Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Leukemia ; 31(6): 1398-1407, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27881872

RESUMEN

Future deep space missions to Mars and near-Earth asteroids will expose astronauts to chronic solar energetic particles (SEP) and galactic cosmic ray (GCR) radiation, and likely one or more solar particle events (SPEs). Given the inherent radiosensitivity of hematopoietic cells and short latency period of leukemias, space radiation-induced hematopoietic damage poses a particular threat to astronauts on extended missions. We show that exposing human hematopoietic stem/progenitor cells (HSC) to extended mission-relevant doses of accelerated high-energy protons and iron ions leads to the following: (1) introduces mutations that are frequently located within genes involved in hematopoiesis and are distinct from those induced by γ-radiation; (2) markedly reduces in vitro colony formation; (3) markedly alters engraftment and lineage commitment in vivo; and (4) leads to the development, in vivo, of what appears to be T-ALL. Sequential exposure to protons and iron ions (as typically occurs in deep space) proved far more deleterious to HSC genome integrity and function than either particle species alone. Our results represent a critical step for more accurately estimating risks to the human hematopoietic system from space radiation, identifying and better defining molecular mechanisms by which space radiation impairs hematopoiesis and induces leukemogenesis, as well as for developing appropriately targeted countermeasures.


Asunto(s)
Radiación Cósmica/efectos adversos , Exposición Profesional/efectos adversos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Traumatismos por Radiación/etiología , Esplenomegalia/etiología , Células Madre/patología , Adulto , Animales , Apoptosis , Astronautas , Carga Corporal (Radioterapia) , Proliferación Celular , Exoma/genética , Femenino , Genoma Humano , Humanos , Técnicas In Vitro , Masculino , Ratones , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Dosis de Radiación , Traumatismos por Radiación/patología , Esplenomegalia/patología , Células Madre/metabolismo , Células Madre/efectos de la radiación , Células Tumorales Cultivadas
2.
Am J Transplant ; 14(4): 820-30, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24592822

RESUMEN

Human cytomegalovirus (HCMV) infection is an important cause of morbidity and mortality among both solid organ and hematopoietic stem cell transplant recipients. Identification of cells throughout the body that can potentially serve as a viral reservoir is essential to dissect mechanisms of cell tropism and latency and to develop novel therapies. Here, we tested and compared the permissivity of liver-, brain-, lung (LNG)- and bone marrow (BM)-derived perivascular mesenchymal stromal cells (MSC) to HCMV infection and their ability to propagate and produce infectious virus. Perivascular MSC isolated from the different organs have in common the expression of CD146 and Stro-1. While all these cells were permissive to HCMV infection, the highest rate of HCMV infection was seen with LNG-MSC, as determined by viral copy number and production of viral particles by these cells. In addition, we showed that, although the supernatants from each of the HCMV-infected cultures contained infectious virus, the viral copy number and the quantity and timing of virus production varied among the various organ-specific MSC. Furthermore, using quantitative polymerase chain reaction, we were able to detect HCMV DNA in BM-MSC isolated from 7 out of 19 healthy, HCMV-seropositive adults, suggesting that BM-derived perivascular stromal cells may constitute an unrecognized natural HCMV reservoir.


Asunto(s)
Infecciones por Citomegalovirus/virología , ADN Viral/genética , Células Madre Mesenquimatosas/virología , Adulto , Médula Ósea/metabolismo , Médula Ósea/virología , Encéfalo/citología , Encéfalo/virología , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Feto/virología , Humanos , Hígado/citología , Hígado/virología , Pulmón/citología , Pulmón/virología , Reacción en Cadena de la Polimerasa , Replicación Viral
3.
J Thromb Haemost ; 8(2): 276-85, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19943872

RESUMEN

BACKGROUND: Large animal models that accurately mimic human hemophilia A (HA) are in great demand for developing and testing novel therapies to treat HA. OBJECTIVES: To re-establish a line of sheep exhibiting a spontaneous bleeding disorder closely mimicking severe human HA, fully characterize their clinical presentation, and define the molecular basis for disease. PATIENTS/METHODS: Sequential reproductive manipulations were performed with cryopreserved semen from a deceased affected ram. The resultant animals were examined for hematologic parameters, clinical symptoms, and responsiveness to human FVIII (hFVIII). The full coding region of sheep FVIII mRNA was sequenced to identify the genetic lesion. RESULTS AND CONCLUSIONS: The combined reproductive technologies yielded 36 carriers and 8 affected animals. The latter had almost non-existent levels of FVIII:C and extremely prolonged aPTT, with otherwise normal hematologic parameters. These animals exhibited bleeding from the umbilical cord, prolonged tail and nail cuticle bleeding time, and multiple episodes of severe spontaneous bleeding, including hemarthroses, muscle hematomas and hematuria, all of which responded to hFVIII. Inhibitors of hFVIII were detected in four treated animals, further establishing the preclinical value of this model. Sequencing identified a premature stop codon and frame-shift in exon 14, providing a molecular explanation for HA. Given the decades of experience using sheep to study both normal physiology and a wide array of diseases and the high homology between human and sheep FVIII, this new model will enable a better understanding of HA and facilitate the development and testing of novel treatments that can directly translate to HA patients.


Asunto(s)
Coagulación Sanguínea/genética , Factor VIII/genética , Hemartrosis/genética , Hemofilia A/genética , Factores de Edad , Envejecimiento , Animales , Secuencia de Bases , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Codón sin Sentido , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exones , Factor VIII/metabolismo , Factor VIII/farmacología , Femenino , Predisposición Genética a la Enfermedad , Hemartrosis/sangre , Hemartrosis/tratamiento farmacológico , Hemartrosis/patología , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Hemofilia A/patología , Humanos , Masculino , Datos de Secuencia Molecular , Tiempo de Tromboplastina Parcial , Fenotipo , ARN Mensajero/sangre , Técnicas Reproductivas Asistidas , Ovinos , Especificidad de la Especie
4.
Blood ; 97(11): 3417-23, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11369632

RESUMEN

The successful transduction of hematopoietic stem cells and long-term (28 months) transgene expression within the hematopoietic system following the direct injection of high-titer retroviral vectors into preimmune fetal sheep was previously demonstrated. The present studies extended these analyses for 40 months postinjection and evaluated whether the longevity of transgene expression in this model system was the result of induction of prenatal tolerance to the transgene product. The intraperitoneal injection of retroviral vectors into preimmune sheep fetuses transduces thymic epithelial cells thought to present antigen and thus define self during immune system development. To directly demonstrate induction of tolerance, postnatal sheep were boosted with purified beta-galactosidase and showed that the peripheral blood lymphocytes from in utero-transduced sheep exhibited significantly lower stimulation indices to transduced autologous cells than did control animals and that the in utero-transduced sheep had a reduced ability to mount an antibody response to the vector-encoded beta-galactosidase protein compared with control sheep. Collectively, our results provide evidence that the direct injection of retroviral vectors into preimmune sheep fetuses induces cellular and humoral tolerance to the vector/transgene products and provide an explanation for the duration and stability of transgene expression seen in this model. These results also suggest that even relatively low levels of gene transfer in utero may render the recipient tolerant to the exogenous gene and thus potentially permit the successful postnatal treatment of the recipient. (Blood. 2001;97:3417-3423)


Asunto(s)
Tolerancia Inmunológica , Transfección , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología , Animales , Formación de Anticuerpos , Antígenos/inmunología , Células Epiteliales/enzimología , Femenino , Feto/inmunología , Expresión Génica , Vectores Genéticos , Humanos , Inmunidad Celular , Inmunización , Embarazo , Retroviridae/genética , Ovinos , Timo/embriología , Timo/enzimología , Factores de Tiempo , Transgenes/genética
5.
Exp Hematol ; 28(6): 642-50, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10880750

RESUMEN

We investigated whether gene transfer into hematopoietic cells could be achieved by direct injection of retroviral vector supernatant into the bone marrow space of newborn sheep. Six sheep (5 weeks old) were injected bilaterally with either 1 mL of G1nBgSvNa8.1 vector supernatant (titer: 1 x 10(7)) in each hip (n = 5) or with 3 mL of the same vector preparation/hip (n = 1). In addition, one 3-month-old sheep was injected unilaterally with 1 mL of the same vector preparation. Blood and marrow of these animals were analyzed for the transgene before injection and at intervals thereafter. At 1 week postinjection, an average of 11.6% of the lymphocytes and 25.5% of the granulocytes/monocytes in the marrow, and an average of 0.9% of the lymphocytes and 1.8% of the granulocytes/monocytes in the blood contained and expressed the LacZ gene. The presence/expression of the transgene has persisted for at least 13 months within the blood and bone marrow of these animals. These findings demonstrate that the direct injection of small volumes of high-titer retroviral supernatant into the bone marrow of newborn sheep results in transduction of hematopoietic cells that persists for at least 13 months postinjection.


Asunto(s)
Animales Recién Nacidos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Células Madre Hematopoyéticas/virología , Retroviridae/genética , Animales , Southern Blotting , Médula Ósea , Supervivencia Celular , Citometría de Flujo , Expresión Génica , Genes Reporteros , Vectores Genéticos/genética , Vectores Genéticos/aislamiento & purificación , Inyecciones , Operón Lac , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/biosíntesis , Retroviridae/aislamiento & purificación , Ovinos , Factores de Tiempo , Transfección , beta-Galactosidasa/biosíntesis
6.
Blood ; 95(11): 3620-7, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10828053

RESUMEN

Both in utero and postnatal hematopoietic stem cell (HSC) transplantation would benefit from the development of approaches that produce increased levels of engraftment or a reduction in the period of time required for reconstitution. We used the in utero model of human-sheep HSC transplantation to investigate ways of improving engraftment and differentiation of donor cells after transplantation. We hypothesized that providing a more suitable microenvironment in the form of human stromal cell progenitors simultaneously with the transplanted human HSC would result in higher rates of engraftment or differentiation of the human cells in this xenogeneic model. The results presented here demonstrate that the cotransplantation of both autologous and allogeneic human bone marrow-derived stromal cell progenitors resulted in an enhancement of long-term engraftment of human cells in the bone marrow of the chimeric animals and in earlier and higher levels of donor cells in circulation both during gestation and after birth. By using marked stromal cells, we have also demonstrated that injected stromal cells alone engraft and remain functional within the sheep hematopoietic microenvironment. Application of this method to clinical HSC transplantation could potentially lead to increased levels of long-term engraftment, a reduction in the time for hematopoietic reconstitution, and a means of delivery of foreign genes to the hematopoietic system.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Hematopoyéticas , Células del Estroma/trasplante , Quimera por Trasplante , Trasplante Heterólogo/inmunología , Animales , Feto , Citometría de Flujo , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Inmunofenotipificación , Proteínas Recombinantes/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ovinos , Células del Estroma/citología , Células del Estroma/inmunología , Transfección , Trasplante Homólogo , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
7.
Exp Hematol ; 28(1): 17-30, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10658673

RESUMEN

OBJECTIVE: We have previously reported that directly injecting low-titer retroviral vector supernatant into pre-immune sheep fetuses resulted in the transfer and long-term expression of the bacterial NeoR gene within the hematopoietic system of these animals for over 5 years. In the present studies, we investigated whether using a higher titer vector would enable more efficient transduction and expression of the transgenes within the hematopoetic cells in sheep injected in utero. MATERIALS AND METHODS: Sixteen pre-immune sheep fetuses were injected intraperitoneally with the G1nBgSvNa8.1 helper-free retroviral vector supernatant encoding the bacterial NeoR and LacZ genes (titer: 1x10(7) cfu/mL). RESULTS: Over the 2-year time course of these studies, the presence and expression of the NeoR and LacZ genes were demonstrated in 12 of the 14 animals evaluated by several immunological and biochemical methods. Seven of the 12 sheep examined by flow cytometric analysis contained > or =6% transduced peripheral blood lymphocytes. Vector distribution was widespread without any detectable pathology. Importantly, PCR analyses and breeding experiments demonstrated that the germ line was not altered. CONCLUSIONS: These studies confirmed that direct injection of an engineered retrovirus is a feasible means of safely delivering foreign genes into a developing fetus and thus achieving long-term expression of the transgenes within the recipient's hematopoietic cells. Furthermore, expression of the NeoR gene from these studies was higher than that reported in our previous study in which a lower titer vector was used.


Asunto(s)
Feto , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Transgenes/genética , Animales , Animales Modificados Genéticamente/genética , Células de la Médula Ósea/química , Células de la Médula Ósea/enzimología , Ensayo de Unidades Formadoras de Colonias , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Granulocitos/citología , Inyecciones Intraperitoneales , Kanamicina Quinasa/genética , Kanamicina Quinasa/metabolismo , Linfocitos/química , Linfocitos/enzimología , Reacción en Cadena de la Polimerasa , Retroviridae/genética , Ovinos , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
8.
Ann N Y Acad Sci ; 872: 220-31; discussion 231-2, 1999 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-10372125

RESUMEN

The fetal sheep competitive engraftment model of human hematopoietic stem cells (HSC) was used to evaluate the in vivo engraftment potential of human bone marrow CD34- Lin- cells. Transplantation of CD34- Lin- cells into primary hosts resulted in the long-term (> 1 year) engraftment and multilineage donor cell/progenitor expression with production of significant numbers of CD34+ cells. Secondary transplantation and limiting dilution studies confirmed the presence in human CD34- fraction of HSC with in vivo long-term engraftment and multilineage differentiation potentials.


Asunto(s)
Antígenos CD34/análisis , Células de la Médula Ósea/citología , Supervivencia de Injerto/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Trasplante Heterólogo/fisiología , Animales , Antígenos CD/análisis , Biomarcadores/análisis , Células de la Médula Ósea/inmunología , Células Cultivadas , Feto , Células Madre Hematopoyéticas/inmunología , Humanos , Ovinos , Trasplante Heterólogo/inmunología
9.
Hum Gene Ther ; 9(11): 1571-85, 1998 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-9694156

RESUMEN

We investigated whether directly injecting retroviral vectors into preimmune fetuses could result in the transfer and long-term expression of exogenous genes. Twenty-nine preimmune sheep fetuses were injected with helper-free retroviral vector preparations. Twenty-two fetuses survived to term, 4 of which were sacrificed at birth. Of the remaining 18 animals, 3 were controls and 15 had received vector preparations. Twelve of these 15 animals demonstrated transduction of hematopoietic cells when blood and marrow were analyzed by neo(r)-specific PCR. Eight experimental sheep have been followed for 5 years, during which time we have consistently observed proviral DNA and G418-resistant hematopoetic progenitors. The G418-resistant colonies were positive when analyzed by neo(r)-specific PCR. neo(r) gene expression was also demonstrated using several immunological and biochemical methods. The transduction of hematopoietic stem cells was confirmed when lambs transplanted with bone marrow from in utero-transduced sheep exhibited neo(r) activity in marrow and blood. Vector distribution was widespread in primary animals without pathology. PCR analysis indicates that the germ line was not altered. These studies demonstrate that direct injection of an engineered retrovirus is a feasible means of safely delivering a foreign gene to a developing fetus and achieving long-term expression without modifying the germ line of the recipient.


Asunto(s)
Enfermedades Fetales/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Kanamicina Quinasa/genética , Kanamicina Quinasa/metabolismo , Retroviridae/genética , Animales , Southern Blotting , Ensayo de Inmunoadsorción Enzimática , Células Precursoras Eritroides/efectos de los fármacos , Femenino , Enfermedades Fetales/genética , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Gentamicinas/farmacología , Trasplante de Células Madre Hematopoyéticas , Hibridación Fluorescente in Situ , Inyecciones Intraperitoneales , Reacción en Cadena de la Polimerasa , Embarazo , Análisis de Secuencia de ADN , Ovinos
10.
Exp Hematol ; 25(12): 1278-85, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9357972

RESUMEN

The human cytomegaloviruses (HCMVs) appear to have the potential to disrupt production of hematopoietic cytokines. We examined the production of granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-8 by cultured and CMV-infected human umbilical vein endothelial cells (HUVECs) and compared this production with that of uninfected cells. Endothelial cells are, among other things, an integral component of human bone marrow stroma, and are responsible for production of factors that modulate the proliferation and differentiation of human hematopoietic progenitors. HCMV infection increased the production of GM-CSF in IL-1-primed HUVECs without altering GM-CSF levels in infected but unprimed HUVECs. However, this same virus was capable of causing increased production of the inhibitory cytokine IL-8. Both the viral pellet and the cleared viral supernatant appeared to contribute equally to the increased IL-8 and GM-CSF production, because each of these preparations alone was capable of exerting only half the effect seen with whole virus preparations. That both live virus and soluble protein factors within the viral stock contributed to the enhancement in GM-CSF and IL-8 production was further confirmed by inactivation with either ultraviolet or heat treatment of the viral stocks. Although the identity of the factor within the HCMV stock that contributes to this effect remains unknown, studies conducted in the presence of neutralizing antibodies or polymyxin B ruled out a role for tumor necrosis factor-alpha, IL-6, or endotoxin, all known inducers of GM-CSF. These studies indicate that HCMVs can exert both direct and indirect effects on the production of the hematopoietic factor GM-CSF and the inflammatory/inhibitory cytokine IL-8.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Interleucina-8/biosíntesis , Células Cultivadas , Citomegalovirus/patogenicidad , Citomegalovirus/efectos de la radiación , Endotelio Vascular , Calor , Humanos , Interferón gamma/farmacología , Interleucina-1/farmacología , Rayos Ultravioleta , Venas Umbilicales
11.
Blood ; 83(2): 370-6, 1994 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8286737

RESUMEN

In an effort to study whether human cytomegalovirus (HCMV) can disrupt the balanced cytokine network that controls human hematopoiesis, we investigated the ability of a laboratory strain HCMV (AD169) to alter the production of interleukin-6 (IL-6) by cultured endothelial cells (HUVECs). ECs are important components of human bone marrow stroma and produce factors that stimulate the proliferation and differentiation of human hematopoietic progenitors. HCMV was able to greatly increase production of both mRNA and protein for IL-6 in unprimed HUVECs. When we discriminated between viral pellet and cleared viral supernatants, the supernatants induced an increase in mRNA at 30 minutes and protein by 2 hours, whereas an increase in IL-6 caused by virus itself did not become evident until 12 hours. The possibility that IL-6 induction was simply caused by the presence in the viral stock of endotoxin, IL-1 alpha, IL-1 beta, tumor necrosis factor alpha, or IL-4, all known inducers of IL-6 in HUVECs, was ruled out by the addition of polymyxin B and appropriate neutralizing antibodies. These findings show that HCMV is capable of directly and indirectly modulating the production by HUVECs of IL-6, one of the cytokines involved in the process of hematopoiesis.


Asunto(s)
Citomegalovirus/patogenicidad , Endotelio Vascular/metabolismo , Interleucina-6/biosíntesis , Células Cultivadas , Humanos , Interleucina-1/farmacología , Interleucina-4/farmacología , Interleucina-6/genética , ARN Mensajero/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA