Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Gene Ther ; 24(9): 590-592, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28485723

RESUMEN

For the last 20 years, academic research has been the major, and often only, driving force behind the spectacular development of gene transfer technology for the therapy of rare genetic diseases. Investors and industry became eventually interested in gene and cell therapy, due to the success of a series of pioneering clinical trials that proved efficacy and safety of last-generation technology, and to favorable orphan drug legislation in both Europe and the United States. Developing this forms of therapy is however complex and requires skills and knowledge not necessary available to the industry, which is better placed to develop processes and products and put them on the market. Cooperation between academia and industry is an opportunity to de-risk innovative approaches and ensure a faster and more economical development of therapies for diseases with high unmet medical needs and low-profit expectations.


Asunto(s)
Centros Médicos Académicos/organización & administración , Industria Farmacéutica/organización & administración , Terapia Genética/métodos , Colaboración Intersectorial , Enfermedades Raras/terapia , Investigación Biomédica/métodos , Investigación Biomédica/organización & administración , Humanos
2.
Gene Ther ; 20(9): 949-57, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23615186

RESUMEN

Transplantation of epithelia derived from keratinocyte stem cells transduced by retroviral vectors is a potential therapy for epidermolysis bullosa (EB), a family of inherited skin adhesion defects. The biosafety characteristics of retroviral vectors in keratinocytes are, however, poorly defined. We developed self-inactivating (SIN) vectors derived from the Moloney murine leukemia (MLV) and the human immunodeficiency (HIV) viruses expressing therapeutic levels of LAMB3, a transgene defective in junctional EB, and tested their integration profile in human primary keratinocytes. The SIN-HIV vector showed the expected preference for transcribed genes while the SIN-MLV vector integrated preferentially in regulatory elements, but showed a significantly lower tendency to target cell growth-related genes, transcription start sites and epigenetically defined promoters compared with a wild-type MLV vector in an epithelial cell context. A quantitative gene expression assay in individual keratinocyte clones showed that MLV-derived vectors deregulate expression of targeted genes at a lower frequency than in hematopoietic cells, and that the SIN-MLV design has the lowest activity compared to both MLV and SIN-HIV vectors. This study indicates that SIN-MLV vectors may have a better safety profile in keratinocyte than in hematopoietic cells, and be a reasonable alternative to lentiviral vectors for gene therapy of inherited skin disorders.


Asunto(s)
Moléculas de Adhesión Celular/genética , Epidermólisis Ampollosa/genética , Epidermólisis Ampollosa/terapia , Vectores Genéticos , Queratinocitos/metabolismo , Virus de la Leucemia Murina de Moloney/genética , Integración Viral , Animales , Moléculas de Adhesión Celular/metabolismo , Epidermólisis Ampollosa/metabolismo , Regulación de la Expresión Génica , Terapia Genética , VIH-1/genética , Células HeLa , Humanos , Ratones , Virus de la Leucemia Murina de Moloney/fisiología , Regiones Promotoras Genéticas , Células 3T3 Swiss , Transducción Genética , Transgenes , Inactivación de Virus , Kalinina
3.
Gene Ther ; 18(7): 674-81, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21368897

RESUMEN

Cutaneous gene therapy can be envisioned through the use of keratinocyte stem cell clones in which retroviral genotoxic risks can be pre-assessed. While transactivation of cellular genes by the retroviral long terminal repeat enhancer has been proven in experimental and clinical settings, the formation of chimeric viral-cellular transcripts originated by the inefficient termination (read-through) of retroviral transcripts remains to be studied in depth. We now demonstrate the widespread presence of viral-cellular fusion transcripts derived from integrated proviruses in keratinocytes transduced with self-inactivating (SIN) retroviral vectors. We have detected high molecular weight RNAs in northern blot analysis of retroviral vector expression in individual cell clones. Characterization of some of these transcripts revealed that they originate from genes located at the proviral integration sites. One class of transcripts corresponds to fusions of the viral vectors with intronic sequences, terminating at cryptic polyadenylation sites located in introns. A second class comprises fusion transcripts with coding sequences of genes at the integration sites. These are generated through splicing from a cryptic, not previously described donor site in the lentiviral vectors to exons of cellular genes, and have the potential to encode unintended open reading frames, although they are downregulated by cellular mechanisms. Our data contribute to a better understanding of the impact of SIN lentiviral vector integration on cellular gene transcription, and will be helpful in improving the design of this type of vectors.


Asunto(s)
Fusión Génica , Terapia Genética/efectos adversos , Queratina-14/genética , Queratinocitos/metabolismo , Lentivirus/genética , Provirus/genética , Inactivación de Virus , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Medición de Riesgo , Transcripción Genética , Transducción Genética , Integración Viral
4.
J Exp Med ; 173(4): 889-97, 1991 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-1706752

RESUMEN

We have investigated the effect of granulocyte colony-stimulating factor (G-CSF) delivery at the site of tumor growth by transducing, via retroviral vector, the human (hu) G-CSF gene into the colon adenocarcinoma C-26 and assaying the ability of transduced cells to form tumors when injected into syngeneic mice. As a control, the same tumor cells were infected with retroviruses engineered to transduce an unrelated gene, the human nerve growth factor receptor, or carry the neomycin resistance gene only. Only cells transduced with the huG-CSF were unable to develop tumors, although huG-CSF was expressed and produced at low level as estimated by both RNA analysis and enzyme-linked immunosorbent assay, indicating that G-CSF can exert an antitumor effect at a physiological dose. Implication of G-CSF as mediator of tumor inhibition was proven by reversing the nontumorigenic phenotype of G-CSF-expressing cells with anti-huG-CSF monoclonal antibody injected at the tumor site. No tumors were formed by injecting C-26 infected cells into nu/nu mice, while neoplastic nodules appeared after injection into sublethally irradiated mice; such tumors, however, regressed when mice normalized their leukocyte counts after irradiation. Tumors were also formed after injection of a mixture of infected and uninfected C-26 cells, although critical delay in tumor formation occurred when infected cells were 10 times more represented in the mixture. Histological examination of tissues surrounding the site of injection showed infiltration of neutrophilic granulocytes, whose number correlated with that of G-CSF-expressing C-26 cells in the injected mixture. These results indicate that G-CSF may have a potent antitumoral activity when released, even at low doses, at the tumor site. The antitumoral effect is mediated by recruitment and targeting of neutrophilic granulocytes to G-CSF-releasing cells.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias del Colon/terapia , Factor Estimulante de Colonias de Granulocitos/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Expresión Génica , Terapia Genética , Factor Estimulante de Colonias de Granulocitos/fisiología , Inmunidad Celular , Técnicas Inmunológicas , Ratones , Ratones Desnudos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Transfección
5.
J Pathol ; 217(2): 217-28, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18855878

RESUMEN

Regenerative medicine refers to innovative therapies aimed at the permanent restoration of diseased tissues and organs. Regeneration of self-renewing tissues requires specific adult stem cells, which need to be genetically modified to correct inherited genetic diseases. Cultures of epithelial stem cells permanently restore severe skin and mucosal defects, and genetically corrected epidermal stem cells regenerate a normal epidermis in patients carrying junctional epidermolysis bullosa. The keratinocyte stem cell is therefore the only cultured stem cell used both in cell therapy and gene therapy clinical protocols. Epithelial stem cell identification, fate and molecular phenotype have been extensively reviewed, but not in relation to tissue regeneration. In this paper we focus on the localization and molecular characterization of human limbal stem cells in relation to corneal regeneration, and the gene therapy of genetic skin diseases by means of genetically modified epidermal stem cells.


Asunto(s)
Células Madre Adultas/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Epitelio Corneal/citología , Terapia Genética/métodos , Animales , Células Epidérmicas , Epitelio Corneal/lesiones , Epitelio Corneal/patología , Humanos , Queratinocitos/citología , Queratinocitos/trasplante , Ratones , Modelos Animales , Regeneración , Enfermedades de la Piel/terapia , Cicatrización de Heridas
6.
Br J Dermatol ; 161(1): 19-24, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19466960

RESUMEN

Gene therapy has the potential to treat devastating inherited diseases for which there is little hope of finding a conventional cure. These include lethal diseases, like immunodeficiencies or several metabolic disorders, or conditions associated with a relatively long life expectancy but poor quality of life and expensive and life-long symptomatic treatments, such as muscular dystrophy, cystic fibrosis and thalassaemia. Skin adhesion defects belong to both groups. For the nonlethal forms, gene therapy, or transplantation of cultured skin derived from genetically corrected epidermal stem cells, represents a very attractive therapeutic option, and potentially a definitive treatment. Recent advances in gene transfer and stem cell culture technology are making this option closer than ever. This paper critically reviews the progress and prospects of gene therapy for epidermolysis bullosa, and the technical and nontechnical factors currently limiting its development.


Asunto(s)
Epidermólisis Ampollosa/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Epidermólisis Ampollosa/genética , Vectores Genéticos , Humanos , Retroviridae , Trasplante de Células Madre
7.
Science ; 279(5356): 1528-30, 1998 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-9488650

RESUMEN

Growth and repair of skeletal muscle are normally mediated by the satellite cells that surround muscle fibers. In regenerating muscle, however, the number of myogenic precursors exceeds that of resident satellite cells, implying migration or recruitment of undifferentiated progenitors from other sources. Transplantation of genetically marked bone marrow into immunodeficient mice revealed that marrow-derived cells migrate into areas of induced muscle degeneration, undergo myogenic differentiation, and participate in the regeneration of the damaged fibers. Genetically modified, marrow-derived myogenic progenitors could potentially be used to target therapeutic genes to muscle tissue, providing an alternative strategy for treatment of muscular dystrophies.


Asunto(s)
Células de la Médula Ósea/fisiología , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Regeneración , Células Madre/fisiología , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Diferenciación Celular , Movimiento Celular , Terapia Genética , Humanos , Ratones , Ratones SCID , Ratones Transgénicos , Fibras Musculares Esqueléticas/citología , Distrofias Musculares/terapia , Células del Estroma/citología , Células del Estroma/fisiología , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
8.
Science ; 251(4999): 1363-6, 1991 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-1848369

RESUMEN

Deficiency of adenosine deaminase (ADA) results in severe combined immunodeficiency (SCID), a candidate genetic disorder for somatic cell gene therapy. Peripheral blood lymphocytes from patients affected by ADA- SCID were transduced with a retroviral vector for human ADA and injected into immunodeficient mice. Long-term survival of vector-transduced human cells was demonstrated in recipient animals. Expression of vector-derived ADA restored immune functions, as indicated by the presence in reconstituted animals of human immunoglobulin and antigen-specific T cells. Retroviral vector gene transfer, therefore, is necessary and sufficient for development of specific immune functions in vivo and has therapeutic potential to correct this lethal immunodeficiency.


Asunto(s)
Adenosina Desaminasa/deficiencia , Síndromes de Inmunodeficiencia/genética , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Terapia Genética , Vectores Genéticos , Humanos , Síndromes de Inmunodeficiencia/terapia , Kanamicina Quinasa , Transfusión de Linfocitos , Linfocitos/fisiología , Ratones , Ratones Mutantes , Oligonucleótidos/química , Fosfotransferasas/genética , Reacción en Cadena de la Polimerasa , Retroviridae/genética
9.
Science ; 270(5235): 470-5, 1995 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-7570000

RESUMEN

Adenosine deaminase (ADA) deficiency results in severe combined immunodeficiency, the first genetic disorder treated by gene therapy. Two different retroviral vectors were used to transfer ex vivo the human ADA minigene into bone marrow cells and peripheral blood lymphocytes from two patients undergoing exogenous enzyme replacement therapy. After 2 years of treatment, long-term survival of T and B lymphocytes, marrow cells, and granulocytes expressing the transferred ADA gene was demonstrated and resulted in normalization of the immune repertoire and restoration of cellular and humoral immunity. After discontinuation of treatment, T lymphocytes, derived from transduced peripheral blood lymphocytes, were progressively replaced by marrow-derived T cells in both patients. These results indicate successful gene transfer into long-lasting progenitor cells, producing a functional multilineage progeny.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Técnicas de Transferencia de Gen , Terapia Genética , Células Madre Hematopoyéticas , Linfocitos , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/administración & dosificación , Adenosina Desaminasa/sangre , Adenosina Desaminasa/uso terapéutico , Formación de Anticuerpos , Secuencia de Bases , Células de la Médula Ósea , Células Cultivadas , Preescolar , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , Humanos , Inmunidad Celular , Transfusión de Linfocitos , Linfocitos/enzimología , Linfocitos/inmunología , Datos de Secuencia Molecular , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología
10.
Science ; 276(5319): 1719-24, 1997 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-9180086

RESUMEN

In allogeneic bone marrow transplantation (allo-BMT), donor lymphocytes play a central therapeutic role in both graft-versus-leukemia (GvL) and immune reconstitution. However, their use is limited by the risk of severe graft-versus-host disease (GvHD). Eight patients who relapsed or developed Epstein-Barr virus-induced lymphoma after T cell-depleted BMT were then treated with donor lymphocytes transduced with the herpes simplex virus thymidine kinase (HSV-TK) suicide gene. The transduced lymphocytes survived for up to 12 months, resulting in antitumor activity in five patients. Three patients developed GvHD, which could be effectively controlled by ganciclovir-induced elimination of the transduced cells. These data show that genetic manipulation of donor lymphocytes may increase the efficacy and safety of allo-BMT and expand its application to a larger number of patients.


Asunto(s)
Trasplante de Médula Ósea , Terapia Genética , Enfermedad Injerto contra Huésped/terapia , Leucemia/terapia , Transfusión de Linfocitos , Timidina Quinasa/genética , Trasplante de Médula Ósea/efectos adversos , Ganciclovir/uso terapéutico , Técnicas de Transferencia de Gen , Enfermedad Injerto contra Huésped/etiología , Humanos , Leucemia/inmunología , Linfocitos/enzimología , Linfoma no Hodgkin/terapia , Trastornos Linfoproliferativos/terapia , Proyectos Piloto , Simplexvirus/enzimología , Simplexvirus/genética , Trasplante Homólogo
11.
Gene Ther ; 15(7): 504-15, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18239607

RESUMEN

Central nervous system (CNS) delivery of anti-inflammatory cytokines, such as interleukin 4 (IL4), holds promise as treatment for multiple sclerosis (MS). We have previously shown that short-term herpes simplex virus type 1-mediated IL4 gene therapy is able to inhibit experimental autoimmune encephalomyelitis (EAE), an animal model of MS, in mice and non-human primates. Here, we show that a single administration of an IL4-expressing helper-dependent adenoviral vector (HD-Ad) into the cerebrospinal fluid (CSF) circulation of immunocompetent mice allows persistent transduction of neuroepithelial cells and long-term (up to 5 months) CNS transgene expression without toxicity. Mice affected by chronic and relapsing EAE display clinical and neurophysiological recovery from the disease once injected with the IL4-expressing HD-Ad vector. The therapeutic effect is due to the ability of IL4 to increase, in inflamed CNS areas, chemokines (CCL1, CCL17 and CCL22) capable of recruiting regulatory T cells (CD4+CD69-CD25+Foxp3+) with suppressant functions. CSF delivery of HD-Ad vectors expressing anti-inflammatory molecules might represent a valuable therapeutic option for CNS inflammatory disorders.


Asunto(s)
Sistema Nervioso Central/inmunología , Terapia Genética/métodos , Interleucina-4/genética , Esclerosis Múltiple/terapia , Linfocitos T Reguladores/inmunología , Adenoviridae/genética , Animales , Sistema Nervioso Central/patología , Quimiocinas/inmunología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Virus Helper/genética , Humanos , Interleucina-4/análisis , Interleucina-4/inmunología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética/métodos
12.
Gene Ther ; 15(3): 233-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17960158

RESUMEN

Inflammation and immune reaction, or pre-existing immunity towards commonly used viral vectors for gene therapy severely impair long-term gene expression in the central nervous system (CNS), impeding the possibility to repeat the therapeutic intervention. Here, we show that injection of a helper-dependent adenoviral (HD-Ad) vector by lumbar puncture into the cerebrospinal fluid (CSF) of non-human primates allows long-term (three months) infection of neuroepithelial cells, also in monkeys bearing a pre-existing anti-adenoviral immunity. Intrathecal injection of the HD-Ad vector was not associated with any sign of systemic or local toxicity, nor by signs of a CNS-specific immune reaction towards the HD-Ad vector. Injection of HD-Ad vectors into the CSF circulation may thus represent a valuable approach for CNS gene therapy allowing for long-term expression and re-administration.


Asunto(s)
Adenoviridae/genética , Líquido Cefalorraquídeo/virología , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Virus Helper/genética , Enfermedad de Parkinson/terapia , Animales , Expresión Génica , Ingeniería Genética , Vectores Genéticos/inmunología , Interleucina-4/genética , Macaca fascicularis , Masculino , Modelos Animales , Células Neuroepiteliales/inmunología , Células Neuroepiteliales/virología , Enfermedad de Parkinson/inmunología , Punción Espinal , Transducción Genética/métodos
13.
Hum Gene Ther ; 18(9): 811-20, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17824830

RESUMEN

Lentiviral vectors are efficiently pseudotyped with RD114-TR, a chimeric envelope glycoprotein made of the extracellular and transmembrane domains of the feline leukemia virus RD114 and the cytoplasmic tail of the murine leukemia virus amphotropic envelope. RD114-TR-pseudotyped vectors may be concentrated by centrifugation, are resistant to complement inactivation, and are suitable for both ex vivo and in vivo gene therapy applications. We analyzed RD114-TR-pseudotyped, HIV-1-derived lentiviral vectors for their ability to transduce human cord blood, bone marrow, and peripheral blood mobilized CD34(+) hematopoietic stem/progenitor cells. Transduction efficiency was analyzed in CD34(+) cells in liquid culture, in CD34(+) clonogenic progenitors in semisolid culture, and in CD34(+) repopulating stem cells after xenotransplantation in NOD-SCID mice. Compared with a standard VSV-G-based packaging system, RD114-TR-pseudotyped particles transduced hematopoietic stem/progenitor cells at lower multiplicity of infection, with lower toxicity and less pseudo-transduction at comparable vector copy number per genome. Potential changes in the CD34(+) cell transcription profile and phenotype on transduction with RD114-TR-pseudotyped vectors was comparatively investigated by microarray analysis. Our study shows that the biology of repopulating hematopoietic stem cells and their progeny is not affected by transduction with RD114-TR-pseudotyped lentiviral vectors. RD114-TR is compatible with the development of lentiviral stable packaging cell lines, and may become the envelope of choice for clinical studies aiming at safe and efficient genetic modification of human hematopoietic stem cells.


Asunto(s)
Vectores Genéticos , Células Madre Hematopoyéticas/fisiología , Lentivirus/genética , Transducción de Señal , Proteínas del Envoltorio Viral/genética , Animales , Antígenos CD34/sangre , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Sangre Fetal/citología , Sangre Fetal/metabolismo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Virus de la Inmunodeficiencia Felina/química , Virus de la Leucemia Murina/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Trasplante Heterólogo
14.
Trends Genet ; 7(10): 329-34, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1685814

RESUMEN

Vertebrate homeobox genes of the Hox family are, like Drosophila homeotic genes, organized in gene clusters and show a strict correspondence, or collinearity, between the order of the genes (3' to 5') within the chromosomal cluster and that of their expression domains (anterior to posterior) in the embryo. Recent data obtained from embryonal carcinoma cells induced to differentiate by retinoic acid cast some light on the molecular mechanisms underlying the collinear expression of the Hox genes.


Asunto(s)
Regulación de la Expresión Génica , Genes Homeobox , Tretinoina/farmacología , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Activación Transcripcional
15.
J Clin Invest ; 78(1): 51-60, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3722384

RESUMEN

Human embryonic development involves transition from yolk sac (YS) to liver (L) hemopoiesis. We report the identification of pluripotent, erythroid, and granulo-macrophage progenitors in YS, L, and blood from human embryos. Furthermore, comprehensive studies are presented on the number of hemopoietic progenitors and precursors, as well as of other cell types, in YS, L, and blood at precisely sequential stages in embryos and early fetuses (i.e., at 4.5-8 wk and 9-10 wk postconception, respectively). Our results provide circumstantial support to a monoclonal hypothesis for human embryonic hemopoiesis, based on migration of stem and early progenitor cells from a generation site (YS) to a colonization site (L) via circulating blood. The YS----L transition is associated with development of the differentiation program in proliferating stem cells: their erythroid progeny shows, therefore, parallel switches of multiple parameters, e.g., morphology (megaloblasts----macrocytes) and globin expression (zeta----alpha, epsilon----gamma).


Asunto(s)
Embrión de Mamíferos/fisiología , Hematopoyesis , Hígado/embriología , Células Madre/fisiología , Saco Vitelino/fisiología , Ensayo de Unidades Formadoras de Colonias , Eritroblastos/análisis , Femenino , Granulocitos/citología , Humanos , Monocitos/citología , Embarazo , Factores de Tiempo
16.
J Clin Invest ; 101(10): 2119-28, 1998 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9593768

RESUMEN

Ex vivo gene therapy of primary myopathies, based on autologous transplantation of genetically modified myogenic cells, is seriously limited by the number of primary myogenic cells that can be isolated, expanded, transduced, and reimplanted into the patient's muscles. We explored the possibility of using the MyoD gene to induce myogenic conversion of nonmuscle, primary cells in a quantitatively relevant fashion. Primary human and murine fibroblasts from skin, muscle, or bone marrow were infected by an E1-deleted adenoviral vector carrying a retroviral long terminal repeat-promoted MyoD cDNA. Expression of MyoD caused irreversible withdrawal from the cell cycle and myogenic differentiation in the majority (from 60 to 90%) of cultured fibroblasts, as defined by activation of muscle-specific genes, fusion into contractile myotubes, and appearance of ultrastructurally normal sarcomagenesis in culture. 24 h after adenoviral exposure, MyoD-converted cultures were injected into regenerating muscle of immunodeficient (severe combined immunodeficiency/beige) mice, where they gave rise to beta-galactosidase positive, centrally nucleated fibers expressing human myosin heavy chains. Fibers originating from converted fibroblasts were indistinguishable from those obtained by injection of control cultures of lacZ-transduced satellite cells. MyoD-converted murine fibroblasts participated to muscle regeneration also in immunocompetent, syngeneic mice. Although antibodies from these mice bound to adenoviral infected cells in vitro, no inflammatory infiltrate was present in the graft site throughout the 3-wk study period. These data support the feasibility of an alternative approach to gene therapy of primary myopathies, based on implantation of large numbers of genetically modified primary fibroblasts massively converted to myogenesis by adenoviral delivery of MyoD ex vivo.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Desarrollo de Músculos , Proteína MioD/genética , Animales , Diferenciación Celular , ADN Viral/genética , Fibroblastos , Expresión Génica/genética , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Músculos/citología , Músculos/ultraestructura , Distrofias Musculares/genética , Distrofias Musculares/terapia , Cadenas Pesadas de Miosina/metabolismo , ARN Mensajero/análisis , Regeneración/fisiología
17.
Mol Cell Biol ; 18(11): 6201-12, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9774637

RESUMEN

Hox proteins control developmental patterns and cell differentiation in vertebrates by acting as positive or negative regulators of still unidentified downstream target genes. The homeodomain and other small accessory sequences encode the DNA-protein and protein-protein interaction functions which ultimately dictate target recognition and functional specificity in vivo. The effector domains responsible for either positive or negative interactions with the cell transcriptional machinery are unknown for most Hox proteins, largely due to a lack of physiological targets on which to carry out functional analysis. We report the identification of the transcriptional activation domains of three human Hox proteins, HOXB1, HOXB3, and HOXD9, which interact in vivo with the autoregulatory and cross-regulatory enhancers of the murine Hoxb-1 and human HOXD9 genes. Activation domains have been defined both in a homologous context, i.e., within a HOX protein binding as a monomer or as a HOX-PBX heterodimer to the specific target, and in a heterologous context, after translocation to the yeast Gal4 DNA-binding domain. Transfection analysis indicates that activation domains can be identified in different regions of the three HOX proteins depending on the context in which they interact with the DNA target. These results suggest that Hox proteins may be multifunctional transcriptional regulators, interacting with different cofactors and/or components of the transcriptional machinery depending on the structure of their target regulatory elements.


Asunto(s)
Proteínas de Homeodominio/química , Proteínas de Saccharomyces cerevisiae , Activación Transcripcional/fisiología , Proteínas de Xenopus , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas de Unión al ADN , Genes Reporteros/genética , Humanos , Datos de Secuencia Molecular , Mutación/genética , Proteínas de Neoplasias , Proteínas Recombinantes de Fusión/genética , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/genética , Transfección/genética
19.
Cancer Res ; 49(24 Pt 1): 6989-96, 1989 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-2582441

RESUMEN

The expression of transferrin receptors (TrfRs) was investigated in acute T-cell leukemia (T-ALL) blasts at the molecular, biochemical, immunological, and functional level. TrfRs, although not detected on quiescent T-cells from normal adults, are constitutively expressed at high level on the blasts from all T-ALL patients and bind normally to transferrin. Their number is modulated by the intracellular iron level, but is independent of exogenous interleukin 2. They also exhibit immunological and biochemical abnormalities, in that: (a) they react preferentially with monoclonal antibodies (MAb) that recognize ligand-binding domains of TrfR (42/6 and 43/31), as compared to MAbs (B3/25, OKT9) that interact with the nonligand binding domains; (b) they have a reduced molecular weight, as compared to TrfR on normal thymocytes and activated T-lymphocytes: this phenomenon is apparently related to a defective glycosylation. It is noteworthy that expression of TrfR was not observed in a large series of other types of acute leukemias, i.e., pre-B, B, and myeloid leukemias, excluding erythroleukemias. The constitutive, high level expression of TrfRs on T-ALL blasts may play a key role in the stepwise progression of this malignancy and particularly provide a proliferative advantage to T-ALL blasts as compared to normal T-lymphocytes. Furthermore, indirect evidence suggests that the glycosylation defect of TrfR on T-ALL blasts contributes to their tumorigenic capacity.


Asunto(s)
Leucemia-Linfoma de Células T del Adulto/metabolismo , Receptores de Transferrina/metabolismo , Linfocitos T/metabolismo , Anticuerpos Monoclonales/inmunología , Northern Blotting , Electroforesis en Gel de Poliacrilamida , Glicosilación , Humanos , Hierro/farmacología , Quelantes del Hierro/farmacología , ARN Mensajero/análisis , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología
20.
Oncogene ; 1(1): 29-35, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2449644

RESUMEN

The murine diploid hematopoietic cell line 32D Cl3 strictly requires interleukin-3 (IL-3) for proliferation. When 32D Cl3 cells are transferred to IL-3-free medium which contains recombinant human granulocyte colony stimulating factor (rhG-CSF), the cell number increases four- to five-fold, and after 14 days the whole cell population is differentiated into morphologically normal and myeloperoxidase- and lactoferrin-positive metamyelocytes and granulocytes. Infection with Abelson murine leukemia virus (A-MuLV) of 32D Cl3 cells growing in the presence of IL-3 induces, within 2 weeks, the appearance of cells that are IL-3-independent for growth. The latter cells lack myeloid, T and B cell markers, and are unable to differentiate, even in the presence of very high doses of rhG-CSF. However, once the 32D Cl3 cells have been exposed to G-CSF, they become resistant to the transforming effects of A-MuLV as judged by the appearance of the IL-3-independent clones. These findings suggest that the ability of Abelson virus to transform immature progenitor cells is due to interference of the v-abl gene product with the mechanisms that control the commitment of the cells to differentiate.


Asunto(s)
Virus de la Leucemia Murina de Abelson/genética , Granulocitos/citología , Interleucina-3/fisiología , Virus de la Leucemia Murina/genética , Oncogenes , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Transformación Celular Viral , Factores Estimulantes de Colonias/farmacología , Factor Estimulante de Colonias de Granulocitos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA