Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
2.
Sci Adv ; 5(9): eaav4620, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31598549

RESUMEN

The control of stem and progenitor cell fate is emerging as a compelling urgency for regenerative medicine. Here, we propose a innovative strategy to gain optical control of endothelial colony-forming cell fate, which represents the only known truly endothelial precursor showing robust in vitro proliferation and overwhelming vessel formation in vivo. We combine conjugated polymers, used as photo-actuators, with the advantages offered by optical stimulation over current electromechanical and chemical stimulation approaches. Light modulation provides unprecedented spatial and temporal resolution, permitting at the same time lower invasiveness and higher selectivity. We demonstrate that polymer-mediated optical excitation induces a robust enhancement of proliferation and lumen formation in vitro. We identify the underlying biophysical pathway as due to light-induced activation of TRPV1 channel. Altogether, our results represent an effective way to induce angiogenesis in vitro, which represents the proof of principle to improve the outcome of autologous cell-based therapy in vivo.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Luz , Neovascularización Fisiológica , Polímeros/farmacología , Canales Catiónicos TRPV/metabolismo , Células Progenitoras Endoteliales/citología , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/efectos de la radiación
3.
Leukemia ; 33(12): 2974-2978, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31363161
4.
Leukemia ; 32(2): 438-449, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28745329

RESUMEN

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hyperplastic megakaryopoiesis and myelofibrosis. We recently described the upregulation of MAF (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog) in PMF CD34+ hematopoietic progenitor cells (HPCs) compared to healthy donor. Here we demonstrated that MAF is also upregulated in PMF compared with the essential thrombocytemia (ET) and polycytemia vera (PV) HPCs. MAF overexpression and knockdown experiments shed some light into the role of MAF in PMF pathogenesis, by demonstrating that MAF favors the megakaryocyte and monocyte/macrophage commitment of HPCs and leads to the increased expression of proinflammatory and profibrotic mediators. Among them, we focused our further studies on SPP1 and LGALS3. We assessed SPP1 and LGALS3 protein levels in 115 PMF, 47 ET and 24 PV patients plasma samples and we found that SPP1 plasma levels are significantly higher in PMF compared with ET and PV patients. Furthermore, in vitro assays demonstrated that SPP1 promotes fibroblasts and mesenchymal stromal cells proliferation and collagen production. Strikingly, clinical correlation analyses uncovered that higher SPP1 plasma levels in PMF patients correlate with a more severe fibrosis degree and a shorter overall survival. Collectively our data unveil that MAF overexpression contributes to PMF pathogenesis by driving the deranged production of the profibrotic mediator SPP1.


Asunto(s)
Médula Ósea/metabolismo , Médula Ósea/patología , Fibrosis/metabolismo , Fibrosis/patología , Osteopontina/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Antígenos CD34/metabolismo , Proliferación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Megacariocitos/metabolismo , Megacariocitos/patología , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Policitemia Vera/metabolismo , Policitemia Vera/patología , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Trombocitemia Esencial/metabolismo , Trombocitemia Esencial/patología
5.
Eur J Histochem ; 60(3): 2695, 2016 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-27734989

RESUMEN

We propose a tool for a rapid high-resolution detection of calcium ions which can be used in parallel with other techniques. We have applied a new approach by  photo-oxidation of diaminobenzidine in presence of the emission of an excited fluorochrome specific for calcium detection. This method combines the selectivity of available fluorophores to the high spatial resolution offered by transmission electron microscopy to detect even fluorescing molecules even when present in low amounts in membrane-bounded organelles. We show in this paper that Mag-Fura 2 photoconversion via diaminobenzidine oxidation is an efficient way for localizing Ca2+ ions at EM level, is easily carried out and reproducible, and can be obtained on a good amount of cells, since the exposition in our conditions is not limited to the direct irradiation of the sample via an objective but obtained with a germicide lamp. The end product is sufficiently electron dense to be detected clearly when present in sufficient amount within a membrane boundary.


Asunto(s)
Señalización del Calcio , Calcio , Colorantes Fluorescentes/química , Fura-2/análogos & derivados , Calcio/análisis , Calcio/metabolismo , Fura-2/química , Células HeLa , Humanos , Microscopía Fluorescente/métodos
7.
Leukemia ; 30(2): 431-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26449662

RESUMEN

A quarter of patients with essential thrombocythemia or primary myelofibrosis carry a driver mutation of CALR, the calreticulin gene. A 52-bp deletion (type 1) and a 5-bp insertion (type 2 mutation) are the most frequent variants. These indels might differentially impair the calcium binding activity of mutant calreticulin. We studied the relationship between mutation subtype and biological/clinical features of the disease. Thirty-two different types of CALR variants were identified in 311 patients. Based on their predicted effect on calreticulin C-terminal, mutations were classified as: (i) type 1-like (65%); (ii) type 2-like (32%); and (iii) other types (3%). Corresponding CALR mutants had significantly different estimated isoelectric points. Patients with type 1 mutation, but not those with type 2, showed abnormal cytosolic calcium signals in cultured megakaryocytes. Type 1-like mutations were mainly associated with a myelofibrosis phenotype and a significantly higher risk of myelofibrotic transformation in essential thrombocythemia. Type 2-like CALR mutations were preferentially associated with an essential thrombocythemia phenotype, low risk of thrombosis despite very-high platelet counts and indolent clinical course. Thus, mutation subtype contributes to determining clinical phenotype and outcomes in CALR-mutant myeloproliferative neoplasms. CALR variants that markedly impair the calcium binding activity of mutant calreticulin are mainly associated with a myelofibrosis phenotype.


Asunto(s)
Calreticulina/genética , Mutación , Mielofibrosis Primaria/genética , Trombocitemia Esencial/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Calcio/metabolismo , Células Cultivadas , Exones , Femenino , Humanos , Punto Isoeléctrico , Masculino , Megacariocitos/metabolismo , Persona de Mediana Edad , Mielofibrosis Primaria/metabolismo , Trombocitemia Esencial/metabolismo
10.
Int J Immunopathol Pharmacol ; 26(3): 809-16, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24067483

RESUMEN

Bronchopulmonary dysplasia (BPD) is a chronic lung disease occurring in very and extremely preterm infants undergoing mechanical ventilation. Given the altered lung vascular growth characterizing BPD, circulating angiogenic cells could be useful biomarkers to predict the risk. The objective of the study was to determine whether the percentages of circulating angiogenic cells (CD34+VEGFR-2+, CD34+CD133+VEGFR-2+, and CD45-CD34+CD133+VEGFR-2+ cells), assessed in the peripheral blood at birth by flow cytometry, could be used as markers for the risk of BPD. In one-hundred and forty-two preterm neonates (gestational age less than 32 weeks and/or birth weight less than 1500 g) admitted to our tertiary care Neonatal Intensive Care Unit between 2006 and 2009, we evaluated the percentages of circulating angiogenic cells at birth, at 7 days, and, in a subset of infants (n=40), at 28 days of life. The main outcome was the correlation between cell counts at birth and the subsequent risk of developing BPD. In our study, all the three cell populations failed to predict the development of BPD or other diseases of prematurity. We suggest that these cells cannot be used as biomarkers in preterm infants, and that research is needed to find other early predictors of BPD.


Asunto(s)
Displasia Broncopulmonar/diagnóstico , Células Madre Hematopoyéticas , Recien Nacido Prematuro/sangre , Recién Nacido de muy Bajo Peso/sangre , Neovascularización Patológica , Antígeno AC133 , Antígenos CD/sangre , Antígenos CD34/sangre , Biomarcadores/sangre , Displasia Broncopulmonar/sangre , Displasia Broncopulmonar/patología , Citometría de Flujo , Edad Gestacional , Glicoproteínas/sangre , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Antígenos Comunes de Leucocito/sangre , Recuento de Leucocitos , Péptidos/sangre , Fenotipo , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Factores de Riesgo , Centros de Atención Terciaria , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre
12.
Curr Med Chem ; 19(34): 5802-18, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22963562

RESUMEN

Endothelial progenitor cells (EPCs) have recently been employed in cell-based therapy (CBT) to promote neovascularization and regeneration of ischemic organs, such as heart and limbs. Furthermore, EPCs may be recruited from bone marrow by growing tumors to drive the angiogenic switch through physical engrafting into the lumen of nascent vessels or paracrine release of pro-angiogenic factors. CBT is hampered by the paucity of EPCs harvested from peripheral blood and suffered from several pitfalls, including the differentiation outcome of transplanted cells and low percentage of engrafted cells. Therefore, CBT will benefit from a better understanding of the signal transduction pathway(s) which govern(s) EPC homing, proliferation and incorporation into injured tissues. At the same time, this information might outline alternative molecular targets to combat tumoral neovascularization. We have recently found that store-operated Ca(2+) entry, a Ca(2+)-permeable membrane pathway that is activated upon depletion of the inositol-1,4,5-trisphosphate-sensitive Ca(2+) pool, is recruited by vascular endothelial growth factor to support proliferation and tubulogenesis in human circulating endothelial colony forming cells (ECFCs). ECFCs are a subgroup of EPCs that circulate in the peripheral blood of adult individuals and are able to proliferate and differentiate into endothelial cells and form capillary networks in vitro and contribute to neovessel formation in vivo. The present review will discuss the relevance of SOCE to ECFC-based cell therapy and will address the pharmacological inhibition of store-dependent Ca(2+) channels as a promising target for anti-angiogenic treatments.


Asunto(s)
Calcio/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Células Madre/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Canales de Calcio/metabolismo , Señalización del Calcio , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Proteína ORAI1 , Células Madre/citología , Molécula de Interacción Estromal 1
13.
Leukemia ; 22(3): 530-7, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18094717

RESUMEN

Circulating endothelial cells (CECs) are associated with neoangiogenesis in various malignant disorders. Using flow cytometry, we studied CECs in 128 patients with myelodysplastic syndrome (MDS). MDS patients had higher CEC levels than controls (P<0.001), and an inverse relationship was found between CECs and international prognostic scoring system risk (r=-0.55, P<0.001). There was a positive correlation between marrow microvessel density and CECs, low-risk patients showing the strongest association (r=0.62, P<0.001). We calculated a progenitor-to-mature CEC ratio, which was higher in MDS patients than in healthy subjects (P<0.001), the highest values were found at diagnosis. CECs assessed by flow cytometry positively correlated with the ability to produce endothelial colony-forming cells in vitro (ECFCs; r=0.57, P=0.021), which was significantly higher in MDS patients than in controls (P=0.011). Fluorescence in situ hybridization analysis showed that a variable proportion of CECs (from 40 to 84%) carried the same chromosomal aberration as the neoplastic clone, while endothelial cells isolated from in vitro assays were negative. This study suggests that CECs reflect the abnormal angiogenesis found in MDS, especially in the early stages of the disease. The increased number of functional endothelial progenitor cells in MDS strengthens the rationale for therapeutic interventions aimed at restoring a normal interaction between hematopoietic progenitors and marrow microenvironment.


Asunto(s)
Células Endoteliales/patología , Síndromes Mielodisplásicos/sangre , Neovascularización Patológica/genética , Anciano , Anciano de 80 o más Años , Médula Ósea/irrigación sanguínea , Recuento de Células , Linaje de la Célula , Aberraciones Cromosómicas , Células Clonales/patología , Ensayo de Unidades Formadoras de Colonias , Progresión de la Enfermedad , Células Endoteliales/química , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/fisiopatología , Neovascularización Patológica/patología , Reacción en Cadena de la Polimerasa , Estudios Prospectivos
14.
Leukemia ; 21(3): 397-402, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17215855

RESUMEN

We investigated genetically affected leukemic cells in FIP1L1-PDGFRA+ chronic eosinophilic leukemia (CEL) and in BCR-ABL1+ chronic myeloid leukemia (CML), two myeloproliferative disorders responsive to imatinib. Fluorescence in situ hybridization specific for BCR-ABL1 and for FIP1L1-PDGFRA was combined with cytomorphology or with lineage-restricted monoclonal antibodies and applied in CML and CEL, respectively. In CEL the amount of FIP1L1-PDGFRA+ cells among CD34+ and CD133+ cells, B and T lymphocytes, and megakaryocytes were within normal ranges. Positivity was found in eosinophils, granulo-monocytes and varying percentages of erythrocytes. In vitro assays with imatinib showed reduced survival of peripheral blood mononuclear cells but no reduction in colony-forming unit growth medium (CFU-GM) growth. In CML the BCR-ABL1 fusion gene was detected in CD34+/CD133+ cells, granulo-monocytes, eosinophils, erythrocytes, megakaryocytes and B-lymphocytes. Growth of both peripheral blood mononuclear cells and CFU-GM was inhibited by imatinib. This study provided evidence for marked differences in the leukemic masses which are targeted by imatinib in CEL or CML, as harboring FIP1L1-PDGFRA or BCR-ABL1.


Asunto(s)
Proteínas de Fusión bcr-abl/análisis , Síndrome Hipereosinofílico/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/enzimología , Proteínas de Fusión Oncogénica/análisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Factores de Escisión y Poliadenilación de ARNm/análisis , Antígeno AC133 , Antígenos CD/análisis , Antígenos CD34/análisis , Antineoplásicos/uso terapéutico , Benzamidas , Linaje de la Célula , Enfermedad Crónica , Células Clonales/enzimología , Resistencia a Medicamentos , Eosinófilos/enzimología , Eritrocitos/enzimología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Glicoforinas/análisis , Glicoproteínas/análisis , Granulocitos/enzimología , Células Madre Hematopoyéticas/enzimología , Humanos , Síndrome Hipereosinofílico/tratamiento farmacológico , Síndrome Hipereosinofílico/enzimología , Síndrome Hipereosinofílico/genética , Mesilato de Imatinib , Inmunofenotipificación , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Subgrupos Linfocitarios/enzimología , Megacariocitos/enzimología , Monocitos/enzimología , Células Mieloides/enzimología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Péptidos/análisis , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Ensayo de Tumor de Célula Madre , Inactivación del Cromosoma X , Factores de Escisión y Poliadenilación de ARNm/antagonistas & inhibidores
15.
Blood ; 98(12): 3249-55, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11719361

RESUMEN

The absolute content of CD34(+) cells in the peripheral blood of 84 patients with myelofibrosis with myeloid metaplasia (MMM) and 23 patients with other Philadelphia-negative (Ph(-)) chronic myeloproliferative disorders (CMDs) was investigated. In MMM, the median absolute number of circulating CD34(+) cells was consistently high (91.6 x 10(6)/L; range, 0-2460 x 10(6)/L). Receiver operating characteristic curve analysis showed that 15 x 10(6)/L as a decision criterion for CD34(+) cells produced an almost complete discrimination between MMM patients out of therapy and other Ph(-) CMDs (positive predictive value, 98.4%; negative predictive value, 85.0%). MMM patients with higher numbers of CD34(+) cells had a significantly longer disease duration (P =.019) and higher spleen volume index (P =.014), liver volume (P =.000), percentage of circulating immature myeloid cells (P =.020), and percentage of myeloid blasts (P =.000). When CD34(+) cells were correlated with the use of Dupriez risk stratification, CD34(+) cells increased significantly from low-risk (median, 68.1 x 10(6)/L) to intermediate-risk (median, 112.8 x 10(6)/L) and high-risk patients (median 666.1 x 10(6)/L) (F = 4.95; P =.009). When CD34(+) cells were correlated with a severity score on the basis of both myeloproliferative and myelodepletive characteristics of the disease, only the myeloproliferation index was significantly associated with CD34(+) cell level (F = 5.7; P =.000). Overall survival and interval to blast transformation from the time of CD34(+) cell analysis were significantly shorter in patients with more than 300 x 10(6)/L CD34(+) cells (P =.005 and.0005, respectively). In conclusion, the absolute number of CD34(+) circulating cells allows MMM to be distinguished from other Ph(-) CMDs; it is strongly associated with the extent of myeloproliferation and predicts evolution toward blast transformation.


Asunto(s)
Antígenos CD34/análisis , Mielofibrosis Primaria/sangre , Mielofibrosis Primaria/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Recuento de Células , Femenino , Humanos , Hidroxiurea/uso terapéutico , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/patología , Mielofibrosis Primaria/tratamiento farmacológico , Pronóstico , Factores de Riesgo , Bazo/patología , Factores de Tiempo
16.
Blood ; 98(12): 3359-66, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11719375

RESUMEN

Adoptive cellular immunotherapy has proven to be a successful approach in preventing and curing cytomegalovirus infection and Epstein-Barr virus-associated lymphomas after bone marrow transplantation. Translation of this approach for preventing leukemia relapse after bone marrow transplantation might require ex vivo priming and long-term maintenance of leukemia blast-specific T cells. To accomplish this goal, procedures were optimized for the in vitro priming of naive CD8 using dendritic cells activated by CD40 ligation, interleukin-12 (IL-12), and IL-7. Using T lymphocytes and dendritic cells obtained from HLA-matched allogeneic bone marrow transplantation donors and leukemia blasts as a source of tumor antigens, anti-acute myeloid leukemia cytotoxic T lymphocytes (CTLs) were induced. In these experiments, it was found that though it is possible to induce CTLs using immature dendritic cells, IL-12, and IL-7, obtaining long-term CTLs requires the presence of CD4 T cells in the priming phase. Using this approach, long-term antileukemia CTL lines could be generated from 4 of 4 bone marrow donors. Because this procedure does not require definition of the target antigen and because it selects responding cells from a virgin T-cell repertoire, its general application is suggested in adoptive immunotherapy and in the definition of tumor rejection antigens.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/terapia , Linfocitos T Citotóxicos/inmunología , Trasplante de Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Células Dendríticas/inmunología , Antígenos HLA/análisis , Histocompatibilidad , Humanos , Interleucina-12/farmacología , Interleucina-7/farmacología , Leucemia Mieloide Aguda/inmunología , Activación de Linfocitos
17.
Hematol J ; 2(1): 26-32, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11920230

RESUMEN

INTRODUCTION: Normal immature hematopoietic progenitors are relatively well preserved in most patients newly diagnosed with chronic myeloid leukemia, but tend to decline rapidly with time. Such exhaustion could reflect a suppressive effect of the Philadelphia positive clone expansion and/or be induced by Interferon-alpha treatment. MATERIALS AND METHODS: A total of 51 CML patients were classified into three groups. Newly diagnosed untreated patients were group A (n=30). Of the 21 treated individuals with Interferon-alpha, for at least 12 months, 15 showed no cytogenetic response (group B) while six showed persisting major/complete response (group C). Patients belonging to groups A and B were mobilized with chemotherapy plus G-CSF while patients of group C received a short course of G-CSF only. RESULTS: Patients responding to IFN-alpha (group C) showed comparable numbers of bone marrow Ph- long-term culture initiating cells to those of newly diagnosed individuals (group A): 8.5 (<1-65)/10(6) MNC vs 10.5 (<1-30), while non-responders had markedly lower numbers: <1 (<1-5). The amount of Ph- LTC-IC collected was significantly lower in patients of group B 1.8 (0-325)x10(2)/kg than in patients of either group A 31.3 (0-952)x10(2)/kg (P<0.002) or group C 109 (8-259)x10(2)/kg (P<0.01). Interestingly, five patients of group B who had 100% Ph+ metaphases, but Ph- progenitors in their bone marrow, mobilized normal amounts of Ph(-) progenitors. CONCLUSION: These findings suggest that the decline of normal hematopoietic progenitors, currently observed in the majority of CML patients, is not induced by IFN-alpha treatment, but it is likely due to the expanding leukemic clone. They also indicate that normal hematopoietic reservoir is consistently preserved in patients given IFN-alpha early after diagnosis and achieving a stable cytogenetic response.


Asunto(s)
Interferón-alfa/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Cromosoma Filadelfia , Células Madre/efectos de los fármacos , Adulto , Células de la Médula Ósea/efectos de los fármacos , Estudios de Casos y Controles , Recuento de Células , Análisis Citogenético , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Interferón-alfa/administración & dosificación , Leucaféresis , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología
18.
Blood ; 96(13): 4111-7, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11110681

RESUMEN

Many mutations of the housekeeping gene encoding glucose-6-phosphate dehydrogenase (G6PD) cause G6PD deficiency in humans. Some underlie severe forms of chronic nonspherocytic hemolytic anemia (CNSHA) for which there is no definitive treatment. By using retroviral vectors pseudotyped with the vesicular stomatitis virus G glycoprotein that harbor the human G6PD (hG6PD) complementary DNA, stable and lifelong expression of hG6PD was obtained in all the hematopoietic tissues of 16 primary bone marrow transplant (BMT) recipient mice and 14 secondary BMT recipients. These findings demonstrate the integration of a functional gene in totipotent stem cells. The average total G6PD in peripheral blood cells of these transplanted mice, measured as enzyme activity, was twice that of untransplanted control mice. This allowed the inference that the amount of G6PD produced by the transduced gene must be therapeutically effective. With the same vectors both the cloning efficiency and the ability to form embryoid bodies were restored in embryonic stem cells, in which the G6PD gene had been inactivated by targeted homologous recombination, thus effectively rescuing their defective phenotype. Finally, expression of normal human G6PD in hG6PD-deficient primary hematopoietic cells and in human hematopoietic cells engrafted in nonobese diabetic/severe combined immunodeficient mice was obtained. This approach could cure severe CNSHA caused by G6PD deficiency.


Asunto(s)
Células de la Médula Ósea/enzimología , Vectores Genéticos/genética , Deficiencia de Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/genética , Células Madre Hematopoyéticas/enzimología , Glicoproteínas de Membrana , Virus de la Leucemia Murina de Moloney/genética , Virus Reordenados/genética , Virus de la Estomatitis Vesicular Indiana/genética , Células 3T3 , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , ADN Complementario/genética , Inducción Enzimática , Prueba de Complementación Genética , Glucosafosfato Deshidrogenasa/metabolismo , Deficiencia de Glucosafosfato Deshidrogenasa/patología , Supervivencia de Injerto , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Provirus/aislamiento & purificación , ARN Viral/aislamiento & purificación , Quimera por Radiación , Células Madre/citología , Células Madre/enzimología , Transcripción Genética , Transfección , Transgenes , Trasplante Heterólogo , Proteínas del Envoltorio Viral/genética
19.
Blood ; 96(13): 4363-5, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11110715

RESUMEN

X-linked sideroblastic anemia (XLSA) is caused by mutations in the erythroid-specific 5-aminolevulinic acid synthase (ALAS2) gene. An elderly woman who presented with an acquired sideroblastic anemia is studied. Molecular analysis revealed that she was heterozygous for a missense mutation in the ALAS2 gene, but she expressed only the mutated gene in reticulocytes. Her 2 daughters and a granddaughter were heterozygous for this mutation, had normal hemoglobin levels, and expressed the normal ALAS2 gene in reticulocytes. A grandson with a previous diagnosis of thalassemia intermedia was found to be hemizygous for the ALAS2 mutation. Treatment with pyridoxine completely corrected the anemia both in the proband and her grandson. All women who were analyzed in this family showed skewed X-chromosome inactivation in leukocytes, which indicated a hereditary condition associated with unbalanced lyonization. Because the preferentially active X chromosome carried the mutant ALAS2 allele, acquired skewing in the elderly likely worsened the genetic condition and abolished the normal ALAS2 allele expression in the proband. (Blood. 2000;96:4363-4365)


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/genética , Compensación de Dosificación (Genética) , Leucocitos/enzimología , Cromosoma X/genética , 5-Aminolevulinato Sintetasa/deficiencia , Adolescente , Adulto , Anciano , Alelos , Anemia Refractaria/diagnóstico , Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/tratamiento farmacológico , Anemia Sideroblástica/enzimología , Transfusión Sanguínea , Diagnóstico Diferencial , Errores Diagnósticos , Femenino , Hematopoyesis , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Linaje , Piridoxina/uso terapéutico , Talasemia/diagnóstico
20.
Br J Haematol ; 108(3): 621-8, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10759722

RESUMEN

The objective of this study was to evaluate the cycling status of cord blood (CB)-derived colony-forming cells (CFC) and long-term culture-initiating cells (LTC-IC), and their recruitment into the S-phase of the cell cycle. By using the cytosine arabinoside (Ara-C) suicide approach, we found that only small proportions of both CFC and LTC-IC were in the S-phase of the cell cycle. These estimates were confirmed by flow cytometric DNA analysis, which showed that 96 +/- 2% of CB-derived CD34+ cells were in G0/G1 and only 1.6 +/- 0.4% in the S-phase. Staining of CD34+ cells with an antistatin monoclonal antibody, a marker of the G0 phase, indicated that among CD34+ cells with a flow cytometric DNA content typical of the G0/G1 phase 68 +/- 7% of cells were in the G0 phase of the cell cycle. Incubation (24 h) with interleukin 3 (IL-3), recombinant human stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) significantly increased the proportion of cells in the S-phase for both CFC and LTC-IC without inducing any loss in numbers. Flow cytometric DNA analysis also showed an increase in CD34+ cells in the S-phase upon continuous exposure to these cytokines. Our findings indicate that: (i) very few CB-derived CFC or LTC-IC were in the S-phase of the cell cycle; (ii) a substantial amount of CD34+ cells with a flow cytometric DNA content typical of the G0/G1 fraction was cycling, as found in the G1 phase of the cell cycle; and (iii) 24-h incubation with IL-3, SCF and G-CSF could drive a proportion of progenitor cells into the S-phase without reducing their number. These data might be useful for gene transfer protocols and the ex vivo expansion of CB-derived progenitor cells.


Asunto(s)
Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Fase S , Antígenos CD34/inmunología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular , Células Cultivadas , Sangre Fetal/inmunología , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/inmunología , Humanos , Interleucina-3/farmacología , Factor 1 de Elongación Peptídica , Proteínas/análisis , Proteínas Recombinantes , Factor de Células Madre/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA