Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Ocul Immunol Inflamm ; 30(7-8): 2047-2054, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34402723

RESUMEN

PURPOSE: To characterize an unusual presentation of infectious posterior uveitis using multimodal imaging, and discuss the clinical decision-making involved in diagnosis and treatment. METHODS: Wide-field fundus photography, swept-source optical coherence tomography (OCT), swept-source OCT angiography, fluorescein angiography, and indocyanine green angiography. RESULTS: This patient presented with cyclical fevers and blurry vision. Fundus examination revealed bilateral optic disc edema, macular intraretinal white spots and many scattered yellow-white chorioretinal lesions. Multimodal imaging characteristics suggested that many of these lesions represent choroidal granulomas. Extensive systemic workup was only notable for borderline elevated Bartonella henselae IgG titers (1:128), however convalescent IgG titers were elevated at 38 days (1:512) supporting the diagnosis of Bartonella chorioretinitis. CONCLUSION: Ocular manifestations of Bartonella henselae infection are varied and may include choroidal granulomas. Multimodal imaging characteristics may help identify etiologies of infectious uveitis. Convalescent titers are important when evaluating patients with suspected Bartonellosis, especially patients with atypical presentations.


Asunto(s)
Enfermedad por Rasguño de Gato , Uveítis Posterior , Humanos , Enfermedad por Rasguño de Gato/complicaciones , Enfermedad por Rasguño de Gato/diagnóstico , Uveítis Posterior/diagnóstico , Uveítis Posterior/etiología , Imagen Multimodal
2.
Blood Adv ; 5(7): 1922-1932, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33821991

RESUMEN

Calreticulin (CALR) is mutated in the majority of JAK2/MPL-unmutated myeloproliferative neoplasms (MPNs). Mutant CALR (CALRdel52) exerts its effect by binding to the thrombopoietin receptor MPL to cause constitutive activation of JAK-STAT signaling. In this study, we performed an extensive mutagenesis screen of the CALR globular N-domain and revealed 2 motifs critical for CALRdel52 oncogenic activity: (1) the glycan-binding lectin motif and (2) the zinc-binding domain. Further analysis demonstrated that the zinc-binding domain was essential for formation of CALRdel52 multimers, which was a co-requisite for MPL binding. CALRdel52 variants incapable of binding zinc were unable to homomultimerize, form CALRdel52-MPL heteromeric complexes, or stimulate JAK-STAT signaling. Finally, treatment with zinc chelation disrupted CALRdel52-MPL complexes in hematopoietic cells in conjunction with preferential eradication of cells expressing CALRdel52 relative to cells expressing other MPN oncogenes. In addition, zinc chelators exhibited a therapeutic effect in preferentially impairing growth of CALRdel52-mutant erythroblasts relative to unmutated erythroblasts in primary cultures of MPN patients. Together, our data implicate zinc as an essential cofactor for CALRdel52 oncogenic activity by enabling CALRdel52 multimerization and interaction with MPL, and suggests that perturbation of intracellular zinc levels may represent a new approach to abrogate the oncogenic activity of CALRdel52 in the treatment of MPNs.


Asunto(s)
Calreticulina , Trastornos Mieloproliferativos , Calreticulina/genética , Humanos , Mutagénesis , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Receptores de Trombopoyetina/genética , Zinc
3.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33351783

RESUMEN

The cohesin complex plays an essential role in chromosome maintenance and transcriptional regulation. Recurrent somatic mutations in the cohesin complex are frequent genetic drivers in cancer, including myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Here, using genetic dependency screens of stromal antigen 2-mutant (STAG2-mutant) AML, we identified DNA damage repair and replication as genetic dependencies in cohesin-mutant cells. We demonstrated increased levels of DNA damage and sensitivity of cohesin-mutant cells to poly(ADP-ribose) polymerase (PARP) inhibition. We developed a mouse model of MDS in which Stag2 mutations arose as clonal secondary lesions in the background of clonal hematopoiesis driven by tet methylcytosine dioxygenase 2 (Tet2) mutations and demonstrated selective depletion of cohesin-mutant cells with PARP inhibition in vivo. Finally, we demonstrated a shift from STAG2- to STAG1-containing cohesin complexes in cohesin-mutant cells, which was associated with longer DNA loop extrusion, more intermixing of chromatin compartments, and increased interaction with PARP and replication protein A complex. Our findings inform the biology and therapeutic opportunities for cohesin-mutant malignancies.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Animales , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Mutantes , Ratones SCID , Ratones Transgénicos , Síndromes Mielodisplásicos/tratamiento farmacológico , Proteínas Nucleares/genética , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto , Cohesinas
4.
Elife ; 82019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31115337

RESUMEN

During their final maturation in the cytoplasm, pre-60S ribosomal particles are converted to translation-competent large ribosomal subunits. Here, we present the mechanism of peptidyltransferase centre (PTC) completion that explains how integration of the last ribosomal proteins is coupled to release of the nuclear export adaptor Nmd3. Single-particle cryo-EM reveals that eL40 recruitment stabilises helix 89 to form the uL16 binding site. The loading of uL16 unhooks helix 38 from Nmd3 to adopt its mature conformation. In turn, partial retraction of the L1 stalk is coupled to a conformational switch in Nmd3 that allows the uL16 P-site loop to fully accommodate into the PTC where it competes with Nmd3 for an overlapping binding site (base A2971). Our data reveal how the central functional site of the ribosome is sculpted and suggest how the formation of translation-competent 60S subunits is disrupted in leukaemia-associated ribosomopathies.


Asunto(s)
Peptidil Transferasas/metabolismo , ARN Ribosómico/metabolismo , Proteínas Ribosómicas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Saccharomyces cerevisiae/metabolismo , Microscopía por Crioelectrón , Peptidil Transferasas/ultraestructura , Subunidades Ribosómicas Grandes de Eucariotas/ultraestructura , Saccharomyces cerevisiae/ultraestructura
5.
Blood ; 131(7): 782-786, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29288169

RESUMEN

Mutations in calreticulin (CALR) are phenotypic drivers in the pathogenesis of myeloproliferative neoplasms. Mechanistic studies have demonstrated that mutant CALR binds to the thrombopoietin receptor MPL, and that the positive electrostatic charge of the mutant CALR C terminus is required for mutant CALR-mediated activation of JAK-STAT signaling. Here we demonstrate that although binding between mutant CALR and MPL is required for mutant CALR to transform hematopoietic cells; binding alone is insufficient for cytokine independent growth. We further show that the threshold of positive charge in the mutant CALR C terminus influences both binding of mutant CALR to MPL and activation of MPL signaling. We find that mutant CALR binds to the extracellular domain of MPL and that 3 tyrosine residues within the intracellular domain of MPL are required to activate signaling. With respect to mutant CALR function, we show that its lectin-dependent function is required for binding to MPL and for cytokine independent growth, whereas its chaperone and polypeptide-binding functionalities are dispensable. Together, our findings provide additional insights into the mechanism of the pathogenic mutant CALR-MPL interaction in myeloproliferative neoplasms.


Asunto(s)
Calreticulina/genética , Calreticulina/metabolismo , Trastornos Mieloproliferativos/genética , Dominios y Motivos de Interacción de Proteínas , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Calreticulina/química , Células Cultivadas , Células HEK293 , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Mutagénesis , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Mapas de Interacción de Proteínas , Receptores de Trombopoyetina/química , Transducción de Señal
6.
Cancer Discov ; 6(4): 368-81, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26951227

RESUMEN

UNLABELLED: Somatic mutations in calreticulin (CALR) are present in approximately 40% of patients with myeloproliferative neoplasms (MPN), but the mechanism by which mutant CALR is oncogenic remains unclear. Here, we demonstrate that expression of mutant CALR alone is sufficient to engender MPN in mice and recapitulates the disease phenotype of patients with CALR-mutant MPN. We further show that the thrombopoietin receptor MPL is required for mutant CALR-driven transformation through JAK-STAT pathway activation, thus rendering mutant CALR-transformed hematopoietic cells sensitive to JAK2 inhibition. Finally, we demonstrate that the oncogenicity of mutant CALR is dependent on the positive electrostatic charge of the C-terminus of the mutant protein, which is necessary for physical interaction between mutant CALR and MPL. Together, our findings elucidate a novel paradigm of cancer pathogenesis and reveal how CALR mutations induce MPN. SIGNIFICANCE: The mechanism by which CALR mutations induce MPN remains unknown. In this report, we show that the positive charge of the CALR mutant C-terminus is necessary to transform hematopoietic cells by enabling binding between mutant CALR and the thrombopoietin receptor MPL.


Asunto(s)
Calreticulina/genética , Transformación Celular Neoplásica/genética , Mutación , Dominios y Motivos de Interacción de Proteínas/genética , Receptores de Trombopoyetina/genética , Animales , Secuencia de Bases , Trasplante de Médula Ósea , Calreticulina/química , Calreticulina/metabolismo , Línea Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Femenino , Mutación del Sistema de Lectura , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Ratones , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Fenotipo , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Trombopoyetina/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Colapso de la Estructura
7.
Nat Med ; 22(3): 288-97, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26878232

RESUMEN

Impaired erythropoiesis in the deletion 5q (del(5q)) subtype of myelodysplastic syndrome (MDS) has been linked to heterozygous deletion of RPS14, which encodes the ribosomal protein small subunit 14. We generated mice with conditional inactivation of Rps14 and demonstrated an erythroid differentiation defect that is dependent on the tumor suppressor protein p53 (encoded by Trp53 in mice) and is characterized by apoptosis at the transition from polychromatic to orthochromatic erythroblasts. This defect resulted in age-dependent progressive anemia, megakaryocyte dysplasia and loss of hematopoietic stem cell (HSC) quiescence. As assessed by quantitative proteomics, mutant erythroblasts expressed higher levels of proteins involved in innate immune signaling, notably the heterodimeric S100 calcium-binding proteins S100a8 and S100a9. S100a8--whose expression was increased in mutant erythroblasts, monocytes and macrophages--is functionally involved in the erythroid defect caused by the Rps14 deletion, as addition of recombinant S100a8 was sufficient to induce a differentiation defect in wild-type erythroid cells, and genetic inactivation of S100a8 expression rescued the erythroid differentiation defect of Rps14-haploinsufficient HSCs. Our data link Rps14 haploinsufficiency in del(5q) MDS to activation of the innate immune system and induction of S100A8-S100A9 expression, leading to a p53-dependent erythroid differentiation defect.


Asunto(s)
Anemia/genética , Calgranulina A/genética , Calgranulina B/genética , Eritropoyesis/genética , Haploinsuficiencia/genética , Síndromes Mielodisplásicos/genética , Proteínas Ribosómicas/genética , Anemia/inmunología , Animales , Western Blotting , Médula Ósea/patología , Calgranulina A/metabolismo , Citocinas/inmunología , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Eritropoyesis/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Madre Hematopoyéticas , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inmunohistoquímica , Hibridación Fluorescente in Situ , Técnicas In Vitro , Espectrometría de Masas , Megacariocitos , Ratones , Ratones Noqueados , Microscopía Confocal , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/patología , Proteína p53 Supresora de Tumor/genética
9.
Cell Rep ; 13(11): 2345-2352, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26686625

RESUMEN

JAK2V617F is the most common oncogenic lesion in patients with myeloproliferative neoplasms (MPNs). Despite the ability of JAK2V617F to instigate DNA damage in vitro, MPNs are nevertheless characterized by genomic stability. In this study, we address this paradox by identifying the DNA helicase RECQL5 as a suppressor of genomic instability in MPNs. We report increased RECQL5 expression in JAK2V617F-expressing cells and demonstrate that RECQL5 is required to counteract JAK2V617F-induced replication stress. Moreover, RECQL5 depletion sensitizes JAK2V617F mutant cells to hydroxyurea (HU), a pharmacological inducer of replication stress and the most common treatment for MPNs. Using single-fiber chromosome combing, we show that RECQL5 depletion in JAK2V617F mutant cells impairs replication dynamics following HU treatment, resulting in increased double-stranded breaks and apoptosis. Cumulatively, these findings identify RECQL5 as a critical regulator of genome stability in MPNs and demonstrate that replication stress-associated cytotoxicity can be amplified specifically in JAK2V617F mutant cells through RECQL5-targeted synthetic lethality.


Asunto(s)
Janus Quinasa 2/metabolismo , RecQ Helicasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Furanos/farmacología , Técnicas de Sustitución del Gen , Inestabilidad Genómica/efectos de los fármacos , Humanos , Hidroxiurea/toxicidad , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Neoplasias/metabolismo , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piridinas/farmacología , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , RecQ Helicasas/genética , Transducción de Señal/efectos de los fármacos
10.
Nat Commun ; 6: 6691, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25849990

RESUMEN

Clonal proliferation in myeloproliferative neoplasms (MPN) is driven by somatic mutations in JAK2, CALR or MPL, but the contribution of inherited factors is poorly characterized. Using a three-stage genome-wide association study of 3,437 MPN cases and 10,083 controls, we identify two SNPs with genome-wide significance in JAK2(V617F)-negative MPN: rs12339666 (JAK2; meta-analysis P=1.27 × 10(-10)) and rs2201862 (MECOM; meta-analysis P=1.96 × 10(-9)). Two additional SNPs, rs2736100 (TERT) and rs9376092 (HBS1L/MYB), achieve genome-wide significance when including JAK2(V617F)-positive cases. rs9376092 has a stronger effect in JAK2(V617F)-negative cases with CALR and/or MPL mutations (Breslow-Day P=4.5 × 10(-7)), whereas in JAK2(V617F)-positive cases rs9376092 associates with essential thrombocythemia (ET) rather than polycythemia vera (allelic χ(2) P=7.3 × 10(-7)). Reduced MYB expression, previously linked to development of an ET-like disease in model systems, associates with rs9376092 in normal myeloid cells. These findings demonstrate that multiple germline variants predispose to MPN and link constitutional differences in MYB expression to disease phenotype.


Asunto(s)
Policitemia Vera/genética , Trombocitemia Esencial/genética , Adulto , Anciano , Alelos , Calreticulina/genética , Estudios de Casos y Controles , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Femenino , Proteínas de Unión al GTP/genética , Frecuencia de los Genes , Genes myb/genética , Predisposición Genética a la Enfermedad , Variación Genética , Genotipo , Proteínas HSP70 de Choque Térmico/genética , Humanos , Janus Quinasa 2/genética , Proteína del Locus del Complejo MDS1 y EV11 , Masculino , Persona de Mediana Edad , Mutación , Trastornos Mieloproliferativos/genética , Factores de Elongación de Péptidos/genética , Polimorfismo de Nucleótido Simple , Proto-Oncogenes/genética , Receptores de Trombopoyetina/genética , Telomerasa/genética , Factores de Transcripción/genética
11.
Blood ; 125(2): 327-35, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25281607

RESUMEN

Signaling mutations (eg, JAK2V617F) and mutations in genes involved in epigenetic regulation (eg, TET2) are the most common cooccurring classes of mutations in myeloproliferative neoplasms (MPNs). Clinical correlative studies have demonstrated that TET2 mutations are enriched in more advanced phases of MPNs such as myelofibrosis and leukemic transformation, suggesting that they may cooperate with JAK2V617F to promote disease progression. To dissect the effects of concomitant Jak2V617F expression and Tet2 loss within distinct hematopoietic compartments in vivo, we generated Jak2V617F/Tet2 compound mutant genetic mice. We found that the combination of Jak2V617F expression and Tet2 loss resulted in a more florid MPN phenotype than that seen with either allele alone. Concordant with this, we found that Tet2 deletion conferred a strong functional competitive advantage to Jak2V617F-mutant hematopoietic stem cells (HSCs). Transcriptional profiling revealed that both Jak2V617F expression and Tet2 loss were associated with distinct and nonoverlapping gene expression signatures within the HSC compartment. In aggregate, our findings indicate that Tet2 loss drives clonal dominance in HSCs, and Jak2V617F expression causes expansion of downstream precursor cell populations, resulting in disease progression through combinatorial effects. This work provides insight into the functional consequences of JAK2V617F-TET2 comutation in MPNs, particularly as it pertains to HSCs.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/patología , Janus Quinasa 2/genética , Trastornos Mieloproliferativos/genética , Proteínas Proto-Oncogénicas/genética , Animales , Dioxigenasas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Mutación
12.
Proc Natl Acad Sci U S A ; 111(42): 15190-5, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288776

RESUMEN

Cancers result from the accumulation of genetic lesions, but the cellular consequences of driver mutations remain unclear, especially during the earliest stages of malignancy. The V617F mutation in the JAK2 non-receptor tyrosine kinase (JAK2V617F) is present as an early somatic event in most patients with myeloproliferative neoplasms (MPNs), and the study of these chronic myeloid malignancies provides an experimentally tractable approach to understanding early tumorigenesis. Introduction of exogenous JAK2V617F impairs replication fork progression and is associated with activation of the intra-S checkpoint, with both effects mediated by phosphatidylinositide 3-kinase (PI3K) signaling. Analysis of clonally derived JAK2V617F-positive erythroblasts from MPN patients also demonstrated impaired replication fork progression accompanied by increased levels of replication protein A (RPA)-containing foci. However, the associated intra-S checkpoint response was impaired in erythroblasts from polycythemia vera (PV) patients, but not in those from essential thrombocythemia (ET) patients. Moreover, inhibition of p53 in PV erythroblasts resulted in more gamma-H2Ax (γ-H2Ax)-marked double-stranded breaks compared with in like-treated ET erythroblasts, suggesting the defective intra-S checkpoint function seen in PV increases DNA damage in the context of attenuated p53 signaling. These results demonstrate oncogene-induced impairment of replication fork progression in primary cells from MPN patients, reveal unexpected disease-restricted differences in activation of the intra-S checkpoint, and have potential implications for the clonal evolution of malignancies.


Asunto(s)
Puntos de Control del Ciclo Celular , Replicación del ADN , Janus Quinasa 2/fisiología , Fase S , Apoptosis , División Celular , Cromosomas/metabolismo , Cromosomas/ultraestructura , Daño del ADN , Reparación del ADN , Diploidia , Fibroblastos/metabolismo , Genotipo , Enfermedades Hematológicas/genética , Humanos , Janus Quinasa 2/genética , Leucemia/metabolismo , Leucemia/patología , Microscopía Fluorescente , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Fosforilación , ARN Interferente Pequeño/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
13.
Int J Oncol ; 45(5): 1997-2004, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25175815

RESUMEN

The Notch1 signaling pathway is essential for hematopoietic development. However, the effects of postnatal activation of Notch1 signaling on hematopoietic system is not yet fully understood. We previously generated ZEG­IC­Notch1 transgenic mice that have a floxed ß­geo/stop signal between a CMV promoter and intracellular domain of Notch1 (IC­Notch1). Constitutively active IC­Notch1 is silent until the introduction of Cre recombinase. In this study, endothelial/hematopoietic specific expression of IC­Notch1 in double transgenic ZEG­IC­Notch1/Tie2­Cre embryos induced embryonic lethality at E9.5 with defects in vascular system but not in hematopoietic system. Inducible IC­Notch1 expression in adult mice was achieved by using tetracycline regulated Cre system. The ZEG­IC­Notch1/Tie2­tTA/tet­O­Cre triple transgenic mice survived embryonic development when maintained on tetracycline. Post­natal withdrawal of tetracycline induced expression of IC­Notch1 transgene in hematopoietic cells of adult mice. The triple transgenic mice displayed extensive T­cell infiltration in multiple organs and T­cell malignancy of lymph nodes. In addition, the protein levels of p53 and alternative reading frame (ARF) were decreased in lymphoma­like neoplasms from the triple transgenic mice while their mRNA expression remained unchanged, suggesting that IC­Notch1 might repress ARF­p53 pathway by a post­transcriptional mechanism. This study demonstrated that activation of constitutive Notch1 signaling after embryonic development alters adult hematopoiesis and induces T­cell malignancy.


Asunto(s)
Transformación Celular Neoplásica/genética , Ganglios Linfáticos/patología , Receptor Notch1/genética , Linfocitos T/inmunología , Animales , Transformación Celular Neoplásica/patología , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Humanos , Integrasas , Ganglios Linfáticos/crecimiento & desarrollo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptor Notch1/biosíntesis , Transducción de Señal/genética , Linfocitos T/patología
14.
Blood ; 123(20): 3139-51, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24692758

RESUMEN

Genomic regions of acquired uniparental disomy (UPD) are common in malignancy and frequently harbor mutated oncogenes. Homozygosity for such gain-of-function mutations is thought to modulate tumor phenotype, but direct evidence has been elusive. Polycythemia vera (PV) and essential thrombocythemia (ET), 2 subtypes of myeloproliferative neoplasms, are associated with an identical acquired JAK2V617F mutation but the mechanisms responsible for distinct clinical phenotypes remain unclear. We provide direct genetic evidence and demonstrate that homozygosity for human JAK2V617F in knock-in mice results in a striking phenotypic switch from an ET-like to PV-like phenotype. The resultant erythrocytosis is driven by increased numbers of early erythroid progenitors and enhanced erythroblast proliferation, whereas reduced platelet numbers are associated with impaired platelet survival. JAK2V617F-homozygous mice developed a severe hematopoietic stem cell defect, suggesting that additional lesions are needed to sustain clonal expansion. Together, our results indicate that UPD for 9p plays a causal role in the PV phenotype in patients as a consequence of JAK2V617F homozygosity. The generation of a JAK2V617F allelic series of mice with a dose-dependent effect on hematopoiesis provides a powerful model for studying the consequences of mutant JAK2 homozygosity.


Asunto(s)
Janus Quinasa 2/genética , Mutación , Policitemia Vera/genética , Trombocitemia Esencial/genética , Animales , Plaquetas/metabolismo , Plaquetas/patología , Eritroblastos/metabolismo , Eritroblastos/patología , Femenino , Técnicas de Sustitución del Gen , Homocigoto , Humanos , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Policitemia Vera/patología , Trombocitemia Esencial/patología , Disomía Uniparental/genética , Disomía Uniparental/patología
15.
PLoS One ; 9(2): e89649, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586935

RESUMEN

The noncluster homeobox gene HOX11/TLX1 (TLX1) is detected at the breakpoint of the t(10;14)(q24;q11) chromosome translocation in patients with T cell acute lymphoblastic leukemia (T-ALL). This translocation results in the inappropriate expression of TLX1 in T cells. The oncogenic potential of TLX1 was demonstrated in IgHµ-TLX1(Tg) mice which develop mature B cell lymphoma after a long latency period, suggesting the requirement of additional mutations to initiate malignancy. To determine whether dysregulation of genes involved in the DNA damage response contributed to tumor progression, we crossed IgHµ-TLX1(Tg) mice with mice deficient in the DNA repair enzyme DNA-PK (Prkdc(Scid/Scid) mice). IgHµ-TLX1(Tg)Prkdc(Scid/Scid) mice developed T-ALL and acute myeloid leukemia (AML) with reduced latency relative to control Prkdc(Scid/Scid) mice. Further analysis of thymi from premalignant mice revealed greater thymic cellularity concomitant with increased thymocyte proliferation and decreased apoptotic index. Moreover, premalignant and malignant thymocytes exhibited impaired spindle checkpoint function, in association with aneuploid karyotypes. Gene expression profiling of premalignant IgHµ-TLX1(Tg)Prkdc(Scid/Scid) thymocytes revealed dysregulated expression of cell cycle, apoptotic and mitotic spindle checkpoint genes in double negative 2 (DN2) and DN3 stage thymocytes. Collectively, these findings reveal a novel synergy between TLX1 and impaired DNA repair pathway in leukemogenesis.


Asunto(s)
Transformación Celular Neoplásica , Proteína Quinasa Activada por ADN/deficiencia , Proteínas de Unión al ADN/deficiencia , Proteínas de Homeodominio/biosíntesis , Proteínas Nucleares/deficiencia , Animales , Reparación del ADN , Humanos , Leucemia Mieloide Aguda/etiología , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Neoplasias del Timo/química , Transcriptoma
16.
Hematology Am Soc Hematol Educ Program ; 2014(1): 268-76, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25696866

RESUMEN

A decade on from the discovery of the JAK2V617F mutation in the majority of patients with myeloproliferative neoplasms (MPNs), JAK2V617F is now firmly installed in the hematology curriculum of medical students and the diagnostic-testing algorithm of clinicians. Furthermore, the oral JAK1/JAK2 inhibitor ruxolitinib, rationally designed to target activated JAK2 signaling in MPN, has been approved by the Food and Drug Administration (FDA) of the United States for the past 3 years for the treatment of intermediate- and advanced-phase myelofibrosis. Notwithstanding this, JAK2V617F continues to stimulate the MPN research community and novel insights into understanding the mechanisms by which JAK2V617F contributes to the pathogenesis of MPN are continually emerging. In this chapter, we focus on recent advances in 4 main areas: (1) the molecular processes coopted by JAK2V617F to induce MPN, (2) the role that JAK2V617F plays in phenotypic diversity in MPN, (3) the functional impact of JAK2V617F on hematopoietic stem cells, and (4) therapeutic strategies to target JAK2V617F. Although great strides have been made, significant deficits still exist in our understanding of the precise mechanisms by which JAK2V617F-mutant hematopoietic stem cells emerge and persist to engender clonal hematopoiesis in MPN and in developing strategies to preferentially target the JAK2V617F-mutant clone therapeutically. Critically, although myelofibrosis remains arguably the greatest clinical challenge in JAK2V617F-mediated MPN, the current understanding of myelofibrosis-specific disease biology remains quite rudimentary. Therefore, many important biological questions pertaining to JAK2V617F will continue to engage and challenge the MPN research community in the coming decade.


Asunto(s)
Janus Quinasa 2/genética , Mutación/genética , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Células Madre Hematopoyéticas/citología , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Terapia Molecular Dirigida , Trastornos Mieloproliferativos/terapia , Fenotipo
17.
Haematologica ; 98(5): 718-21, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633544

RESUMEN

Subclones homozygous for JAK2V617F are more common and larger in patients with polycythemia vera compared to essential thrombocythemia, but their role in determining phenotype remains unclear. We genotyped 4564 erythroid colonies from 59 patients with polycythemia vera or essential thrombocythemia to investigate whether the proportion of JAK2V617F -homozygous precursors, compared to heterozygous precursors, is associated with clinical or demographic features. In polycythemia vera, a higher proportion of homozygous-mutant precursors was associated with more extreme blood counts at diagnosis, consistent with a causal role for homozygosity in polycythemia vera pathogenesis. Larger numbers of homozygous-mutant colonies were associated with older age, and with male gender in polycythemia vera but female gender in essential thrombocythemia. These results suggest that age promotes development or expansion of homozygous-mutant clones and that gender modulates the phenotypic consequences of JAK2V617F homozygosity, thus providing a potential explanation for the long-standing observations of a preponderance of men with polycythemia vera but of women with essential thrombocythemia.


Asunto(s)
Homocigoto , Janus Quinasa 2/genética , Mutación , Policitemia Vera/sangre , Policitemia Vera/genética , Trombocitemia Esencial/sangre , Trombocitemia Esencial/genética , Factores de Edad , Índices de Eritrocitos , Femenino , Humanos , Recuento de Leucocitos , Masculino , Modelos Genéticos , Recuento de Plaquetas , Factores Sexuales
18.
Blood ; 120(13): 2704-7, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22898600

RESUMEN

Subclones homozygous for JAK2V617F are more common in polycythemia vera (PV) than essential thrombocythemia (ET), but their prevalence and significance remain unclear. The JAK2 mutation status of 6495 BFU-E, grown in low erythropoietin conditions, was determined in 77 patients with PV or ET. Homozygous-mutant colonies were common in patients with JAK2V617F-positive PV and were surprisingly prevalent in JAK2V617F-positive ET and JAK2 exon 12-mutated PV. Using microsatellite PCR to map loss-of-heterozygosity breakpoints within individual colonies, we demonstrate that recurrent acquisition of JAK2V617F homozygosity occurs frequently in both PV and ET. PV was distinguished from ET by expansion of a dominant homozygous subclone, the selective advantage of which is likely to reflect additional genetic or epigenetic lesions. Our results suggest a model in which development of a dominant JAK2V617F-homzygous subclone drives erythrocytosis in many PV patients, with alternative mechanisms operating in those with small or undetectable homozygous-mutant clones.


Asunto(s)
Homocigoto , Janus Quinasa 2/genética , Mutación/genética , Policitemia Vera/genética , Policitemia/patología , Trombocitemia Esencial/genética , Genes Dominantes , Heterocigoto , Humanos , Repeticiones de Microsatélite , Policitemia/genética , Reacción en Cadena de la Polimerasa , Pronóstico , Recurrencia
19.
Immunity ; 36(4): 529-41, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22520846

RESUMEN

Genetic alterations affecting members of the Janus kinase (JAK) family have been discovered in a wide array of cancers and are particularly prominent in hematological malignancies. In this review, we focus on the role of such lesions in both myeloid and lymphoid tumors. Oncogenic JAK molecules can activate a myriad of canonical downstream signaling pathways as well as directly interact with chromatin in noncanonical processes, the interplay of which results in a plethora of diverse biological consequences. Deciphering these complexities is shedding unexpected light on fundamental cellular mechanisms and will also be important for improved diagnosis, identification of new therapeutic targets, and the development of stratified approaches to therapy.


Asunto(s)
Quinasas Janus/genética , Quinasas Janus/metabolismo , Leucemia/genética , Linfoma/genética , Animales , Cromatina/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Leucemia/metabolismo , Leucemia/patología , Linfoma/metabolismo , Linfoma/patología , Ratones , Mutación , Transducción de Señal/genética
20.
Dis Model Mech ; 4(3): 311-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21558064

RESUMEN

In 2005, several groups identified a single gain-of-function point mutation in the JAK2 kinase that was present in the majority of patients with myeloproliferative neoplasms (MPNs). Since this discovery, much effort has been dedicated to understanding the molecular consequences of the JAK2V617F mutation in the haematopoietic system. Three waves of mouse models have been produced recently (bone marrow transplantation, transgenic and targeted knock-in), which have facilitated the understanding of the molecular pathogenesis of JAK2V617F-positive MPNs, providing potential platforms for designing and validating novel therapies in humans. This Commentary briefly summarises the first two types of mouse models and then focuses on the more recently generated knock-in models.


Asunto(s)
Neoplasias de la Médula Ósea/enzimología , Modelos Animales de Enfermedad , Quinasas Janus/metabolismo , Trastornos Mieloproliferativos/enzimología , Animales , Neoplasias de la Médula Ósea/patología , Trasplante de Médula Ósea , Humanos , Ratones , Mutación/genética , Trastornos Mieloproliferativos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA