Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Endocrine ; 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38801599

RESUMO

INTRODUCTION: Micro- and macrovascular complications are common among persons with type 2 diabetes. Recently there has been growing interest to investigate the potential of circulating small non-coding RNAs (sncRNAs) as contributors to the development of diabetic complications. In this study we investigate to what extent circulating sncRNAs levels associate with prevalent diabetic kidney disease (DKD) in persons with type 2 diabetes. METHODS: Plasma sncRNAs levels were determined using small RNA-seq, allowing detection of miRNAs, snoRNAs, piRNAs, tRNA fragments, and various other sncRNA classes. We tested for differentially expressed sncRNAs in persons with type 2 diabetes, with DKD (n = 69) or without DKD (n = 405). In secondary analyses, we also tested the association with eGFR, albuminuria (UACR), and the plasma proteome. RESULTS: In total seven sncRNAs were negatively associated with prevalent DKD (all PFDR ≤ 0.05). Including one microRNA (miR-143-5p), five snoRNAs (U8, SNORD118, SNORD24, SNORD107, SNORD87) and a piRNA (piR-019825 | DQ597218). Proteomic analyses showed that the seven sncRNAs, and especially the piRNA piR-019825, were associated with plasma levels of 24 proteins of which several have known associations with kidney function including TNF sR-I (TNFRFS1A), DAN (NBL1) and cystatin C (CST3). CONCLUSION: We have identified novel small non-coding RNAs, primarily from classes other than microRNAs, that are associated with diabetic kidney disease. Our results show that the involvement of small non-coding RNAs in DKD goes beyond the already known microRNAs and also involves other classes of sncRNA, in particular snoRNAs and the piRNA piR-019825, that have never been studied before in relation to kidney function.

2.
Resuscitation ; 194: 110086, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38097106

RESUMO

BACKGROUND: During stabilisation of preterm infants at birth, a face mask is used to provide respiratory support. However, application of these masks may activate cutaneous stretch receptors of the trigeminal nerve, causing apnoea and bradycardia. This study investigated the amount of force exerted on the face mask during non-invasive ventilation of preterm infants at birth and whether the amount of exerted force is associated with apnoea and bradycardia. METHODS: A prospective observational study was performed in preterm infants born <32 weeks of gestation who were stabilised at birth. During the first 10 minutes of respiratory support, we measured breathing and heart rate as well as the amount of force exerted on a face mask using a custom-made pressure sensor placed on top of the face mask. RESULTS: Thirty infants were included (median (IQR) gestational age(GA) 28+3 (27+0-30+0) weeks, birthweight 1104 (878-1275) grams). The median exerted force measured was 297 (198-377) grams, ranging from 0 to 1455 grams. Significantly more force was exerted on the face mask during positive pressure ventilation when compared to CPAP (410 (256-556) vs 286 (190-373) grams, p = 0.009). In a binary logistic regression model, higher forces were associated with an increased risk of apnoea (OR = 1.607 (1.556-1.661), p < 0.001) and bradycardia (OR = 1.140 (1.102-1.180), p < 0.001) during the first 10 minutes of respiratory support at birth. CONCLUSION: During mask ventilation, the median exerted force on a face mask was 297 grams with a maximum of 1455 grams. Higher exerted forces were associated apnoea and bradycardia during the first 10 minutes of respiratory support at birth.


Assuntos
Apneia , Recém-Nascido Prematuro , Recém-Nascido , Humanos , Apneia/etiologia , Máscaras/efeitos adversos , Bradicardia/etiologia , Respiração com Pressão Positiva
3.
Stem Cell Res ; 67: 103031, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36702081

RESUMO

We previously generated a doxycycline-inducible H2B-mTurq2 reporter in hiPSCs to track cells and study cell division and apoptosis. To improve visualization of cycling cells, we introduced a ubiquitously transcribed mScarletI-Geminin (GMMN) (1-110) into the previously untargeted second AAVS1 allele. Fusion to the N-terminal part of GMNN provided tightly controlled mScarletI expression during the cell cycle. mScarletI fluorescence increased gradually from the S-phase through the M-phase of the cell cycle and was lost at the metaphase-anaphase transition. The resulting hiPSC reporter line generated, which we named ProLiving, is a valuable tool to study cell division and cell cycle characteristics in living hiPSC-derived cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Geminina/genética , Geminina/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Ciclo Celular , Divisão Celular , Proteínas de Ciclo Celular/genética
4.
Stem Cell Res ; 57: 102592, 2021 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-34775202

RESUMO

An induced pluripotent stem cell (iPSC) line, in which a H2B-fluorescent protein fusion is temporally expressed, is a valuable tool to track cells and study cell divisions and apoptosis. To this end we introduced a 3rd generation "all-in-one" doxycycline-inducible H2B-mTurquoise2 vector into the AAVS1 locus of PAX3-Venus iPSCs via CRISPR/Cas9. H2B-mTurquoise2 expression is absent but readily induced by doxycycline allowing quantification of cell divisions and imaging of living cells. Besides being a universal reporter in iPSC-based differentiation and toxicity assays, the generated pluripotent and genomically normal LUMCi041-A-2 line is particularly suited to study PAX3-positive stages of development.

5.
Gene Ther ; 19(9): 899-906, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22011643

RESUMO

Human adenoviruses have a great potential as anticancer agents. One strategy to improve their tumor-cell specificity and anti-tumor efficacy is to include tumor-specific targeting ligands in the viral capsid. This can be achieved by fusion of polypeptide-targeting ligands with the minor capsid protein IX. Previous research suggested that protein IX-mediated targeting is limited by inefficient release of protein IX-fused ligands from their cognate receptors in the endosome. This thwarts endosomal escape of the virus particles. Here we describe that the targeted transduction of tumor cells is augmented by a cathepsin-cleavage site between the protein IX anchor and the HER2/neu-binding ZH Affibody molecule as ligand. The cathepsin-cleavage site did not interfere with virus production and incorporation of the Affibody molecules in the virus capsid. Virus particles harboring the cleavable protein IX-ligand fusion in their capsid transduced the HER2/neu-positive SKOV-3 ovarian carcinoma cells with increased efficiency in monolayer cultures, three-dimensional spheroid cultures and in SKOV-3 tumors grown on the chorioallantoic membrane of embryonated chicken eggs. These data show that inclusion of a cathepsin-cleavage sequence between protein IX and a high-affinity targeting ligand enhances targeted transduction. This modification further augments the applicability of protein IX as an anchor for coupling tumor-targeting ligands.


Assuntos
Adenovírus Humanos/genética , Proteínas do Capsídeo/metabolismo , Catepsinas/química , Vetores Genéticos , Ligantes , Transdução Genética , Linhagem Celular Tumoral , Marcação de Genes , Humanos , Neoplasias/terapia , Proteínas Recombinantes de Fusão/química
6.
Gene Ther ; 15(24): 1567-78, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18650851

RESUMO

Human Orthoreovirus Type 3 Dearing is not pathogenic to humans and has been evaluated clinically as an oncolytic agent. Its transduction efficiency and the tumor cell selectivity may be enhanced by incorporating ligands for alternative receptors. However, the genetic modification of reoviruses has been difficult, and genetic targeting of reoviruses has not been reported so far. Here we describe a technique for generating genetically targeted reoviruses. The propagation of wild-type reoviruses on cells expressing a modified sigma 1-encoding segment embedded in a conventional RNA polymerase II transcript leads to substitution of the wild-type genome segment by the modified version. This technique was used for generating reoviruses that are genetically targeted to an artificial receptor expressed on U118MG cells. These cells lack the junction adhesion molecule-1 and therefore resist infection by wild-type reoviruses. The targeted reoviruses were engineered to carry the ligand for this receptor at the C terminus of the sigma 1 spike protein. This demonstrates that the C terminus of the sigma 1 protein is a suitable locale for the insertion of oligopeptide ligands and that targeting of reoviruses is feasible. The genetically targeted viruses can be propagated using the modified U118MG cells as helper cells. This technique may be applicable for the improvement of human reoviruses as oncolytic agents.


Assuntos
Moléculas de Adesão Celular/genética , Terapia Genética/métodos , Orthoreovirus Mamífero 3/genética , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Sequência de Aminoácidos , Efeito Espectador , Linhagem Celular Tumoral , Sobrevivência Celular , Clonagem Molecular , Marcação de Genes , Engenharia Genética , Humanos , Fragmentos de Imunoglobulinas , Moléculas de Adesão Juncional , Ligantes , Dados de Sequência Molecular , Neoplasias/patologia , Neoplasias/virologia , Alinhamento de Sequência , Transdução Genética/métodos
7.
Gene Ther ; 15(13): 978-89, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18323790

RESUMO

Adenovirus vectors have great potential in cancer gene therapy. Targeting of cancer-testis (CT) antigens, which are specifically presented at the surface of tumor cells by human leukocyte antigen (HLA) class I molecules, is an attractive option. In this study, a single-chain T-cell receptor (scTCR) directed against the CT antigen melanoma-associated antigen (MAGE)-A1 in complex with the HLA class I molecule of haplotype HLA-A1 is fused with the C terminus of the adenovirus minor capsid protein IX. Propagation of a protein-IX (pIX)-gene-deleted human adenovirus 5 (HAdV-5) vector on cells that constitutively express the pIXscTCR fusion protein yielded viral particles with the pIXscTCR fusion protein incorporated in their capsid. Generated particles specifically transduced melanoma cell lines expressing the HLA-A1/MAGE-A1 target complex with at least 10-fold higher efficiency than control viruses. Whereas loading of HLA-A1-positive cells with MAGE-A1 peptides leads to enhanced transduction of the cells, the efficiency of virus transduction is strongly reduced if the HLA-A1 molecules are not accessible at the target cell. Taken together, these data provide proof of principle that pIXscTCR fusions can be used to target HAdV-5 vectors to tumor cells expressing intracellular CT antigens.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Melanoma/terapia , Transdução Genética/métodos , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Citometria de Fluxo , Marcação de Genes , Engenharia Genética , Vetores Genéticos/genética , Antígeno HLA-A1/imunologia , Humanos , Masculino , Melanoma/imunologia , Melanoma/metabolismo , Antígenos Específicos de Melanoma , Proteínas de Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética
8.
Cancer Gene Ther ; 13(6): 584-91, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16456552

RESUMO

The cellular immune response against transgene-encoded neoantigens is a potential hurdle in gene therapy applications where long-term expression of transgenes is desired. Here a new optimized derivative of the herpes simplex virus 1-thymidine-kinase (HSV1-TK) gene is described. The HSV-TK gene is frequently used in experimental studies on gene-directed enzyme prodrug therapy. In the optimized gene, the HSV-TK coding region is fused with the codons for the Gly-Ala repeat of the Epstein-Barr virus nuclear-antigen 1 to prevent proteasomal degradation of the HSV-TK. To measure the protective effect in vitro, a model cytotoxic T lymphocyte epitope derived from the ovalbumin was inserted in the TK. Cells expressing the GAr-modified TK do not present TK-derived peptides in the major histocompatibility complex. Furthermore, conservative nucleotide substitutions were introduced, which prevent splicing, as well as mutations that render the TK-expressing cells more sensitive to ganciclovir (GCV). The GAr HSV-TK fusion protein is fully functional in vitro. This HSV-TK gene may be especially useful in those gene therapy applications where an immune response against the transgene-encoded product would frustrate the treatment.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Simplexvirus/enzimologia , Timidina Quinase/genética , Timidina Quinase/imunologia , Apresentação de Antígeno , Sequência de Bases , Clonagem Molecular , Antígenos Nucleares do Vírus Epstein-Barr/genética , Humanos , Dados de Sequência Molecular , Receptores Imunológicos/metabolismo , Proteínas Recombinantes de Fusão/genética , Linfócitos T Citotóxicos/metabolismo
9.
Org Lett ; 3(11): 1645-8, 2001 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-11405676

RESUMO

The synthesis and photophysical characterization of a series of free base porphyrin-containing polyamide dendrimers terminated with anthraquinone groups (FbP-Ga-AQ(n)(), a = 1-3, n = 12, 36, 108) are described. Substantial quenching (58-75%) of the porphyrin fluorescence of FbP-Ga-AQ(n)() is observed when compared to the analogous ethyl-terminated dendrimers (FbP-Ga-Et(n)()) in steady-state fluorescence experiments and is attributed to intramolecular electron transfer. Time-resolved fluorescence experiments were fit to 2-3 exponentials, indicating multiple orientations for electron transfer, consistent with the flexible nature of these dendrimers.

10.
Nucleic Acids Res ; 28(23): E104, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11095700

RESUMO

Bacteriophage Mu has one of the best studied, most efficient and largest transposition machineries of the prokaryotic world. To harness this attractive integration machinery for use in mammalian cells, we cloned the coding sequences of the phage factors MuA and MuB in a eukaryotic expression cassette and fused them to a FLAG epitope and a SV40-derived nuclear localization signal. We demonstrate that these N-terminal extensions were sufficient to target the Mu proteins to the nucleus, while their function in Escherichia coli was not impeded. In vivo transposition in mammalian cells was analysed by co-transfection of the MuA and MuB expression vectors with a donor construct, which contained a miniMu transposon carrying a Hygromycin-resistance marker (Hyg(R)). In all co-transfections, a significant but moderate (up to 2.7-fold) increase in Hyg(R) colonies was obtained if compared with control experiments in which the MuA vector was omitted. To study whether the increased efficiency was the result of bona fide Mu transposition, integrated vector copies were cloned from 43 monoclonal and one polyclonal cell lines. However, in none of these clones, the junction between the vector and the chromosomal DNA was localized precisely at the border of the Att sites. From our data we conclude that expression of MuA and MuB increases the integration of miniMu vectors in mammalian cells, but that this increase is not the result of bona fide Mu-induced transposition.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Bacteriófago mu/genética , Linhagem Celular Transformada , Elementos de DNA Transponíveis/genética , DNA Recombinante , Proteínas de Ligação a DNA/genética , Escherichia coli/genética , Escherichia coli/virologia , Expressão Gênica , Teste de Complementação Genética , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Oligopeptídeos , Peptídeos/genética , Plasmídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Transposases/genética , Células Tumorais Cultivadas , Proteínas Virais/genética
11.
Gene Ther ; 7(16): 1410-6, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10981668

RESUMO

The host-immune response against adenoviruses forms a major obstacle for their use as gene therapy vectors for treatment of genetic defects. None the less, they are the preferred vectors for in vivo gene transfer in experimental gene therapy protocols for cancer. In this article we demonstrate the antitumor efficacy of adenovirus-mediated transfer of human interleukin-2 cDNA in the rat-CC531 model for hepatic metastases of colorectal cancer: intratumoral administration of 10 plaque-forming units of the hlL-2-expressing adenoviral vector, AdCAIL-2, resulted in a cessation of tumor growth in 80% of the injected tumors. In control groups receiving AdCnull, a vector with the same viral backbone, but lacking transgene expression, none of the tumors responded. However, intratumoral treatment with this vector significantly enhanced tumor regression induced by systemic IL-2 protein treatment, which was used as a positive control. In addition we show, by performing delayed-type of hypersensitivity assays, that AdCnull when injected intratumorally enhances recognition of tumor antigens by T lymphocytes to the same extent as intratumoral treatment with the IL-2-expressing vector. The replication-deficient adenoviruses appear to have a therapeutic advantage in cytokine-mediated immunotherapy: even adenovirus vectors that do not express a transgene, show adjuvant activity and stimulate an antitumor immune response.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/imunologia , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Hepáticas Experimentais/terapia , Linfócitos T/imunologia , Animais , Neoplasias Colorretais/imunologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/análise , Antígenos de Histocompatibilidade Classe II/análise , Humanos , Molécula 1 de Adesão Intercelular/análise , Interleucina-2/genética , Neoplasias Hepáticas Experimentais/imunologia , Ativação Linfocitária , Ratos , Ratos Endogâmicos , Células Tumorais Cultivadas
12.
Gene Ther ; 6(5): 873-81, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10505113

RESUMO

Recombinant adenovirus vectors are popular tools for gene transfer and gene therapy. However biosafety constraints require that all handling of the vectors and vector-containing samples is restricted to dedicated containment laboratories, unless they had undergone a validated virus-inactivation procedure, which decontaminates the samples from any active virus. In this study we evaluated the feasibility of photodynamic treatment (PDT) with visible light to inactivate recombinant adenovirus vectors in biological samples, with minimum associated effects on other biological activities. Several photosensitizers were tested for their capacity to inactivate a model human adenovirus vector, AdCMVLuc, upon illumination. Four photosensitizers (methylene blue (MB), rose bengal (RB), uroporphyrin (UP) and aluminum phthalocynine tetrasulphonate (AIPcS4)) could inactivate the adenovirus, as measured by expression of the luciferase reporter gene and by plaque assay. Of these, MB demonstrated to be the most effective sensitizer in phosphate-buffered saline (PBS), giving > 7 log10 inactivation of the adenovirus. DNA isolated from MB- and light-treated virions was inefficient as a template for transcription. Furthermore, Southern blot analysis revealed fragmentation of the viral DNA. Based on its preference for DNA, MB is suited for adenovirus inactivation in blood plasma. Spiking experiments in which AdCMVLuc was added to plasma samples demonstrated a reduction (> 4 log10-fold) of reporter gene expression to almost background levels. In contrast to MB, photodynamic treatment with RB, UP or AIPcS4 did not lead to DNA damage. Although alterations of the viral capsid could not be detected, the binding pattern of the particles to target cells was significantly changed. Taken together, our data demonstrate that PDT is an efficient, convenient and useful method for the inactivation of adenovirus vectors in biological samples.


Assuntos
Adenoviridae/genética , DNA Viral/efeitos da radiação , Terapia Genética/métodos , Vetores Genéticos , Luz , Ativação Viral/efeitos da radiação , Adenoviridae/ultraestrutura , Fragmentação do DNA , Estudos de Viabilidade , Humanos , Azul de Metileno , Microscopia Eletrônica , Fármacos Fotossensibilizantes
13.
Med Ref Serv Q ; 18(3): 65-78, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-11299599
14.
Hum Gene Ther ; 9(13): 1909-17, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9741429

RESUMO

The presence of replication-competent adenoviruses (RCAs) in batches of replication-defective adenovirus (Ad) vectors is a major problem for the application of these vectors in gene therapy. RCAs are generated by recombination between sequences in the Ad vector and homologous Ad sequences in the helper cells, resulting in the acquisition by the vector of early region 1. To prevent the formation of RCAs, we have developed helper cell lines, which we named PER, and matched Ad vectors that do not have sequence overlap. PER cells contain the Ad serotype 5 (Ad5) E1A- and E1B-encoding sequences (Ad5 nucleotides 459-3510) under the control of the human phosphoglycerate kinase (PGK) promoter. We demonstrate that PER cells synthesize high levels of the Ad5 E1A and E1B proteins. The yields from PER cells of E1-deleted Ads are similar to those obtained from earlier helper cells, such as 911 and 293 cells. Propagation of matched Ad vectors, which lack Ad5 nucleotides 459-3510, in one of the PER clones, PER.C6, does not result in the generation of RCAs, in contrast to propagation in 293 cells. We conclude that the combination of PER.C6 cells and nonoverlapping E1-deleted adenoviral vectors eliminates the problem of RCA generation by homologous recombination, and allows cost-effective production of safe, clinical-grade batches of recombinant Ad vectors.


Assuntos
Adenovírus Humanos/genética , Proteínas do Capsídeo , Linhagem Celular Transformada/virologia , Vetores Genéticos/genética , Vírus Auxiliares/genética , Replicação Viral/genética , Proteínas E1A de Adenovirus/análise , Proteínas E1A de Adenovirus/genética , Proteínas E1B de Adenovirus/análise , Proteínas E1B de Adenovirus/genética , Adenovírus Humanos/fisiologia , Capsídeo/biossíntese , Linhagem Celular Transformada/citologia , Temperatura Alta , Humanos , Retina/citologia , Vírion , Cultura de Vírus
15.
Gene Ther ; 5(4): 451-8, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9614568

RESUMO

The use of so-called 'suicide' genes to activate prodrugs has been effective in animal models for several solid tumor types and is now in phase I and II clinical trials. We have exploited adenovirus vectors (Ad) for transfer and expression of the herpes simplex virus thymidine kinase (HSVtk) gene to render rat colorectal liver metastases sensitive to the anti-herpetic agent ganciclovir (GCV). The efficacy and toxicity of this enzyme-prodrug combination were tested after in situ transduction of rat colorectal tumor cells and after intraportal administration of the vector Ad.CMV.TK. Our results demonstrate the validity of the approach but reveal that hepatic expression of HSVtk, both in tumor bearing and in tumor-free rats, provokes severe liver dysfunction and mortality upon GCV administration. These data show, that in contrast to the common assumption, normally non-mitotic tissues too, can be affected by adenovirus-mediated HSVtk transfer and subsequent GCV treatment. Given the hepatotropic nature of systemically administered adenovirus type 2- and 5-derived vectors, it will be essential to monitor liver functions of patients included in all gene therapy trials involving adenoviral vectors with the HSVtk gene.


Assuntos
Técnicas de Transferência de Genes/efeitos adversos , Terapia Genética/efeitos adversos , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Fígado/fisiopatologia , Adenoviridae , Animais , Antimetabólitos/efeitos adversos , Antimetabólitos/uso terapêutico , Neoplasias Colorretais/fisiopatologia , Ganciclovir/efeitos adversos , Ganciclovir/uso terapêutico , Genes Virais , Terapia Genética/métodos , Vetores Genéticos/efeitos adversos , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/fisiopatologia , Ratos , Ratos Endogâmicos , Simplexvirus/enzimologia , Timidina Quinase/genética
16.
Gene Ther ; 4(1): 55-62, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9068796

RESUMO

Targeted gene delivery is essential for gene therapy involving in vivo gene transfer. In the present study we analyzed the efficiency and tissue-specificity of gene transfer into the liver with recombinant adenoviruses. Adenovirus vectors carrying the E. coli lacZ gene (Ad.RSV.beta-gal) and the firefly luciferase gene (AdCMV-luc) as reporters were administered to the liver of adult Wistar rats, either via infusion into the portal vein (intraportal infusion; IPI) or via perfusion of the vascularity isolated liver (isolated liver perfusion; ILP). Ex vivo liver perfusion experiments with Ad.RSV. beta-gal were used to optimize the conditions for hepatic gene transfer. Ex vivo perfusion of rat livers with 2 x 10(9) plaque forming units (p.f.u) Ad RSV.beta-gal was sufficient to infect about 20% of the liver parenchymal cells. Perfusion with chelating agents (1 mM EGTA, or 2 mM EDTA) prior to the administration of the vector increased the efficiency to at least 40%. Similar efficiencies were obtained in experiments with liver lobes of Rhesus monkeys. In vivo administration of AdCMV-luc via ILP resulted in a significantly more efficient (P = 0.028) and also more reproducible gene transfer when compared to IPI. Although detectable in both groups, extrahepatic luciferase expression was considerably reduced in the ILP group. Our data demonstrate that IPL can be used for efficient and reproducible liver-specific gene delivery. Therefore, we think that the perfusion of vascularly isolated organs is useful as a modality for the tissue-specific administration of recombinant adenovirus vectors.


Assuntos
Adenoviridae , Marcação de Genes , Técnicas de Transferência de Genes , Fígado , Animais , Expressão Gênica , Masculino , Perfusão , Sistema Porta , Ratos , Ratos Wistar , Transgenes , beta-Galactosidase/genética
17.
Mol Cell Biol ; 16(8): 4264-72, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8754827

RESUMO

Expression of the human blood-clotting factor VIII (FVIII) cDNA is hampered by the presence of sequences located in the coding region that repress transcription. We have previously identified a 305-bp fragment within the FVIII cDNA that is involved in the repression (R.C. Hoeben, F.J. Fallaux, S.J. Cramer, D.J.M. van den Wollenberg, H. van Ormondt, E. Briet, and A.J. van der Eb, Blood 85:2447-2454, 1995). Here, we show that this 305-bp region of FVIII cDNA contains sequences that resemble the yeast (Saccharomyces cerevisiae) autonomously replicating sequence consensus. Two of these DNA elements coincide with AT-rich sequences that are often found in matrix attachment regions or scaffold-attached regions. One of these elements, consisting of nucleotides 1569 to 1600 of the FVIII cDNA (nucleotide numbering is according to the system of Wood et al. (W.I. Wood, D.J. Capon, C.C. Simonsen, D.L. Eaton, J. Gitschier, D. Keyt, P.H. Seeburg, D.H. Smith, P. Hollingshead, K.L. Wion, et al., Nature [London] 312:330-337,1984), binds a nuclear factor in vitro but loses this capacity after four of its base pairs have been changed. A synthetic heptamer of this segment can repress the expression of a chloramphenicol acetyltransferase (CAT) reporter gene and also loses this capacity upon mutation. Furthermore, we demonstrate that repression by FVIII sequences can be relieved by sodium butyrate. We demonstrate that the synthetic heptamer (FVIII nucleotides 1569 to 1600), when placed upstream of the Moloney murine leukemia virus long terminal repeat promoter that drives the CAT reporter, can render the CAT reporter inducible by butyrate. This effect was absent when the same element was mutated. The stimulatory effect of butyrate could not be attributed to butyrate-responsive elements in the studied long terminal repeat promoters. Our data provide a functional characterization of the sequences that repress expression of the FVIII cDNA. These data also suggest a link between transcriptional repression by FVIII cDNA elements and the stimulatory effect of butyrate on FVIII cDNA expression.


Assuntos
Butiratos/farmacologia , Fator VIII/genética , Regulação da Expressão Gênica , RNA Mensageiro/genética , Proteínas de Saccharomyces cerevisiae , Proteínas de Schizosaccharomyces pombe , Fatores de Transcrição , Sequência de Bases , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica/efeitos dos fármacos
18.
Hum Gene Ther ; 7(2): 215-22, 1996 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-8788172

RESUMO

Currently, the preferred host for the production of early region-1 (E1)-deleted recombinant adenoviruses (rAdV) is cell line 293, which was generated by transformation of human embryonic kidney cells by sheared adenovirus 5 (Ad5) DNA. To develop alternative hosts for the production of rAdV, we generated adenovirus-transformed human cell lines by transformation of human embryonic retinoblasts (HER) with a plasmid containing base pairs 79-5789 of the Ad5 genome. One of the established HER cell lines, which we called 911, exhibited favorable growth characteristics and was chosen for further study. This cell line is demonstrated to have several characteristics in common with the well-known 293 cell line: The 911 cell line is highly transfectable, and exhibits similar frequencies of homologous recombination. However, it has additional characteristics that make it a useful alternative for 293. The 911 cells perform particularly well in plaque assays. Upon infection with E1-deleted adenoviruses, plaques become apparent in monolayers of 911 cells already after 3-4 days versus 4-10 days in monolayers of 293 cells, thereby reducing the time required for quantitative plaque assays. Furthermore, yields of E1-deleted adenovirus vectors up to three times as high as those achieved with 293 cells can be obtained with 911 cells. Finally, the Ad5-DNA content of the 911 cell line is completely known. These features make the 911 cell line a useful alternative for the construction, propagation, and titration of E1-deleted recombinant adenoviruses.


Assuntos
Adenovírus Humanos/genética , Linhagem Celular Transformada/virologia , Vetores Genéticos , Vírus Auxiliares/fisiologia , Proteínas E1 de Adenovirus/genética , Adenovírus Humanos/fisiologia , Divisão Celular , Linhagem Celular Transformada/citologia , Fator IX/genética , Fator VIII/genética , Genoma Viral , Vírus Auxiliares/genética , Humanos , Recombinação Genética , Retina/citologia , Retina/embriologia , Deleção de Sequência , Transfecção , Ensaio de Placa Viral , Vírion , Cultura de Vírus , Replicação Viral
19.
Blood ; 85(9): 2447-54, 1995 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-7727775

RESUMO

Hemophilia A is caused by a deficiency of factor-VIII procoagulant (fVIII) activity. The current treatment by frequent infusions of plasma-derived fVIII concentrates is very effective but has the risk of transmittance of blood-borne viruses (human immunodeficiency virus [HIV], hepatitis viruses). Use of recombinant DNA-derived fVIII as well as gene therapy could make hemophilia treatment independent of blood-derived products. So far, the problematic production of the fVIII protein and the low titers of the fVIII retrovirus stocks have prevented preclinical trials of gene therapy for hemophilia A in large-animal models. We have initiated a study of the mechanisms that oppose efficient fVIII synthesis. We have established that fVIII cDNA contains sequences that dominantly inhibit its own expression from retroviral as well as from plasmid vectors. The inhibition is not caused by instability of the fVIII mRNA (t1/2, > or = 6 hours) but rather to repression at the level of transcription. A 305-bp fragment is identified that is involved in but not sufficient for repression. This fragment does not overlap the region recently identified by Lynch et al (Hum Gene Ther 4:259, 1993) as a dominant inhibitor of RNA accumulation. The repression is mediated by a cellular factor (or factors) and is independent of the orientation of the element in the transcription unit, giving the repressor element the hallmarks of a transcriptional silencer.


Assuntos
Fator VIII/biossíntese , Regulação da Expressão Gênica , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica , Sequência de Aminoácidos , Linhagem Celular , DNA Complementar/genética , DNA Recombinante , Fator VIII/genética , Expressão Gênica , Meia-Vida , Células HeLa , Humanos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/biossíntese
20.
Hum Gene Ther ; 4(2): 179-86, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8494927

RESUMO

Hemophilia A is caused by the lack of functional blood-clotting factor VIII. We have used retrovirus-mediated gene transfer to generate various cell lines, rodent as well as human, that secrete the human factor VIII protein. To study whether transplantation of genetically modified fibroblasts is a feasible approach for gene therapy of hemophilia A, we implanted the factor VIII-secreting cells into immune-deficient mice. Implantation of factor VIII-secreting primary human skin fibroblasts resulted in long-term persistence of the transplanted cells; cells recovered from the implants up to 2 months post-implantation still had the capacity to secrete factor VIII when regrown in tissue culture. However, we were unable to detect any human factor VIII in plasma samples of the recipient mice. The absence of human factor VIII in the recipients' plasma is shown to be due neither to (epigenetic) inactivation of the retroviral vector in vivo, nor to inability of the stationary cells to secrete factor VIII protein. However, we did note a rapid clearing of the human factor VIII: CAg from plasma upon intravenous injection of plasma-derived human factor VIII in mice (t1/2 < 60 min vs. 10 hr in humans). This phenomenon can fully explain the apparent absence of human factor VIII in the recipients' plasma.


Assuntos
Fator VIII/metabolismo , Fibroblastos/transplante , Terapia Genética , Hemofilia A/terapia , Hospedeiro Imunocomprometido , Animais , Células Cultivadas , Fator VIII/genética , Fibroblastos/metabolismo , Meia-Vida , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...