Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 144(7): 729-741, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38805639

RESUMEN

ABSTRACT: Loss of long-term hematopoietic stem cell (LT-HSC) function ex vivo hampers the success of clinical protocols that rely on culture. However, the kinetics and mechanisms through which this occurs remain incompletely characterized. In this study, through time-resolved single-cell RNA sequencing, matched in vivo functional analysis, and the use of a reversible in vitro system of early G1 arrest, we defined the sequence of transcriptional and functional events that occur during the first ex vivo division of human LT-HSCs. We demonstrated that the sharpest loss in LT-HSC repopulation capacity happens early on, between 6 and 24 hours of culture, before LT-HSCs commit to cell cycle progression. During this time window, LT-HSCs adapt to the culture environment, limit the global variability in gene expression, and transiently upregulate gene networks involved in signaling and stress responses. From 24 hours, LT-HSC progression past early G1 contributes to the establishment of differentiation programs in culture. However, contrary to the current assumptions, we demonstrated that the loss of HSC function ex vivo is independent of cell cycle progression. Finally, we showed that targeting LT-HSC adaptation to culture by inhibiting the early activation of JAK/STAT signaling improves HSC long-term repopulating function ex vivo. Collectively, our study demonstrated that controlling early LT-HSC adaptation to ex vivo culture, for example, via JAK inhibition, is critically important to improve HSC gene therapy and expansion protocols.


Asunto(s)
Ciclo Celular , Células Madre Hematopoyéticas , Humanos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Cultivadas , Transducción de Señal , Diferenciación Celular , Técnicas de Cultivo de Célula/métodos , Adaptación Fisiológica
2.
Mol Ther ; 27(10): 1706-1717, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31526597

RESUMEN

The field of cell and gene therapy (GT) is expanding rapidly and there is undoubtedly a wave of enthusiasm and anticipation for what these treatments could achieve next. Here we assessed the worldwide landscape of GT assets currently in early clinical development (clinical trial phase 1/2 or about to enter clinical trial). We included all gene therapies, i.e., strategies that modify an individual's protein make-up by introducing exogenous nucleic acid or nucleic acid modifiers, regardless of delivery. Unmodified cell therapies, oncology therapies (reviewed elsewhere), and vaccine programs (distinct therapeutic strategy) were not included. Using a December 31, 2018 cutoff date, we identified 336 gene therapies being developed for 138 different indications covering 165 genetic targets. In all, we found that the early clinical GT landscape comprises a very disparate group of drug candidates in terms of indications, organizations, and delivery methods. We also highlight interesting trends, revealing the evolution of the field toward in vivo therapies and adeno-associated virus vector-based delivery systems. It will be interesting to witness what proportion of this current list effectively translates into new medicines.


Asunto(s)
Sistemas de Liberación de Medicamentos/clasificación , Terapia Genética/métodos , Ensayos Clínicos como Asunto , Vectores Genéticos/administración & dosificación , Humanos , Terapia Molecular Dirigida
3.
Int J Mol Sci ; 20(18)2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491876

RESUMEN

Fabry disease (FD) is caused by mutations in the GLA gene that encodes lysosomal α-galactosidase-A (α-gal-A). A number of pathogenic mechanisms have been proposed and these include loss of mitochondrial respiratory chain activity. For FD, gene therapy is beginning to be applied as a treatment. In view of the loss of mitochondrial function reported in FD, we have considered here the impact of loss of mitochondrial respiratory chain activity on the ability of a GLA lentiviral vector to increase cellular α-gal-A activity and participate in cross correction. Jurkat cells were used in this study and were exposed to increasing viral copies. Intracellular and extracellular enzyme activities were then determined; this in the presence or absence of the mitochondrial complex I inhibitor, rotenone. The ability of cells to take up released enzyme was also evaluated. Increasing transgene copies was associated with increasing intracellular α-gal-A activity but this was associated with an increase in Km. Release of enzyme and cellular uptake was also demonstrated. However, in the presence of rotenone, enzyme release was inhibited by 37%. Excessive enzyme generation may result in a protein with inferior kinetic properties and a background of compromised mitochondrial function may impair the cross correction process.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Mitocondrias/genética , Mitocondrias/metabolismo , alfa-Galactosidasa/biosíntesis , Línea Celular , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Enfermedad de Fabry/genética , Enfermedad de Fabry/metabolismo , Dosificación de Gen , Expresión Génica , Humanos , Células Jurkat , Lisosomas/metabolismo , Mitocondrias/efectos de los fármacos , Transducción Genética , Transgenes , alfa-Galactosidasa/genética
4.
Mol Ther ; 25(8): 1790-1804, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28550974

RESUMEN

Lentiviral vector genomic RNA requires sequences that partially overlap wild-type HIV-1 gag and env genes for packaging into vector particles. These HIV-1 packaging sequences constitute 19.6% of the wild-type HIV-1 genome and contain functional cis elements that potentially compromise clinical safety. Here, we describe the development of a novel lentiviral vector (LTR1) with a unique genomic structure designed to prevent transfer of HIV-1 packaging sequences to patient cells, thus reducing the total HIV-1 content to just 4.8% of the wild-type genome. This has been achieved by reconfiguring the vector to mediate reverse-transcription with a single strand transfer, instead of the usual two, and in which HIV-1 packaging sequences are not copied. We show that LTR1 vectors offer improved safety in their resistance to remobilization in HIV-1 particles and reduced frequency of splicing into human genes. Following intravenous luciferase vector administration to neonatal mice, LTR1 sustained a higher level of liver transgene expression than an equivalent dose of a standard lentivirus. LTR1 vectors produce reverse-transcription products earlier and start to express transgenes significantly quicker than standard lentiviruses after transduction. Finally, we show that LTR1 is an effective lentiviral gene therapy vector as demonstrated by correction of a mouse hemophilia B model.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , VIH-1/genética , ARN Viral , Secuencias Reguladoras de Ácido Ribonucleico , Transducción Genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Factor IX/genética , Expresión Génica , Orden Génico , Genes Reporteros , Terapia Genética , Genoma Viral , Duplicado del Terminal Largo de VIH , Hemofilia B/sangre , Hemofilia B/genética , Hemofilia B/terapia , Humanos , Ratones , Provirus/genética , Recombinación Genética , Transgenes , Replicación Viral/genética
5.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L258-L267, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27979861

RESUMEN

Air-liquid interface (ALI) culture of primary airway epithelial cells enables mucociliary differentiation providing an in vitro model of the human airway, but their proliferative potential is limited. To extend proliferation, these cells were previously transduced with viral oncogenes or mouse Bmi-1 + hTERT, but the resultant cell lines did not undergo mucociliary differentiation. We hypothesized that use of human BMI-1 alone would increase the proliferative potential of bronchial epithelial cells while retaining their mucociliary differentiation potential. Cystic fibrosis (CF) and non-CF bronchial epithelial cells were transduced by lentivirus with BMI-1 and then their morphology, replication kinetics, and karyotype were assessed. When differentiated at ALI, mucin production, ciliary function, and transepithelial electrophysiology were measured. Finally, shRNA knockdown of DNAH5 in BMI-1 cells was used to model primary ciliary dyskinesia (PCD). BMI-1-transduced basal cells showed normal cell morphology, karyotype, and doubling times despite extensive passaging. The cell lines underwent mucociliary differentiation when cultured at ALI with abundant ciliation and production of the gel-forming mucins MUC5AC and MUC5B evident. Cilia displayed a normal beat frequency and 9+2 ultrastructure. Electrophysiological characteristics of BMI-1-transduced cells were similar to those of untransduced cells. shRNA knockdown of DNAH5 in BMI-1 cells produced immotile cilia and absence of DNAH5 in the ciliary axoneme as seen in cells from patients with PCD. BMI-1 delayed senescence in bronchial epithelial cells, increasing their proliferative potential but maintaining mucociliary differentiation at ALI. We have shown these cells are amenable to genetic manipulation and can be used to produce novel disease models for research and dissemination.


Asunto(s)
Bronquios/citología , Diferenciación Celular , Cilios/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Moco/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Animales , Dineínas Axonemales/metabolismo , Proliferación Celular , Forma de la Célula , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Dineínas/metabolismo , Impedancia Eléctrica , Fenómenos Electrofisiológicos , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patología , Síndrome de Kartagener/fisiopatología , Cariotipificación , Ratones , Microtúbulos/metabolismo , Modelos Biológicos , Fenotipo , Transducción Genética
6.
FASEB J ; 29(9): 3876-88, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26062602

RESUMEN

Several acute monogenic diseases affect multiple body systems, causing death in childhood. The development of novel therapies for such conditions is challenging. However, improvements in gene delivery technology mean that gene therapy has the potential to treat such disorders. We evaluated the ability of the AAV9 vector to mediate systemic gene delivery after intravenous administration to perinatal mice and late-gestation nonhuman primates (NHPs). Titer-matched single-stranded (ss) and self-complementary (sc) AAV9 carrying the green fluorescent protein (GFP) reporter gene were intravenously administered to fetal and neonatal mice, with noninjected age-matched mice used as the control. Extensive GFP expression was observed in organs throughout the body, with the epithelial and muscle cells being particularly well transduced. ssAAV9 carrying the WPRE sequence mediated significantly more gene expression than its sc counterpart, which lacked the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) sequence. To examine a realistic scale-up to larger models or potentially patients for such an approach, AAV9 was intravenously administered to late-gestation NHPs by using a clinically relevant protocol. Widespread systemic gene expression was measured throughout the body, with cellular tropisms similar to those observed in the mouse studies and no observable adverse events. This study confirms that AAV9 can safely mediate systemic gene delivery in small and large animal models and supports its potential use in clinical systemic gene therapy protocols.


Asunto(s)
Dependovirus , Feto , Vectores Genéticos , Proteínas Fluorescentes Verdes , Transducción Genética/métodos , Tropismo Viral , Animales , Femenino , Feto/citología , Feto/embriología , Feto/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Vectores Genéticos/farmacología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Haplorrinos , Ratones , Embarazo
7.
Stem Cells ; 30(6): 1134-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22367629

RESUMEN

Human mesenchymal stem cells (hMSCs) have been shown to have potential in regenerative approaches in bone and blood. Most protocols rely on their in vitro expansion prior to clinical use. However, several groups including our own have shown that hMSCs lose proliferation and differentiation ability with serial passage in culture, limiting their clinical applications. Cellular prion protein (PrP) has been shown to enhance proliferation and promote self-renewal of hematopoietic, mammary gland, and neural stem cells. Here we show, for the first time, that expression of PrP decreased in hMSC following ex vivo expansion. When PrP expression was knocked down, hMSC showed significant reduction in proliferation and differentiation. In contrast, hMSC expanded in the presence of small molecule 3/689, a modulator of PrP expression, showed retention of PrP expression with ex vivo expansion and extended lifespan up to 10 population doublings. Moreover, cultures produced a 300-fold increase in the number of cells generated. These cells showed a 10-fold increase in engraftment levels in bone marrow 5 weeks post-transplant. hMSC treated with 3/689 showed enhanced protection from DNA damage and enhanced cell cycle progression, in line with data obtained by gene expression profiling. Moreover, upregulation of superoxide dismutase-2 (SOD2) was also observed in hMSC expanded in the presence of 3/689. The increase in SOD2 was dependent on PrP expression and suggests increased scavenging of reactive oxygen species as mechanism of action. These data point to PrP as a good target for chemical intervention in stem cell regenerative medicine.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Priones/biosíntesis , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Lentivirus/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosforilación , Priones/genética , Transfección
8.
Nat Med ; 12(3): 348-53, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16491086

RESUMEN

Retroviral and lentiviral vector integration into host-cell chromosomes carries with it a finite chance of causing insertional mutagenesis. This risk has been highlighted by the induction of malignancy in mouse models, and development of lymphoproliferative disease in three individuals with severe combined immunodeficiency-X1 (refs. 2,3). Therefore, a key challenge for clinical therapies based on retroviral vectors is to achieve stable transgene expression while minimizing insertional mutagenesis. Recent in vitro studies have shown that integration-deficient lentiviral vectors can mediate stable transduction. With similar vectors, we now show efficient and sustained transgene expression in vivo in rodent ocular and brain tissues. We also show substantial rescue of clinically relevant rodent models of retinal degeneration. Therefore, the high efficiency of gene transfer and expression mediated by lentiviruses can be harnessed in vivo without a requirement for vector integration. For therapeutic application to postmitotic tissues, this system substantially reduces the risk of insertional mutagenesis.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Lentivirus/genética , Animales , Encéfalo/citología , Proteínas Portadoras , Electrorretinografía , Proteínas del Ojo/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Epitelio Pigmentado Ocular/citología , Ratas , Retina/citología , Células Tumorales Cultivadas , Integración Viral/genética , cis-trans-Isomerasas
9.
Mol Ther ; 20(8): 1610-21, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22547151

RESUMEN

Mucopolysaccharidosis IIIA (MPS IIIA or Sanfilippo disease) is a neurodegenerative disorder caused by a deficiency in the lysosomal enzyme sulfamidase (SGSH), catabolizing heparan sulfate (HS). Affected children present with severe behavioral abnormalities, sleep disturbances, and progressive neurodegeneration, leading to death in their second decade. MPS I, a similar neurodegenerative disease accumulating HS, is treated successfully with hematopoietic stem cell transplantation (HSCT) but this treatment is ineffectual for MPS IIIA. We compared HSCT in MPS IIIA mice using wild-type donor cells transduced ex vivo with lentiviral vector-expressing SGSH (LV-WT-HSCT) versus wild-type donor cell transplant (WT-HSCT) or lentiviral-SGSH transduced MPS IIIA cells (LV-IIIA-HSCT). LV-WT-HSCT results in 10% of normal brain enzyme activity, near normalization of brain HS and GM2 gangliosides, significant improvements in neuroinflammation and behavioral correction. Both WT-HSCT and LV-IIIA-HSCT mediated improvements in GM2 gangliosides and neuroinflammation but were less effective at reducing HS or in ameliorating abnormal HS sulfation and had no significant effect on behavior. This suggests that HS may have a more significant role in neuropathology than neuroinflammation or GM2 gangliosides. These data provide compelling evidence for the efficacy of gene therapy in conjunction with WT-HSCT for neurological correction of MPS IIIA where conventional transplant is ineffectual.


Asunto(s)
Terapia Genética/métodos , Células Madre Hematopoyéticas/fisiología , Mucopolisacaridosis/patología , Mucopolisacaridosis/terapia , Animales , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Inmunohistoquímica , Ratones
10.
Nature ; 443(7109): E5; discussion E6-7, 2006 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-16988660

RESUMEN

The gene IL2RG encodes the gamma-chain of the interleukin-2 receptor and is mutated in patients with X-linked severe combined immune deficiency (X-SCID). Woods et al. report the development of thymus tumours in a mouse model of X-SCID after correction by lentiviral overexpression of IL2RG and claim that these were caused by IL2RG itself. Here we find that retroviral overexpression of IL2RG in human CD34+ cells has no effect on T-cell development, whereas overexpression of the T-cell acute lymphoblastic leukaemia (T-ALL) oncogene LMO2 leads to severe abnormalities. Retroviral expression of IL2RG may therefore not be directly oncogenic--rather, the restoration of normal signalling by the interleukin-7 receptor to X-SCID precursor cells allows progression of T-cell development to stages that are permissive for the pro-leukaemic effects of ectopic LMO2.


Asunto(s)
Transformación Celular Neoplásica , Terapia Genética/efectos adversos , Receptores de Interleucina-2/metabolismo , Linfocitos T/metabolismo , Linfocitos T/patología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas con Dominio LIM , Metaloproteínas/genética , Metaloproteínas/metabolismo , Ratones , Proteínas Proto-Oncogénicas , Receptores de Interleucina-2/genética
11.
Mol Ther ; 19(4): 703-10, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21364536

RESUMEN

Lentiviral vectors with self-inactivating (SIN) long terminal repeats (LTRs) are promising for safe and sustained transgene expression in dividing as well as quiescent cells. As genome organization and transcription substantially differs between actively dividing and postmitotic cells in vivo, we hypothesized that genomic vector integration preferences might be distinct between these biological states. We performed integration site (IS) analyses on mouse dividing cells (fibroblasts and hematopoietic progenitor cells (HPCs)) transduced ex vivo and postmitotic cells (eye and brain) transduced in vivo. As expected, integration in dividing cells occurred preferably into gene coding regions. In contrast, postmitotic cells showed a close to random frequency of integration into genes and gene spare long interspersed nuclear elements (LINE). Our studies on the potential mechanisms responsible for the detected differences of lentiviral integration suggest that the lowered expression level of Psip1 reduce the integration frequency in vivo into gene coding regions in postmitotic cells. The motif TGGAA might represent one of the factors for preferred lentiviral integration into mouse and rat Satellite DNA. These observations are highly relevant for the correct assessment of preclinical biosafety studies, indicating that lentiviral vectors are well suited for safe and effective clinical gene transfer into postmitotic tissues.


Asunto(s)
Vectores Genéticos/genética , Lentivirus/genética , Mitosis/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , ADN Satélite/genética , Femenino , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Ratas , Secuencias Repetidas Terminales/genética , Factores de Transcripción/genética , Integración Viral/genética
12.
Mol Ther ; 19(11): 2031-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21862999

RESUMEN

Vector-associated side effects in clinical gene therapy have provided insights into the molecular mechanisms of hematopoietic regulation in vivo. Surprisingly, many retrovirus insertion sites (RIS) present in engrafted cells have been found to cluster nonrandomly in close association with specific genes. Our data demonstrate that these genes directly influence the in vivo fate of hematopoietic cell clones. Analysis of insertions thus far has been limited to individual clinical studies. Here, we studied >7,000 insertions retrieved from various studies. More than 40% of all insertions found in engrafted gene-modified cells were clustered in the same genomic areas covering only 0.36% of the genome. Gene classification analyses displayed significant overrepresentation of genes associated with hematopoietic functions and relevance for cell growth and survival in vivo. The similarity of insertion distributions indicates that vector insertions in repopulating cells cluster in predictable patterns. Thus, insertion analyses of preclinical in vitro and murine in vivo studies as well as vector insertion repertoires in clinical trials yielded concerted results and mark a small number of interesting genomic loci and genes that warrants further investigation of the biological consequences of vector insertions.


Asunto(s)
Gammaretrovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Genoma , Integración Viral , Animales , Mapeo Cromosómico , Redes Reguladoras de Genes , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Primates , Trasplantes , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
13.
J Clin Invest ; 118(9): 3143-50, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18688286

RESUMEN

X-linked SCID (SCID-X1) is amenable to correction by gene therapy using conventional gammaretroviral vectors. Here, we describe the occurrence of clonal T cell acute lymphoblastic leukemia (T-ALL) promoted by insertional mutagenesis in a completed gene therapy trial of 10 SCID-X1 patients. Integration of the vector in an antisense orientation 35 kb upstream of the protooncogene LIM domain only 2 (LMO2) caused overexpression of LMO2 in the leukemic clone. However, leukemogenesis was likely precipitated by the acquisition of other genetic abnormalities unrelated to vector insertion, including a gain-of-function mutation in NOTCH1, deletion of the tumor suppressor gene locus cyclin-dependent kinase 2A (CDKN2A), and translocation of the TCR-beta region to the STIL-TAL1 locus. These findings highlight a general toxicity of endogenous gammaretroviral enhancer elements and also identify a combinatorial process during leukemic evolution that will be important for risk stratification and for future protocol design.


Asunto(s)
Cromosomas Humanos X , Terapia Genética/efectos adversos , Terapia Genética/métodos , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Inmunodeficiencia Combinada Grave/terapia , Proteínas Adaptadoras Transductoras de Señales , Antineoplásicos/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/genética , Estudios de Seguimiento , Humanos , Lactante , Proteínas con Dominio LIM , Masculino , Metaloproteínas/genética , Modelos Biológicos , Mutagénesis , Leucemia-Linfoma Linfoblástico de Células T Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Proteínas Proto-Oncogénicas , Receptor Notch1/genética , Receptores de Interleucina-2/genética , Inmunodeficiencia Combinada Grave/complicaciones
14.
Mol Ther ; 18(12): 2104-11, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20736928

RESUMEN

The transforming growth factor-ß (TGFß) family plays a critical regulatory role in repair and coordination of remodeling after cutaneous wounding. TGFß1-mediated chemotaxis promotes the recruitment of fibroblasts to the wound site and their resultant myofibroblastic transdifferentiation that is responsible for elastic fiber deposition and wound closure. TGFß3 has been implicated in an antagonistic role regulating overt wound closure and promoting ordered dermal remodeling. We generated a mutant form of TGFß3 (mutTGFß3) by ablating its binding site for the latency-associated TGFß binding protein (LTBP-1) in order to improve bioavailability and activity. The mutated cytokine is secreted as the stable latency-associated peptide (LAP)-associated form and is activated by normal intracellular and extracellular mechanisms including integrin-mediated activation but is not sequestered. We show localized intradermal transduction using a lentiviral vector expressing the mutTGFß3 in a mouse skin wounding model reduced re-epithelialization density and fibroblast/myofibroblast transdifferentiation within the wound area, both indicative of reduced scar tissue formation.


Asunto(s)
Terapia Genética , Factor de Crecimiento Transformador beta3/genética , Cicatrización de Heridas/fisiología , Animales , Cicatriz/patología , Vectores Genéticos/genética , Células HEK293 , Humanos , Lentivirus/genética , Ratones , Mutación , Piel/patología , Cicatrización de Heridas/genética
15.
J Clin Invest ; 117(8): 2225-32, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17671652

RESUMEN

Recent reports have challenged the notion that retroviruses and retroviral vectors integrate randomly into the host genome. These reports pointed to a strong bias toward integration in and near gene coding regions and, for gammaretroviral vectors, around transcription start sites. Here, we report the results obtained from a large-scale mapping of 572 retroviral integration sites (RISs) isolated from cells of 9 patients with X-linked SCID (SCID-X1) treated with a retrovirus-based gene therapy protocol. Our data showed that two-thirds of insertions occurred in or very near to genes, of which more than half were highly expressed in CD34(+) progenitor cells. Strikingly, one-fourth of all integrations were clustered as common integration sites (CISs). The highly significant incidence of CISs in circulating T cells and the nature of their locations indicate that insertion in many gene loci has an influence on cell engraftment, survival, and proliferation. Beyond the observed cases of insertional mutagenesis in 3 patients, these data help to elucidate the relationship between vector insertion and long-term in vivo selection of transduced cells in human patients with SCID-X1.


Asunto(s)
Gammaretrovirus , Terapia Genética , Vectores Genéticos , Genoma Humano , Linfopoyesis/genética , Integración Viral/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Antígenos CD34 , Proliferación Celular , Supervivencia Celular/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Mutagénesis Insercional , Sitios de Carácter Cuantitativo , Linfocitos T , Factores de Tiempo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
16.
J Clin Invest ; 117(8): 2241-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17671654

RESUMEN

We treated 10 children with X-linked SCID (SCID-X1) using gammaretrovirus-mediated gene transfer. Those with sufficient follow-up were found to have recovered substantial immunity in the absence of any serious adverse events up to 5 years after treatment. To determine the influence of vector integration on lymphoid reconstitution, we compared retroviral integration sites (RISs) from peripheral blood CD3(+) T lymphocytes of 5 patients taken between 9 and 30 months after transplantation with transduced CD34(+) progenitor cells derived from 1 further patient and 1 healthy donor. Integration occurred preferentially in gene regions on either side of transcription start sites, was clustered, and correlated with the expression level in CD34(+) progenitors during transduction. In contrast to those in CD34(+) cells, RISs recovered from engrafted CD3(+) T cells were significantly overrepresented within or near genes encoding proteins with kinase or transferase activity or involved in phosphorus metabolism. Although gross patterns of gene expression were unchanged in transduced cells, the divergence of RIS target frequency between transduced progenitor cells and post-thymic T lymphocytes indicates that vector integration influences cell survival, engraftment, or proliferation.


Asunto(s)
Complejo CD3 , Gammaretrovirus , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Linfocitos T/inmunología , Integración Viral , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Adulto , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Niño , Preescolar , Femenino , Estudios de Seguimiento , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Células Madre Hematopoyéticas/inmunología , Humanos , Lactante , Masculino , Transducción Genética , Trasplante Autólogo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
17.
Analyst ; 135(1): 157-64, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20024196

RESUMEN

The bio-electrospray technique has been recently pioneered to manipulate living, immortalised and primary cells, including a wide range of stem cells. Studies have demonstrated that the creation of viable, fully functional in vitro microenvironments is possible using this technique. By modifying the bio-electrospray procedure (referred to as cell electrospinning), a variety of microenvironment morphologies have been fabricated. Because bio-electrospraying of biological material is a relatively new technique, it is important to determine if there are any unwanted consequences to the manipulated cells as a result of the procedure. Here, we establish the validity of the process using a heterogeneous, living population of hematopoietic stem/progenitor cells, using a functional in vitro assay and in vivo mouse model to investigate for side-effects that previous in vitro assays may not have detected. Our studies demonstrate that these bio-protocols have no obvious negative effects, thus indicating significant promise for utility in biological sciences and for a plethora of healthcare applications.


Asunto(s)
Separación Celular/instrumentación , Células Madre Hematopoyéticas/citología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Separación Celular/métodos , Femenino , Trasplante de Células Madre Hematopoyéticas , Ratones
18.
Mol Ther ; 17(4): 632-40, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19174762

RESUMEN

The treatment of rheumatoid arthritis remains suboptimal; thus there is considerable interest in the development of strategies that mediate tolerance to autoantigens. Using lentiviral gene transfer in vivo, we expressed the immunodominant epitope of collagen type II (CII) on major histocompatibility complex class II molecules (MHC II) in a mouse model of destructive arthritis. A sequence corresponding to amino acids 259-270 of CII was fused into the class II-associated invariant chain peptide (CLIP) position of the invariant chain to achieve efficient binding to MHC II. Transduction of cloned cells and primary antigen-presenting cells (APCs) in vitro demonstrated successful presentation of the peptide on MHC II, and a physiological glycosylation pattern. Compared with controls, mice intravenously injected with lentiviral vectors encoding this epitope displayed significantly less frequent, less severe, and less destructive arthritis, decreased lymphocyte proliferation in response to restimulation with CII, and lower CII-specific antibody levels. This was associated with an increased production of transforming growth factor-beta (TGF-beta) in vitro. We suggest that overexpression of the immunodominant CII epitope on MHC II induces T cell production of TGF-beta and leads to inhibition of arthritis by means of both antigen-specific and bystander mechanisms. Thus, antigen-specific tolerance induction using lentiviral gene delivery can ameliorate arthritis.


Asunto(s)
Artritis Experimental/terapia , Colágeno Tipo II/inmunología , Terapia Genética , Tolerancia Inmunológica , Lentivirus/genética , Transfección , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Autoanticuerpos/inmunología , Secuencia de Bases , Trasplante de Médula Ósea , Proliferación Celular , Cartilla de ADN , Vectores Genéticos , Glicosilación , Ratones , Reacción en Cadena de la Polimerasa , Índice de Severidad de la Enfermedad , Bazo/patología , Linfocitos T Reguladores/inmunología
19.
Mol Ther Methods Clin Dev ; 19: 47-57, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-32995359

RESUMEN

Stable suspension producer cell lines for the production of vesicular stomatitis virus envelope glycoprotein (VSVg)-pseudotyped lentiviral vectors represent an attractive alternative to current widely used production methods based on transient transfection of adherent 293T cells with multiple plasmids. We report here a method to rapidly generate such producer cell lines from 293T cells by stable transfection of a single DNA construct encoding all lentiviral vector components. The resulting suspension cell lines yield titers as high as can be achieved with transient transfection, can be readily scaled up in single-use stirred-tank bioreactors, and are genetically and functionally stable in extended cell culture. By removing the requirement for efficient transient transfection during upstream processing of lentiviral vectors and switching to an inherently scalable suspension cell culture format, we believe that this approach will result in significantly higher batch yields than are possible with current manufacturing processes and enable better patient access to medicines based on lentiviral vectors.

20.
Mol Ther ; 16(3): 590-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18180772

RESUMEN

Gene therapy for X-linked severe combined immunodeficiency (SCID-X1) has proven highly effective for long-term restoration of immunity in human subjects. However, the development of lymphoproliferative complications due to dysregulated proto-oncogene expression has underlined the necessity for developing safer vector systems. To reduce the potential for insertional mutagenesis, we have evaluated the efficacy of self-inactivating (SIN) gammaretroviral vectors in cellular and in vivo models of SCID-X1. Vectors incorporating an internal human elongation factor-1alpha regulatory element were capable of fully restoring the lymphoid differentiation potential of gammac-deficient lineage negative cells. Multilineage lymphoid reconstitution of a murine model was achieved at a similar level to that achieved by a conventional long-terminal repeat (LTR)-regulated vector used in previous clinical trials. Functional proliferative responses to mitogenic stimuli were also restored, and serum immunoglobulin levels were normalized. The reduced mutagenic potential conferred by SIN vector configurations and alternative non-LTR-based regulatory elements, together with proven efficacy in correction of cellular defects provides an important platform for development of the next phase of clinical trials for SCID-X1.


Asunto(s)
Gammaretrovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Northern Blotting , Diferenciación Celular , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Reacción en Cadena de la Polimerasa , Proto-Oncogenes Mas , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA