings strongly suggest a role for HDAC9 in B-NHL and establish a novel GEM model for the study of lymphomagenesis and, potentially, preclinical testing of therapeutic approaches based on histone deacetylase inhibitors.


Asunto(s)
Linfocitos B/enzimología , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/genética , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/genética , Proteínas Represoras/genética , Acetilación , Animales , Linfocitos B/patología , Ciclo Celular/genética , Perfilación de la Expresión Génica , Reordenamiento Génico de Cadena Pesada de Linfocito B/genética , Células HeLa , Histona Desacetilasas/metabolismo , Humanos , Linfoma de Células B/patología , Trastornos Linfoproliferativos/patología , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
5.
Cancer Biomark ; 17(4): 383-389, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27662324

RESUMEN

BACKGROUND: Hematopoietic malignancies are a group of blood cell disorders characterized by abnormal hematopoietic proliferation. OBJECTIVE: The identification of specific clinicopathologic characteristics and tumor-related gene status provides critical information on potential therapeutic targets. METHODS: The specimens were tested with immunohistochemistry, flow cytometry, RT-PCR and fragment analysis. RESULTS: In this study, a patient with a long history of tobacco use was reported with a diagnosis of simultaneous low-grade B-cell lymphoproliferative disorder (LPD) and myeloproliferative neoplasm (MPN). Mutational analysis revealed that JAK2 V617F mutation and CALR mutation with 52bp deletion were present in this patient. CONCLUSION: These results suggest that lymphoproliferative and myeloproliferative neoplasms may coexist, although the pathogenetic mechanism of coexisting hematologic requires further investigation. Additionally, the data indicate that JAK2 V617F and CALR mutations are not mutually exclusive and the actual frequency of simultaneous JAK2 V617F and CALR mutations is unknown. Whether the coexistence of these mutations imposes any biological or clinical significance awaits further investigation.


Asunto(s)
Calreticulina/genética , Janus Quinasa 2/genética , Trastornos Linfoproliferativos/genética , Mutación , Trastornos Mieloproliferativos/genética , Humanos , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/metabolismo , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/metabolismo
6.
Carcinogenesis ; 36(11): 1354-62, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26363032

RESUMEN

Arginase (ARG) is a metabolic enzyme present in two isoforms that hydrolyze l-arginine to urea and ornithine. In humans, ARG isoform 1 is also expressed in cells of the myeloid lineage. ARG activity promotes tumour growth and inhibits T lymphocyte activation. However, the two ARG transgenic mouse lines produced so far failed to show such effects. We have generated, in two different genetic backgrounds, transgenic mice constitutively expressing ARG1 under the control of the CD68 promoter in macrophages and monocytes. Both heterozygous and homozygous transgenic mice showed a relevant increase in mortality at early age, compared with wild-type siblings (67/267 and 48/181 versus 8/149, respectively, both P < 0.005). This increase was due to high incidence of haematologic malignancies, in particular myeloid leukaemia, myeloid dysplasia, lymphomas and disseminated intravascular coagulation (DIC), diseases that were absent in wild-type mice. Atrophy of lymphoid organs due to reduction in T-cell compartment was also detected. Our results indicate that ARG activity may participate in the pathogenesis of lymphoproliferative and myeloproliferative disorders, suggest the involvement of alterations of L-arginine metabolism in the onset of DIC and confirm a role for the enzyme in regulating T-cell homeostasis.


Asunto(s)
Arginasa/metabolismo , Trastornos Linfoproliferativos/enzimología , Monocitos/enzimología , Trastornos Mieloproliferativos/enzimología , Animales , Arginasa/genética , Linaje de la Célula , Femenino , Expresión Génica , Activación de Linfocitos , Trastornos Linfoproliferativos/patología , Masculino , Ratones Transgénicos , Trastornos Mieloproliferativos/patología , Linfocitos T/enzimología , Linfocitos T/inmunología
7.
J Exp Med ; 211(13): 2537-47, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25488983

RESUMEN

Class IA phosphatidylinositol 3-kinases (PI3K), which generate PIP3 as a signal for cell growth and proliferation, exist as an intracellular complex of a catalytic subunit bound to a regulatory subunit. We and others have previously reported that heterozygous mutations in PIK3CD encoding the p110δ catalytic PI3K subunit cause a unique disorder termed p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency (PASLI) disease. We report four patients from three families with a similar disease who harbor a recently reported heterozygous splice site mutation in PIK3R1, which encodes the p85α, p55α, and p50α regulatory PI3K subunits. These patients suffer from recurrent sinopulmonary infections and lymphoproliferation, exhibit hyperactive PI3K signaling, and have prominent expansion and skewing of peripheral blood CD8(+) T cells toward terminally differentiated senescent effector cells with short telomeres. The PIK3R1 splice site mutation causes skipping of an exon, corresponding to loss of amino acid residues 434-475 in the inter-SH2 domain. The mutant p85α protein is expressed at low levels in patient cells and activates PI3K signaling when overexpressed in T cells from healthy subjects due to qualitative and quantitative binding changes in the p85α-p110δ complex and failure of the C-terminal region to properly inhibit p110δ catalytic activity.


Asunto(s)
Empalme Alternativo/genética , Genes Dominantes , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Trastornos Linfoproliferativos/genética , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Adolescente , Adulto , Formación de Anticuerpos , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , Dominio Catalítico , Diferenciación Celular , Preescolar , Fosfatidilinositol 3-Quinasa Clase Ia , Activación Enzimática , Exones/genética , Femenino , Heterocigoto , Humanos , Síndromes de Inmunodeficiencia/inmunología , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/inmunología , Masculino , Datos de Secuencia Molecular , Linaje , Fosfatidilinositol 3-Quinasas/química , Estructura Terciaria de Proteína , Eliminación de Secuencia , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Telómero/metabolismo
8.
J Clin Immunol ; 34(8): 892-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25339095

RESUMEN

In patients with underlying immunodeficiency, Epstein-Barr virus (EBV) may lead to severe immune dysregulation manifesting as fatal mononucleosis, lymphoma, lymphoproliferative disease (LPD), lymphomatoid granulomatosis, hemophagocytic lymphohistiocytosis (HLH) and dysgammaglobulinemia. Several newly discovered primary immunodeficiencies (STK4, CD27, MAGT1, CORO1A) have been described in recent years; our group and collaborators were able to reveal the pathogenicity of mutations in the Interleukin-2-inducible T-cell Kinase (ITK) in a cohort of nine patients with most patients presenting with massive EBV B-cell lymphoproliferation. This review summarizes the clinical and immunological findings in these patients. Moreover, we describe the functional consequences of the mutations and draw comparisons with the extensively investigated function of ITK in vitro and in the murine model.


Asunto(s)
Interleucina-2/metabolismo , Trastornos Linfoproliferativos/enzimología , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Linfocitos T/enzimología , Edad de Inicio , Animales , Linfocitos B/enzimología , Linfocitos B/patología , Modelos Animales de Enfermedad , Humanos , Trastornos Linfoproliferativos/genética , Linfocitos T/patología
10.
Bull Exp Biol Med ; 155(5): 676-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24288737

RESUMEN

Enzymes of the cytochrome P450 and GSTP1 families play a pivotal role in the metabolism of a wide variety of antitumor drugs and polymorphisms of genes encoding for metabolizing enzymes can affect drug efficacy and toxicity. We studied the associations between functionally significant gene polymorphisms CYP2C8, CYP2C9, CYP2C19, CYP3A5, and GSTP1 and clinical response to chemotherapy in patients with chronic lymphoproliferative diseases. Significant correlations with chemotherapy resistance were observed for CYP2C8 3 (OR=7.05; CI 95%=1.76-29.55) and CYP2C9 2 polymorphisms (OR=4.1; CI 95%=1.03-16.81). No significant association between chemotherapy resistance and other examined polymorphisms were found.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/genética , Resistencia a Antineoplásicos/genética , Inactivación Metabólica/genética , Trastornos Linfoproliferativos/genética , Polimorfismo Genético , Alelos , Antineoplásicos/uso terapéutico , Enfermedad Crónica , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP2C9 , Femenino , Frecuencia de los Genes , Genotipo , Heterocigoto , Homocigoto , Humanos , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/patología , Masculino , Persona de Mediana Edad
11.
Am J Transplant ; 13(4): 883-890, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23398911

RESUMEN

Posttransplant lymphoproliferative disorder (PTLD)-associated Epstein-Barr virus (EBV)+ B cell lymphomas are serious complications of solid organ and bone marrow transplantation. The EBV protein LMP2a, a B cell receptor (BCR) mimic, provides survival signals to virally infected cells through Syk tyrosine kinase. Therefore, we explored whether Syk inhibition is a viable therapeutic strategy for EBV-associated PTLD. We have shown that R406, the active metabolite of the Syk inhibitor fostamatinib, induces apoptosis and cell cycle arrest while decreasing downstream phosphatidylinositol-3'-kinase (PI3K)/Akt signaling in EBV+ B cell lymphoma PTLD lines in vitro. However, Syk inhibition did not inhibit or delay the in vivo growth of solid tumors established from EBV-infected B cell lines. Instead, we observed tumor growth in adjacent inguinal lymph nodes exclusively in fostamatinib-treated animals. In contrast, direct inhibition of PI3K/Akt significantly reduced tumor burden in a xenogeneic mouse model of PTLD without evidence of tumor growth in adjacent inguinal lymph nodes. Taken together, our data indicate that Syk activates PI3K/Akt signaling which is required for survival of EBV+ B cell lymphomas. PI3K/Akt signaling may be a promising therapeutic target for PTLD, and other EBV-associated malignancies.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trastornos Linfoproliferativos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Aminopiridinas , Animales , Apoptosis , Linfocitos B/metabolismo , Ciclo Celular , Línea Celular Tumoral , Activación Enzimática , Herpesvirus Humano 4 , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Ganglios Linfáticos/patología , Linfoma de Células B/enzimología , Linfoma de Células B/virología , Trastornos Linfoproliferativos/virología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Morfolinas , Oxazinas/farmacología , Complicaciones Posoperatorias , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas , Transducción de Señal , Quinasa Syk , Trasplante Heterólogo
12.
J Immunol ; 190(1): 147-58, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23209318

RESUMEN

Mice expressing a germline mutation in the phospholipase C-γ1-binding site of linker for activation of T cells (LAT) show progressive lymphoproliferation and ultimately die at 4-6 mo age. The hyperactivated T cells in these mice show defective TCR-induced calcium flux but enhanced Ras/ERK activation, which is critical for disease progression. Despite the loss of LAT-dependent phospholipase C-γ1 binding and activation, genetic analysis revealed RasGRP1, and not Sos1 or Sos2, to be the major Ras guanine exchange factor responsible for ERK activation and the lymphoproliferative phenotype in these mice. Analysis of isolated CD4(+) T cells from LAT-Y136F mice showed altered proximal TCR-dependent kinase signaling, which activated a Zap70- and LAT-independent pathway. Moreover, LAT-Y136F T cells showed ERK activation that was dependent on Lck and/or Fyn, protein kinase C-θ, and RasGRP1. These data demonstrate a novel route to Ras activation in vivo in a pathological setting.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Linfocitos T CD4-Positivos/inmunología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Activación de Linfocitos/inmunología , Trastornos Linfoproliferativos/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas de la Membrana/genética , Fosfolipasa C gamma , Fosfoproteínas/genética , Animales , Linfocitos T CD4-Positivos/enzimología , Progresión de la Enfermedad , Mutación de Línea Germinal/inmunología , Activación de Linfocitos/genética , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/genética , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Fosfolipasa C gamma/fisiología
13.
J Immunol ; 189(8): 4005-13, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22984075

RESUMEN

The ability of the transmembrane adaptor protein linker for activation of T cells (LAT) to regulate T cell development, activation, survival, and homeostasis depends upon phosphorylation of its multiple tyrosine residues. The mutation of tyrosine 136 on LAT abrogates its interaction with phospholipase C-γ1, causing severe ramifications on TCR-mediated signaling. Mice harboring this mutation, LATY136F mice, have significantly impaired thymocyte development; however, they rapidly develop a fatal lymphoproliferative disease marked by the uncontrolled expansion of Th2-skewed CD4(+) T cells, high levels of IgE and IgG1, and autoantibody production. In this study, we assessed the contribution of multiple signaling pathways in LATY136F disease development. The deletion of the critical signaling proteins Gads and RasGRP1 caused a further block in thymocyte development, but, over time, could not prevent CD4(+) T cell hyperproliferation. Also, restoring signaling through the NF-κB and NFAT pathways was unable to halt the development of disease. However, expression of a constitutively active Raf transgene enhanced lymphoproliferation, indicating a role for the Ras-MAPK pathway in LAT-mediated disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas de la Membrana/fisiología , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Enfermedades Autoinmunes/genética , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Trastornos Linfoproliferativos/genética , Sistema de Señalización de MAP Quinasas/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Receptores de Antígenos de Linfocitos T/deficiencia , Receptores de Antígenos de Linfocitos T/genética , Quinasas raf/biosíntesis , Quinasas raf/genética
14.
Int Arch Allergy Immunol ; 158(4): 418-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22487848

RESUMEN

IL-2-inducible T-cell kinase (ITK) deficiency is a rare inherited immunodeficiency disease characterized by homozygous mutations in the ITK gene and the inability to control Epstein-Barr virus (EBV) infection leading to EBV-associated lymphoproliferative disorders of B cell origin. Many aspects of its clinical presentation and immunologic phenotype are still unclear to clinicians. We report on a 14-year-old female patient with complaints of an 8-month history of cough and fever. Imaging studies revealed diffuse pulmonary nodules and mediastinal lymphadenopathy. Transbronchial lung biopsy showed nonmalignant polyclonal B cell proliferation. High titers of EBV DNA were detected by PCR analysis in bronchoalveolar lavage fluid, bone marrow, and blood. Genomic analysis revealed a homozygous single base pair deletion in exon 5 of the ITK gene (c.468delT) in this patient. Treatment with rituximab (anti-CD20 mab) resulted in complete clinical remission with resolution of pulmonary lesions and a negative EBV titer in serum. All patients with EBV-associated lymphoproliferative disorders should be analyzed for mutations in ITK.


Asunto(s)
Infecciones por Virus de Epstein-Barr/enzimología , Neumonía Viral/enzimología , Proteínas Tirosina Quinasas/genética , Adolescente , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Linfocitos B/virología , Líquido del Lavado Bronquioalveolar/virología , Tos/diagnóstico , Tos/tratamiento farmacológico , Tos/enzimología , Tos/patología , Tos/virología , ADN Viral/análisis , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/patología , Femenino , Fiebre/diagnóstico , Fiebre/tratamiento farmacológico , Fiebre/enzimología , Fiebre/patología , Fiebre/virología , Humanos , Factores Inmunológicos/uso terapéutico , Pulmón/diagnóstico por imagen , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Pulmón/virología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Trastornos Linfoproliferativos/diagnóstico por imagen , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/virología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/patología , Mutación Puntual , Rituximab , Tomografía Computarizada por Rayos X
15.
Am J Surg Pathol ; 33(5): 720-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19145199

RESUMEN

Cyclin-dependent protein kinase 6 (CDK6), in cooperation with cyclin Ds, drives cell cycle progression from G1 to S phase through phosphorylation and subsequent inactivation of retinoblastoma 1 protein. Alteration of this pathway results in both nonhematologic and hematologic malignancies, which include a small subset of B-cell lymphoproliferative disorders (BLPDs). We identified 5 cases of BLPD that carried CDK6 chromosomal translocations and characterized their clinical, pathologic, immunophenotypic, and genetic features. Common clinical characteristics included marked neoplastic lymphocytosis, systemic lymphadenopathy, splenomegaly, and bone marrow involvement. Three patients were diagnosed with low-grade B-cell lymphoma and had an indolent clinical course, and 2 patients (one who transformed to large B-cell lymphoma, and the other who was initially diagnosed with a high-grade B-cell lymphoma) had an aggressive clinical course. Immunophenotypically, the neoplastic B cells expressed CD5, CDK6, and cytoplasmic retinoblastoma 1 protein in all cases, expressed phospho-RB, p27kip1, and cyclin D2 in most cases, and uniformly lacked expression of all other cyclins. In 4 cases, the CDK6 translocation partner was kappa immunoglobulin light-chain gene; and in the fifth case, the CDK6 translocation partner was unknown. These distinct clinicopathologic and cytogenetic features distinguish the CDK6 translocation-associated BLPDs (CDK6-BLPDs) from other mature B-cell lymphomas.


Asunto(s)
Linfocitos B/enzimología , Quinasa 6 Dependiente de la Ciclina/genética , Cadenas kappa de Inmunoglobulina/genética , Linfoma de Células B/genética , Trastornos Linfoproliferativos/genética , Translocación Genética , Anciano , Anciano de 80 o más Años , Linfocitos B/inmunología , Linfocitos B/patología , Antígenos CD5/análisis , Ciclina D2 , Quinasa 6 Dependiente de la Ciclina/análisis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/análisis , Bases de Datos como Asunto , Femenino , Genotipo , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Péptidos y Proteínas de Señalización Intracelular/análisis , Linfoma de Células B/enzimología , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Linfoma de Células B/terapia , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/terapia , Masculino , Persona de Mediana Edad , Fenotipo , Fosforilación , Proteína de Retinoblastoma/análisis , Resultado del Tratamiento
16.
Blood ; 113(12): 2746-54, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19139084

RESUMEN

The tyrosine kinase JAK3 plays a well-established role during normal lymphocyte development and is constitutively phosphorylated in several lymphoid malignancies. However, its contribution to lymphomagenesis remains elusive. In this study, we used the newly identified activating JAK3A572V mutation to elucidate the effect of constitutive JAK3 signaling on murine lymphopoiesis. In a bone marrow transplantation model, JAK3A572V induces an aggressive, fatal, and transplantable lymphoproliferative disorder characterized by the expansion of CD8(+)TCRalphabeta(+)CD44(+)CD122(+)Ly-6C(+) T cells that closely resemble an effector/memory T-cell subtype. Compared with wild-type counterparts, these cells show increased proliferative capacities in response to polyclonal stimulation, enhanced survival rates with elevated expression of Bcl-2, and increased production of interferon-gamma (IFNgamma) and tumor necrosis factor-alpha (TNFalpha), correlating with enhanced cytotoxic abilities against allogeneic target cells. Of interest, the JAK3A572V disease is epidermotropic and produces intraepidermal microabscesses. Taken together, these clinical features are reminiscent of those observed in an uncommon but aggressive subset of CD8(+) human cutaneous T-cell lymphomas (CTCLs). However, we also observed a CD4(+) CTCL-like phenotype when cells are transplanted in an MHC-I-deficient background. These data demonstrate that constitutive JAK3 activation disrupts T-cell homeostasis and induces lymphoproliferative diseases in mice.


Asunto(s)
Linfocitos T CD8-positivos/patología , Janus Quinasa 3/fisiología , Linfopoyesis/fisiología , Trastornos Linfoproliferativos/etiología , Mutación Puntual , Proteínas Recombinantes de Fusión/fisiología , Subgrupos de Linfocitos T/patología , Animales , Antígenos Ly/análisis , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/química , Inducción Enzimática , Humanos , Receptores de Hialuranos/análisis , Subunidad beta del Receptor de Interleucina-2/análisis , Janus Quinasa 3/biosíntesis , Janus Quinasa 3/genética , Linfoma Cutáneo de Células T/patología , Linfopoyesis/genética , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/patología , Ratones , Ratones Endogámicos C57BL , Quimera por Radiación , Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Piel/patología , Subgrupos de Linfocitos T/química
17.
Cancer Cell ; 13(6): 467-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18538729

RESUMEN

The introduction of tyrosine kinase inhibitors in the treatment of BCR-ABL1-rearranged malignancies has revolutionized therapy, but the prognosis for acute leukemias remains suboptimal. In this issue of Cancer Cell, Bueno et al. (2008) add a new dimension to the regulation of ABL1 expression. The authors demonstrate that ABL1 is a direct target of miR-203, miR-203 is silenced by genetic and epigenetic mechanisms in hematopoietic malignancies expressing either ABL1 or BCR-ABL1, and restoration of miR-203 expression reduces ABL1 and BCR-ABL1 levels and inhibits cell proliferation. These findings may have broad implications for mechanisms underlying malignant transformation in hematopoietic and other malignancies.


Asunto(s)
Metilación de ADN , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Trastornos Linfoproliferativos/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Linfoma de Células T/genética , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Ratones , MicroARNs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Regulación hacia Arriba
18.
Cancer Cell ; 13(6): 496-506, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18538733

RESUMEN

The mammalian genome contains several hundred microRNAs that regulate gene expression through modulation of target mRNAs. Here, we report a fragile chromosomal region lost in specific hematopoietic malignancies. This 7 Mb region encodes about 12% of all genomic microRNAs, including miR-203. This microRNA is additionally hypermethylated in several hematopoietic tumors, including chronic myelogenous leukemias and some acute lymphoblastic leukemias. A putative miR-203 target, ABL1, is specifically activated in these hematopoietic malignancies in some cases as a BCR-ABL1 fusion protein (Philadelphia chromosome). Re-expression of miR-203 reduces ABL1 and BCR-ABL1 fusion protein levels and inhibits tumor cell proliferation in an ABL1-dependent manner. Thus, miR-203 functions as a tumor suppressor, and re-expression of this microRNA might have therapeutic benefits in specific hematopoietic malignancies.


Asunto(s)
Proliferación Celular , Metilación de ADN , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Trastornos Linfoproliferativos/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Regiones no Traducidas 3' , Animales , Azacitidina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromosomas Humanos Par 14 , Cromosomas de los Mamíferos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Pérdida de Heterocigocidad , Linfoma de Células T/genética , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Fenilbutiratos/farmacología , Cromosoma Filadelfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Regiones Promotoras Genéticas , Factores de Tiempo , Transfección , Regulación hacia Arriba , Irradiación Corporal Total
19.
Cancer Biol Ther ; 6(12): 1837-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18087222

RESUMEN

Ataxia telangiectasia (A-T) is a disorder characterized by cerebellar degeneration, immunodeficiency, genomic instability and genetic predisposition to lymphoid malignancies with translocations involving antigen receptor loci. The Ataxia Telangiectasia Mutated gene encodes the ATM kinase, a central transducer of DNA damage signals. Until recently, the etiology of the lymphoid phenotype in A-T patients and the mechanisms by which ATM ensures normal repair of DNA double strand break (DSB) intermediates during antigen receptor diversification reactions remained poorly understood. Last year, Bredemeyer et al. (Nature 2006; 442:466-70) demonstrated that ATM stabilizes chromosomal V(D)J recombination DSB intermediates, facilitates DNA end joining and prevents broken DNA ends from participating in chromosome deletions, inversions and translocations. A more recent study by Callen et al. (Cell 2007; 130:63-75) highlighted the importance of ATM-mediated checkpoints in blocking the long-term persistence and transmission of un-repaired DSBs in developing lymphocytes. Collectively, these results have provided complementary mechanistic insights into ATM functions in V(D)J recombination that can account for the lymphoid tumor-prone phenotype associated with A-T.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Aberraciones Cromosómicas , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Reordenamiento Génico/fisiología , Genes cdc , Proteínas de Homeodominio/fisiología , Humanos , Linfocitos/citología , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/fisiopatología , Modelos Inmunológicos , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptores de Antígenos/genética , Recombinación Genética/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , VDJ Recombinasas/fisiología
20.
J Biol Chem ; 282(21): 15833-42, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17400555

RESUMEN

The clinical immunosuppressant FTY720 is a sphingosine analogue that, once phosphorylated by sphingosine kinase 2 (Sphk2), is an agonist of multiple receptor subtypes for sphingosine 1-phosphate. Short exposures to FTY720 afford long term protection in lymphoproliferative and autoimmune disease models, presumably by inducing apoptosis in subsets of cells essential for pathogenesis. Sphingosine itself is pro-apoptotic, and apoptosis induced with FTY720 or sphingosine is thought to proceed independently of their phosphorylation. Following chemical mutagenesis of Jurkat cells we isolated mutants that are selectively resistant to FTY720 analogue AAL(R), as well as natural sphingolipid bases, including sphingosine. Cells lacking functional Sphk2 were resistant to apoptosis induced with AAL(R), indicating that apoptosis proceeds through AAL(R) phosphorylation. Phosphorylation of AAL(R) was also required for induction of lymphocyte apoptosis in mice, as apoptosis was not induced with the non-phosphorylatable chiral analogue, AAL(S). Apoptosis was induced in the spleen but not the thymus of mice administered 1 mg/kg AAL(R), correlating with levels of AAL(R)-phosphate (AFD(R)) in organ extracts. AFD(R) did not induce apoptosis when added to the cell culture medium, indicating that it induces apoptosis through an intracellular target. NBD-labeled AAL(R) localized to the endoplasmic reticulum, and AAL(R) treatment resulted in elevated cytosolic calcium, Bax redistribution from cytosol to mitochondrial and endoplasmic reticulum membranes, and caspase-independent mitochondrial permeabilization in Jurkat cells. We therefore describe an apoptotic pathway triggered by intracellular accumulation of sphingolipid base phosphates and suggest that sphingoid base substrates for Sphk2 acting intracellularly could be useful in the treatment of lymphoproliferative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Inmunosupresores/farmacología , Trastornos Linfoproliferativos/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Apoptosis/genética , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/genética , Señalización del Calcio/genética , Caspasas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Retículo Endoplásmico/enzimología , Clorhidrato de Fingolimod , Células HeLa , Humanos , Células Jurkat , Trastornos Linfoproliferativos/tratamiento farmacológico , Trastornos Linfoproliferativos/genética , Ratones , Ratones Noqueados , Mitocondrias , Mutagénesis , Especificidad de Órganos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Esfingolípidos/metabolismo , Esfingolípidos/farmacología , Esfingosina/farmacología , Bazo/enzimología , Timo/enzimología , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA