Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Gene Ther ; 24(9): 590-592, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28485723

RESUMO

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.


Assuntos
Centros Médicos Acadêmicos/organização & administração , Indústria Farmacêutica/organização & administração , Terapia Genética/métodos , Colaboração Intersetorial , Doenças Raras/terapia , Pesquisa Biomédica/métodos , Pesquisa Biomédica/organização & administração , Humanos
2.
Gene Ther ; 20(9): 949-57, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23615186

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/genética , Epidermólise Bolhosa/genética , Epidermólise Bolhosa/terapia , Vetores Genéticos , Queratinócitos/metabolismo , Vírus da Leucemia Murina de Moloney/genética , Integração Viral , Animais , Moléculas de Adesão Celular/metabolismo , Epidermólise Bolhosa/metabolismo , Regulação da Expressão Gênica , Terapia Genética , HIV-1/genética , Células HeLa , Humanos , Camundongos , Vírus da Leucemia Murina de Moloney/fisiologia , Regiões Promotoras Genéticas , Células Swiss 3T3 , Transdução Genética , Transgenes , Inativação de Vírus , Calinina
3.
Gene Ther ; 18(7): 674-81, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21368897

RESUMO

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.


Assuntos
Fusão Gênica , Terapia Genética/efeitos adversos , Queratina-14/genética , Queratinócitos/metabolismo , Lentivirus/genética , Provírus/genética , Inativação de Vírus , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Humanos , Medição de Risco , Transcrição Gênica , Transdução Genética , Integração Viral
4.
Br J Dermatol ; 161(1): 19-24, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19466960

RESUMO

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.


Assuntos
Epidermólise Bolhosa/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Epidermólise Bolhosa/genética , Vetores Genéticos , Humanos , Retroviridae , Transplante de Células-Tronco
5.
J Pathol ; 217(2): 217-28, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18855878

RESUMO

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.


Assuntos
Células-Tronco Adultas/citologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Epitélio Corneano/citologia , Terapia Genética/métodos , Animais , Células Epidérmicas , Epitélio Corneano/lesões , Epitélio Corneano/patologia , Humanos , Queratinócitos/citologia , Queratinócitos/transplante , Camundongos , Modelos Animais , Regeneração , Dermatopatias/terapia , Cicatrização
6.
Gene Ther ; 15(7): 504-15, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18239607

RESUMO

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.


Assuntos
Sistema Nervoso Central/imunologia , Terapia Genética/métodos , Interleucina-4/genética , Esclerose Múltipla/terapia , Linfócitos T Reguladores/imunologia , Adenoviridae/genética , Animais , Sistema Nervoso Central/patologia , Quimiocinas/imunologia , Quimiotaxia de Leucócito , Modelos Animais de Doenças , Feminino , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Vírus Auxiliares/genética , Humanos , Interleucina-4/análise , Interleucina-4/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética/métodos
7.
Gene Ther ; 15(3): 233-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17960158

RESUMO

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.


Assuntos
Adenoviridae/genética , Líquido Cefalorraquidiano/virologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vírus Auxiliares/genética , Doença de Parkinson/terapia , Animais , Expressão Gênica , Engenharia Genética , Vetores Genéticos/imunologia , Interleucina-4/genética , Macaca fascicularis , Masculino , Modelos Animais , Células Neuroepiteliais/imunologia , Células Neuroepiteliais/virologia , Doença de Parkinson/imunologia , Punção Espinal , Transdução Genética/métodos
8.
Hum Gene Ther ; 18(9): 811-20, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17824830

RESUMO

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.


Assuntos
Vetores Genéticos , Células-Tronco Hematopoéticas/fisiologia , Lentivirus/genética , Transdução de Sinais , Proteínas do Envelope Viral/genética , Animais , Antígenos CD34/sangue , Antígenos CD34/imunologia , Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Vírus da Imunodeficiência Felina/química , Vírus da Leucemia Murina/genética , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Transplante Heterólogo
10.
Blood ; 98(9): 2664-72, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11675336

RESUMO

Use of oncoretroviral vectors in gene therapy for hemoglobinopathies has been impeded by low titer vectors, genetic instability, and poor expression. Fifteen self- inactivating (SIN) lentiviral vectors using 4 erythroid promoters in combination with 4 erythroid enhancers with or without the woodchuck hepatitis virus postregulatory element (WPRE) were generated using the enhanced green fluorescent protein as a reporter gene. Vectors with high erythroid-specific expression in cell lines were tested in primary human CD34(+) cells and in vivo in the murine bone marrow (BM) transplantation model. Vectors containing the ankyrin-1 promoter showed high-level expression and stable proviral transmission. Two vectors containing the ankyrin-1 promoter and 2 erythroid enhancers (HS-40 plus GATA-1 or HS-40 plus 5-aminolevulinate synthase intron 8 [I8] enhancers) and WPRE expressed at levels higher than the HS2/beta-promoter vector in bulk unilineage erythroid cultures and individual erythroid blast-forming units derived from human BM CD34(+) cells. Sca1(+)/lineage(-) Ly5.1 mouse hematopoietic cells, transduced with these 2 ankyrin-1 promoter vectors, were injected into lethally irradiated Ly5.2 recipients. Eleven weeks after transplantation, high-level expression was seen from both vectors in blood (63%-89% of red blood cells) and erythroid cells in BM (70%-86% engraftment), compared with negligible expression in myeloid and lymphoid lineages in blood, BM, spleen, and thymus (0%-4%). The I8/HS-40-containing vector encoding a hybrid human beta/gamma-globin gene led to 43% to 113% human gamma-globin expression/copy of the mouse alpha-globin gene. Thus, modular use of erythroid-specific enhancers/promoters and WPRE in SIN-lentiviral vectors led to identification of high-titer, stably transmitted vectors with high-level erythroid-specific expression for gene therapy of red cell diseases.


Assuntos
Células Precursoras Eritroides/metabolismo , Vetores Genéticos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Lentivirus/genética , Transdução Genética/métodos , Animais , Antígenos CD34 , Células da Medula Óssea , Transplante de Medula Óssea , Elementos Facilitadores Genéticos , Expressão Gênica , Regulação Viral da Expressão Gênica , Vetores Genéticos/normas , Proteínas de Fluorescência Verde , Vírus da Hepatite B da Marmota/genética , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Modelos Animais , Regiões Promotoras Genéticas , Processamento Pós-Transcricional do RNA/genética , Transdução Genética/normas , gama-Globulinas/genética , gama-Globulinas/metabolismo
12.
J Biol Chem ; 276(23): 20506-15, 2001 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-11278854

RESUMO

Regionally restricted expression patterns of Hox genes in developing embryos rely on auto-, cross-, and para-regulatory transcriptional elements. One example is the Hoxb1 auto-regulatory element (b1-ARE), which drives expression of Hoxb1 in the fourth rhombomere of the hindbrain. We previously showed that HOXB1 and PBX1 activate transcription from the b1-ARE by binding to sequences required for the expression of a reporter gene in rhombomere 4 in vivo. We now report that in embryonal carcinoma cells, which retain characteristics of primitive neuroectodermal cells, the b1-ARE displays higher basal and HOX/PBX-induced activities than in other cell backgrounds. We have identified a bipartite-binding site for SOX/OCT heterodimers within the b1-ARE that accounts for its cell context-specific activity and is required for maximal transcriptional activity of HOX/PBX complexes in embryonal carcinoma cells. Furthermore, we found that in an embryonal carcinoma cell background, HOXB1 has a significantly higher transcriptional activity than its paralog HOXA1. We map the determinants for this differential activity within the HOXB1 N-terminal transcriptional activation domain. By using analysis in transgenic and HOXA1 mutant mice, we extended these findings on the differential activities of HOXA1 and HOXB1 in vivo, and we demonstrated that they are important for regulating aspects of HOXB1 expression in the hindbrain. We found that mutation of the SOX/OCT site and targeted inactivation of Hoxa1 both impair the response of the b1-ARE to retinoic acid in transgenic mice. Our results show that Hoxa1 is the primary mediator of the response of b1-ARE to retinoic acid in vivo and that this function is dependent on the binding of SOX/OCT heterodimers to the b1-ARE. These results uncover novel functional differences between Hox paralogs and their modulators.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Sondas de DNA , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Fatores de Transcrição/fisiologia , Ativação Transcricional/fisiologia , Tretinoína/farmacologia
13.
Hum Gene Ther ; 11(16): 2283-7, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11084687

RESUMO

Junctional epidermolysis bullosa (JEB) is a group of severe, inherited skin diseases caused by mutations in the genes encoding laminin 5 or other components of the hemidesmosome. Since human epidermis is a self-renewing tissue, gene therapy of JEB requires the stable integration of the transgene into the genome of the epidermal stem cell. Human epidermal stem cells can indeed be cultivated and stably transduced with replication-defective retroviral vectors, allowing full phenotypic correction of the adhesion properties of JEB keratinocytes. Epidermal stem cells generate cohesive sheets of stratified epithelium suitable for the permanent coverage of massive skin defects, and genetically modified epidermal sheets maintain long-term expression of the transgene after transplantation on immunodeficient animals. Moreover, we have developed a clinical procedure that allows transplantation of cultured epidermal sheets on large body areas under local anesthesia and without cicatricial outcomes. Thus, (1) the possibility of cultivating lining epithelia, (2) the availability of noninvasive surgical procedures that allow the grafting of large skin areas, and (3) the demonstration of sustained transgene expression in vitro and in vivo by epidermal stem cells, prompt us to propose the implementation of a phase I/II clinical trial aimed at the ex vivo gene therapy of selected JEB patients. The aim of the trial is to validate the ex vivo procedure in a clinical setting, to prove its overall safety, and to analyze critical issues such as long-term survival of the genetically modified implant, immune response against the transgene product, and persistence of transgene expression at therapeutic levels.


Assuntos
Epiderme/metabolismo , Epidermólise Bolhosa Juncional/genética , Epidermólise Bolhosa Juncional/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Células-Tronco/citologia , Células Cultivadas , Humanos , Queratinócitos/citologia , Mutação , Retroviridae/genética , Transgenes , Transplante Heterólogo
15.
Hum Gene Ther ; 10(10): 1607-17, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10428206

RESUMO

Myogenic cells have a limited life span in culture, which prevents expansion at clinically relevant levels, and seriously limits any potential use in cell replacement or ex vivo gene therapy. We developed a strategy for reversibly immortalizing human primary myogenic cells, based on retrovirus-mediated integration of a wild-type SV40 large-T antigen (Tag), excisable by means of the Cre-Lox recombination system. Myogenic cells were transduced with a vector (LTTN-LoxP) expressing the SV40 Tag under the control of an LTR modified by the insertion of a LoxP site in the U3 region. Clonal isolates of Tag-positive cells showed modified growth characteristics and a significantly extended life span, while maintaining a full myogenic potential. Transient expression of Cre recombinase, delivered by transfection or adenoviral vector transduction, allowed excision of the entire provirus with up to >90% efficiency. Cultures of Cre-treated (Tag-) or untreated (Tag+) myogenic cells were genetically labeled with a lacZ retroviral vector, and injected into the regenerating muscle of SCID/bg immunodeficient mice. Tag- cells underwent terminal differentiation in vivo, giving rise to clusters of beta-Gal+ hybrid fibers with an efficiency comparable to that of control untransduced cells. Tag+ cells could not be detected after injection. Neither Tag+ nor Tag- cells formed tumor in this xenotransplantation model. Reversible immortalization by Tag therefore allows the expansion of primary myogenic cells in culture without compromising their ability to differentiate in vivo, and could represent a safe method by which to increase the availability of these cells for clinical application.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Integrases , Vírus da Leucemia Murina de Moloney , Proteínas Virais , Células 3T3 , Adulto , Animais , Diferenciação Celular , Divisão Celular , Transformação Celular Viral , Células Cultivadas , Pré-Escolar , Humanos , Camundongos , Músculos/citologia , Oncogenes
16.
Blood ; 93(10): 3276-85, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10233879

RESUMO

Targeted expression to specific tissues or cell lineages is a necessary feature of a gene therapy vector for many clinical applications, such as correction of hemoglobinopathies or thalassemias by transplantation of genetically modified hematopoietic stem cells. We developed retroviral vectors in which the constitutive viral enhancer in the U3 region of the 3' LTR is replaced by an autoregulatory enhancer of the erythroid-specific GATA-1 transcription factor gene. The replaced enhancer is propagated to the 5' LTR upon integration into the target cell genome. The modified vectors were used to transduce human hematopoietic cell lines, cord blood-derived CD34(+) stem/progenitor cells, and murine bone marrow repopulating stem cells. The expression of appropriate reporter genes (triangle upLNGFR, EGFP) was analyzed in the differentiated progeny of transduced stem cells in vitro, in liquid culture as well as in clonogenic assay, and in vivo, after bone marrow transplantation in lethally irradiated mice. The GATA-1 autoregulatory enhancer effectively restricts the expression of the LTR-driven proviral transcription unit to the erythroblastic progeny of both human progenitors and mouse-repopulating stem cells. Packaging of viral particles, integration into the target genome, and stability of the integrated provirus are not affected by the LTR modification. Enhancer replacement is therefore an effective strategy to target expression of a retroviral transgene to a specific progeny of transduced hematopoietic stem cells.


Assuntos
Proteínas de Ligação a DNA/genética , Eritroblastos/citologia , Células-Tronco Hematopoéticas/fisiologia , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Linhagem Celular , Proteínas de Ligação a DNA/biossíntese , Elementos Facilitadores Genéticos , Eritroblastos/fisiologia , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA1 , Genes Reporter , Terapia Genética/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/citologia , Humanos , Células K562 , Leucemia Eritroblástica Aguda , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Receptor de Fator de Crescimento Neural , Receptores de Fator de Crescimento Neural/biossíntese , Receptores de Fator de Crescimento Neural/genética , Sequências Reguladoras de Ácido Nucleico , Retroviridae , Fatores de Transcrição/biossíntese , Transfecção , Células Tumorais Cultivadas , Células U937
17.
Genes Dev ; 13(8): 946-53, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10215622

RESUMO

Nuclear localization of the Extradenticle (EXD) and PBX1 proteins is regionally restricted during Drosophila and mammalian development. We studied the subcellular localization of EXD, PBX, and their partners Homothorax (HTH) and PREP1, in different cell contexts. HTH and PREP1 are cytoplasmic and require association with EXD/PBX for nuclear localization. EXD and PBX1 are nuclear in murine fibroblasts but not in Drosophila Schneider cells, in which they are actively exported to the cytoplasm. Coexpression of EXD/PBX with HTH/PREP1 causes nuclear localization of their heterodimers in both cell contexts. We propose that heterodimerization with HTH/PREP induces nuclear translocation of EXD and PBX1 in specific cell contexts by blocking their nuclear export.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Drosophila , Camundongos , Sinais de Localização Nuclear , Fator de Transcrição 1 de Leucemia de Células Pré-B , Frações Subcelulares
18.
Mol Cell Biol ; 18(11): 6201-12, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9774637

RESUMO

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.


Assuntos
Proteínas de Homeodomínio/química , Proteínas de Saccharomyces cerevisiae , Ativação Transcricional/fisiologia , Proteínas de Xenopus , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas de Ligação a DNA , Genes Reporter/genética , Humanos , Dados de Sequência Molecular , Mutação/genética , Proteínas de Neoplasias , Proteínas Recombinantes de Fusão/genética , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/genética , Transfecção/genética
19.
J Clin Invest ; 101(10): 2119-28, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9593768

RESUMO

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.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Desenvolvimento Muscular , Proteína MyoD/genética , Animais , Diferenciação Celular , DNA Viral/genética , Fibroblastos , Expressão Gênica/genética , Terapia Genética/métodos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Músculos/citologia , Músculos/ultraestrutura , Distrofias Musculares/genética , Distrofias Musculares/terapia , Cadeias Pesadas de Miosina/metabolismo , RNA Mensageiro/análise , Regeneração/fisiologia
20.
EMBO J ; 17(5): 1423-33, 1998 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-9482739

RESUMO

The human transcription factor, UEF3, is important in regulating the activity of the urokinase plasminogen activator (uPA) gene enhancer. The UEF3 DNA target site is a regulatory element in the promoters of several growth factor and protease genes. We reported previously that purified UEF3 is a complex of several subunits. In this paper we report the cloning of the cDNA of one of the subunits which encodes for a novel human homeodomain protein, which we have termed Prep1. The Prep1 homeodomain belongs to the TALE class of homeodomains, is most closely related to those of the TGIF and Meis1 proteins, and like these, recognizes a TGACAG motif. We further identify the other UEF3 subunit as a member of the Pbx protein family. Unlike other proteins known to interact with Pbx, Prep1 forms a stable complex with Pbx independent of DNA binding. Heterodimerization of Prep1 and Pbx results in a strong DNA binding affinity towards the TGACAG target site of the uPA promoter. Overall, these data indicate that Prep1 is a stable intracellular partner of Pbx in vivo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , Reações Cruzadas , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Dimerização , Regulação da Expressão Gênica/genética , Células HeLa , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Humanos , Dados de Sequência Molecular , Peso Molecular , Proteína Meis1 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes de Fusão , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Fatores de Transcrição/genética , Ativador de Plasminogênio Tipo Uroquinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA