Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 110(4): 681-690, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36996813

RESUMEN

The blood-brain barrier (BBB) is an essential gatekeeper for the central nervous system and incidence of neurodevelopmental disorders (NDDs) is higher in infants with a history of intracerebral hemorrhage (ICH). We discovered a rare disease trait in thirteen individuals, including four fetuses, from eight unrelated families associated with homozygous loss-of-function variant alleles of ESAM which encodes an endothelial cell adhesion molecule. The c.115del (p.Arg39Glyfs∗33) variant, identified in six individuals from four independent families of Southeastern Anatolia, severely impaired the in vitro tubulogenic process of endothelial colony-forming cells, recapitulating previous evidence in null mice, and caused lack of ESAM expression in the capillary endothelial cells of damaged brain. Affected individuals with bi-allelic ESAM variants showed profound global developmental delay/unspecified intellectual disability, epilepsy, absent or severely delayed speech, varying degrees of spasticity, ventriculomegaly, and ICH/cerebral calcifications, the latter being also observed in the fetuses. Phenotypic traits observed in individuals with bi-allelic ESAM variants overlap very closely with other known conditions characterized by endothelial dysfunction due to mutation of genes encoding tight junction molecules. Our findings emphasize the role of brain endothelial dysfunction in NDDs and contribute to the expansion of an emerging group of diseases that we propose to rename as "tightjunctionopathies."


Asunto(s)
Encefalopatías , Moléculas de Adhesión Celular , Malformaciones del Sistema Nervioso , Trastornos del Neurodesarrollo , Animales , Ratones , Alelos , Encefalopatías/genética , Moléculas de Adhesión Celular/genética , Células Endoteliales/metabolismo , Hemorragias Intracraneales/genética , Malformaciones del Sistema Nervioso/genética , Trastornos del Neurodesarrollo/genética , Uniones Estrechas/genética , Humanos
2.
Blood ; 137(15): 2085-2089, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33238000

RESUMEN

Aberrant megakaryopoiesis is a hallmark of the myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies originating from hematopoietic stem cells, leading to an increase in mature blood cells in the peripheral blood. Sialylated derivatives of the glycan structure ß4-N-acetyllactosamine (Galß1,4GlcNAc or type-2 LacNAc, hereafter referred to as LacNAc) regulate platelet life span, hepatic thrombopoietin (TPO) production, and thrombopoiesis. We found increased TPO plasma levels in MPNs with high allele burden of the mutated clones. Remarkably, platelets isolated from MPNs had a significant increase in LacNAc expression that correlated with the high allele burden regardless of the underlying identified mutation. Megakaryocytes derived in vitro from these patients showed an increased expression of the B4GALT1 gene encoding ß-1,4-galactosyltransferase 1 (ß4GalT1). Consistently, megakaryocytes from MPN showed increased LacNAc expression relative to healthy controls, which was counteracted by the treatment with a Janus kinase 1/2 inhibitor. Altered expression of B4GALT1 in mutant megakaryocytes can lead to the production of platelets with aberrant galactosylation, which in turn promote hepatic TPO synthesis regardless of platelet mass. Our findings provide a new paradigm for understanding aberrant megakaryopoiesis in MPNs and identify ß4GalT1 as a potential actionable target for therapy.


Asunto(s)
Plaquetas/patología , Galactosa/metabolismo , Galactosiltransferasas/genética , Trastornos Mieloproliferativos/genética , Trombopoyetina/sangre , Plaquetas/metabolismo , Galactosa/análisis , Galactosiltransferasas/metabolismo , Humanos , Megacariocitos/metabolismo , Megacariocitos/patología , Mutación , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/metabolismo , Trombopoyetina/metabolismo , Regulación hacia Arriba
3.
Acta Haematol ; 146(1): 14-25, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36349787

RESUMEN

INTRODUCTION: About 15% of people with a myeloproliferative neoplasm (MPN) are identified as MPN, unclassifiable using the 2016 WHO classification. METHODS: We tested whether persons with platelet concentration ≥450 × 10E+9/L, bone marrow megakaryocyte morphology typical of prefibrotic/early myelofibrosis (pre-MF), and no minor criteria of pre-MF should be classified as a distinct MPN subtype, clonal megakaryocyte dysplasia with isolated thrombocytosis (CMD-IT). RESULTS: 139 subjects meet these criteria who we compared with primary myelofibrosis (PMF) including 402 with pre-MF and 521 with overt myelofibrosis. CMD-IT subjects were more likely female and younger. They had lower frequencies of JAK2V617F compared with persons with PMF (55% vs. 70%; p < 0.001) and higher frequencies of CALR mutations (37% vs. 17%; p < 0.001). They also had lower frequency of variations associated with JAK2V617F susceptibility, JAK2 46/1 (35% vs. 47%; p = 0.021), and VEGFA rs3025039 (12% vs. 17%; p = 0.030). Subjects with CMD-IT had lower incidences of thrombotic events compared with those with pre-MF (9.7% vs. 26%; p < 0.001) and longer survival (median, not reached vs. 23 years; HR = 0.34 (0.10, 0.30); p < 0.001). CONCLUSION: Our data indicate CMD-IT is a distinct MPN subtype and should be included in the classification of myeloid neoplasms.


Asunto(s)
Trastornos Mieloproliferativos , Neoplasias , Mielofibrosis Primaria , Trombocitemia Esencial , Trombocitosis , Femenino , Humanos , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/genética , Megacariocitos , Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/genética , Mutación , Trastornos Mieloproliferativos/diagnóstico , Trastornos Mieloproliferativos/genética , Trombocitosis/genética , Fenotipo , Janus Quinasa 2/genética , Calreticulina/genética
4.
Blood ; 135(2): 133-144, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31697806

RESUMEN

Approximately one-fourth of patients with essential thrombocythemia or primary myelofibrosis carry a somatic mutation of the calreticulin gene (CALR), the gene encoding for calreticulin. A 52-bp deletion (type I mutation) and a 5-bp insertion (type II mutation) are the most frequent genetic lesions. The mechanism(s) by which a CALR mutation leads to a myeloproliferative phenotype has been clarified only in part. We studied the interaction between calreticulin and store-operated calcium (Ca2+) entry (SOCE) machinery in megakaryocytes (Mks) from healthy individuals and from patients with CALR-mutated myeloproliferative neoplasms (MPNs). In Mks from healthy subjects, binding of recombinant human thrombopoietin to c-Mpl induced the activation of signal transducer and activator of transcription 5, AKT, and extracellular signal-regulated kinase 1/2, determining inositol triphosphate-dependent Ca2+ release from the endoplasmic reticulum (ER). This resulted in the dissociation of the ER protein 57 (ERp57)-mediated complex between calreticulin and stromal interaction molecule 1 (STIM1), a protein of the SOCE machinery that leads to Ca2+ mobilization. In Mks from patients with CALR-mutated MPNs, defective interactions between mutant calreticulin, ERp57, and STIM1 activated SOCE and generated spontaneous cytosolic Ca2+ flows. In turn, this resulted in abnormal Mk proliferation that was reverted using a specific SOCE inhibitor. In summary, the abnormal SOCE regulation of Ca2+ flows in Mks contributes to the pathophysiology of CALR-mutated MPNs. In perspective, SOCE may represent a new therapeutic target to counteract Mk proliferation and its clinical consequences in MPNs.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Megacariocitos/patología , Mutación , Trastornos Mieloproliferativos/patología , Canales de Calcio Activados por la Liberación de Calcio/genética , Estudios de Casos y Controles , Humanos , Megacariocitos/metabolismo , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
5.
Acta Haematol ; 145(1): 30-37, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34280924

RESUMEN

INTRODUCTION: In 1991, we reported 18 persons with a clinical-pathologic entity and termed atypical myeloproliferative disorder because they did not meet the contemporary diagnostic criteria for a myeloproliferative neoplasm. We sought to gain further knowledge on this disease entity. METHODS: This retrospective cohort study included consecutive subjects registered in the database of the Center for the Study of Myelofibrosis in Pavia, Italy, from 1998 to 2020 (June), and diagnosed with atypical myeloproliferative disorder according to our adjudicated criteria. We studied clinical, histological, cytogenetic, and molecular covariates and risks of thrombosis, disease progression, and death. Data were compared with those of concurrent subjects with prefibrotic myelofibrosis. RESULTS: Fifteen new subjects with atypical myeloproliferative disorder were identified. Seven were male. Median age was 50 years (IQR, 41-54 years). Thirteen were diagnosed with a synchronous symptomatic or incidentally detected thrombotic event. The bone marrow showed megakaryocyte hyperplasia with dysplasia. JAK2V617F was present in 10 subjects and CALR mutation in one. No other somatic mutations were identified in next generation sequencing. After a median follow-up of 101 months (IQR, 40-160 months), no subject had disease progression or blast transformation. Incidence of post-diagnosis or recurrent thrombosis was 3.9 events (95% confidence interval, 3.5-4.0) and 5.0 events (4.6-5.6) per 100 person-years. Features of subjects with atypical myeloproliferative disorder differed markedly from those of 546 subjects with prefibrotic myelofibrosis. CONCLUSION: Our data indicate that these 15 persons have a distinct myeloproliferative neoplasm. We propose naming this new disorder clonal megakaryocyte dysplasia with normal blood values.


Asunto(s)
Calreticulina , Neoplasias Hematológicas , Janus Quinasa 2 , Megacariocitos , Mutación Missense , Trastornos Mieloproliferativos , Adulto , Sustitución de Aminoácidos , Médula Ósea/metabolismo , Médula Ósea/patología , Calreticulina/genética , Calreticulina/metabolismo , Femenino , Neoplasias Hematológicas/sangre , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Persona de Mediana Edad , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Estudios Retrospectivos , Trombosis
6.
J Cell Mol Med ; 25(16): 8074-8086, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34288391

RESUMEN

Second trimester foetal human amniotic fluid-derived stem cells (hAFS) have been shown to possess remarkable cardioprotective paracrine potential in different preclinical models of myocardial injury and drug-induced cardiotoxicity. The hAFS secretome, namely the total soluble factors released by cells in their conditioned medium (hAFS-CM), can also strongly sustain in vivo angiogenesis in a murine model of acute myocardial infarction (MI) and stimulates human endothelial colony-forming cells (ECFCs), the only truly recognized endothelial progenitor, to form capillary-like structures in vitro. Preliminary work demonstrated that the hypoxic hAFS secretome (hAFS-CMHypo ) triggers intracellular Ca2+ oscillations in human ECFCs, but the underlying mechanisms and the downstream Ca2+ -dependent effectors remain elusive. Herein, we found that the secretome obtained by hAFS undergoing hypoxic preconditioning induced intracellular Ca2+ oscillations by promoting extracellular Ca2+ entry through Transient Receptor Potential Vanilloid 4 (TRPV4). TRPV4-mediated Ca2+ entry, in turn, promoted the concerted interplay between inositol-1,4,5-trisphosphate- and nicotinic acid adenine dinucleotide phosphate-induced endogenous Ca2+ release and store-operated Ca2+ entry (SOCE). hAFS-CMHypo -induced intracellular Ca2+ oscillations resulted in the nuclear translocation of the Ca2+ -sensitive transcription factor p65 NF-κB. Finally, inhibition of either intracellular Ca2+ oscillations or NF-κB activity prevented hAFS-CMHypo -induced ECFC tube formation. These data shed novel light on the molecular mechanisms whereby hAFS-CMHypo induces angiogenesis, thus providing useful insights for future therapeutic strategies against ischaemic-related myocardial injury.


Asunto(s)
Líquido Amniótico/metabolismo , Calcio/metabolismo , Medios de Cultivo Condicionados/química , Células Endoteliales/fisiología , FN-kappa B/metabolismo , Secretoma , Células Madre/citología , Líquido Amniótico/química , Células Cultivadas , Células Endoteliales/citología , Humanos , FN-kappa B/genética , Transporte de Proteínas , Transducción de Señal , Células Madre/metabolismo
7.
J Cell Physiol ; 236(1): 688-705, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32583526

RESUMEN

Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most recently discovered Ca2+ -releasing messenger that increases the intracellular Ca2+ concentration by mobilizing the lysosomal Ca2+ store through two-pore channels 1 (TPC1) and 2 (TPC2). NAADP-induced lysosomal Ca2+ release regulates multiple endothelial functions, including nitric oxide release and proliferation. A sizeable acidic Ca2+ pool endowed with TPC1 is also present in human endothelial colony-forming cells (ECFCs), which represent the only known truly endothelial precursors. Herein, we sought to explore the role of the lysosomal Ca2+ store and TPC1 in circulating ECFCs by harnessing Ca2+ imaging and molecular biology techniques. The lysosomotropic agent, Gly-Phe ß-naphthylamide, and nigericin, which dissipates the proton gradient which drives Ca2+ sequestration by acidic organelles, caused endogenous Ca2+ release in the presence of a replete inositol-1,4,5-trisphosphate (InsP3 )-sensitive endoplasmic reticulum (ER) Ca2+ pool. Likewise, the amount of ER releasable Ca2+ was reduced by disrupting lysosomal Ca2+ content. Liposomal delivery of NAADP induced a transient Ca2+ signal that was abolished by disrupting the lysosomal Ca2+ store and by pharmacological and genetic blockade of TPC1. Pharmacological manipulation revealed that NAADP-induced Ca2+ release also required ER-embedded InsP3 receptors. Finally, NAADP-induced lysosomal Ca2+ release was found to trigger vascular endothelial growth factor-induced intracellular Ca2+ oscillations and proliferation, while it did not contribute to adenosine-5'-trisphosphate-induced Ca2+ signaling. These findings demonstrated that NAADP-induced TPC1-mediated Ca2+ release can selectively be recruited to induce the Ca2+ response to specific cues in circulating ECFCs.


Asunto(s)
Canales de Calcio/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , NADP/análogos & derivados , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Retículo Endoplásmico/metabolismo , Células Endoteliales/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , NADP/metabolismo , NADP/farmacología , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Blood ; 134(18): 1498-1509, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31515250

RESUMEN

Prior studies have reported high response rates with recombinant interferon-α (rIFN-α) therapy in patients with essential thrombocythemia (ET) and polycythemia vera (PV). To further define the role of rIFN-α, we investigated the outcomes of pegylated-rIFN-α2a (PEG) therapy in ET and PV patients previously treated with hydroxyurea (HU). The Myeloproliferative Disorders Research Consortium (MPD-RC)-111 study was an investigator-initiated, international, multicenter, phase 2 trial evaluating the ability of PEG therapy to induce complete (CR) and partial (PR) hematologic responses in patients with high-risk ET or PV who were either refractory or intolerant to HU. The study included 65 patients with ET and 50 patients with PV. The overall response rates (ORRs; CR/PR) at 12 months were 69.2% (43.1% and 26.2%) in ET patients and 60% (22% and 38%) in PV patients. CR rates were higher in CALR-mutated ET patients (56.5% vs 28.0%; P = .01), compared with those in subjects lacking a CALR mutation. The median absolute reduction in JAK2V617F variant allele fraction was -6% (range, -84% to 47%) in patients achieving a CR vs +4% (range, -18% to 56%) in patients with PR or nonresponse (NR). Therapy was associated with a significant rate of adverse events (AEs); most were manageable, and PEG discontinuation related to AEs occurred in only 13.9% of subjects. We conclude that PEG is an effective therapy for patients with ET or PV who were previously refractory and/or intolerant of HU. This trial was registered at www.clinicaltrials.gov as #NCT01259856.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón-alfa/uso terapéutico , Policitemia Vera/tratamiento farmacológico , Polietilenglicoles/uso terapéutico , Trombocitemia Esencial/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Hidroxiurea , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
9.
Hematol Oncol ; 39(1): 123-128, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32979286

RESUMEN

We analyzed cytogenetic data at diagnosis in 395 primary myelofibrosis (PMF) patients to evaluate any possible association between karyotype and WHO 2017 classification and its impact on prognosis. All the cases were diagnosed and followed at five Italian Hematological Centers between November 1983 and December 2016. An abnormal karyotype (AK) was found in 69 patients and clustered differently according to bone marrow fibrosis grade as it was found in 31 (27.0%) cases with overt fibrotic and 38 (13.6%) with pre-fibrotic PMF (p = 0.001). Sex, anemia, thrombocytopenia, circulating blasts ≥1%, higher lactate dehydrogenase, and International Prognostic Scoring System risk classes were all significantly associated with karyotype. At a median follow-up of >6 years, 101 deaths were recorded. Survival was different between AK and normal karyotype (NK) patients with an estimated median overall survival (OS) of 11.6 and 25.7 years, respectively (p = 0.0148). In conclusion, in our cohort around 20% of patients had an AK, more frequently in subjects with an advanced bone marrow fibrosis grade and clinical-laboratory features indicative of a more aggressive disease. This study shows that an AK confers a more severe clinical phenotype and impacts adversely on OS, thus representing an additional parameter to be considered in the evaluation of PMF prognosis.


Asunto(s)
Cariotipo Anormal , Mielofibrosis Primaria , Anciano , Médula Ósea/patología , Análisis Citogenético , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Italia , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/patología , Pronóstico , Factores de Riesgo , Tasa de Supervivencia
10.
J Autoimmun ; 112: 102486, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32482487

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune multisystem disease featured by an increased cardiovascular risk that may lead to premature patient's death. It has been demonstrated that SLE patients suffer from early onset endothelial dysfunction which is due to the impairment of endogenous vascular repair mechanisms. Vascular integrity and homeostasis are maintained by endothelial progenitor cells (EPCs), which are mobilized in response to endothelial injury to replace damaged endothelial cells. Two main EPCs subpopulations exist in peripheral blood: endothelial colony forming cells (ECFCs), which represent truly endothelial precursors and can physically engraft within neovessels, and myeloid angiogenic cells (MACs), which sustain angiogenesis in a paracrine manner. Emerging evidence indicates that ECFCs/MACs are down-regulated and display compromised angiogenic activity in SLE, thereby contributing to the pathogenesis of this disease. Intracellular calcium (Ca2+) signaling plays a crucial role in maintaining vascular integrity by stimulating migration, proliferation and tube formation in both ECFCs and MACs. Herein, we illustrate the evidences that support the role played by EPCs dysfunction in SLE. Subsequently, we discuss about the hypothesis that the Ca2+ handling machinery is compromised in SLE-derived ECFCs and MACs, thereby resulting in their reduced pro-angiogenic activity. Finally, we speculate about the proposal to exploit intracellular Ca2+ signaling to improve ECFCs' reparative phenotype and suggest this strategy as a new approach to treat SLE patients.


Asunto(s)
Señalización del Calcio/inmunología , Células Progenitoras Endoteliales/metabolismo , Lupus Eritematoso Sistémico/inmunología , Células Mieloides/metabolismo , Neovascularización Patológica/inmunología , Animales , Calcio/metabolismo , Movimiento Celular/inmunología , Proliferación Celular , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/inmunología , Humanos , Lupus Eritematoso Sistémico/patología , Células Mieloides/inmunología , Neovascularización Patológica/patología , Comunicación Paracrina/inmunología , Transducción de Señal/inmunología
11.
Int J Mol Sci ; 21(19)2020 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-33036489

RESUMEN

Cardiovascular disease (CVD) comprises a range of major clinical cardiac and circulatory diseases, which produce immense health and economic burdens worldwide. Currently, vascular regenerative surgery represents the most employed therapeutic option to treat ischemic disorders, even though not all the patients are amenable to surgical revascularization. Therefore, more efficient therapeutic approaches are urgently required to promote neovascularization. Therapeutic angiogenesis represents an emerging strategy that aims at reconstructing the damaged vascular network by stimulating local angiogenesis and/or promoting de novo blood vessel formation according to a process known as vasculogenesis. In turn, circulating endothelial colony-forming cells (ECFCs) represent truly endothelial precursors, which display high clonogenic potential and have the documented ability to originate de novo blood vessels in vivo. Therefore, ECFCs are regarded as the most promising cellular candidate to promote therapeutic angiogenesis in patients suffering from CVD. The current briefly summarizes the available information about the origin and characterization of ECFCs and then widely illustrates the preclinical studies that assessed their regenerative efficacy in a variety of ischemic disorders, including acute myocardial infarction, peripheral artery disease, ischemic brain disease, and retinopathy. Then, we describe the most common pharmacological, genetic, and epigenetic strategies employed to enhance the vasoreparative potential of autologous ECFCs by manipulating crucial pro-angiogenic signaling pathways, e.g., extracellular-signal regulated kinase/Akt, phosphoinositide 3-kinase, and Ca2+ signaling. We conclude by discussing the possibility of targeting circulating ECFCs to rescue their dysfunctional phenotype and promote neovascularization in the presence of CVD.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Progenitoras Endoteliales/metabolismo , Isquemia/metabolismo , Isquemia/terapia , Trasplante de Células Madre , Animales , Biomarcadores , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Quimiotaxis , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Células Progenitoras Endoteliales/citología , Regulación de la Expresión Génica , Humanos , Isquemia/etiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Fenotipo , Transducción de Señal , Trasplante de Células Madre/métodos
12.
Blood ; 129(24): 3227-3236, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28351937

RESUMEN

The 2016 revision of the World Health Organization (WHO) classification of myeloproliferative neoplasms defines 2 stages of primary myelofibrosis (PMF): prefibrotic/early (pre-PMF) and overt fibrotic (overt PMF) phase. In this work, we studied the clinical and molecular features of patients belonging to these categories of PMF. The diagnosis of 661 PMF patients with a bone marrow biopsy at presentation was revised according to modern criteria; clinical information and annotation of somatic mutations in both driver and selected nondriver myeloid genes were available for all patients. Compared with pre-PMF, overt PMF was enriched in patients with anemia, thrombocytopenia, leukopenia, higher blast count, symptoms, large splenomegaly, and unfavorable karyotype. The different types of driver mutations were similarly distributed between the 2 categories, whereas selected mutations comprising the high mutation risk (HMR) category (any mutations in ASXL1, SRSF2, IDH1/2, EZH2) were more represented in overt PMF. More patients with overt PMF were in higher International Prognostic Scoring System risk categories at diagnosis, and the frequency increased during follow-up, suggesting greater propensity to disease progression compared with pre-PMF. Median survival was significantly shortened in overt PMF (7.2 vs 17.6 years), with triple negativity for driver mutations and presence of HMR mutations representing independent predictors of unfavorable outcome. The findings of this "real-life" study indicate that adherence to 2016 WHO criteria allows for identification of 2 distinct categories of patients with PMF where increased grades of fibrosis are associated with more pronounced disease manifestations, adverse mutation profile, and worse outcome, overall suggesting they might represent a phenotypic continuum.


Asunto(s)
Mutación , Mielofibrosis Primaria , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/mortalidad , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Tasa de Supervivencia , Organización Mundial de la Salud
13.
Cytotherapy ; 21(9): 958-972, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31279696

RESUMEN

BACKGROUND: The Wilms tumor antigen 1 (WT1) is over-expressed in a vast majority of adult and childhood acute leukemia and myelodysplastic syndromes, being lowly or transiently expressed in normal tissues and hematopoietic stem cells (HSCs). A number of HLA-restricted WT1 epitopes are immunogenic, allowing the in vitro induction of WT1-specific cytotoxic T lymphocytes (CTLs) from patients and healthy donors. AIM: The aim of the study was to investigate the feasibility of producing WT1-specific CTLs suitable for somatic cell therapy to prevent or treat relapse in children with acute myeloid or lymphoblastic leukemia given haploidentical HSC transplantation (haplo-HSCT). METHODS: For WT1-specific CTL production, donor-derived either peripheral blood mononuclear cells (PBMCs) or CD8+ lymphocytes were stimulated with WT1 peptide-loaded donor dendritic cells in the presence of interleukin (IL)-7 and IL-12. Effector cells were re-stimulated once with irradiated donor PBMCs pulsed with WT1-peptides, and then expanded in an antigen-independent way. RESULTS: WT1-specific CTLs, displaying high-level cytotoxicity against patients' leukemia blasts and negligible activity against patients' non-malignant cells, were obtained from both PBMCs and CD8+ lymphocytes. WT1-specific CTLs obtained from PBMCs showed a better expansion capacity and better anti-leukemia activity than those obtained from CD8+ lymphocytes, even though the difference was not statistically significant. In CTLs derived from PBMCs, both CD8+ and CD4+ subpopulations displayed strong anti-leukemia cytotoxic activity. DISCUSSION: Results of this pre-clinical study pave the way to a somatic cell therapy approach aimed at preventing or treating relapse in children given haplo-HSCT for WT1-positive leukemia.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia/inmunología , Leucemia/terapia , Linfocitos T Citotóxicos/inmunología , Donantes de Tejidos , Proteínas WT1/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Estudios de Factibilidad , Femenino , Células Madre Hematopoyéticas/inmunología , Humanos , Interferón gamma/biosíntesis , Masculino , Péptidos/metabolismo , Fenotipo , Trasplante Haploidéntico
14.
Am J Pathol ; 187(8): 1879-1892, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28728747

RESUMEN

Primary myelofibrosis is characterized by the development of fibrosis in the bone marrow that contributes to ineffective hematopoiesis. Bone marrow fibrosis is the result of a complex and not yet fully understood interaction among megakaryocytes, myeloid cells, fibroblasts, and endothelial cells. Here, we report that >30% of the endothelial cells in the small vessels of the bone marrow and spleen of patients with primary myelofibrosis have a mesenchymal phenotype, which is suggestive of the process known as endothelial-to-mesenchymal transition (EndMT). EndMT can be reproduced in vitro by incubation of cultured endothelial progenitor cells or spleen-derived endothelial cells with inflammatory cytokines. Megakaryocytes appear to be implicated in this process, because EndMT mainly occurs in the microvessels close to these cells, and because megakaryocyte-derived supernatant fluid can reproduce the EndMT switch in vitro. Furthermore, EndMT is an early event in a JAK2-V617F knock-in mouse model of primary myelofibrosis. Overall, these data show for the first time that microvascular endothelial cells in the bone marrow and spleen of patients with primary myelofibrosis show functional and morphologic changes that are associated to the mesenchymal phenotype.


Asunto(s)
Médula Ósea/patología , Mielofibrosis Primaria/patología , Bazo/patología , Animales , Modelos Animales de Enfermedad , Humanos , Megacariocitos/patología , Ratones
15.
Am J Hematol ; 93(5): 615-622, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29359451

RESUMEN

Splenic hematopoiesis is a major feature in the course of myelofibrosis (MF). In fact, the spleen of patients with MF contains malignant hematopoietic stem cells retaining a complete differentiation program, suggesting both a pivotal role of the spleen in maintaining the disease and a tight regulation of hematopoiesis by the splenic microenvironment, in particular by mesenchymal stromal cells (MSCs). Little is known about splenic MSCs (Sp-MSCs), both in normal and in pathological context. In this work, we have in vitro expanded and characterized Sp-MSCs from 25 patients with MF and 13 healthy subjects (HS). They shared similar phenotype, growth kinetics, and differentiation capacity. However, MF Sp-MSCs expressed significant lower levels of nestin, and favored megakaryocyte (Mk) differentiation in vitro at a larger extent than their normal counterpart. Moreover, they showed a significant upregulation of matrix metalloprotease 2 (MMP2) and fibronectin 1 (FN1) genes both at mRNA expression and at protein level, and, finally, developed genetic abnormalities which were never detected in HS-derived Sp-MSCs. Our data point toward the existence of a defective splenic niche in patients with MF that could be responsible of some pathological features of the disease, including the increased trafficking of CD34+ cells and the expansion of the megakaryocytic lineage.


Asunto(s)
Células Madre Mesenquimatosas/patología , Mielofibrosis Primaria/patología , Bazo/patología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD34 , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Femenino , Fibronectinas/metabolismo , Hematopoyesis , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Megacariocitos/patología , Persona de Mediana Edad , Nestina/metabolismo , Adulto Joven
16.
Genes Chromosomes Cancer ; 56(1): 51-58, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27553422

RESUMEN

Shwachman-Diamond syndrome (SDS) (OMIM 260400) is a rare autosomal recessive disease characterized by exocrine pancreatic insufficiency, skeletal, and hematological abnormalities and bone marrow (BM) dysfunction. Mutations in the SBDS gene cause SDS. Clonal chromosome anomalies are often present in BM, i(7)(q10) and del(20q) being the most frequent ones. We collected 6 SDS cases with del(20q): a cluster of imprinted genes, including L3MBTL1 and SGK2 is present in the deleted region. Only the paternal allele is expressed for these genes. Based on these data, we made the hypothesis that the loss of this region, in relation to parental origin of deletion, may be of relevance for the hematological phenotype. By comparing hematological data of our 6 cases with a group of 20 SDS patients without evidence of del(20q) in BM, we observed a significant difference for Hb levels (P < 0.012), and a difference slightly above the significance level for RBC counts (P < 0.053): in both cases the values were higher in patients with del(20q). We also report preliminary evidence for an increased number of BFU-E colonies in cases with paternal deletion, data on the presence of the deletion in colonies and in mature circulating lymphocytes. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos Par 20/genética , Insuficiencia Pancreática Exocrina/genética , Impresión Genómica , Proteínas Inmediatas-Precoces/genética , Lipomatosis/genética , Proteínas Serina-Treonina Quinasas/genética , Eliminación de Secuencia , Biomarcadores de Tumor , Aberraciones Cromosómicas , Estudios de Seguimiento , Humanos , Mutación/genética , Estadificación de Neoplasias , Fenotipo , Pronóstico , Proteínas Represoras , Estudios Retrospectivos , Síndrome de Shwachman-Diamond , Proteínas Supresoras de Tumor
17.
J Cell Biochem ; 118(11): 3722-3729, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28374913

RESUMEN

Nicotinic acid adenine dinucleotide phosphate (NAADP) is the newest discovered intracellular second messengers, which is able to release Ca2+ stored within endolysosomal (EL) vesicles. NAADP-induced Ca2+ signals mediate a growing number of cellular functions, ranging from proliferation to muscle contraction and differentiation. Recently, NAADP has recently been shown to regulate angiogenesis by promoting endothelial cell growth. It is, however, still unknown whether NAADP stimulates proliferation also in endothelial progenitor cells, which are mobilized in circulation after an ischemic insult to induce tissue revascularization. Herein, we described a novel approach to prepare NAADP-containing liposomes, which are highly cell membrane permeable and are therefore amenable for stimulating cell activity. Accordingly, NAADP-containing liposomes evoked an increase in intracellular Ca2+ concentration, which was inhibited by NED-19, a selective inhibitor of NAADP-induced Ca2+ release. Furthermore, NAADP-containing liposomes promoted EPC proliferation, a process which was inhibited by NED-19 and BAPTA, a membrane permeable intracellular Ca2+ buffer. Therefore, NAADP-containing liposomes stand out as a promising tool to promote revascularization of hypoxic/ischemic tissues by favoring EPC proliferation. J. Cell. Biochem. 118: 3722-3729, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/metabolismo , NADP/análogos & derivados , Neovascularización Fisiológica/efectos de los fármacos , Adulto , Carbolinas/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/metabolismo , Células Endoteliales/citología , Femenino , Humanos , Liposomas , Masculino , NADP/farmacología , Piperazinas/metabolismo
18.
Br J Cancer ; 116(3): 335-343, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28072764

RESUMEN

BACKGROUND: Juvenile myelomonocytic leukaemia (JMML) and chronic myelomonocytic leukaemia (CMML) are myelodysplastic myeloproliferative (MDS/MPN) neoplasms with unfavourable prognosis and without effective chemotherapy treatment. Trabectedin is a DNA minor groove binder acting as a modulator of transcription and interfering with DNA repair mechanisms; it causes selective depletion of cells of the myelomonocytic lineage. We hypothesised that trabectedin might have an antitumour effect on MDS/MPN. METHODS: Malignant CD14+ monocytes and CD34+ haematopoietic progenitor cells were isolated from peripheral blood/bone marrow mononuclear cells. The inhibition of CFU-GM colonies and the apoptotic effect on CD14+ and CD34+ induced by trabectedin were evaluated. Trabectedin's effects were also investigated in vitro on THP-1, and in vitro and in vivo on MV-4-11 cell lines. RESULTS: On CMML/JMML cells, obtained from 20 patients with CMML and 13 patients with JMML, trabectedin - at concentration pharmacologically reasonable, 1-5 nM - strongly induced apoptosis and inhibition of growth of haematopoietic progenitors (CFU-GM). In these leukaemic cells, trabectedin downregulated the expression of genes belonging to the Rho GTPases pathway (RAS superfamily) having a critical role in cell growth and cytoskeletal dynamics. Its selective activity on myelomonocytic malignant cells was confirmed also on in vitro THP-1 cell line and on in vitro and in vivo MV-4-11 cell line models. CONCLUSIONS: Trabectedin could be good candidate for clinical studies in JMML/CMML patients.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Dioxoles/uso terapéutico , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Leucemia Mielomonocítica Juvenil/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Tetrahidroisoquinolinas/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/patología , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patología , Ratones , Ratones Desnudos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Trabectedina , Ensayo de Tumor de Célula Madre
19.
Stem Cells ; 34(4): 1123-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26748484

RESUMEN

Extracellular matrix (ECM) components initiate crucial biochemical and biomechanical cues that are required for bone marrow homeostasis. In our research, we prove that a peri-cellular matrix composed primarily of type III and type IV collagens, and fibronectin surrounds human megakaryocytes in the bone marrow. The data we collected support the hypothesis that bone marrow megakaryocytes possess a complete mechanism to synthesize the ECM components, and that thrombopoietin is a pivotal regulator of this new function inducing transforming growth factor-ß1 (TGF-ß1) release and consequent activation of the downstream pathways, both in vitro and in vivo. This activation results in a dose dependent increase of ECM component synthesis by megakaryocytes, which is reverted upon incubation with JAK and TGF-ß1 receptor specific inhibitors. These data are pivotal for understanding the central role of megakaryocytes in creating their own regulatory niche within the bone marrow environment.


Asunto(s)
Matriz Extracelular/metabolismo , Megacariocitos/metabolismo , Trombopoyetina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Médula Ósea/crecimiento & desarrollo , Médula Ósea/metabolismo , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Matriz Extracelular/genética , Sangre Fetal/citología , Sangre Fetal/metabolismo , Fibronectinas/metabolismo , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Megacariocitos/efectos de los fármacos , Ratones , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Trombopoyetina/genética , Factor de Crecimiento Transformador beta1/genética
20.
Am J Hematol ; 92(2): 187-195, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27880982

RESUMEN

Splanchnic vein thrombosis (SVT) is one of the vascular complications of myeloproliferative neoplasms (MPN). We designed a phase 2 clinical trial to evaluate safety and efficacy of ruxolitinib in reducing splenomegaly and improving disease-related symptoms in patients with MPN-associated SVT. Patients diagnosed with myelofibrosis (12 cases), polycythemia vera (5 cases) and essential thrombocythemia (4 cases) received ruxolitinib for 24 weeks in the core study period. Spleen volume was assessed by magnetic resonance imaging (MRI) and splanchnic vein circulation by echo-Doppler analysis. Nineteen patients carried JAK2V617F, one had MPLW515L, and one CALRL367fs*46 mutation. Eighteen patients had spleno-portal-mesenteric thrombosis, two had Budd-Chiari syndrome, and one had both sites involved; 16 patients had esophageal varices. Ruxolitinib was well tolerated with hematological toxicities consistent with those of patients without SVT and no hemorrhagic adverse events were recorded. After 24 weeks of treatment, spleen volume reduction ≥35% by MRI was achieved by 6/21 (29%) patients, and a ≥50% spleen length reduction by palpation at any time up to week 24 was obtained by 13/21 (62%) patients. At week 72, 8 of the 13 (62%) patients maintained the spleen response by palpation. No significant effect of treatment on esophageal varices or in splanchnic circulation was observed. MPN-related symptoms, evaluated by MPN-symptom assessment form (SAF) TSS questionnaire, improved significantly during the first 4 weeks and remained stable up to week 24. In conclusion, this trial shows that ruxolitinib is safe in patients with MPN-associated SVT, and effective in reducing spleen size and disease-related symptoms.


Asunto(s)
Quinasas Janus/antagonistas & inhibidores , Trastornos Mieloproliferativos/tratamiento farmacológico , Pirazoles/efectos adversos , Pirazoles/uso terapéutico , Circulación Esplácnica/efectos de los fármacos , Trombosis de la Vena/prevención & control , Adulto , Anciano , Esquema de Medicación , Femenino , Humanos , Quinasas Janus/genética , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/complicaciones , Nitrilos , Recuento de Plaquetas , Pirazoles/administración & dosificación , Pirimidinas , Esplenomegalia/prevención & control , Resultado del Tratamiento , Trombosis de la Vena/sangre , Trombosis de la Vena/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA