Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 11(21): 1599-605, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15334112

RESUMO

We describe here a clonal approach for efficient and robust construction of recombinant adenoviral genomes that holds certain advantages over existing approaches. Transgenes of interest are cloned into a small, conditionally replicating plasmid containing the left end of a recombinant adenoviral genome, encompassing pIX coding regions. Transformation of this plasmid into recombination-competent Escherichia coli bearing a plasmid containing the right end of a recombinant adenoviral genome, commencing from pIX coding regions, yields a stable co-integrated plasmid encoding a full adenoviral genome, by virtue of shared homology in pIX coding regions contained in both plasmids. The recombination process yielding the full adenoviral plasmid requires only one step, and always results in the formation of only the desired recombinant adenoviral genome. Thus, no screening is required to identify the correct plasmid encoding the desired recombinant adenoviral genome. In addition, the plasmid encoding the right-hand side of the adenoviral genome is itself incapable of producing contaminating adenovirus. We have successfully employed this approach to generate over 200 recombinant adenoviruses, obtaining only the desired recombinant adenoviral species each time. The process is amenable to medium-to-high-throughput parallel construction of adenoviral genomes, and as such should aid efforts aimed towards high-throughput functional annotation of therapeutic gene targets, which aim to leverage the benefits of adenoviruses as gene delivery and expression vectors.


Assuntos
Adenoviridae/genética , Engenharia Genética , Vetores Genéticos/genética , Genoma Viral , Recombinação Genética , Animais , Clonagem Molecular , Escherichia coli/genética , Luciferases de Vaga-Lume/genética , Plasmídeos , Transgenes
2.
Cytogenet Genome Res ; 98(2-3): 160-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12697998

RESUMO

The ABCA subfamily of ATP-binding cassette (ABC) transporters includes eleven members to date. In this study, we describe a new, unusually large gene on chromosome 7p12.3, ABCA13. This gene spans over 450 kb and is split into 62 exons. The predicted ABCA13 protein consists of 5,058 ami- no acid residues making it the largest ABC protein described to date. Like the other ABCA subfamily members, ABCA13 contains a hydrophobic, predicted transmembrane segment at the N-terminus, followed by a large hydrophilic region. In the case of ABCA13, the hydrophilic region is unexpectedly large, more than 3,500 amino acids, encoded by 30 exons, two of which are 4.8 and 1.7 kb in length. These two large exons are adjacent to each other and are conserved in the mouse Abca13 gene. Tissue profiling of the major transcript reveals the highest expression in human trachea, testis, and bone marrow. The expression of the gene was also determined in 60 tumor cell lines and the highest expression was detected in the SR leukemia, SNB-19 CNS tumor and DU-145 prostate tumor cell lines. ABCA13 has high similarity with other ABCA subfamily genes which are associated with human inherited diseases: ABCA1 with the cholesterol transport disorders Tangier disease and familial hypoalphalipoproteinemia, and ABCA4 with several retinal degeneration disorders. The ABCA13 gene maps to chromosome 7p12.3, a region that contains an inherited disorder affecting the pancreas (Shwachman-Diamond syndrome) as well as a locus involved in T-cell tumor invasion and metastasis (INM7), and therefore is a positional candidate for these pathologies.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Cromossomos Humanos Par 7 , Transportadores de Cassetes de Ligação de ATP/biossíntese , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Sequência Conservada , DNA Complementar/isolamento & purificação , Éxons , Humanos , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , RNA Mensageiro/biossíntese , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Células Tumorais Cultivadas
3.
Cytogenet Cell Genet ; 92(3-4): 264-70, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11435699

RESUMO

We report here the genomic and transcriptional characterization in mouse and man of a novel transporter of the ABCA subclass, named ABCA7. As it is the case for other ABCA genes, the predicted protein encoded by ABCA7 is a full symmetric transporter, highly conserved across species. The ABCA7 gene maps to human chromosome 19 and to the homologous region at band B4-C1 on mouse chromosome 10. The preferential expression of ABCA7 in the spleen, thymus, and fetal liver is consistent with the finding, in both human and mouse promoter, of sites targeted by lymphomyeloid-specific transcription factors. This suggests that ABCA7 may play a pivotal role in the developmental specification of hematopoietic cell lineages.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Éxons/genética , Íntrons/genética , Regiões Promotoras Genéticas/genética , Transportadores de Cassetes de Ligação de ATP/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Cromossomos Humanos Par 19/genética , Sequência Conservada/genética , DNA Complementar/genética , Humanos , Hibridização in Situ Fluorescente , Fígado/embriologia , Fígado/metabolismo , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , RNA Mensageiro/análise , RNA Mensageiro/genética , Mapeamento de Híbridos Radioativos , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Baço/metabolismo , Timo/metabolismo
4.
Circulation ; 99(1): 105-10, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-9884386

RESUMO

BACKGROUND: Apolipoprotein (apo) A-I is the major component of HDL, and it displays antiatherogenic properties. METHODS AND RESULTS: The human apoA-I gene has been transferred into different mouse models by use of a recombinant adenovirus under the control of an RSV-LTR promoter (AV RSV apoA-I). Administration of AV RSV apoA-I to C57BL/6 mice resulted in moderate expression of human apoA-I for 3 weeks, leading to a transient elevation (40% at day 11 after injection) of HDL cholesterol concentration. In contrast, administration of AV RSV apoA-I to human apoA-I-transgenic mice induced a large increase of human apoA-I and HDL cholesterol concentrations (300% and 360%, respectively, at day 14 after injection) for 10 weeks, indicating that an immune response to the transgene was one major hurdle for long-term duration of expression. Recombinant adenovirus expressing human apolipoprotein A-I (AV RSV apoA-I) was also injected into human apoA-I-transgenic/apoE-deficient mice, which are prone to develop atherosclerosis. Over a 6-week period, overexpression of human apoA-I inhibited fatty streak lesion formation by 56% in comparison with control. CONCLUSIONS: Somatic gene transfer of human apoA-I prevents the development of atherosclerosis in the mouse model.


Assuntos
Apolipoproteína A-I/genética , Arteriosclerose/terapia , Técnicas de Transferência de Genes , Adenoviridae/genética , Animais , Apolipoproteínas E/deficiência , Modelos Animais de Doenças , Progressão da Doença , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Valores de Referência
5.
Clin Invest Med ; 21(4-5): 172-85, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9800066

RESUMO

The authors previously demonstrated that the gene for human lipoprotein lipase (hLPL), an enzyme crucial to the breakdown of triglyceride (TG)-rich dietary fats, corrects the hypertriglyceridemia in lipoprotein lipase (LPL)-deficient knockout mice after adenoviral (Ad)-mediated LPL gene transfer. They have now extended their observations to primary cultured mouse hepatocytes and intact animals of normal LPL genotype, and confirm effective overexpression of hLPL from the liver and a sustained TG-lowering effect in plasma over 60 days. A typical first-generation Ad-vector containing the hLPL cDNA (Ad-LPL) resulted in efficient gene transfer into isolated mouse hepatocytes and significant de novo synthesis of active hLPL protein. In this experiment, 5 x 10(9) viral particles (5 x 10(7) pfu) of either Ad-LPL or an Ad-LacZ control vector were injected into CD1 mice of normal LPL genotype. Hepatic expression of hLPL was confirmed at Day 7 postinjection by in situ hybridization and direct measurement of LPL in the liver. This correlated with a total LPL activity (human + mouse) in postheparin plasma (PHP) of 1020.5 standard deviation [SD] 93.6 mU/mL, versus 479.5 SD 129.7 mU/mL (p < 0.001) in Ad-LacZ controls at Day 7. Respective hLPL activity comprised 49% of the total. Significantly raised levels of hLPL protein mass persisted until Day 60. Corresponding plasma TGs decreased to 39% of Ad-LacZ controls at Day 7, and, despite absent hLPL activity from Day 28 on, serum TGs remained significantly lower in Ad-LPL mice up to Day 42. Fast phase liquid chromatography analysis showed a dramatic depletion in TG-rich lipoproteins, mainly very low density lipoproteins (VLDL) and chylomicron fractions. Therefore, Ad-mediated overexpression of hepatic LPL was found to significantly decrease plasma TG levels unrelated to primary LPL deficiency.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Lipólise/genética , Lipase Lipoproteica/genética , Fígado/enzimologia , Animais , Células Cultivadas , Quilomícrons/sangue , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Hibridização In Situ , Lipase Lipoproteica/metabolismo , Lipoproteínas/sangue , Fígado/virologia , Masculino , Camundongos , Camundongos Endogâmicos , Triglicerídeos/sangue
6.
Biochim Biophys Acta ; 1391(3): 329-36, 1998 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-9555083

RESUMO

Rat hepatocytes cocultured with rat liver epithelial cells (RLEC) were used to investigate the influence of all-trans retinoic acid (RA) on the regulation of apolipoproteins (Apo) A-I and A-II gene expression, the major protein constituent of high-density lipoproteins. In contrast to rat hepatocytes in conventional primary culture, Apo A-I and Apo A-II gene expression remained high and stable for several days in parenchymal cells in coculture. Treatment of cocultured rat hepatocytes with RA resulted in a specific decrease in Apo A-I mRNA levels whereas no marked difference in Apo A-II mRNA levels was observed. Such a negative effect of RA was already detected as early as 2 days of treatment and was effective for the entire experimental period (6 days). As controls, RARbeta mRNA levels increased whereas those of GAPDH mRNA were not affected by the RA treatment. The decrease in Apo A-I mRNA levels was associated with lower amounts of Apo A-I secreted in the culture medium within day 1 of treatment. This effect required active transcription and protein synthesis. These results show that, contrary to primary pure hepatocyte cultures and hepatoma cell lines, cocultures of rat hepatocytes reproduce the in vivo results suggesting that only well differentiated hepatocytes may correctly respond to RA. Furthermore, they demonstrate that RA can directly act on hepatocytes and differently affect Apo A-I and Apo A-II gene expression.


Assuntos
Apolipoproteína A-I/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Tretinoína/farmacologia , Albuminas/efeitos dos fármacos , Albuminas/metabolismo , Animais , Apolipoproteína A-I/biossíntese , Apolipoproteína A-I/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
7.
Arterioscler Thromb Vasc Biol ; 17(11): 2532-9, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9409224

RESUMO

Humans homozygous or heterozygous for mutations in the lipoprotein lipase (LPL) gene demonstrate significant disturbances in plasma lipoproteins, including raised triglyceride (TG) and reduced HDL cholesterol levels. In this study we explored the feasibility of adenovirus-mediated gene replacement therapy for LPL deficiency. A total of 5 x 10(9) plaque-forming units (pfu) of an E1/E3-deleted adenovirus expressing either human LPL (Ad-LPL) or the bacterial beta-galactosidase gene (Ad-LacZ) as a control were administered to mice heterozygous for targeted disruption in the LPL gene (n = 57). Peak expression of total postheparin plasma LPL activity was observed at day 7 in Ad-LPL mice versus Ad-LacZ controls (834 +/- 133 vs 313 +/- 89 mU/mL, P < .01), and correlated with human-specific LPL activity (522 +/- 219 mU/mL) and mass (9214 +/- 782 ng/mL), a change that was significant to 14 and 42 days, respectively. At day 7, plasma TGs were significantly reduced relative to Ad-LacZ mice (0.17 +/- 0.07 vs 1.90 +/- 0.89 mmol/L, P < .01) but returned to endogenous levels by day 42. Ectopic liver expression of human LPL was confirmed by in situ hybridization analysis and from raised LPL activity and mass in liver homogenates. Analysis of plasma lipoprotein composition revealed a marked decrease in VLDL-derived TGs. Severely impaired oral and intravenous fat-load tolerance in LPL-deficient mice was subsequently corrected after Ad-LPL administration and closely paralleled that observed in wild-type mice. These findings suggest that liver-targeted adenovirus-mediated LPL gene transfer offers an effective means for transient correction of altered lipoprotein metabolism and impaired fat tolerance due to LPL deficiency.


Assuntos
Terapia Genética , Hiperlipoproteinemia Tipo I/terapia , Hipertrigliceridemia/terapia , Lipase Lipoproteica/genética , Adenoviridae/genética , Animais , Gorduras na Dieta , Vetores Genéticos/genética , Heterozigoto , Humanos , Hipertrigliceridemia/etiologia , Lipase Lipoproteica/sangue , Lipase Lipoproteica/fisiologia , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes de Fusão/fisiologia
8.
Biochim Biophys Acta ; 1358(1): 103-12, 1997 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-9296527

RESUMO

Previous studies have indicated that in HepG2 cells HDL3-signalling involves glycosylphosphatidylinositol (GPI) anchored proteins. HDL3-binding to HepG2 cells was found to be enhanced by cellular preincubation with PI-PLC inhibitors and sensitive to a cellular preincubation with exogenous PI-PLC, suggesting that HDL3 binds directly on GPI-anchored proteins to initiate signaling. Moreover HDL3-binding was found to be partly inhibited by antibodies against the HDL-binding protein (AbHBP). HDL3, when binding to HepG2 cells, promoted the release in the culture medium of a 110 kDa protein that binds AbHBP, while a cellular preincubation with antibodies against the inositol-phosphoglycan (IPG) moiety of GPI-anchor (AbIPG), used to block lipolytic cleavage of the GPI-anchor, inhibits HDL3-induced release of the 110 kDa protein in the culture medium. In [3H]-PC prelabeled HepG2 cells, AbHBP were found to stimulate PC-hydrolysis and DAG generation within 5 min as did HDL3 stimulation. Cellular preincubation with AbIPG was found to inhibit only the HDL3-signal and not the AbHBP-signal, while a prior cellular pretreatment with PI-PLC from Bacillus cereus was found to inhibit the HDL3-and AbHBP-signal. Moreover cellular preincubation with AbHBP for 1 h at 37 degrees C was found to inhibit HDL3-signalling pathways. Our results suggest that in HepG2 cells a 110 kDa protein, which could be HBP, can be anchored to the membrane via GPI, and can function in HDL3-signalling pathways as binding sites.


Assuntos
Proteínas de Transporte , Glicosilfosfatidilinositóis/metabolismo , Lipoproteínas HDL/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA , Receptores de Lipoproteínas/metabolismo , Anticorpos/farmacologia , Sítios de Ligação , Humanos , Proteínas de Membrana/imunologia , Fosfatidilcolinas/metabolismo , Fosfolipases/antagonistas & inibidores , Receptores de Lipoproteínas/imunologia , Transdução de Sinais , Células Tumorais Cultivadas
9.
Genes Dev ; 11(13): 1674-89, 1997 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9224717

RESUMO

gax, a diverged homeobox gene expressed in vascular smooth muscle cells (VSMCs), is down-regulated in vitro by mitogen stimulation and in vivo in response to vascular injury that leads to cellular proliferation. Recombinant Gax protein microinjected into VSMCs and fibroblasts inhibited the mitogen-induced entry into S-phase when introduced either during quiescence or early stages of G1. Overexpression of gax with a replication-defective adenovirus vector resulted in G0/G1 cell cycle arrest of VSMCs and fibroblasts. The gax-induced growth inhibition correlated with a p53-independent up-regulation of the cyclin-dependent kinase inhibitor p21. Gax overexpression also led to an association of p21 with cdk2 complexes and a decrease in cdk2 activity. Fibroblasts deficient in p21 were not susceptible to a reduction in cdk2 activity or growth inhibition by gax overexpression. Localized delivery of the virus to denuded rat carotid arteries significantly reduced neointima formation and luminal narrowing. These data indicate that gax overexpression can inhibit cell proliferation in a p21-dependent manner and can modulate injury-induced changes in vessel wall morphology that result from excessive cellular proliferation.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclinas/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas Musculares/fisiologia , Adenoviridae/genética , Adenoviridae/fisiologia , Angioplastia com Balão/efeitos adversos , Animais , Lesões das Artérias Carótidas , Divisão Celular , Linhagem Celular , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Regulação da Expressão Gênica , Vetores Genéticos , Proteínas de Homeodomínio/genética , Humanos , Proteínas Musculares/genética , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteína Supressora de Tumor p53/genética
10.
Circulation ; 96(2): 408-11, 1997 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9244204

RESUMO

BACKGROUND: Gene delivery of the thymidine kinase (tk) gene combined with ganciclovir (GCV) limits intimal hyperplasia after abrasion of normal arteries. However, the low efficiency of adenoviral-mediated gene transfer to atherosclerotic arteries has raised concerns about the applicability of this strategy to the prevention of restenosis. METHODS AND RESULTS: A replication-defective adenoviral vector expressing tk (Ad-RSVtk) demonstrated selective toxicity toward GCV-treated arterial smooth muscle cells, with oligonucleolytic cleavage suggesting apoptosis. In vivo, after demonstration of tk expression after Ad-RSVtk delivery, the combination of Ad-RSVtk followed by GCV was tested in a rabbit model of angioplasty of atheromatous iliac arteries. Angioplasty (8 atm, 20 minutes) was performed by use of a hydrogel balloon coated with Ad-RSVtk (4x10(9) plaque forming units). GCV was infused (25 mg.kg(-1) I.V. BID) from days 2 through 7 after angioplasty in 8 of 12 rabbits. Four weeks later, morphometric analysis demonstrated a reduced intima-to-media ratio in the group receiving combination therapy compared with Ad-RSVtk alone (3.0+/-1.2 versus 5.2+/-0.5, P<.018). GCV per se had no effect on intimal hyperplasia after arterial injury. CONCLUSIONS: In vitro, Ad-RSVtk demonstrates selective toxicity toward GCV-treated arterial smooth muscle cells involving apoptosis. In vivo, GCV conditions reduction of neointimal formation after percutaneous delivery of Ad-RSVtk during angioplasty of atheromatous arteries.


Assuntos
Angioplastia com Balão , Aorta/patologia , Arteriosclerose/terapia , Terapia Genética , Timidina Quinase/genética , Animais , Arteriosclerose/fisiopatologia , Morte Celular/genética , Modelos Animais de Doenças , Ganciclovir , Técnicas de Transferência de Genes , Coelhos , Recidiva , Túnica Íntima/patologia
11.
Proc Natl Acad Sci U S A ; 94(4): 1414-9, 1997 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-9037067

RESUMO

A two-step gene replacement procedure was developed that generates infectious adenoviral genomes through homologous recombination in Escherichia coli. As a prerequisite, a human adenovirus serotype 5 (Ad5)-derived genome was first introduced as a PacI restriction fragment into an incP-derived replicon which, in contrast to ColE1-derivatives (e.g., pBR322 or pUC plasmids), is functional in a polA mutant of E. coli. Any modification can be introduced at will following two consecutive homologous recombinations between the incP/Ad5 replicon and the ColE1 plasmid. The overall procedure requires only the in vitro engineering of the ColE1-derivative by flanking the desired modification with small stretches of identical sequences. In the first step, a cointegrate between the tetracycline-resistant incP/Ad5 replicon and the kanamycin-resistant ColE1-derivative is selected by growing the polA host in the presence of both antibiotics. Resolution of this cointegrate is further selected in sucrose growth conditions due to the loss of a conditional suicide marker (the sacB gene of Bacillus subtilis) present in the ColE1 plasmid, leading to unmodified and modified incP/Ad5 replicons that can be differentiated upon restriction analysis. Consecutive rounds of this two-step cloning procedure allowed the introduction of multiple independent modifications within the virus genome, with no requirement for an intermediate virus. The potential of this procedure is demonstrated by the recovery of several E1E3E4-deleted adenoviruses following transfection of the corresponding E. coli-derived genomes in IGRP2 cells.


Assuntos
Adenovírus Humanos/genética , Clonagem Molecular/métodos , Engenharia Genética/métodos , Genoma Viral , Recombinação Genética , Proteínas E1 de Adenovirus/genética , Proteínas E3 de Adenovirus/genética , Adenovírus Humanos/crescimento & desenvolvimento , Adenovírus Humanos/patogenicidade , Escherichia coli/genética , Marcadores Genéticos , Resistência a Canamicina , Plasmídeos/genética , Replicon , Resistência a Tetraciclina
12.
Biochemistry ; 36(8): 2243-9, 1997 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-9047326

RESUMO

Studies assessing fatty streak formation in mice have revealed that human apolipoprotein A-I (apoAI) transgenic mice (TgAI) have 15-fold less atherosclerosis susceptibility than combined human apolipoprotein A-I/human apolipoprotein A-II (apoAI:AII) transgenics (TgAI:AII) and 40-fold less than nontransgenic control mice. In order to examine the biochemical mechanisms underlying those in vivo observations, we have compared in vitro properties of serum from the different groups of animals for participation in cholesterol efflux, LCAT activation, and pre-beta particle formation. Analysis of cholesterol efflux from both Fu5AH hepatoma and Ob1771 adipose cells revealed serum from the TgAI to be the most efficient in promoting efflux. The two-dimensional electrophoresis of mouse serum shows that control mice have exclusively apoAI in alpha particles. TgAI and TgAI:AII mice have 30 and 38% of total apoAI in particles with pre-beta electrophoretic mobility, respectively. The distribution of cell-derived cholesterol between these apoAI-containing lipoprotein subspecies after 1 and 60 min of incubation with Fu5AH hepatoma cells was examined. This revealed after a 1 min incubation 66 +/- 8 and 83 +/- 9% of the counts in particles with pre-beta mobility for TgAI and TgAI:AII mice, respectively; while after 60 min of incubation, only 6 +/- 2% of counts remained in pre-beta particles from the TgAI and 30 +/- 3% for the TgAI:AII. This suggests faster movement of cholesterol from pre-beta to alpha particles in plasma from the TgAI. Consistent with this is the observation that LCAT activity with both exogenous and endogenous substrate increased in the TgAI versus the TgAI:AII mice. The previously observed decrease in fatty streak formation in the TgAI versus the TgAI:AII and control mice is consistent with the in vitro studies presented here and suggests that HDL containing human apoAI is a more effective participant in the postulated early steps in reverse cholesterol transport than HDL containing both human apoAI and human apoAII, and/or murine HDL.


Assuntos
Apolipoproteína A-II/sangue , Apolipoproteína A-I/sangue , Colesterol/sangue , Fosfatidilcolina-Esterol O-Aciltransferase/sangue , Animais , Apolipoproteína A-I/genética , Apolipoproteína A-II/genética , Transporte Biológico , Linhagem Celular , Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos
13.
Hum Gene Ther ; 8(2): 205-14, 1997 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9017424

RESUMO

Gene therapy to deliver and express a corrective lipoprotein lipase (LPL) gene may improve the lipid profile and reduce the morbidity and potential atherogenic risk from hypertriglyceridemia and dyslipoproteinemia in patients with complete or partial LPL deficiency. We have used an E1-/E3- adenoviral vector, with an RSV-driven human LPL cDNA expression cassette (Ad-RSV-LPL), to achieve high ectopic LPL gene expression in the human hepatoma cell line HepG2, an accepted hepatocellular model of lipoprotein metabolism. Ad-RSV-LPL transduction of HepG2 cells with a multiplicity of infection (moi) between 12.5 and 100 yielded dose-dependent increments in LPL mass and activity. Peak levels of LPL protein of 2,032.1 +/- 274.5 ng/10(5) cells per ml (mol 100) correlated with increased activity of 92.7 +/- 22.6 mU/10(5) cells per ml relative to negligible LPL levels in Ad-RSV-LacZ (beta-galactosidase) controls. Exogenous LPL expression over a 5-day period peaked at day 3. Susceptibility to inhibition by 1 M NaCl and an anti-LPL monoclonal antibody confirmed that lipase activity was indeed derived from human LPL. Hydrolysis, by LPL-overexpressing HepG2 cells, of TG carried in very-low-density lipoprotein (VLDL) showed that greater than 50% of the triglycerides (TG) disappeared after 4 hr of incubation. These results were compatible with FPLC evidence of a marked reduction in VLDL-TG. These results provide strong in vitro evidence that adenoviral-mediated ectopic expression of the human LPL gene could render hepatic cells capable of VLDL catabolism and thus support the possibility for in vivo adenoviral vector-mediated liver-targeted LPL gene therapy.


Assuntos
Adenoviridae/genética , Carcinoma Hepatocelular/metabolismo , Vetores Genéticos/genética , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Proteínas E1 de Adenovirus/genética , Proteínas E3 de Adenovirus/genética , Vírus do Sarcoma Aviário/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , DNA Complementar/genética , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Hidrólise , Lipoproteínas VLDL/química , Lipoproteínas VLDL/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Triglicerídeos/química , Triglicerídeos/metabolismo , Células Tumorais Cultivadas
14.
Cardiovasc Res ; 35(3): 536-46, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9415299

RESUMO

OBJECTIVES: The expression of gax, an anti-proliferative homeobox gene, is rapidly downregulated in vascular smooth muscle cells (VSMCs) following arterial injury. Here we performed percutaneous adenovirus-mediated gene transfer into the iliac arteries of normal rabbits using a channel balloon catheter to assess the effects of gax overexpression on neointima formation, lumen diameter, reendothelialization and functional vasomotion. METHODS: A channel balloon catheter was used to perform both the arterial injury and local gene delivery. In each animal both iliac arteries were randomly assigned to receive either an adenovirus expressing the gax gene (Ad-Gax) or the beta-galactosidase gene (Ad-beta gal). In a second group of animals arteries were randomly assigned to receive either Ad-beta gal or saline. RESULTS: At one month, angiography revealed 36% less luminal narrowing in the Ad-Gax-treated arteries relative to the Ad-beta gal-treated control arteries. Histological analysis revealed that the intimal/medial ratio (I/M) was reduced by 56% in the Ad-Gax group. Endothelium-dependent vasomotion was not affected by the gax gene transfer. In the second group, no statistically significant differences were found between the saline and the Ad-beta gal-treated vessels for any of these parameters. CONCLUSIONS: Percutaneous adenovirus delivery of the gax gene to rabbit iliac arteries following endothelial denudation and vessel wall injury reduces neointimal hyperplasia and luminal stenosis, but does not affect endothelium-dependent vasomotion. This study demonstrates that a VSMC transcription factor can potentially be utilized for the development of a molecular therapy for vascular disorders.


Assuntos
Adenoviridae , Angioplastia com Balão , Arteriopatias Oclusivas/prevenção & controle , Técnicas de Transferência de Genes , Genes Homeobox , Artéria Ilíaca , Animais , Arteriopatias Oclusivas/terapia , Divisão Celular , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos , Humanos , Músculo Liso Vascular/citologia , Coelhos , Recidiva , Túnica Íntima/citologia
15.
Circulation ; 94(9): 2177-84, 1996 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8901669

RESUMO

BACKGROUND: Overexpression of human lecithin-cholesterol acyltransferase (LCAT) in transgenic mice results in an increase of the antiatherogenic HDLs. METHODS AND RESULTS: To investigate the potential use of LCAT for gene therapy, a recombinant adenovirus was constructed in which the human LCAT cDNA was expressed under the control of the human cytomegalovirus immediate/early promoter followed by a chimeric intron (AdCMV human LCAT). Human apolipoprotein (apo) A-I transgenic mice infected with AdCMV human LCAT by intravenous injection accumulated reactive LCAT in the plasma. LCAT activity was increased 201-fold in the plasma of mice infected with 1 x 10(6) pfu AdCMV human LCAT, from 45 +/- 2 to 9068 +/- 812 nmol.mL-1.h-1, in comparison with basal LCAT activity measured in control mice, 5 days after injection. Plasma HDL cholesterol levels rose from 117 +/- 12 to 797 +/- 48 mg/dL, and plasma human apo A-I concentrations increased from 247 +/- 14 to 616 +/- 17 mg/dL, in AdCMV human LCAT infected mice compared with control mice. HDL particles were larger and had a different electrophoretic mobility. Studies of cholesterol efflux by incubation of serum with cholesterol-loaded Fu5AH cells showed that serum from AdCMV human LCAT-infected mice promoted a significantly higher efflux than did that of the controls. CONCLUSIONS: These data establish the potential of this approach for treatment of subjects with LCAT gene defects as well as patients with low plasma levels of apo A-I and HDL cholesterol.


Assuntos
Adenoviridae , Técnicas de Transferência de Genes , Terapia Genética , Fosfatidilcolina-Esterol O-Aciltransferase/genética , Doença de Tangier/genética , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/genética , Carcinoma Hepatocelular , Ésteres do Colesterol/sangue , HDL-Colesterol/sangue , Eletroforese , Ácidos Graxos/análise , Expressão Gênica/fisiologia , Humanos , Lipoproteínas/sangue , Masculino , Camundongos , Camundongos Transgênicos , Peso Molecular , Mutação/genética , Fosfolipídeos/sangue , Fosfolipídeos/química , Triglicerídeos/sangue , Células Tumorais Cultivadas/metabolismo
16.
Circulation ; 94(4): 713-7, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8772693

RESUMO

BACKGROUND: Prospective epidemiological studies support the hypothesis that high levels of high-density lipoprotein (HDL) cholesterol and apolipoprotein (apo) A-I limit atherosclerosis development. However, more data from studies with animal models of atherosclerosis that resemble the human disease are required to demonstrate the effect of apo A-I in the inhibition of atherogenesis. The rabbit is a good animal model for human atherosclerosis. METHODS AND RESULTS: Human apo A-I-transgenic rabbits have been produced, and we have evaluated the effect of apo A-I on the development of atherosclerosis in transgenic rabbits fed a cholesterol-rich diet for 14 weeks. Plasma cholesterol levels of atherogenic apo B-containing lipoproteins were similar for transgenic and control rabbits (> 1000 mg/dL), while plasma levels of HDL cholesterol in the transgenic group were always about twice that of the control group (68 +/- 11 versus 37 +/- 3 mg/dL at 14 weeks; P < .001). At the end of the experiment, the amount of aortic surface area covered by lesions as well as the amount of lipid accumulation in the aorta were significantly less in transgenic rabbits compared with the control group (15 +/- 12% versus 30 +/- 8%, P < .0027 for the surface area of the thoracic aorta; 116 +/- 31 versus 247 +/- 39 mumol/g aorta, P < .0068 for cholesterol content in total aorta). CONCLUSIONS: Overexpression of human apo A-I in rabbits inhibits the development of atherosclerosis in this animal model that resembles, in many respects, human atherosclerosis.


Assuntos
Apolipoproteína A-I/biossíntese , Apolipoproteínas/sangue , Arteriosclerose/prevenção & controle , Arteriosclerose/fisiopatologia , Colesterol na Dieta , Análise de Variância , Animais , Animais Geneticamente Modificados , Aorta/química , Aorta/metabolismo , Apolipoproteína A-I/sangue , Apolipoproteína A-I/genética , Arteriosclerose/sangue , Colesterol/sangue , Colesterol/metabolismo , Dieta Aterogênica , Humanos , Neoplasias Hepáticas Experimentais , Músculo Liso Vascular/química , Músculo Liso Vascular/metabolismo , Coelhos , Ratos , Valores de Referência
17.
J Virol ; 70(1): 559-65, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8523570

RESUMO

Transgene expression after the administration of recombinant adenovirus with E1 deleted is constantly transient. It is admitted that E1A-substituting activities of cellular or viral origin allow viral antigen synthesis and trigger cytotoxic lymphocyte-mediated clearance of the recipient cells. Our approach to solving this problem relies on the additional deletion of the E4 region from the vector backbone as this region upregulates viral gene expression at both transcriptional and posttranscriptional levels. As a prerequisite to the construction of E1 E4 doubly defective adenoviruses, we investigated the possibility of transcomplementing both functions within a single cell. In particular, the distal ORF6+ORF7 segment from the E4 locus of adenovirus type 5 was cloned under the control of the dexamethasone-inducible mouse mammary tumor virus long terminal repeat. Following transfection into 293 cells, clone IGRP2 was retained and characterized as it can rescue the growth defect of all E1+ E4- adenoviral deletants tested. DNA and RNA analysis experiments verified that the mouse mammary tumor virus promoter drives the expression of the ORF6+ORF7 unit and permits its bona fide alternative splicing, generating ORF6/7 mRNA in addition to the ORF6-expressing primary transcript. Importantly, IGRP2 cells sustain cell confluence for a period longer than that of 293 parental cells and allow the plaque purification of E1- or E4- defective viruses. The dual expression of E1 and E4 regulatory genes within IGRP2 cells is demonstrated by the construction, plaque purification, and helper-free propagation of recombinant lacZ-encoding doubly defective adenoviruses harboring different E4 deletions. In addition, the emergence, if any, of replicative particles during viral propagation in this novel packaging cell line will be drastically impaired as only a limited segment of E4 has been integrated.


Assuntos
Proteínas E1 de Adenovirus/genética , Proteínas E4 de Adenovirus/genética , Adenovírus Humanos/genética , Proteínas do Capsídeo , Adenovírus Humanos/crescimento & desenvolvimento , Animais , Sequência de Bases , Capsídeo/metabolismo , Linhagem Celular , Clonagem Molecular , Deleção de Genes , Genes Virais , Teste de Complementação Genética , Humanos , Cinética , Vírus do Tumor Mamário do Camundongo/genética , Dados de Sequência Molecular , Fases de Leitura Aberta , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
18.
Biochem Biophys Res Commun ; 206(2): 652-8, 1995 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-7826384

RESUMO

The mechanism of inhibition of protein phosphatase-1 catalytic subunit (PP-1c) by recombinant DARPP-32 and synthetic peptides was studied. DARPP-32 was expressed in Escherichia coli as a non-fusion protein using a pEt-3a plasmid, purified to homogeneity and shown to have physicochemical properties similar to those of the protein purified from bovine brain. Recombinant DARPP-32 phosphorylated on threonine-34 by cAMP-dependent protein kinase inhibited PP-1c with an IC50 approximately 0.5 nM, comparable to that obtained with bovine DARPP-32. Non-phosphorylated DARPP-32, and mutated forms in which threonine-34 was replaced by an alanine or a glutamic acid, inhibited PP-1c with an IC50 approximately 1 microM. Surface plasmon resonance analysis showed binding of PP-1c to nonphospho- and phospho-DARPP-32-(8-38) synthetic peptides with apparent Kd values of 1.2 and 0.3 microM, respectively, supporting the existence of an interaction between non-phosphorylated DARPP-32 and PP-1c that is increased by phosphorylation of DARPP-32 at threonine-34. These results suggest a model in which DARPP-32 interacts with PP-1c by at least two low affinity sites, the combination of which is responsible for the high affinity (nM) inhibition.


Assuntos
Proteínas do Tecido Nervoso/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosfopeptídeos/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina , Escherichia coli , Cinética , Dados de Sequência Molecular , Músculo Esquelético/enzimologia , Mutagênese Sítio-Dirigida , Miocárdio/enzimologia , Oligodesoxirribonucleotídeos , Fragmentos de Peptídeos/síntese química , Fosfopeptídeos/síntese química , Fosfoproteínas/farmacologia , Fosforilação , Ligação Proteica , Proteína Fosfatase 1 , Coelhos , Proteínas Recombinantes/farmacologia , Spodoptera
19.
20.
Arterioscler Thromb ; 14(10): 1657-64, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7918317

RESUMO

We studied the pharmacological potential of retinoids to modulate apolipoprotein (apo) A-I and apoA-II gene expression and production in vitro in the human cell line HepG2 as well as in primary cultures of adult rat hepatocytes and in vivo in the rat. In HepG2 cells, addition of all-trans retinoic acid (RA) doubled apoA-I mRNA within 24 hours and protein secreted in the culture medium after 48 hours. The induction of apoA-I mRNA by RA was completely blocked by actinomycin D, suggesting that RA acts at the transcriptional level in HepG2 cells. In primary cultures of rat hepatocytes, addition of RA increased apoA-I mRNA in a dose- and time-dependent manner as well as the secretion of apoA-I protein. Similar changes in apoA-I mRNA were observed with 9-cis RA. However, in vivo, hepatic apoA-I mRNA levels decreased after a single administration of RA at 10 mg/kg and remained low after prolonged treatment or at a higher dose, and serum apoA-I concentrations did not change. Furthermore, RA treatment did not substantially affect apoA-II mRNA levels or protein secretion either in vitro or in vivo. As a control, RA receptor-beta mRNA levels increased after RA both in vitro and in vivo. In conclusion, RA treatment selectively induces apoA-I and not apoA-II expression in vitro but not in vivo. These results therefore show additional regulatory effects of RA on apoA-I gene expression in vivo and raise questions about the usefulness of RA in the treatment of atherosclerosis.


Assuntos
Apolipoproteína A-II/genética , Apolipoproteína A-I/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/fisiologia , Tretinoína/farmacologia , Animais , Apolipoproteína A-I/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Masculino , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do Ácido Retinoico/genética , Estereoisomerismo , Tretinoína/química , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA