Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 107(10): 4716-21, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133734

RESUMEN

To analyze the in vivo structure of antigen-specific immunological synapses during an effective immune response, we established brain tumors expressing the surrogate tumor antigen ovalbumin and labeled antigen-specific anti-glioma T cells using specific tetramers. Using these techniques, we determined that a significant number of antigen-specific T cells were localized to the brain tumor and surrounding brain tissue and a large percentage could be induced to express IFNgamma when exposed to the specific ovalbumin-derived peptide epitope SIINFEKL. Detailed morphological analysis of T cells immunoreactive for tetramers in direct physical contact with tumor cells expressing ovalbumin indicated that the interface between T cells and target tumor cells displayed various morphologies, including Kupfer-type immunological synapses. Quantitative analysis of adjacent confocal optical sections was performed to determine if the higher frequency of antigen-specific antiglioma T cells present in animals that developed an effective antitumor immune response could be correlated with a specific immunological synaptic morphology. Detailed in vivo quantitative analysis failed to detect an increased proportion of immunological synapses displaying the characteristic Kupfer-type morphology in animals mounting a strong and effective antitumor immune response as compared with those experiencing a clinically ineffective response. We conclude that an effective cytolytic immune response is not dependent on an increased frequency of Kupfer-type immunological synapses between T cells and tumor cells.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioma/inmunología , Sinapsis Inmunológicas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antivirales/farmacología , Western Blotting , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Femenino , Citometría de Flujo , Ganciclovir/farmacología , Terapia Genética/métodos , Glioma/patología , Glioma/terapia , Herpesvirus Humano 1/enzimología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Ovalbúmina/genética , Ovalbúmina/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/metabolismo , Timidina Quinasa/genética
2.
Nat Med ; 5(11): 1256-63, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10545991

RESUMEN

The long-term consequences of adenovirus-mediated conditional cytotoxic gene therapy for gliomas remain uncharacterized. We report here detection of active brain inflammation 3 months after successful inhibition of syngeneic glioma growth. The inflammatory infiltrate consisted of activated macrophages/microglia and astrocytes, and T lymphocytes positive for leucosyalin, CD3 and CD8, and included secondary demyelination. We detected strong widespread herpes simplex virus 1 thymidine kinase immunoreactivity and vector genomes throughout large areas of the brain. Thus, patient evaluation and the design of clinical trials in ongoing and future gene therapy for brain glioblastoma must address not only tumor-killing efficiency, but also long-term active brain inflammation, loss of myelin fibers and persistent transgene expression.


Asunto(s)
Neoplasias Encefálicas/terapia , Encefalitis/etiología , Terapia Genética/efectos adversos , Glioma/terapia , Adenoviridae/genética , Animales , Astrocitos/inmunología , Secuencia de Bases , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Ensayos Clínicos como Asunto , Cartilla de ADN , Encefalitis/inmunología , Ganciclovir/efectos adversos , Ganciclovir/uso terapéutico , Vectores Genéticos , Glioma/inmunología , Glioma/patología , Herpesvirus Humano 1/enzimología , Humanos , Linfocitos/inmunología , Activación de Macrófagos , Microglía/inmunología , Vaina de Mielina/metabolismo , Ratas , Timidina Quinasa/genética , Transgenes , Células Tumorales Cultivadas
3.
J Virol ; 83(4): 2004-10, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073729

RESUMEN

Increased transgene expression per vector genome is an important goal in the optimization of viral vectors for gene therapy. Herein we demonstrate that herpes simplex virus type 1 (HSV1) thymidine kinase (TK) gene sequences (1,131 bp) fused to the 3' end of lacZ increase transgene expression from high-capacity adenoviral vectors (HCAd), but not from first-generation (Ad) vectors. The woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), in contrast, increased transgene expression levels from Ad but not HCAd vectors. The differential activity of the HSV1 TK gene and WPRE sequences was detected both in vitro and in vivo and suggests potentially different mechanisms of action or the interaction of these elements with vector genomic sequences.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Herpesvirus Humano 1/enzimología , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo , Expresión Génica , Virus de la Hepatitis B de la Marmota/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
5.
Nat Biotechnol ; 19(6): 582-5, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11385466

RESUMEN

Helper-dependent (HD), high-capacity adenoviruses are one of the most efficient and safe gene therapy vectors, capable of mediating long-term expression. Currently, the most widely used system for HD vector production avoids significant contamination with helper virus by using producer cells stably expressing a nuclear-targeted Cre recombinase and an engineered first-generation helper virus with parallel loxP sites flanking its packaging signal. The system requires a final, density-based separation of HD and residual helper viruses by ultracentrifugation to reduce contaminating helper virus to low levels. This separation step hinders large-scale production of clinical-grade HD virus. By using a very efficient recombinase, in vitro-evolved FLPe (ref. 14), to excise the helper virus packaging signal in the producer cells, we have developed a scalable HD vector production method. FLP has previously been shown to mediate maximum levels of excision close to 100% compared to 80% for Cre (ref. 15). Utilizing a common HD plasmid backbone, the FLPe-based system reproducibly yielded HD virus with the same low levels of helper virus contamination before any density-based separation by ultracentrifugation. This should allow large-scale production of HD vectors using column chromatography-based virus purification.


Asunto(s)
Adenoviridae/genética , ADN Nucleotidiltransferasas/metabolismo , Vectores Genéticos , Virus Helper/metabolismo , Animales , Biotecnología/métodos , Encéfalo/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Humanos , Luciferasas/metabolismo , Modelos Genéticos , Plásmidos/metabolismo , Ratas , Transfección , beta-Galactosidasa/metabolismo
6.
Hum Gene Ther ; 17(5): 531-44, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16716110

RESUMEN

First-generation adenoviral (Ad) and high-capacity adenoviral (HC-Ad) vectors are efficient delivery vehicles for transferring therapeutic transgenes in vivo into tissues/organs. The initial successes reported with adenoviral vectors in preclinical trials have been limited by immune-related adverse side effects. This has been, in part, attributed to the use of poorly characterized preparations of adenoviral vectors and also to the untoward immune adverse side effects elicited when high doses of these vectors were used. HC-Ads have several advantages over Ads, including the lack of viral coding sequences, which after infection and uncoating, makes them invisible to the host's immune system. Another advantage is their large cloning capacity (up to approximately 35 kb). However, accurate characterization of HC-Ad vectors, and of contaminating replication-competent adenovirus (RCA) or helper virus, is necessary before these preparations can be used safely in clinical trials. Consequently, the development of accurate, simple, and reproducible methods to standardize and validate adenoviral preparations for the presence of contaminant genomes is required. By using a molecular method that allows accurate, reproducible, and simultaneous determination of HC-Ad, contaminating helper virus, and RCA genome copy numbers based on real-time quantitative PCR, we demonstrate accurate detection of these three genomic entities, within CsCl-purified vector stocks, total DNA isolated from cells transduced in vitro, and from brain tissue infected in vivo. This approach will allow accurate assessment of the levels and biodistribution of HC-Ad and improve the safety and efficacy of clinical trials.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/normas , Genoma Viral , Reacción en Cadena de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa/normas , Adenoviridae/aislamiento & purificación , Animales , Secuencia de Bases , Bioensayo , Encéfalo/virología , Línea Celular , ADN Viral/metabolismo , Femenino , Vectores Genéticos/efectos adversos , Virus Helper/genética , Virus Helper/aislamiento & purificación , Ratones
7.
Curr Top Microbiol Immunol ; 288: 149-73, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15648178

RESUMEN

Foot-and-mouth disease virus (FMDV) is genetically and phenotypically variable. As a typical RNA virus, FMDV follows a quasispecies dynamics, with the many biological implications of such a dynamics. Mutant spectra provide a reservoir of FMDV variants, and minority subpopulations may become dominant in response to environmental demands or as a result of statistical fluctuations in population size. Accumulation of mutations in the FMDV genome occurs upon subjecting viral populations to repeated bottleneck events and upon viral replication in the presence of mutagenic base or nucleoside analogs. During serial bottleneck passages, FMDV survive during extended rounds of replication maintaining low average relative fitness, despite linear accumulation of mutations in the consensus genomic sequence. The critical event is the occurrence of a low frequency of compensatory mutations. In contrast, upon replication in the presence of mutagens, the complexity of mutant spectra increases, apparently no compensatory mutations can express their fitness-enhancing potential, and the virus can cross an error threshold for maintenance of genetic information, resulting in virus extinction. Low relative fitness and low viral load favor FMDV extinction in cell culture. The comparison of the molecular basis of resistance to extinction upon bottleneck passage and extinction by enhanced mutagenesis is providing new insights in the understanding of quasispecies dynamics. Such a comparison is contributing to the development of new antiviral strategies based on the transition of viral replication into error catastrophe.


Asunto(s)
Evolución Biológica , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/efectos de los fármacos , Mutación , Ribavirina/farmacología
8.
Adv Pharmacol ; 76: 147-73, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27288077

RESUMEN

Malignant brain tumors are one of the most lethal cancers. They originate from glial cells which infiltrate throughout the brain. Current standard of care involves surgical resection, radiotherapy, and chemotherapy; median survival is currently ~14-20 months postdiagnosis. Given that the brain immune system is deficient in priming systemic immune responses to glioma antigens, we proposed to reconstitute the brain immune system to achieve immunological priming from within the brain. Two adenoviral vectors are injected into the resection cavity or remaining tumor. One adenoviral vector expresses the HSV-1-derived thymidine kinase which converts ganciclovir into a compound only cytotoxic to dividing glioma cells. The second adenovirus expresses the cytokine fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L differentiates precursors into dendritic cells and acts as a chemokine that attracts dendritic cells to the brain. HSV-1/ganciclovir killing of tumor cells releases tumor antigens that are taken up by dendritic cells within the brain tumor microenvironment. Tumor killing also releases HMGB1, an endogenous TLR2 agonist that activates dendritic cells. HMGB1-activated dendritic cells, loaded with glioma antigens, migrate to cervical lymph nodes to stimulate a systemic CD8+ T cells cytotoxic immune response against glioma. This immune response is specific to glioma tumors, induces immunological memory, and does neither cause brain toxicity nor autoimmune responses. An IND was granted by the FDA on 4/7/2011. A Phase I, first in person trial, to test whether reengineering the brain immune system is potentially therapeutic is ongoing.


Asunto(s)
Neoplasias Encefálicas/terapia , Colina/metabolismo , Terapia Genética/métodos , Glioma/terapia , Adenoviridae/genética , Animales , Células Dendríticas/metabolismo , Vectores Genéticos , Humanos , Proteínas de la Membrana/genética , Timidina Quinasa/genética , Microambiente Tumoral
9.
Circulation ; 104(21): 2595-601, 2001 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11714656

RESUMEN

BACKGROUND: Extracellular matrix (ECM) remodeling is central to the development of restenosis after coronary angioplasty (PTCA). As a regulator of ECM deposition by vascular cells, substantial evidence implicates transforming growth factor-beta1 (TGF-beta1) in the pathogenesis of restenosis. We investigated the effects of intracoronary expression of a transgenic antagonist of TGF-beta1 on luminal loss after PTCA. METHODS AND RESULTS: Porcine coronary arteries were randomized to receive a recombinant adenovirus expressing a secreted form of TGF-beta type II receptor (Ad5-RIIs), an adenovirus expressing beta-galactosidase (Ad5-lacZ), or vehicle only by intramural injection at the site of PTCA. Computerized morphometry 28 days after angioplasty revealed a greater minimum luminal area in Ad5-RIIs-injected arteries (1.71+/-0.12 mm(2)) than in the Ad5-lacZ (1.33+/-0.13 mm(2)) or vehicle-only (1.08+/-0.17 mm(2); P=0.010 by ANOVA) groups. This was accompanied by greater areas within the internal (P=0.013) and external (P=0.031) elastic laminae in Ad5-RIIs-treated vessels. Adventitial collagen content at the site of injury was increased in the Ad5-RIIs group, in contrast to decreases in the Ad5-lacZ and vehicle-only groups (P=0.004). CONCLUSIONS: Adenovirus-mediated antagonism of TGF-beta1 at the site of PTCA reduces luminal loss after PTCA by inhibiting constrictive remodeling. Antagonism of TGF-beta1 stimulates the formation of a dense collagenous adventitia, which prevents constrictive remodeling by acting as an external scaffold. These findings demonstrate the potential of gene therapy-mediated antagonism of TGF-beta1 as prophylactic therapy for restenosis.


Asunto(s)
Adenoviridae/genética , Angioplastia Coronaria con Balón/efectos adversos , Colágeno/metabolismo , Reestenosis Coronaria/terapia , Terapia Genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Animales , Células Cultivadas , Constricción Patológica/metabolismo , Constricción Patológica/patología , Constricción Patológica/terapia , Reestenosis Coronaria/etiología , Reestenosis Coronaria/metabolismo , Reestenosis Coronaria/patología , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Medios de Cultivo Condicionados/farmacología , Vectores Genéticos , Inflamación/etiología , Músculo Liso Vascular/metabolismo , Proteínas Serina-Treonina Quinasas , ARN Mensajero/biosíntesis , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Porcinos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1 , beta-Galactosidasa/metabolismo
10.
Pharmacol Ther ; 98(1): 71-108, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12667889

RESUMEN

Glioblastoma (GB) is the most common subtype of primary brain tumor in adults. These tumors are highly invasive, very aggressive, and often infiltrate critical neurological areas within the brain. The mean survival time after diagnosis of GB has remained unchanged during the last few decades, in spite of advances in surgical techniques, radiotherapy, and also chemotherapy; patients' survival ranges from 9 to 12 months after initial diagnosis. In the same time frame, with our increasing understanding and knowledge of the physiopathology of several cancers, meaningful advances have been made in the treatment and control of several cancers, such as breast, prostate, and hematopoietic malignancies. Although a number of the genetic lesions present in GB have been elucidated and our understanding of the progressions of this cancer has increased dramatically over the last few years, it has not yet been possible to harness this information towards developing effective cures. In this review, we will focus on the classical ways in which GB is currently being treated, and will introduce a novel therapeutic modality, i.e., gene therapy, which we believe will be used in combination with classical treatment strategies to prolong the life-span of patients and to ultimately be able to control and/or cure these brain tumors. We will discuss the use of several vector systems that are needed to introduce the therapeutic genes within either the tumor mass, if these are not resectable, or the tumor bed, after successful tumor resection. We also discuss different therapeutic modalities that could be exploited using gene therapy, i.e., conditional cytotoxic approach, direct cytotoxicity, immunotherapy, inhibition of angiogenesis, and the use of pro-apoptotic genes. The advantages and disadvantages of each of the current vector systems available to transfer genes into the CNS are also discussed. With the advances in molecular techniques, both towards the elucidation of the physiopathology of GB and the development of novel, more efficient and less toxic vectors to deliver putative therapeutic genes into the CNS, it should be possible to develop new rationale and effective therapeutic approaches to treat this devastating cancer.


Asunto(s)
Neoplasias Encefálicas/terapia , Animales , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirugía , Ensayos Clínicos como Asunto , Terapia Genética/métodos , Glioma/tratamiento farmacológico , Glioma/radioterapia , Glioma/cirugía , Glioma/terapia , Humanos
11.
Brain Res Mol Brain Res ; 137(1-2): 1-10, 2005 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-15950755

RESUMEN

We have investigated the in vivo dynamics of an adenovirus-based, LacZ expressing vector, RAd36, at different doses, when injected unilaterally into the corpus striatum of normal rats. We have further investigated the characteristics of this vector in the presence of a 6-OHDA lesion of the nigrostriatal pathway. The dopamine-depleting lesion had an effect on both the number and the distribution of cells transduced by the adenoviral vector. The lesioned side of the brain contained significantly greater numbers of beta-galactosidase positive cells than the unlesioned side at 3 days, 1 week and 4 weeks post-injection and the distribution of transduced cells was altered by the presence of a dopamine lesion. We conclude that the increased levels of transgene expression seen in the lesioned hemisphere are due to a change in the diffusion characteristics of the injected vector in the lesioned hemisphere. These results indicate that, when investigating the use of virus-based vectors, ultimately for use in gene therapies in the CNS, the in vivo dynamics of the vector need to be assessed not only in the normal brain, but also in the pathological brain state such as animal models of target diseases.


Asunto(s)
Adenoviridae/genética , Regulación Viral de la Expresión Génica/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Trastornos Parkinsonianos/terapia , Transgenes/genética , Animales , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Cuerpo Estriado/cirugía , Modelos Animales de Enfermedad , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Genes Reporteros/genética , Vectores Genéticos/uso terapéutico , Operón Lac/genética , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Oxidopamina , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/metabolismo , Ratas , Ratas Sprague-Dawley , Sustancia Negra/metabolismo , Sustancia Negra/fisiopatología , Simpaticolíticos , Transfección/métodos
12.
Brain Res Bull ; 68(1-2): 31-41, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16325002

RESUMEN

The poor survival of dopamine grafts in Parkinson's disease is one of the main obstacles to the widespread application of this therapy. One hypothesis is that implanted neurons, once removed from the embryonic environment, lack the differentiation factors needed to develop the dopaminergic phenotype. In an effort to improve the numbers of dopamine neurons surviving in the grafts, we have investigated the potential of adenoviral vectors to deliver the differentiation factor sonic hedgehog or the glial cell line-derived neurotrophic factor GDNF to dopamine-rich grafts in a rat model of Parkinson's disease. Adenoviral vectors containing sonic hedgehog, GDNF, or the marker gene LacZ were injected into the dopamine depleted striatum of hemiparkinsonian rats. Two weeks later, ventral mesencephalic cell suspensions were prepared from embryos of donor ages E12, E13, E14 or E15 and implanted into the vector-transduced striatum. Pre-treatment with the sonic hedgehog vector produced a three-fold increase in the numbers of tyrosine hydroxylase-positive (presumed dopaminergic) cells in grafts derived from E12 donors, but had no effect on E13-E15 grafts. By contrast, pre-treatment with the GDNF vector increased yields of dopamine cells in grafts derived from E14 and E15 donors but had no effect on grafts from younger donors. The results indicate that provision of both trophic and differentiation factors can enhance the yields of dopamine neurons in ventral mesencephalic grafts, but that the two factors differ in the age and stage of embryonic development at which they have maximal effects.


Asunto(s)
Trasplante de Tejido Fetal/métodos , Terapia Genética/métodos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Mesencéfalo/trasplante , Enfermedad de Parkinson/terapia , Transactivadores/genética , Adenoviridae/genética , Anfetamina/farmacología , Animales , Recuento de Células , Modelos Animales de Enfermedad , Dopamina/fisiología , Femenino , Edad Gestacional , Proteínas Hedgehog , Actividad Motora/efectos de los fármacos , Oxidopamina , Enfermedad de Parkinson/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Simpaticolíticos , Simpatomiméticos/farmacología
13.
Trends Endocrinol Metab ; 12(2): 58-64, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11167123

RESUMEN

The main objectives of pituitary tumour treatment are to restore normal function of the pituitary gland and prevent tumour recurrences. In spite of the success of current therapies in the treatment of relatively small tumours, new therapeutic alternatives need to be explored for large invasive tumours, tumour recurrences postsurgery, and when intolerance to drug treatment develops. Gene therapy, which uses nucleic acids as drugs, is a very attractive alternative to classic therapeutic modalities. With the development of efficient gene delivery vectors, which allow widespread distribution and long-term transgene expression with limited side effects, the clinical implementation of gene therapy for the treatment of pituitary tumours will become a reality within the next five to ten years.


Asunto(s)
Terapia Genética , Neoplasias Hipofisarias/terapia , Adenoviridae/genética , Animales , Terapia Combinada , Marcación de Gen , Terapia Genética/efectos adversos , Vectores Genéticos , Herpesvirus Humano 1/genética , Humanos , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/cirugía , Retroviridae/genética
14.
Hum Gene Ther ; 12(7): 839-46, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11339900

RESUMEN

The utility of first-generation adenovirus vectors for long-term gene transfer in humans is limited by preexisting antiadenoviral immunity. We demonstrate here that new-generation high-capacity adenovirus vectors (HC-Ads) can efficiently transduce the brain and mediate stable transgene expression for at least 2 months, even in the presence of a preexisting antiadenoviral immune response. First-generation vector-mediated transduction was almost completely abolished in preimmunized animals within 60 days of the vector injection. Levels of HC-Ad-mediated transduction by 3 days postinjection were not significantly affected by preimmunization, were reduced within 14 days to 56% of those levels seen in nonimmunized animals, and remained stable until day 60 postinjection. Acute brain inflammation elicited by the HC-Ad vector injection was more transient, and was reduced in intensity compared with brain inflammation elicited by the first-generation vector injection in immunized animals. Inflammation was significantly higher in all immunized animals than in nonimmunized animals. Our results show that preexisting antiadenoviral immunity does not significantly reduce initial HC-Ad-mediated infection of the brain and is not a barrier to stable HC-Ad vector-mediated transduction of the CNS. Although input HC-Ad capsid proteins injected into the brain may contain transient targets for a brain-infiltrating cellular adenovirus-specific immune response, this fails to eliminate transgene expression. Thus HC-Ads show promise for gene therapy of chronic brain disease.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Encéfalo/metabolismo , Vectores Genéticos/genética , Transgenes/genética , Animales , Encéfalo/inmunología , Encéfalo/patología , Expresión Génica , Terapia Genética/métodos , Sueros Inmunes/inmunología , Inmunización , Inmunohistoquímica , Inflamación/inmunología , Inflamación/patología , Pruebas de Neutralización , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Transducción Genética , Transgenes/inmunología
15.
Hum Gene Ther ; 10(10): 1641-8, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10428209

RESUMEN

Fas ligand (FasL) is a cytokine, produced by activated T cells and NK cells, that triggers apoptosis of Fas-positive target cells including human glioma cells. As shown here, in vitro infection of rat F98 and human LN18 glioma cell lines with recombinant adenovirus (rAd) expressing FasL cDNA under control of the cytomegalovirus promoter (rAd-CMV-FasL) induced striking cytotoxicity in Fas-positive glioma cell lines but not in the Fas-negative F98 glioma subline F98/ZH. The extent of FasL-mediated cytotoxic effects outranged the expectations based on expression of beta-galactosidase (beta-Gal) by F98 cells infected with a control virus expressing the lacZ gene (rAd-CMV-lacZ). The detection of FasL bioactivity in supernatants of infected cells provides evidence of a bystander mechanism involving the cytotoxic action of FasL on uninfected cells. In F98 tumor-bearing rats, infection with rAd-CMV-FasL increased the mean survival time by 50% compared with infection with rAd-CMV-lacZ or untreated controls. These data suggest that viral vector transduction of the FasL gene could be part of a successful glioma gene therapy.


Asunto(s)
Adenovirus Humanos , Terapia Genética , Vectores Genéticos , Glioma/terapia , Glicoproteínas de Membrana/genética , Animales , Resistencia a Medicamentos , Proteína Ligando Fas , Expresión Génica , Terapia Genética/métodos , Humanos , Masculino , Glicoproteínas de Membrana/biosíntesis , Ratones , Neoplasias Experimentales/terapia , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas
16.
Endocrinology ; 142(1): 464-76, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11145611

RESUMEN

Adenoviral vectors have been identified as useful tools for gene transfer to the pituitary gland with the aim of providing therapeutic treatments for pituitary diseases. Although successful adenovirus-mediated gene transfer to the pituitary has been shown, the duration of transgene expression, local immune responses and consequences on circulating pituitary hormone levels have not been investigated. These are critical not only for the successful implementation of these gene transfer techniques both for physiological and/or therapeutic applications but also for assessing the safety of these approaches. We have therefore assessed duration and levels of transgene expression 3 days, 14 days, 1, 2, and 3 months after delivery of adenoviruses expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK), under the control of the major immediate early human cytomegalovirus (RAd-hCMV/TK) or human PRL (RAd-hPrl/TK) promoters, to the anterior pituitary (AP) gland in situ. The presence of vector genome and cellular immune infiltrates within the AP gland were also studied along with the levels of circulating anti-adenovirus neutralizing antibodies and AP hormones in sera. Ubiquitous or cell-type specific expression of HSV1-TK within the AP gland was seen from RAd-hCMV/TK and RAd-hPrl/TK respectively at all time points, although a reduction in expression was seen over time. PCR amplification of HSV1-TK specific sequences showed the persistence of adenoviral genomes for up to 3 months. Analysis of the AP showed the presence of a virus-induced inflammation that peaked around day 14 and was resolved between 2-3 months. ED1-positive macrophages, CD8-positive T-cells and CD161-positive NK cells were identified up to 1 month after virus administration. A virus-induced humoral immune response was also present as anti-adenovirus neutralizing antibodies were detected from 14 days after virus administration. Levels of circulating pituitary hormones were unaffected by virus administration with the exception of the stress hormone ACTH which was increased at 3 days but normalized by 14 days. In conclusion, our data indicates that adenovirus-mediated delivery to the AP gland in situ may be a useful tool for the treatment of pituitary diseases as no major cytotoxicity or disruption of AP hormonal functions are seen. Despite of this, further developments to this approach still need to be made to combat the reduced transgene expression seen over time and the induction of virus-induced immune responses.


Asunto(s)
Técnicas de Transferencia de Gen , Adenohipófisis/fisiología , Hormonas Adenohipofisarias/análisis , Timidina Quinasa/análisis , Timidina Quinasa/genética , Adenoviridae , Animales , Anticuerpos Heterófilos/análisis , Formación de Anticuerpos , Linfocitos T CD8-positivos/inmunología , Citomegalovirus/genética , Vectores Genéticos , Humanos , Inmunidad Celular , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Masculino , Adenohipófisis/inmunología , Hormonas Adenohipofisarias/sangre , Prolactina/genética , Regiones Promotoras Genéticas , Ratas , Ratas Endogámicas BUF , Simplexvirus/genética
17.
Endocrinology ; 142(6): 2521-32, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11356701

RESUMEN

To further develop our understanding of anterior pituitary (AP) function and to aid the development of gene therapy strategies for the treatment of pituitary diseases, adenovirus (Ad)-mediated gene transfer to the AP gland will be a useful tool. Although successful widespread gene transfer within the AP has been achieved using first generation Ads the ability to control transgene expression would be very beneficial when studying AP regulatory functions and delivering a potentially therapeutic gene into the AP gland. A dual adenoviral vector system encoding for cell type-specific and regulatable transcription units was developed to achieve transcriptionally targeted transgenesis within specific cell populations in the adult AP gland. To achieve regulatable transgene expression within predetermined AP cells, the tetracycline-responsive transcriptional elements have been engineered to be under the control of human, lactotroph-specific PRL (hPRL) promoter elements within a dual adenoviral vector system. The inducibility, cell type specificity, and levels of transgene expression were characterized in vitro and in vivo and compared with the strong ubiquitous beta-actin/human cytomegalovirus (CAG) promoter. Inducible expression of the marker gene beta-galactosidase under the control of the hPRL promoter was restricted to lactotrophic tumor cell lines and lactotrophic cells within primary AP cultures. Lactotroph cell type specificity and inducible transgene expression were also observed within the AP gland in vivo, and this could be switched on or off. Administration of doxycycline abrogated transgene expression both in vitro and in vivo. Our results also provide evidence that an excess of trans-activator is needed to achieve maximal transgene expression. Our data indicate that combined transcriptional and inducible transgenesis can be achieved using adenoviral vectors that allow spatial and temporal restriction of transgene expression within the adult AP gland in vivo.


Asunto(s)
Regulación de la Expresión Génica , Adenohipófisis/metabolismo , Prolactina/genética , Adenoviridae/genética , Animales , Animales Modificados Genéticamente , Línea Celular , Células Cultivadas , Cricetinae , Doxiciclina/farmacología , Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , Inmunohistoquímica , Riñón , Masculino , Regiones Promotoras Genéticas , Ratas , Ratas Endogámicas BUF , Proteínas Recombinantes de Fusión , Elementos de Respuesta , Tetraciclina/farmacología , Transactivadores , Transfección , Células Tumorales Cultivadas , beta-Galactosidasa/genética
18.
Endocrinology ; 142(2): 795-801, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11159852

RESUMEN

Ablative therapies for pituitary tumors commonly cause irreversible damage to normal pituitary cells. Toxin gene therapy should therefore ideally be targeted to specific cell types to avoid collateral cell damage. To evaluate cell-type-specific adenoviral gene transfer in the intact pituitary gland we have used stereotaxic transcranial delivery of recombinant adenoviruses in the sheep with continuous assessment of endocrine function. Adenoviral ss-galactosidase expression was driven either by the human cytomegalovirus (hCMV) promoter or the human PRL gene promoter. The hCMV promoter directed adenoviral ss-galactosidase expression in all pituitary cell types, but the PRL promoter restricted this exclusively to lactotropic cells, indicating that this promoter conferred appropriate cell type specificity in the context of adenoviral transduction in vivo. Serial measurements of plasma hormones showed no disruption of endocrine function over 7 days after intrapituitary injection. In summary, this work shows cell type-specific expression of an adenoviral transgene in the mixed cell population of the intact pituitary gland in vivo in a large animal model and indicates that stereotaxic intrapituitary delivery does not disrupt normal endocrine function.


Asunto(s)
Expresión Génica , Técnicas de Transferencia de Gen , Hipófisis/fisiología , Técnicas Estereotáxicas , Transgenes/fisiología , Adenoviridae/genética , Animales , Femenino , Terapia Genética , Humanos , Ovinos
19.
Endocrinology ; 138(5): 2184-94, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9112418

RESUMEN

Adenovirus vectors have recently been used to transfer genes into a variety of cell types, including neurons, glial cells, Schwann cells, and epithelial cells. To evaluate the efficiency of gene transfer into pituitary cells using viral vectors, we used replication-deficient recombinant adenovirus vectors (RAds) encoding beta-galactosidase driven by various viral promoters. We tested the ability of RAds to infect and express beta-galactosidase within the different identified cell populations of the anterior pituitary anterior pituitary gland and also in tumor cells of anterior pituitary origin, i.e. GH3 and AtT20 cells. Our results demonstrate that transgenes encoded by RAds are expressed within all cell types of the adenohypophysis in vitro and also within AtT20 and GH3 endocrine tumor cells. Our long term expression studies indicate that long term expression with low cytotoxicity can be achieved, but that the longevity of transgene expression from RAds depends on the proliferative status of the target cells. Slowly dividing cells (endocrine population) express transgenes for longer than actively dividing cells (tumor cells and nonendocrine anterior pituitary cells). The ability of anterior pituitary cells to secrete ACTH or LH through the regulated secretory pathway decreased after infection with RAds at high multiplicity of infection (> or = 20 plaque-forming units/target cell), whereas cell viability was not affected. We also demonstrate that a higher percentage of cells expressed the transgene beta-galactosidase when we infected actively dividing GH3 cells compared with the infection of growth-arrested GH3 cells. This could reflect differential virus entry or differential activity of the individual promoters during different stages of the cell cycle. This work demonstrates that high efficiency gene transfer into all pituitary cell types can be achieved with RAds, and that this system can be exploited to characterize and experimentally manipulate pituitary-specific gene expression. The higher efficiency of infection and transgene expression in actively dividing cells compared to that in their growth-arrested counterparts could also be exploited for the treatment of pituitary adenomas that do not respond to classical treatment strategies, using suicide or cytotoxic gene therapy.


Asunto(s)
Adenoviridae/genética , ADN Recombinante , Expresión Génica , Adenohipófisis/metabolismo , Hormonas Hipofisarias/metabolismo , Neoplasias Hipofisarias/metabolismo , Animales , Ciclo Celular , Citometría de Flujo , Técnicas de Transferencia de Gen , Vectores Genéticos , Masculino , Ratas , Ratas Wistar , Células Tumorales Cultivadas , beta-Galactosidasa/genética
20.
Endocrinology ; 141(9): 3493-505, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10965923

RESUMEN

The use of pituitary cell type-specific promoters is a powerful molecular tool to achieve pituitary cell type-specific transcriptional targeting of transgenes encoded by viral vectors. It has recently been proposed that transcriptional targeting of therapeutic genes could be harnessed as a gene therapy strategy for the treatment of pituitary disease. We describe the successful use of the human PRL promoter (hPrl) encoded within recombinant adenovirus vectors to target transgene expression of Herpes Simplex Virus Type 1-Thymidine Kinase (HSV1-TK) or beta-galactosidase to lactotrophic cells in vitro and in vivo. Functionally, the restriction of expression of HSV1-TK to lactotrophic tumor cells, using the hPrl promoter, resulted in the cell type-specific induction of apoptosis in the lactotrophic GH3 tumor cell line, in the presence of ganciclovir (GCV). In the corticotrophic AtT20 cell line, we detected neither HSV1-TK expression, nor apoptosis in the presence of GCV. The hPrl promoter encoded within a recombinant adenoviral vector also restricted transgene expression to lactotrophic cells in primary anterior pituitary (AP) cultures, and importantly, within the anterior pituitary gland in vivo. When the HSV1-TK driven by hPrl promoter was used in an in vivo model ofestrogen/sulpiride lactotroph induced hyperplasia within the AP in situ, the treatment was not effective in either reducing the weight of the gland, the number of lactotrophic cells within the transduced area in vivo, or the circulating PRL levels. This is in contrast to the human cytomegalovirus promoter (hCMV) driving expression of HSV1-TK in the same experimental paradigm, which was effective in reducing pituitary weight and circulating PRL levels. Our results have important implications in the design of gene therapy strategies for pituitary tumors. We demonstrate that both the choice of the in vivo animal model, i.e. adenoma in the AP gland in situ, and the particular gene therapy strategy chosen, i.e. use of strong ubiquitous promoters vs. weaker but cell type-specific promoters, determine the experimental therapeutic outcome.


Asunto(s)
Adenoviridae/genética , Antipsicóticos/farmacología , Estrógenos/farmacología , Marcación de Gen/métodos , Vectores Genéticos/genética , Adenohipófisis/citología , Sulpirida/farmacología , Transcripción Genética/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Galactosidasas/genética , Herpesvirus Humano 1/enzimología , Hiperplasia/inducido químicamente , Hiperplasia/patología , Inmunohistoquímica , Indicadores y Reactivos , Adenohipófisis/patología , Hormonas Adenohipofisarias/sangre , Ratas , Ratas Endogámicas BUF , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA