Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 136(18): 2051-2064, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-32726410

RESUMEN

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) that leads to progressive bone marrow (BM) fibrosis. Although the cellular mutations involved in the pathogenesis of PMF have been extensively investigated, the sequential events that drive stromal activation and fibrosis by hematopoietic-stromal cross-talk remain elusive. Using an unbiased approach and validation in patients with MPN, we determined that the differential spatial expression of the chemokine CXCL4/platelet factor-4 marks the progression of fibrosis. We show that the absence of hematopoietic CXCL4 ameliorates the MPN phenotype, reduces stromal cell activation and BM fibrosis, and decreases the activation of profibrotic pathways in megakaryocytes, inflammation in fibrosis-driving cells, and JAK/STAT activation in both megakaryocytes and stromal cells in 3 murine PMF models. Our data indicate that higher CXCL4 expression in MPN has profibrotic effects and is a mediator of the characteristic inflammation. Therefore, targeting CXCL4 might be a promising strategy to reduce inflammation in PMF.


Asunto(s)
Médula Ósea/patología , Fibrosis/patología , Inflamación/patología , Trastornos Mieloproliferativos/complicaciones , Factor Plaquetario 4/metabolismo , Mielofibrosis Primaria/patología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Fibrosis/etiología , Fibrosis/inmunología , Fibrosis/metabolismo , Humanos , Inflamación/etiología , Inflamación/inmunología , Inflamación/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Megacariocitos , Ratones , Ratones Noqueados , Mutación , Factor Plaquetario 4/genética , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/inmunología , Mielofibrosis Primaria/metabolismo
2.
Ann Hematol ; 100(6): 1463-1471, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33903952

RESUMEN

Myelodysplastic syndrome (MDS) with isolated deletion of chromosome 5q (MDS del5q) is a distinct subtype of MDS with quite favorable prognosis and excellent response to treatment with lenalidomide. Still, a relevant percentage of patients do not respond to lenalidomide and even experience progression to acute myeloid leukemia (AML). In this study, we aimed to investigate whether global DNA methylation patterns could predict response to lenalidomide. Genome-wide DNA methylation analysis using Illumina 450k methylation arrays was performed on n=51 patients with MDS del5q who were uniformly treated with lenalidomide in a prospective multicenter trial of the German MDS study group. To study potential direct effects of lenalidomide on DNA methylation, 17 paired samples pre- and post-treatment were analyzed. Our results revealed no relevant effect of lenalidomide on methylation status. Furthermore, methylation patterns prior to therapy could not predict lenalidomide response. However, methylation clustering identified a group of patients with a trend towards inferior overall survival. These patients showed hypermethylation of several interesting target genes, including genes of relevant signaling pathways, potentially indicating the evaluation of novel therapeutic targets.


Asunto(s)
Antineoplásicos/uso terapéutico , Metilación de ADN/efectos de los fármacos , Lenalidomida/uso terapéutico , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Femenino , Humanos , Lenalidomida/farmacología , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
3.
Eur J Haematol ; 106(4): 520-528, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33460496

RESUMEN

OBJECTIVES: Myeloproliferative neoplasms (MPN) comprising polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) follow a bi-phasic course of disease with fibrotic and/or blastic progression. At presentation in the chronic phase, currently there are only insufficient tools to predict the risk of progression in individual cases. METHODS: In this study, chronic phase MPN (16 PMF, 11 PV, and 11 MPN unclassified) with blastic transformation during course of disease (n = 38, median follow-up 5.3 years) were analyzed by high-throughput sequencing. MPN cases with a comparable follow-up period and without evidence of blast increase served as control (n = 63, median follow-up 5.8 years). RESULTS: Frequent ARCH/CHIP-associated mutations (TET2, ASXL1, DNMT3A) found at presentation were not significantly associated with blastic transformation. By contrast, mutations of SRSF2, U2AF1, and IDH1/2 at first presentation were frequently observed in the progression cohort (13/38, 34.2%) and were completely missing in the control group without blast transformation during follow-up (P = .0007 for SRSF2; P = .0063 for U2AF1 and IDH1/2). CONCLUSION: Unlike frequent ARCH/CHIP alterations (TET2, ASXL1, DNMT3A), mutations in SRSF2, IDH1/2, and U2AF1 when manifest already at first presentation provide an independent risk factor for rapid blast transformation of MPN.


Asunto(s)
Epigénesis Genética , Genes Modificadores , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Oncogenes , Factores de Empalme de ARN/genética , Activación Transcripcional , Anciano , Crisis Blástica , Preescolar , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Persona de Mediana Edad , Factores de Empalme Serina-Arginina , Factor de Empalme U2AF/genética
4.
PLoS Pathog ; 13(9): e1006639, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28938025

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the infectious cause of the highly vascularized tumor Kaposi's sarcoma (KS), which is characterized by proliferating spindle cells of endothelial origin, extensive neo-angiogenesis and inflammatory infiltrates. The KSHV K15 protein contributes to the angiogenic and invasive properties of KSHV-infected endothelial cells. Here, we asked whether K15 could also play a role in KSHV lytic replication. Deletion of the K15 gene from the viral genome or its depletion by siRNA lead to reduced virus reactivation, as evidenced by the decreased expression levels of KSHV lytic proteins RTA, K-bZIP, ORF 45 and K8.1 as well as reduced release of infectious virus. Similar results were found for a K1 deletion virus. Deleting either K15 or K1 from the viral genome also compromised the ability of KSHV to activate PLCγ1, Erk1/2 and Akt1. In infected primary lymphatic endothelial (LEC-rKSHV) cells, which have previously been shown to spontaneously display a viral lytic transcription pattern, transfection of siRNA against K15, but not K1, abolished viral lytic replication as well as KSHV-induced spindle cell formation. Using a newly generated monoclonal antibody to K15, we found an abundant K15 protein expression in KS tumor biopsies obtained from HIV positive patients, emphasizing the physiological relevance of our findings. Finally, we used a dominant negative inhibitor of the K15-PLCγ1 interaction to establish proof of principle that pharmacological intervention with K15-dependent pathways may represent a novel approach to block KSHV reactivation and thereby its pathogenesis.


Asunto(s)
Herpesvirus Humano 8/fisiología , Sarcoma de Kaposi/virología , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Western Blotting , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Sarcoma de Kaposi/metabolismo , Activación Viral/fisiología , Latencia del Virus/fisiología
5.
Haematologica ; 103(10): 1593-1603, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30076180

RESUMEN

Pathological erythropoiesis with consequent anemia is a leading cause of symptomatic morbidity in internal medicine. The etiologies of anemia are complex and include reactive as well as neoplastic conditions. Clonal expansion of erythroid cells in the bone marrow may result in peripheral erythrocytosis and polycythemia but can also result in anemia when clonal cells are dysplastic and have a maturation arrest that leads to apoptosis and hinders migration, a constellation typically seen in the myelodysplastic syndromes. Rarely, clonal expansion of immature erythroid blasts results in a clinical picture resembling erythroid leukemia. Although several mechanisms underlying normal and abnormal erythropoiesis and the pathogenesis of related disorders have been deciphered in recent years, little is known about specific markers and targets through which prognosis and therapy could be improved in anemic or polycythemic patients. In order to discuss new markers, targets and novel therapeutic approaches in erythroid disorders and the related pathologies, a workshop was organized in Vienna in April 2017. The outcomes of this workshop are summarized in this review, which includes a discussion of new diagnostic and prognostic markers, the updated WHO classification, and an overview of new drugs used to stimulate or to interfere with erythropoiesis in various neoplastic and reactive conditions. The use and usefulness of established and novel erythropoiesis-stimulating agents for various indications, including myelodysplastic syndromes and other neoplasms, are also discussed.


Asunto(s)
Anemia/metabolismo , Células Eritroides/metabolismo , Eritropoyesis , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Policitemia/metabolismo , Adulto , Anemia/patología , Células Eritroides/patología , Humanos , Neoplasias/patología , Policitemia/patología
6.
Ann Hematol ; 96(6): 887-894, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28374162

RESUMEN

Transfusion-dependent patients with low- or intermediate-1-risk myelodysplastic syndrome, <5% bone marrow (BM) blasts and isolated 5q-deletion received lenalidomide within the German MDS study group phase-II clinical trial LE-MON-5 (EudraCT:2008-001866-10) of the University of Duesseldorf, Germany. Cytogenetic monitoring was performed by chromosome banding analyses (CBA) of BM cells and fluorescence in situ hybridization (FISH) analyses of peripheral blood (PB) mononuclear CD34+ cells using extended probe panels. Out of 144 patients screened for study enrollment, 24% failed to meet inclusion criteria due to cytogenetic findings. Eighty-seven patients were followed with a median observation time of 30 months. Cytogenetic response detected by FISH and CBA in 74 and 66% of patients, respectively, was predictive for hematologic response as well as of high prognostic relevance. After 2 years, AML rate was 8% for all patients. Karyotype evolution was detected in 21 (FISH)-34% (CBA) of patients associated with significantly shorter AML-free survival. Disease progression was first detectable on the cytogenetic level on average 5-6 months before recurrence of transfusion dependence. Our data show for the first time in a prospective setting that a cytogenetic monitoring from the PB helps to early identify treatment failure and progressive disease in lenalidomide-treated patients to improve clinical management. TRIAL REGISTRATION: EudraCT:2008-001866-10.


Asunto(s)
Síndromes Mielodisplásicos/tratamiento farmacológico , Talidomida/análogos & derivados , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/uso terapéutico , Antígenos CD34/sangre , Bandeo Cromosómico , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Supervivencia sin Enfermedad , Femenino , Alemania , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Lenalidomida , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/genética , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/genética , Pronóstico , Talidomida/uso terapéutico , Insuficiencia del Tratamiento
7.
Ann Hematol ; 95(5): 783-91, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26891877

RESUMEN

The novel FMS-like tyrosine kinase 3 (FLT3)-N676K point mutation within the FLT3 kinase domain-1 was recently identified in 6 % of de novo acute myeloid leukemia (AML) patients with inv(16). Because FLT3-N676K was encountered almost exclusively in inv(16) AML, we investigated the transforming potential of FLT3-N676K, the cooperation between FLT3-N676K and core binding factor ß-smooth muscle myosin heavy chain (CBFß-SMMHC) (encoded by the inv(16) chimeric gene CBFB-MYH11) in inducing acute leukemia, and tested the sensitivity of FLT3-N676K-positive leukemic cells to FLT3 inhibitors. Retroviral expression of FLT3-N676K in myeloid 32D cells induced AML in syngeneic C3H/HeJ mice (n = 11/13, median latency 58 days), with a transforming activity similar to FLT3-internal tandem duplication (ITD) (n = 8/8), FLT3-TKD D835Y (n = 8/9), and FLT3-ITD-N676K (n = 9/9) mutations. Three out of 14 (21.4 %) C57BL/6J mice transplanted with FLT3-N676K-transduced primary hematopoietic progenitor cells developed acute leukemia (latency of 68, 77, and 273 days), while no hematological malignancy was observed in the control groups including FLT3-ITD. Moreover, co-expression of FLT3-N676K/CBFß-SMMHC did not promote acute leukemia in three independent experiments (n = 16). In comparison with FLT3-ITD, FLT3-N676K induced much higher activation of FLT3 and tended to trigger stronger phosphorylation of MAPK and AKT. Importantly, leukemic cells carrying the FLT3-N676K mutant in the absence of an ITD mutation were highly sensitive to FLT3 inhibitors AC220 and crenolanib, and crenolanib even retained activity against the AC220-resistant FLT3-ITD-N676K mutant. Taken together, the FLT3-N676K mutant is potent to transform murine hematopoietic stem/progenitor cells in vivo. This is the first report of acute leukemia induced by an activating FLT3 mutation in C57BL/6J mice. Moreover, further experiments investigating molecular mechanisms for leukemogenesis induced by FLT3-N676K mutation and clinical evaluation of FLT3 inhibitors in FLT3-N676K-positive AML seem warranted.


Asunto(s)
Leucemia Experimental/genética , Mutación Missense , Mutación Puntual , Tirosina Quinasa 3 Similar a fms/genética , Sustitución de Aminoácidos , Animales , Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Benzotiazoles/uso terapéutico , Trasplante de Médula Ósea , Transformación Celular Neoplásica/genética , Regulación Leucémica de la Expresión Génica , Predisposición Genética a la Enfermedad , Vectores Genéticos , Humanos , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/enzimología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Células Madre Neoplásicas/trasplante , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/fisiología , Compuestos de Fenilurea/uso terapéutico , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional/genética , Quimera por Radiación , Retroviridae , Secuencias Repetidas en Tándem , Transgenes , Ensayo de Tumor de Célula Madre , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/fisiología
8.
PLoS Pathog ; 9(11): e1003737, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244164

RESUMEN

Kaposi's sarcoma (KS) is a mesenchymal tumour, which is caused by Kaposi's sarcoma herpesvirus (KSHV) and develops under inflammatory conditions. KSHV-infected endothelial spindle cells, the neoplastic cells in KS, show increased invasiveness, attributed to the elevated expression of metalloproteinases (MMPs) and cyclooxygenase-2 (COX-2). The majority of these spindle cells harbour latent KSHV genomes, while a minority undergoes lytic reactivation with subsequent production of new virions and viral or cellular chemo- and cytokines, which may promote tumour invasion and dissemination. In order to better understand KSHV pathogenesis, we investigated cellular mechanisms underlying the lytic reactivation of KSHV. Using a combination of small molecule library screening and siRNA silencing we found a STE20 kinase family member, MAP4K4, to be involved in KSHV reactivation from latency and to contribute to the invasive phenotype of KSHV-infected endothelial cells by regulating COX-2, MMP-7, and MMP-13 expression. This kinase is also highly expressed in KS spindle cells in vivo. These findings suggest that MAP4K4, a known mediator of inflammation, is involved in KS aetiology by regulating KSHV lytic reactivation, expression of MMPs and COX-2, and, thereby modulating invasiveness of KSHV-infected endothelial cells.


Asunto(s)
Células Endoteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Herpesvirus Humano 8/fisiología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Sarcoma de Kaposi/metabolismo , Activación Viral/fisiología , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Células Endoteliales/patología , Células Endoteliales/virología , Femenino , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Metaloproteinasa 13 de la Matriz/biosíntesis , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/patología
9.
Biol Blood Marrow Transplant ; 20(6): 812-5, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24589549

RESUMEN

We correlate regression of bone marrow fibrosis (BMF) on day 30 and 100 after dose- reduced allogeneic stem cell transplantation (allo-SCT) in 57 patients with primary or post-essential thrombocythemia/polycythemia vera myelofibrosis with graft function and survival. The distribution of International Prognostic Scoring System (IPSS) risk score categories was 1 patient with low risk, 5 patients with intermediate-1 risk, 18 patients with intermediate-2 risk, and 33 patients with high risk. Before allo-SCT, 41 patients (72%) were classified as XXX [myclofibrosis (MF)]-3 and 16 (28%) were classified as MF-2 according to the World Health Organization criteria. At postengraftment day +30 (±10 days), 21% of the patients had near-complete or complete regression of BMF (MF-0/-1), and on day +100 (±20 days), 54% were MF-0/-1. The 5-year overall survival rate at day +100 was 96% in patients with MF-0/-1 and 57% for those with MF-2/-3 (P = .04). There was no difference in BMF regression at day +100 between IPSS high-risk and low/intermediate-risk patients. Complete donor cell chimerism at day +100 was seen in 81% of patients with MF-0/-1 and in 31% of those with MF-2/-3. Patients with MF-2/-3 at day +100 were more likely to be transfusion-dependent for either RBCs (P = .014) or platelets (P = .018). Rapid BMF regression after reduced-intensity conditioning allo-SCT resulted in a favorable survival independent of IPSS risk score at transplantation.


Asunto(s)
Médula Ósea/patología , Trasplante de Células Madre Hematopoyéticas/métodos , Mielofibrosis Primaria/terapia , Acondicionamiento Pretrasplante/métodos , Adolescente , Adulto , Anciano , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trasplante Homólogo , Adulto Joven
10.
Genes Chromosomes Cancer ; 52(4): 423-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23340989

RESUMEN

Suv39h1 mediates heterochromatin formation in pericentric and telomeric regions by trimethylation of lysine 9 of histone 3 (H3K9me3). Yet, its role in the induction of chromosomal instability is poorly understood. We established a leukemia model by retrovirally expressing Myc in wild-type and histone methyltransferase Suv39h1-deficient hematopoietic cells and characterized the resulting leukemias for chromosomal instability. All mice that received cells overexpressing Myc developed myeloid leukemia with a median survival of 44 days posttransplantation. Myc-overexpressing wild-type leukemias demonstrated clones with numerical chromosomal aberrations (5/16). In secondary transplantations of these leukemic cells, structural changes, mostly end-to-end fusions of chromosomes, appeared (10/12). In contrast, leukemic cells overexpressing Myc with reduced or no Suv39h1 expression had a normal karyotype in primary, secondary, and tertiary transplantations (16/16). Myc-transduced Suv39h1-deficient cells showed less critically short telomeres (P < 0.05) compared with Myc-transduced wild-type bone marrow cells. Gene expression analysis showed upregulation of genes involved in the alternative lengthening of telomeres (ALT) mechanism. Thus, we hypothesize that loss of Suv39h1 implies activation of the ALT mechanism, in turn ensuring telomere length and stability. Our data show for the first time that Suv39h1 deficiency may prevent chromosomal instability by more efficient telomere stabilization in hematopoietic bone marrow cells overexpressing Myc.


Asunto(s)
Inestabilidad Cromosómica , Leucemia Mieloide/genética , Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Represoras/genética , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hibridación Fluorescente in Situ , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Masculino , Metiltransferasas/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cariotipificación Espectral , Telómero/genética , Homeostasis del Telómero/genética , Acortamiento del Telómero/genética
11.
Blood ; 117(14): 3737-47, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21289307

RESUMEN

Thpo/Mpl signaling plays an important role in the maintenance of hematopoietic stem cells (HSCs) in addition to its role in megakaryopoiesis. Patients with inactivating mutations in Mpl develop thrombocytopenia and aplastic anemia because of progressive loss of HSCs. Yet, it is unknown whether this loss of HSCs is an irreversible process. In this study, we used the Mpl knockout (Mpl(-/-)) mouse model and expressed Mpl from newly developed lentiviral vectors specifically in the physiologic Mpl target populations, namely, HSCs and megakaryocytes. After validating lineage-specific expression in vivo using lentiviral eGFP reporter vectors, we performed bone marrow transplantation of transduced Mpl(-/-) bone marrow cells into Mpl(-/-) mice. We show that restoration of Mpl expression from transcriptionally targeted vectors prevents lethal adverse reactions of ectopic Mpl expression, replenishes the HSC pool, restores stem cell properties, and corrects platelet production. In some mice, megakaryocyte counts were atypically high, accompanied by bone neo-formation and marrow fibrosis. Gene-corrected Mpl(-/-) cells had increased long-term repopulating potential, with a marked increase in lineage(-)Sca1(+)cKit(+) cells and early progenitor populations in reconstituted mice. Transcriptome analysis of lineage(-)Sca1(+)cKit(+) cells in Mpl-corrected mice showed functional adjustment of genes involved in HSC self-renewal.


Asunto(s)
Anemia Aplásica/genética , Anemia Aplásica/terapia , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/fisiología , Lentivirus/genética , Receptores de Trombopoyetina/genética , Regeneración/genética , Anemia Aplásica/patología , Anemia Aplásica/fisiopatología , Animales , Linaje de la Célula/genética , Células Cultivadas , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Receptores de Trombopoyetina/metabolismo , Receptores de Trombopoyetina/fisiología
12.
Ann Hematol ; 92(5): 587-94, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23307597

RESUMEN

Laboratory mice play an outstanding role in modeling human development and disease. In contrast to human leukemia, the spleen is involved in almost all cases, and the bone marrow is only variably involved in murine models. Although mice have been used for medical research for over 100 years, there are only few reports with a small number of cases looking at morphology and quantitative composition of murine hematopoietic cells in the bone marrow of non-transplanted animals of most strains. To our knowledge, there is not even a single report describing the splenogram in C57BL/6J mice, one of the most commonly used strains for medical research. The present study illustrates the morphology of the hematopoietic cells in the bone marrow and spleen of non-treated C57BL/6J mice and establishes the murine myelogram from the largest healthy C57BL/6J cohort reported to date. Furthermore, we present the first murine splenogram described for C57BL/6J mice. Our study supports the acceptance of the presence of >5 % blast cells as providing clear evidence of abnormality in bone marrow like in humans. In addition, we are the first to show <1 % blast cells in the normal spleen. Interestingly, classical dysplastic changes were rare in normal healthy mice. Our study of the bone marrow and spleen of healthy non-transplanted animals provides reference ranges of each cell type and for the myeloid/erythroid ratio, which can be used to interpret preclinical gene therapy data, leukemogenesis, and hematopoiesis studies, and may improve the quality of such analyses.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Bazo/citología , Animales , Médula Ósea/fisiología , Células de la Médula Ósea/clasificación , Recuento de Células , Forma de la Célula , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/fisiología , Granulocitos/citología , Granulocitos/fisiología , Salud , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/clasificación , Macrófagos/citología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/fisiología
13.
Ann Hematol ; 92(5): 595-604, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23307598

RESUMEN

Gene therapy has proven its potential to cure diseases of the hematopoietic system, but potential adverse reactions related to insertional mutagenesis by integrating gene vectors and chromosomal instability in long-lived repopulating cells have emerged as a major limitation. Preclinical gene therapy in murine models is a powerful model for assessment of gene marking efficiency and adverse reactions. However, changes in the hematologic composition after transplantation with retrovirally modified hematopoietic stem cells have not been well investigated in large cohorts of animals by systematic cytological analyses. In the present study, cytological analyses of bone marrow and spleen were performed in a large cohort (n = 58) of C57BL/6J mice over an extended observation period after gene marking. Interestingly, we observed hematological malignancies in four out of 30 animals transplanted with dLNGFR (truncated form of the human p75 low-affinity nerve growth factor receptor) and tCD34 modified stem/progenitor cells. Our data demonstrate that cytological analysis provides important information for diagnosis of hematological disorders and thus should be included in preclinical studies and performed in each investigated animal. Together with histological analysis, flow cytometric analysis, and other analyses, the quality and predictive value of preclinical gene therapy studies will be improved.


Asunto(s)
Células de la Médula Ósea/patología , Estudios de Evaluación como Asunto , Terapia Genética/efectos adversos , Neoplasias Hematológicas/diagnóstico , Células Madre Hematopoyéticas/patología , Bazo/patología , Animales , Técnicas Citológicas/normas , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Valor Predictivo de las Pruebas , Mejoramiento de la Calidad , Resultado del Tratamiento
14.
Br J Haematol ; 157(1): 75-85, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22280409

RESUMEN

To define a prognostic model for predicting outcome of reduced-intensity allogeneic stem cell transplantation (RIC-ASCT) for myelofibrosis we evaluated 150 homogeneously treated patients and developed a new risk score for overall survival (OS). In a multivariate Cox model for OS, only JAK2 V617F wild-type, age ≥57 years and constitutional symptoms were independently predictive for OS (Hazard Ratio: 2·02; 2·43 and 2·80 respectively). Depending on the presence of one, two or all of these factors, HR of death was 3·08; 4·70 and 16·61 respectively (P < 0·001). This score was compared to the Lille, Cervantes, International Prognostic Scoring System (IPSS), dynamic IPSS (DIPSS) and modified European Blood and Marrow Transplantation Group (EBMT) scores. Lille score correlated significantly with OS but discriminated poorly between the intermediate and high-risk groups (5-year OS 56% and 51% respectively). IPSS and DIPSS correlated significantly with OS but differences between intermediate-1 and intermediate-2 groups were not significant (5-year OS 78% vs. 78% and 70%, 60% respectively). Modified EBMT and Cervantes models did not predict OS post-ASCT. In conclusion, a simple model which includes: age, JAK2 V617F-status and constitutional symptoms can clearly separate distinct risk groups and can be used in addition to the Lille model to predict OS after RIC-ASCT for myelofibrosis.


Asunto(s)
Modelos Biológicos , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/terapia , Trasplante de Células Madre , Acondicionamiento Pretrasplante , Adulto , Anciano , Sustitución de Aminoácidos , Supervivencia sin Enfermedad , Femenino , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Persona de Mediana Edad , Mutación Missense , Mielofibrosis Primaria/enzimología , Mielofibrosis Primaria/genética , Medición de Riesgo , Tasa de Supervivencia , Factores de Tiempo , Trasplante Homólogo
15.
Am J Pathol ; 178(2): 494-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21281782

RESUMEN

A key event in the progression of glomerular disease is podocyte loss that leads to focal and segmental glomerulosclerosis (FSGS). Because adult podocytes are postmitotic cells, podocyte replacement by bone marrow-derived progenitors could prevent podocytopenia and FSGS. This study uses double immunofluorescence for Wilms' tumor-1 and enhanced green fluorescent protein (eGFP) to examine whether an eGFP-positive bone marrow transplant can replace podocytes under normal circumstances and in 3 different rat models of FSGS: puromycin aminoglycoside nephropathy, subtotal nephrectomy, and uninephrectomy. Bone marrow engraftment was successful, with more than 70% eGFP-positive cells and virtually normal histologic findings. No bone marrow transplant-derived podocytes were found in four control rats after transplantation, in nine rats at up to 10 weeks after puromycin aminoglycoside nephropathy induction, in three rats 23 days after subtotal nephrectomy, and in six rats up to 21 days after uninephrectomy. A total of 2200 glomeruli with 14,474 podocytes were evaluated in all groups. Thus, podocyte replacement by bone marrow-derived cells does not contribute to podocyte turnover in rats, even in models of podocyte damage. This is in contrast to previous studies in mice, in which bone marrow-derived podocytes were found. Further studies will address this discrepancy, which could be explained by species differences or by predominant podocyte regeneration from a parietal epithelial cell niche.


Asunto(s)
Técnicas de Ablación , Células de la Médula Ósea/citología , Enfermedades Renales/patología , Enfermedades Renales/cirugía , Riñón/cirugía , Podocitos/citología , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/cirugía , Riñón/patología , Podocitos/metabolismo , Puromicina , Ratas , Ratas Wistar , Células Madre/metabolismo
16.
Leukemia ; 36(3): 675-686, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732858

RESUMEN

With an incidence of ~50%, the absence or reduced protein level of p53 is much more common than TP53 mutations in acute myeloid leukemia (AML). AML with FLT3-ITD (internal tandem duplication) mutations has an unfavorable prognosis and is highly associated with wt-p53 dysfunction. While TP53 mutation in the presence of FLT3-ITD does not induce AML in mice, it is not clear whether p53 haploinsufficiency or loss cooperates with FLT3-ITD in the induction of AML. Here, we generated FLT3-ITD knock-in; p53 knockout (heterozygous and homozygous) double-transgenic mice and found that both alterations strongly cooperated in the induction of cytogenetically normal AML without increasing the self-renewal potential. At the molecular level, we found the strong upregulation of Htra3 and the downregulation of Lin28a, leading to enhanced proliferation and the inhibition of apoptosis and differentiation. The co-occurrence of Htra3 overexpression and Lin28a knockdown, in the presence of FLT3-ITD, induced AML with similar morphology as leukemic cells from double-transgenic mice. These leukemic cells were highly sensitive to the proteasome inhibitor carfilzomib. Carfilzomib strongly enhanced the activity of targeting AXL (upstream of FLT3) against murine and human leukemic cells. Our results unravel a unique role of p53 haploinsufficiency or loss in the development of FLT3-ITD + AML.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Haploinsuficiencia , Leucemia Mieloide Aguda/genética , Proteína p53 Supresora de Tumor/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Duplicación de Gen , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL , Mutación
18.
Haematologica ; 96(2): 319-22, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21109690

RESUMEN

In patients with low and intermediate risk myelodysplastic syndrome and deletion 5q (del(5q)) treated with lenalidomide, monitoring of cytogenetic response is mandatory, since patients without cytogenetic response have a significantly increased risk of progression. Therefore, we have reviewed cytogenetic data of 302 patients. Patients were analyzed by karyotyping and fluorescence in situ hybridization. In 85 patients, del(5q) was only detected by karyotyping. In 8 patients undergoing karyotypic evolution, the del(5q) and additional chromosomal aberrations were only detected by karyotyping. In 3 patients, del(5q) was only detected by fluorescence in situ hybridization, but not by karyotyping due to a low number of metaphases. Karyotyping was significantly more sensitive than fluorescence in situ hybridization in detecting the del(5q) clone. In conclusion, to optimize therapy control of myelodysplastic syndrome patients with del(5q) treated with lenalidomide and to identify cytogenetic non-response or progression as early as possible, fluorescence in situ hybridization alone is inadequate for evaluation. Karyotyping must be performed to optimally evaluate response.


Asunto(s)
Antineoplásicos/uso terapéutico , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Talidomida/análogos & derivados , Estudios de Cohortes , Estudios de Seguimiento , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Lenalidomida , Síndromes Mielodisplásicos/tratamiento farmacológico , Pronóstico , Inducción de Remisión , Factores de Riesgo , Tasa de Supervivencia , Talidomida/uso terapéutico
19.
Ann Hematol ; 90(3): 307-13, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20842500

RESUMEN

The microRNA/miR deregulation in BCR-ABL-negative myelodysplastic-myeloproliferative neoplasms (MDS/MPN) is not known. Myelopoiesis-associated miR-10a, miR-17-5p, miR-155, miR-223 and miR-424 were analysed by real-time polymerase chain reaction (PCR) in bone marrow cells of atypical chronic myeloid leukaemia (aCML, n = 7) and chronic myelomonocytic leukaemia (CMML, n = 8) and were compared to BCR-ABL-positive chronic myelogenous leukaemia (CML, n = 10) and non-neoplastic haematopoiesis (n = 10). Down-regulation of miR-10a was found in CMML but also in CML (each p < 0.05, versus controls). Overexpression of miR-424 was detected in aCML (p < 0.05, versus CML and controls). Despite different compositions of bone marrow cells, expression of myelopoiesis-associated microRNA shows mainly similar patterns in aCML and its main differential diagnosis CMML and does not allow discrimination of these two MDS/MPN entities. Therefore, the link of deregulated microRNA expression to disease-related phenotype and the underlying molecular mechanism are still unknown.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/genética , Leucemia Mielomonocítica Crónica/genética , MicroARNs/biosíntesis , Mielopoyesis/genética , Adulto , Anciano , Anciano de 80 o más Años , Células de la Médula Ósea/patología , Regulación hacia Abajo , Femenino , Proteínas de Fusión bcr-abl/biosíntesis , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/patología , Leucemia Mielomonocítica Crónica/patología , Masculino , MicroARNs/genética , Persona de Mediana Edad
20.
Ann Hematol ; 90(1): 33-40, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20625903

RESUMEN

Primary myelofibrosis (PMF) is a chronic myeloproliferative neoplasm showing aberrant bone marrow remodeling with increased angiogenesis, progressive matrix accumulation, and fibrosis development. Thrombospondins (TSP) are factors sharing pro-fibrotic and anti-angiogenic properties, and have not been addressed in PMF before. We investigated the expression of TSP-1 and TSP-2 in PMF related to the stage of myelofibrosis (n = 51) and in individual follow-up biopsies by real-time PCR, immunohistochemistry, and confocal laser scanning microscopy (CLSM). TSP-1 was significantly overexpressed (p < 0.05) in all stages of PMF when compared to controls. Individual follow-up biopsies showed involvement of TSP-1 during progressive myelofibrosis. TSP-2 was barely detectable but 40% of cases with advanced myelofibrosis showed a strong expression. Megakaryocytes and interstitial proplatelet formations were shown to be the relevant source for TSP-1 in PMF. Stroma cells like endothelial cells and fibroblasts showed no TSP-1 labeling when double-immunofluorescence staining and CLSM were applied. Based on its dual function, TSP-1 in PMF is likely to be a mediator within a pro-fibrotic environment which discriminates from ET cases. On the other hand, TSP-1 is a factor acting (ineffectively) against exaggerated angiogenesis. Both features suggest TSP-1 to be a biomarker for monitoring a PMF-targeted therapy.


Asunto(s)
Megacariocitos/metabolismo , Mielofibrosis Primaria/diagnóstico , Trombocitemia Esencial/diagnóstico , Trombospondina 1/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/análisis , Biomarcadores/metabolismo , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Estudios de Casos y Controles , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Megacariocitos/química , Megacariocitos/patología , Persona de Mediana Edad , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Sistema de Registros , Trombocitemia Esencial/genética , Trombocitemia Esencial/metabolismo , Trombocitemia Esencial/patología , Trombospondina 1/análisis , Trombospondina 1/genética , Trombospondina 1/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA