Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
2.
J Allergy Clin Immunol ; 142(3): 904-913.e3, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29355678

RESUMEN

BACKGROUND: Mutations in the perforin 1 (PRF1) gene account for up to 58% of familial hemophagocytic lymphohistiocytosis syndromes. The resulting defects in effector cell cytotoxicity lead to hypercytokinemia and hyperactivation with inflammation in various organs. OBJECTIVE: We sought to determine whether autologous gene-corrected T cells can restore cytotoxic function, reduce disease activity, and prevent hemophagocytic lymphohistiocytosis (HLH) symptoms in in vivo models. METHODS: We developed a gammaretroviral vector to transduce murine CD8 T cells in the Prf-/- mouse model. To verify functional correction of Prf-/- CD8 T cells in vivo, we used a lymphocytic choriomeningitis virus (LCMV) epitope-transfected murine lung carcinoma cell tumor model. Furthermore, we challenged gene-corrected and uncorrected mice with LCMV. One patient sample was transduced with a PRF1-encoding lentiviral vector to study restoration of cytotoxicity in human cells. RESULTS: We demonstrated efficient engraftment and functional reconstitution of cytotoxicity after intravenous administration of gene-corrected Prf-/- CD8 T cells into Prf-/- mice. In the tumor model infusion of Prf-/- gene-corrected CD8 T cells eliminated the tumor as efficiently as transplantation of wild-type CD8 T cells. Similarly, mice reconstituted with gene-corrected Prf-/- CD8 T cells displayed complete protection from the HLH phenotype after infection with LCMV. Patients' cells showed correction of cytotoxicity in human CD8 T cells after transduction. CONCLUSION: These data demonstrate the potential application of T-cell gene therapy in reconstituting cytotoxic function and protection against HLH in the setting of perforin deficiency.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Coriomeningitis Linfocítica/terapia , Linfohistiocitosis Hemofagocítica/terapia , Perforina/genética , Animales , Línea Celular Tumoral , Preescolar , Terapia Genética , Humanos , Virus de la Coriomeningitis Linfocítica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Cytotherapy ; 19(6): 710-720, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28428057

RESUMEN

BACKGROUND AIMS: Natural killer (NK) cells have the potential to become a successful immunotherapy as they can target malignant cells without being direct effectors of graft-versus-host disease. Our group has previously shown that large numbers of functional NK cells can be differentiated in vitro from umbilical cord blood (CB) CD34+ cells. To produce a clinically relevant and effective immunotherapy, we hypothesized that it is essential that the NK cells are able to proliferate and persist in vivo while maintaining an optimal activation status and killing capacity. METHODS: We evaluated the proliferation capacity, telomere length and terminal differentiation markers expressed by NK cells differentiated in vitro. We also determined how their cytotoxicity compared with peripheral blood (PB) NK cells and CBNK cells when targeting patient acute myeloid leukemia (AML) blasts and solid tumor cell lines. RESULTS: We found that the differentiated NK cells could respond to interleukin-2 and proliferate in vitro. Telomere length was significantly increased, whereas CD57 expression was significantly reduced compared with PBNK cells. The cytotoxicity of the differentiated NK cells was equivalent to that of the PBNK and CBNK cell controls, and priming consistently led to higher levels of killing of patient leukemic blasts and solid tumor cell lines in vitro. Interestingly, this activation step was not required to observe killing of patient AML blasts in vivo. CONCLUSION: We are able to generate NK cells from CBCD34+ cells in high numbers, allowing for multiple infusions of highly cytotoxic NK cells that have potential to further proliferate in vivo, making them a desirable product for application as an immunotherapy in the clinic.


Asunto(s)
Antígenos CD34/metabolismo , Sangre Fetal/citología , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Humanos , Interleucina-2/farmacología , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Leucemia Mieloide Aguda/terapia
4.
Blood ; 124(16): 2514-22, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25185261

RESUMEN

Epstein-Barr virus (EBV)-associated posttransplant lymphoma (PTLD) is a major cause of morbidity/mortality after hematopoietic stem cell (SCT) or solid organ (SOT) transplant. Adoptive immunotherapy with EBV-specific cytotoxic lymphocytes (CTLs), although effective in SCT, is less successful after SOT where lifelong immunosuppression therapy is necessary. We have genetically engineered EBV-CTLs to render them resistant to calcineurin (CN) inhibitor FK506 through retroviral transfer of a calcineurin A mutant (CNA12). Here we examined whether or not FK506-resistant EBV-CTLs control EBV-driven tumor progression in the presence of immunosuppression in a xenogeneic mouse model. NOD/SCID/IL2rγ(null) mice bearing human B-cell lymphoma were injected with autologous CTLs transduced with either CNA12 or eGFP in the presence/absence of FK506. Adoptive transfer of autologous CNA12-CTLs induced dramatic lymphoma regression despite the presence of FK506, whereas eGFP-CTLs did not. CNA12-CTLs persisted longer, homed to the tumor, and expanded more than eGFP-CTLs in mice treated with FK506. Mice receiving CNA12-CTLs and treated with FK506 survived significantly longer than control-treated animals. Our results demonstrate that CNA12-CTL induce regression of EBV-associated tumors in vivo despite ongoing immunosuppression. Clinical application of this novel approach may enhance the efficacy of adoptive transfer of EBV-CTL in SOT patients developing PTLD without the need for reduction in immunosuppressive therapy.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Terapia Genética , Inmunoterapia Adoptiva , Linfoma/terapia , Linfoma/virología , Linfocitos T Citotóxicos/trasplante , Linfocitos T Citotóxicos/virología , Animales , Calcineurina/genética , Inhibidores de la Calcineurina/farmacología , Resistencia a Medicamentos , Ingeniería Genética/métodos , Terapia Genética/métodos , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Inmunosupresores/farmacología , Inmunoterapia Adoptiva/métodos , Linfoma/genética , Linfoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Tacrolimus/farmacología , Transducción Genética
5.
Stem Cells ; 33(1): 122-32, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25186828

RESUMEN

Unmatched allogeneic in utero stem cell transplantation (IUSCT) produces poor engraftment unless the fetus has congenital immunodeficiency, probably because of maternal and fetal immune responses to injected cells. We studied the functional hematopoietic potential of transduced green fluorescent protein (GFP+) sheep amniotic fluid (AF) stem cells, before and after autologous IUSCT. CD34+ cells were selected from first trimester sheep AF, transduced overnight, and injected intravenously into NOD-SCID-gamma (NSG) mice. At 3 months, primary recipient bone marrow (BM) was injected into secondary NSG recipients. GFP+ cells were detected in the hematopoietic organs and peripheral blood of primary and secondary recipients at 3 months. Autologous IUSCT (transduced GFP+CD34+AF) was performed in fetal sheep. Six months postnatally, lamb BM was injected into secondary NSG recipients. GFP+ cells were detected in the peripheral blood of primary and secondary recipients. This confirms the hematopoietic potential of AF stem cells supporting the concept of autologous IUSCT to treat congenital hematopoietic disease.


Asunto(s)
Líquido Amniótico/citología , Líquido Amniótico/metabolismo , Antígenos CD34/biosíntesis , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Femenino , Feto/cirugía , Ratones , Ratones Endogámicos NOD , Ratones SCID , Embarazo , Ovinos , Trasplante Autólogo , Trasplante Heterólogo
6.
Mol Ther ; 23(4): 737-45, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25523759

RESUMEN

Defects in perforin lead to the failure of T and NK cell cytotoxicity, hypercytokinemia, and the immune dysregulatory condition known as familial hemophagocytic lymphohistiocytosis (FHL). The only curative treatment is allogeneic hematopoietic stem cell transplantation which carries substantial risks. We used lentiviral vectors (LV) expressing the human perforin gene, under the transcriptional control of the ubiquitous phosphoglycerate kinase promoter or a lineage-specific perforin promoter, to correct the defect in different murine models. Following LV-mediated gene transfer into progenitor cells from perforin-deficient mice, we observed perforin expression in mature T and NK cells, and there was no evidence of progenitor cell toxicity when transplanted into irradiated recipients. The resulting perforin-reconstituted NK cells showed partial recovery of cytotoxicity, and we observed full recovery of cytotoxicity in polyclonal CD8(+) T cells. Furthermore, reconstituted T cells with defined antigen specificity displayed normal cytotoxic function against peptide-loaded targets. Reconstituted CD8(+) lymphoblasts had reduced interferon-γ secretion following stimulation in vitro, suggesting restoration of normal immune regulation. Finally, upon viral challenge, mice with >30% engraftment of gene-modified cells exhibited reduction of cytokine hypersecretion and cytopenias. This study demonstrates the potential of hematopoietic stem cell gene therapy as a curative treatment for perforin-deficient FHL.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/metabolismo , Células Asesinas Naturales/inmunología , Linfohistiocitosis Hemofagocítica/terapia , Perforina/genética , Animales , Linfohistiocitosis Hemofagocítica/inmunología , Ratones , Ratones Transgénicos , Fenotipo
7.
Blood ; 121(7): 1073-6, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23223356

RESUMEN

X-linked lymphoproliferative disease (XLP1) arises from mutations in the gene encoding SLAM-associated protein (SAP) and leads to abnormalities of NKT-cell development, NK-cell cytotoxicity, and T-dependent humoral function. Curative treatment is limited to allogeneic hematopoietic stem cell (HSC) transplantation. We tested whether HSC gene therapy could correct the multilineage defects seen in SAP(-/-) mice. SAP(-/-) murine HSCs were transduced with lentiviral vectors containing either SAP or reporter gene before transplantation into irradiated recipients. NKT-cell development was significantly higher and NK-cell cytotoxicity restored to wild-type levels in mice receiving the SAP vector in comparison to control mice. Baseline immunoglobulin levels were significantly increased and T-dependent humoral responses to NP-CGG, including germinal center formation, were restored in SAP-transduced mice.We demonstrate for the first time that HSC gene transfer corrects the cellular and humoral defects in SAP(-/-) mice providing proof of concept for gene therapy in XLP1.


Asunto(s)
Terapia Genética/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/terapia , Animales , Linaje de la Célula , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas , Inmunidad Celular , Inmunidad Humoral , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/inmunología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Ratones , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria
8.
Blood ; 121(13): 2542-52, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23305739

RESUMEN

Megakaryocytes give rise to platelets via extension of proplatelet arms, which are released through the vascular sinusoids into the bloodstream. Megakaryocytes and their precursors undergo varying interactions with the extracellular environment in the bone marrow during their maturation and positioning in the vascular niche. We demonstrate that podosomes are abundant in primary murine megakaryocytes adherent on multiple extracellular matrix substrates, including native basement membrane. Megakaryocyte podosome lifetime and density, but not podosome size, are dependent on the type of matrix, with podosome lifetime dramatically increased on collagen fibers compared with fibrinogen. Podosome stability and dynamics depend on actin cytoskeletal dynamics but not matrix metalloproteases. However, podosomes degrade matrix and appear to be important for megakaryocytes to extend protrusions across a native basement membrane. We thus demonstrate for the first time a fundamental requirement for podosomes in megakaryocyte process extension across a basement membrane, and our results suggest that podosomes may have a role in proplatelet arm extension or penetration of basement membrane.


Asunto(s)
Membrana Basal/fisiología , Extensiones de la Superficie Celular/fisiología , Matriz Extracelular/metabolismo , Megacariocitos/fisiología , Animales , Membrana Basal/metabolismo , Plaquetas/metabolismo , Plaquetas/fisiología , Extensiones de la Superficie Celular/metabolismo , Células Cultivadas , Fibrinógeno/metabolismo , Células HEK293 , Humanos , Recién Nacido , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miosina Tipo II/metabolismo
9.
Mol Ther ; 22(3): 607-622, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24256635

RESUMEN

Gene transfer into autologous hematopoietic stem cells by γ-retroviral vectors (gRV) is an effective treatment for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). However, current gRV have significant potential for insertional mutagenesis as reported in clinical trials for other primary immunodeficiencies. To improve the efficacy and safety of ADA-SCID gene therapy (GT), we generated a self-inactivating lentiviral vector (LV) with a codon-optimized human cADA gene under the control of the short form elongation factor-1α promoter (LV EFS ADA). In ADA(-/-) mice, LV EFS ADA displayed high-efficiency gene transfer and sufficient ADA expression to rescue ADA(-/-) mice from their lethal phenotype with good thymic and peripheral T- and B-cell reconstitution. Human ADA-deficient CD34(+) cells transduced with 1-5 × 10(7) TU/ml had 1-3 vector copies/cell and expressed 1-2x of normal endogenous levels of ADA, as assayed in vitro and by transplantation into immune-deficient mice. Importantly, in vitro immortalization assays demonstrated that LV EFS ADA had significantly less transformation potential compared to gRV vectors, and vector integration-site analysis by nrLAM-PCR of transduced human cells grown in immune-deficient mice showed no evidence of clonal skewing. These data demonstrated that the LV EFS ADA vector can effectively transfer the human ADA cDNA and promote immune and metabolic recovery, while reducing the potential for vector-mediated insertional mutagenesis.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Agammaglobulinemia/inmunología , Agammaglobulinemia/terapia , Vectores Genéticos/efectos adversos , Lentivirus/genética , Factor 1 de Elongación Peptídica/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/inmunología , Adenosina Desaminasa/metabolismo , Agammaglobulinemia/genética , Agammaglobulinemia/patología , Animales , Linfocitos B/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/patología , Linfocitos T/inmunología , Transducción Genética , Integración Viral
10.
J Exp Med ; 204(9): 2213-24, 2007 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-17724125

RESUMEN

Specific mutations in the human gene encoding the Wiskott-Aldrich syndrome protein (WASp) that compromise normal auto-inhibition of WASp result in unregulated activation of the actin-related protein 2/3 complex and increased actin polymerizing activity. These activating mutations are associated with an X-linked form of neutropenia with an intrinsic failure of myelopoiesis and an increase in the incidence of cytogenetic abnormalities. To study the underlying mechanisms, active mutant WASp(I294T) was expressed by gene transfer. This caused enhanced and delocalized actin polymerization throughout the cell, decreased proliferation, and increased apoptosis. Cells became binucleated, suggesting a failure of cytokinesis, and micronuclei were formed, indicative of genomic instability. Live cell imaging demonstrated a delay in mitosis from prometaphase to anaphase and confirmed that multinucleation was a result of aborted cytokinesis. During mitosis, filamentous actin was abnormally localized around the spindle and chromosomes throughout their alignment and separation, and it accumulated within the cleavage furrow around the spindle midzone. These findings reveal a novel mechanism for inhibition of myelopoiesis through defective mitosis and cytokinesis due to hyperactivation and mislocalization of actin polymerization.


Asunto(s)
Actinas/metabolismo , Citocinesis , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Mitosis , Neutropenia/metabolismo , Neutropenia/patología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Aberraciones Cromosómicas , Cromosomas Humanos , Citocinesis/efectos de los fármacos , ADN , Depsipéptidos/farmacología , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Mitosis/efectos de los fármacos , Proteínas Mutantes/metabolismo , Poliploidía , Proteínas Recombinantes de Fusión/metabolismo , Transgenes
11.
Hum Mol Genet ; 20(15): 2905-13, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21593218

RESUMEN

We present a novel and efficient non-integrating gene expression system in human embryonic stem cells (hESc) utilizing human artificial chromosomes (HAC), which behave as autonomous endogenous host chromosomes and segregate correctly during cell division. HAC are important vectors for investigating the organization and structure of the kinetochore, and gene complementation. HAC have so far been obtained in immortalized or tumour-derived cell lines, but never in stem cells, thus limiting their potential therapeutic application. In this work, we modified the herpes simplex virus type 1 amplicon system for efficient transfer of HAC DNA into two hESc. The deriving stable clones generated green fluorescent protein gene-expressing HAC at high frequency, which were stably maintained without selection for 3 months. Importantly, no integration of the HAC DNA was observed in the hESc lines, compared with the fibrosarcoma-derived control cells, where the exogenous DNA frequently integrated in the host genome. The hESc retained pluripotency, differentiation and teratoma formation capabilities. This is the first report of successfully generating gene expressing de novo HAC in hESc, and is a significant step towards the genetic manipulation of stem cells and potential therapeutic applications.


Asunto(s)
Cromosomas Artificiales Humanos/metabolismo , Células Madre Embrionarias/metabolismo , Neuronas/citología , Neuronas/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Cromosomas Artificiales Humanos/genética , Células Madre Embrionarias/citología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Herpesvirus Humano 1/genética , Humanos
12.
Blood ; 118(9): 2492-501, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21690559

RESUMEN

Rearrangement of the cytoskeleton in T cells plays a critical role in the organization of a complex signaling interface referred to as immunologic synapse (IS). Surprisingly, the contribution of antigen presenting cells, in particular dendritic cells (DCs), to the structure and function of the IS has not been investigated in as much detail. We have used a natural model of cytoskeletal dysfunction caused by deficiency of the Wiskott-Aldrich syndrome protein (WASp) to explore the contribution of the DC cytoskeleton to IS formation and to T-cell priming. In an antigen-specific system, T-DC contacts were found to be less stable when DCs alone lacked WASp, and associated with multiple defects of IS structure. As a consequence, DCs were unable to support normal IL-12 secretion, and events downstream of TCR signaling were abrogated, including increased calcium flux, microtubule organizing center (MTOC) polarization, phosphorylation of ZAP-70, and T-cell proliferation. Formation of an effective signaling interface is therefore dependent on active cytoskeletal rearrangements in DCs even when T cells are functionally competent. Deficiency of DC-mediated activities may contribute significantly to the varied immunodysregulation observed in patients with WAS, and also in those with limited myeloid reconstitution after allogeneic hematopoietic stem cell transplantation.


Asunto(s)
Citoesqueleto/ultraestructura , Células Dendríticas/ultraestructura , Sinapsis Inmunológicas/ultraestructura , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Señalización del Calcio/inmunología , Movimiento Celular , Cruzamientos Genéticos , Genes Reporteros , Prueba de Complementación Genética , Humanos , Sinapsis Inmunológicas/inmunología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Linfocitos T/ultraestructura , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/inmunología
13.
Haematologica ; 98(8): 1300-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23445877

RESUMEN

The Wiskott-Aldrich syndrome protein is an essential cytoskeleton regulator found in cells of the hematopoietic lineage and controls the motility of leukocytes. The impact of WAS gene deficiency on the mobilization of hematopoietic progenitor/stem cells in circulation has remained unexplored but information would be pertinent in the context of autologous gene therapy of Wiskott-Aldrich syndrome. The response to granulocyte-colony stimulating factor mobilization was investigated in a murine WAS knock-out model of the disease, by measuring hematologic parameters, circulation and engraftment of hematopoietic progenitor/stem cells. In the steady-state, adult WAS knock-out mice have B-cell lymphopenia, marked neutrophilia, increased counts of circulating hematopoietic progenitor cells and splenomegaly, presumably caused by the retention of hematopoietic progenitor cells due to high levels of splenic CXCL12. In spite of these anomalies, the administration of granulocyte-colony-stimulating factor mobilizes progenitor/stem cells in WAS knock-out mice to the same level and with the same kinetics as in wild-type control mice. Mobilized peripheral blood cells from WAS knock-out mice can be transduced and are able to engraft into lethally-irradiated hosts reconstituting multiple lineages of cells and providing more effective radio-protection than mobilized cells from wild-type control mice. Surprisingly, the homing and the peripheral blood recovery of B lymphocytes was influenced by the background of the host. Thus, in the absence of Wiskott-Aldrich syndrome protein, effective mobilization is achieved but partial correction may occur as a result of an abnormal hematopoietic environment.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/terapia , Animales , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Proteínas Recombinantes/farmacología
14.
Mol Ther ; 20(7): 1400-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22434141

RESUMEN

Some gene therapy strategies are compromised by the levels of gene expression required for therapeutic benefit, and also by the breadth of cell types that require correction. We designed a lentiviral vector system in which a transgene is under the transcriptional control of the short form of constitutively acting elongation factor 1α promoter (EFS) combined with essential elements of the locus control region of the ß-globin gene (ß-LCR). We show that the ß-LCR can upregulate EFS activity specifically in erythroid cells but does not alter EFS activity in myeloid or lymphoid cells. Experiments using the green fluorescent protein (GFP) reporter or the human adenosine deaminase (ADA) gene demonstrate 3-7 times upregulation in vitro but >20 times erythroid-specific upregulation in vivo, the effects of which were sustained for 1 year. The addition of the ß-LCR did not alter the mutagenic potential of the vector in in vitro mutagenesis (IM) assays although microarray analysis showed that the ß-LCR upregulates ~9% of neighboring genes. This vector design therefore combines the benefits of multilineage gene expression with high-level erythroid expression, and has considerable potential for correction of multisystem diseases including certain lysosomal storage diseases through a hematopoietic stem cell (HSC) gene therapy approach.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Región de Control de Posición , Factor 1 de Elongación Peptídica/genética , Factores de Elongación de Péptidos/genética , Globinas beta/genética , Adenosina Desaminasa/genética , Animales , Línea Celular , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Células Madre Hematopoyéticas , Humanos , Células Jurkat , Lentivirus/genética , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Células U937 , Regulación hacia Arriba
15.
Mol Ther ; 20(10): 1953-67, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22760542

RESUMEN

Induced pluripotent stem cells (iPSCs) with potential for therapeutic applications can be derived from somatic cells via ectopic expression of a set of limited and defined transcription factors. However, due to risks of random integration of the reprogramming transgenes into the host genome, the low efficiency of the process, and the potential risk of virally induced tumorigenicity, alternative methods have been developed to generate pluripotent cells using nonintegrating systems, albeit with limited success. Here, we show that c-KIT+ human first-trimester amniotic fluid stem cells (AFSCs) can be fully reprogrammed to pluripotency without ectopic factors, by culture on Matrigel in human embryonic stem cell (hESC) medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA). The cells share 82% transcriptome identity with hESCs and are capable of forming embryoid bodies (EBs) in vitro and teratomas in vivo. After long-term expansion, they maintain genetic stability, protein level expression of key pluripotency factors, high cell-division kinetics, telomerase activity, repression of X-inactivation, and capacity to differentiate into lineages of the three germ layers, such as definitive endoderm, hepatocytes, bone, fat, cartilage, neurons, and oligodendrocytes. We conclude that AFSC can be utilized for cell banking of patient-specific pluripotent cells for potential applications in allogeneic cellular replacement therapies, pharmaceutical screening, and disease modeling.


Asunto(s)
Líquido Amniótico/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ácido Valproico/farmacología , Líquido Amniótico/citología , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Genoma Humano , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación , Cinética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Análisis de Secuencia de ADN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma , Transgenes , Inactivación del Cromosoma X/efectos de los fármacos
16.
Br J Haematol ; 158(6): 778-87, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22816563

RESUMEN

In vitro exposure of haematopoietic stem and progenitor cells (HSPC) to cytokines in expansion or gene therapy protocols reduces homing and engraftment in vivo. We have previously reported that this is related in part to altered tissue specificity of short-term homing, leading to loss of cells in non-haematopoietic tissues. Here we demonstrate that defective engraftment persists when cultured HSPC are transplanted by intrabone injection. Changes in engraftment function occur within 24 h of cytokine exposure, and are evident when engraftment is analysed solely in the injected bone. A novel ex vivo model of the bone marrow was developed, in which the attachment of infused HSPC in rodent long bones is reduced following culture with cytokines. Finally, cultured HSPC demonstrated reduced adhesion to N-cadherin, osteopontin and vascular cell-adhesion molecule-1, ligands present in bone marrow niches. These changes in adhesive function occur rapidly, and are not related to downregulation of the relevant receptors. Our findings suggest that cytokine exposure of adult human HSPC results in altered adhesion within bone marrow niches, further leading to reduced engraftment potential in vivo.


Asunto(s)
Médula Ósea/patología , Adhesión Celular/fisiología , Factores Estimulantes de Colonias/farmacología , Supervivencia de Injerto/fisiología , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Nicho de Células Madre/fisiología , Adulto , Animales , Antígenos CD34/análisis , Células Cultivadas/efectos de los fármacos , Células Cultivadas/trasplante , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Organismos Libres de Patógenos Específicos
17.
Haematologica ; 97(5): 687-91, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22133775

RESUMEN

Podosomes are actin-based adhesions involved in migration of cells that have to cross tissue boundaries such as myeloid cells. The Wiskott Aldrich Syndrome Protein regulates de novo actin polymerization during podosome formation and it is cleaved by the protease calpain during podosome disassembly. The mechanisms that may induce the Wiskott Aldrich Syndrome Protein cleavage by calpain remain undetermined. We now report that in myeloid cells, tyrosine phosphorylation of the Wiskott Aldrich Syndrome Protein-tyrosine291 (Human)/tyrosine293 (mouse) not only enhances Wiskott Aldrich Syndrome Protein-mediated actin polymerization but also promotes its calpain-dependent degradation during podosome disassembly. We also show that activation of the Wiskott Aldrich Syndrome Protein leading to podosome formation occurs independently of tyrosine phosphorylation in spleen-derived dendritic cells. We conclude that tyrosine phosphorylation of the Wiskott Aldrich Syndrome Protein integrates dynamics of actin and cell adhesion proteins during podosome disassembly required for mobilization of myeloid cells during the immune response.


Asunto(s)
Citoesqueleto de Actina/fisiología , Calpaína/metabolismo , Estructuras de la Membrana Celular/metabolismo , Tirosina/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Animales , Adhesión Celular , Movimiento Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Técnica del Anticuerpo Fluorescente , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Mieloides/metabolismo , Fosforilación , Unión Proteica
18.
Mol Ther ; 19(1): 122-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20978475

RESUMEN

X-linked chronic granulomatous disease (X-CGD) is a primary immunodeficiency caused by mutations in the CYBB gene encoding the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase catalytic subunit gp91(phox). A recent clinical trial for X-CGD using a spleen focus-forming virus (SFFV)-based γ-retroviral vector has demonstrated clear therapeutic benefits in several patients although complicated by enhancer-mediated mutagenesis and diminution of effectiveness over time due to silencing of the viral long terminal repeat (LTR). To improve safety and efficacy, we have designed a lentiviral vector that directs transgene expression primarily in myeloid cells. To this end, we created a synthetic chimeric promoter that contains binding sites for myeloid transcription factors CAAT box enhancer-binding family proteins (C/EBPs) and PU.1, which are highly expressed during granulocytic differentiation. As predicted, the chimeric promoter regulated higher reporter gene expression in myeloid than in nonmyeloid cells, and in human hematopoietic progenitors upon granulocytic differentiation. In a murine model of stem cell gene therapy for X-CGD, the chimeric vector resulted in high levels of gp91(phox) expression in committed myeloid cells and granulocytes, and restored normal NADPH-oxidase activity. These findings were recapitulated in human neutrophils derived from transduced X-CGD CD34(+) cells in vivo, and suggest that the chimeric promoter will have utility for gene therapy of myeloid lineage disorders such as CGD.


Asunto(s)
Catepsina G/genética , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/terapia , Células Mieloides/fisiología , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Recombinantes de Fusión/genética , Transgenes , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Genes Ligados a X , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Granulocitos/metabolismo , Enfermedad Granulomatosa Crónica/enzimología , Enfermedad Granulomatosa Crónica/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Datos de Secuencia Molecular , Mutagénesis/genética , Células Mieloides/metabolismo , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Virus Formadores de Foco en el Bazo/genética , Virus Formadores de Foco en el Bazo/metabolismo , Células Madre/metabolismo , Secuencias Repetidas Terminales , Transactivadores/metabolismo
19.
Proc Natl Acad Sci U S A ; 106(37): 15738-43, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19805221

RESUMEN

The Wiskott-Aldrich syndrome protein (WASp) is a key cytoskeletal regulator in hematopoietic cells. Covalent modification of a conserved tyrosine by phosphorylation has emerged as an important potential determinant of activity, although the physiological significance remains uncertain. In a murine knockin model, mutation resulting in inability to phosphorylate Y293 (Y293F) mimicked many features of complete WASp-deficiency. Although a phosphomimicking mutant Y293E conferred enhanced actin-polymerization, the cellular phenotype was similar due to functional dysregulation. Furthermore, steady-state levels of Y293E-WASp were markedly reduced compared to wild-type WASp and Y293F-WASp, although partially recoverable by treatment of cells with proteasome inhibitors. Consequently, tyrosine phosphorylation plays a critical role in normal activation of WASp in vivo, and is indispensible for multiple tasks including proliferation, phagocytosis, chemotaxis, and assembly of adhesion structures. Furthermore, it may target WASp for proteasome-mediated degradation, thereby providing a default mechanism for self-limiting stimulation of the Arp2/3 complex.


Asunto(s)
Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Línea Celular , Movimiento Celular , Chlorocebus aethiops , Hematopoyesis , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Fagocitosis , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tirosina/química , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/química , Proteína del Síndrome de Wiskott-Aldrich/genética
20.
J Exp Med ; 202(10): 1399-410, 2005 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-16287709

RESUMEN

Tumors have evolved elaborate mechanisms for evading immune detection, such as production of immunoinhibitory cytokines and down-regulation of major histocompatibility complex (MHC) expression. We have studied PAX3-FKHR as an example of an oncogenic fusion protein associated with an aggressive metastatic cancer. We show that PAX3-FKHR alters expression of genes that are normally regulated by Janus kinase/signal transducer and activator of transcription (STAT) signaling pathways. This occurs as a result of a specific interaction between PAX3-FKHR and the STAT3 transcription factor, which results in a dramatic reduction in tumor MHC expression, and an alteration in local cytokine concentrations to inhibit surrounding inflammatory cells and immune detection. Collectively, these data show that an oncogenic transcription factor can promote tumor growth and tissue invasion while inhibiting local inflammatory and immune responses. This is the first time that an immunomodulatory role has been described for an oncogenic fusion protein.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Mediadores de Inflamación/fisiología , Proteínas de Fusión Oncogénica/fisiología , Factores de Transcripción Paired Box/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/inmunología , Regulación hacia Abajo/genética , Humanos , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/metabolismo , Transcripción Genética/fisiología , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA