Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biotechnol Bioeng ; 118(9): 3348-3358, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33624832

RESUMEN

Single-domain antibodies (sdAbs) offer the affinity and therapeutic value of conventional antibodies, with increased stability and solubility. Unlike conventional antibodies, however, sdAbs do not benefit from a platform manufacturing process. While successful production of a variety of sdAbs has been shown in numerous hosts, purification methods are often molecule specific or require affinity tags, which generally cannot be used in clinical manufacturing due to regulatory concerns. Here, we have developed a broadly applicable production and purification process for sdAbs in Komagataella phaffii (Pichia pastoris) and demonstrated the production of eight different sdAbs at a quality appropriate for nonclinical studies. We developed a two-step, integrated purification process without the use of affinity resins and showed that modification of a single process parameter, pH of the bridging buffer, was required for the successful purification of a variety of sdAbs. Further, we determined that this parameter can be predicted based only on the biophysical characteristics of the target molecule. Using these methods, we produced nonclinical quality sdAbs as few as 5 weeks after identifying the product sequence. Nonclinical studies of three different sdAbs showed that molecules produced using our platform process conferred protection against viral shedding of rotavirus or H1N1 influenza and were equivalent to similar molecules produced in Escherichia coli and purified using affinity tags.


Asunto(s)
Anticuerpos Antivirales , Subtipo H1N1 del Virus de la Influenza A/inmunología , Rotavirus/inmunología , Saccharomycetales/crecimiento & desarrollo , Anticuerpos de Cadena Única , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/aislamiento & purificación
2.
Mol Cell Neurosci ; 67: 75-83, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26051800

RESUMEN

Transforming growth factor beta 1 (TGF-beta1), an anti-inflammatory cytokine, has been shown to have pro-neurogenic effects on adult Neural Stem Cells (aNSC) from the dentate gyrus and in vivo models. Here, we expanded the observation of the pro-neurogenic effect of TGF-beta1 on aNSC from the subventricular zone (SVZ) of adult rats and performed a functional genomic analysis to identify candidate genes to mediate its effect. 10 candidate genes were identified by microarray analysis and further validated by qRT-PCR. Of these, Fibulin-2 was increased 477-fold and its inhibition by siRNA blocks TGF-beta1 pro-neurogenic effect. Curiously, Fibulin-2 was not expressed by aNSC but by a GFAP-positive population in the culture, suggesting an indirect mechanism of action. TGF-beta1 also induced Fibulin-2 in the SVZ in vivo. Interestingly, 5 out of the 10 candidate genes identified are known to interact with integrins, paving the way for exploring their functional role in adult neurogenesis. In conclusion, we have identified 10 genes with putative pro-neurogenic effects, 5 of them related to integrins and provided proof that Fibulin-2 is a major mediator of the pro-neurogenic effects of TGF-beta1. These data should contribute to further exploring the molecular mechanism of adult neurogenesis of the genes identified and the involvement of the integrin pathway on adult neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Factor de Crecimiento Transformador beta1/farmacología , Células Madre Adultas/citología , Células Madre Adultas/efectos de los fármacos , Animales , Astrocitos/metabolismo , Proteínas de Unión al Calcio/genética , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Integrinas/metabolismo , Ventrículos Laterales/citología , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta1/metabolismo
3.
Proc Natl Acad Sci U S A ; 109(20): 7835-40, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22547816

RESUMEN

Following antigen recognition on target cells, effector T cells establish immunological synapses and secrete cytokines. It is thought that T cells secrete cytokines in one of two modes: either synaptically (i.e., toward antigenic target cells) or multidirectionally, affecting a wider population of cells. This paradigm predicts that synaptically secreted cytokines such as IFN-γ will preferentially signal to antigenic target cells contacted by the T cell through an immunological synapse. Despite its physiological significance, this prediction has never been tested. We developed a live-cell imaging system to compare the responses of target cells and nonantigenic bystanders to IFN-γ secreted by CD8+, antigen-specific, cytotoxic T cells. Both target cells and surrounding nontarget cells respond robustly. This pattern of response was detected even at minimal antigenic T-cell stimulation using low doses of antigenic peptide, or altered peptide ligands. Although cytotoxic immunological synapses restrict killing to antigenic target cells, the effects of IFN-γ are more widespread.


Asunto(s)
Sinapsis Inmunológicas/inmunología , Interferón gamma/metabolismo , Linfocitos T Citotóxicos/inmunología , Adenoviridae , Análisis de Varianza , Astrocitos/inmunología , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Procesamiento de Imagen Asistido por Computador , Interferón gamma/inmunología , Microscopía/métodos
4.
BMC Vet Res ; 10: 8, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24401205

RESUMEN

BACKGROUND: Bovine herpesvirus type 1 (BoHV-1) is the causative agent of respiratory and genital tract infections; causing a high economic loss in all continents. Use of marker vaccines in IBR eradication programs is widely accepted since it allows for protection of the animals against the disease while adding the possibility of differentiating vaccinated from infected animals.The aim of the present study was the development and evaluation of safety and efficacy of a glycoprotein E-deleted (gE-) BoHV-1 marker vaccine strain (BoHV-1ΔgEßgal) generated by homologous recombination, replacing the viral gE gene with the ß-galactosidase (ßgal) gene. RESULTS: In vitro growth kinetics of the BoHV-1ΔgEßgal virus was similar to BoHV-1 LA. The immune response triggered by the new recombinant strain in cattle was characterized both as live attenuated vaccine (LAV) and as an inactivated vaccine. BoHV-1ΔgEßgal was highly immunogenic in both formulations, inducing specific humoral and cellular immune responses. Antibody titers found in animals vaccinated with the inactivated vaccine based on BoHV-1ΔgEßgal was similar to the titers found for the control vaccine (BoHV-1 LA). In the same way, titers of inactivated vaccine groups were significantly higher than any of the LAV immunized groups, independently of the inoculation route (p < 0.001). Levels of IFN-γ were significantly higher (p < 0.001) in those animals that received the LAV compared to those that received the inactivated vaccine. BoHV-1ΔgEßgal exhibited an evident attenuation when administered as a LAV; no virus was detected in nasal secretions of vaccinated or sentinel animals during the post-vaccination period. BoHV-1ΔgEßgal, when used in either formulation, elicited an efficient immune response that protected animals against challenge with virulent wild-type BoHV-1. Also, the deletion of the gE gene served as an immunological marker to differentiate vaccinated animals from infected animals. All animals vaccinated with the BoHV-1ΔgE ßgal strain were protected against disease after challenge and shed significantly less virus than control calves, regardless of the route and formulation they were inoculated. CONCLUSIONS: Based on its attenuation, immunogenicity and protective effect after challenge, BoHV-1ΔgEßgal virus is an efficient and safe vaccine candidate when used either as inactivated or as live attenuated forms.


Asunto(s)
Infecciones por Herpesviridae/veterinaria , Herpesvirus Bovino 1/metabolismo , Proteínas Virales/metabolismo , Vacunas Virales/inmunología , Animales , Bovinos , Línea Celular , Perros , Femenino , Eliminación de Gen , Regulación Viral de la Expresión Génica/fisiología , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/inmunología , Embarazo , Complicaciones Infecciosas del Embarazo/prevención & control , Complicaciones Infecciosas del Embarazo/veterinaria , Complicaciones Infecciosas del Embarazo/virología , Vacunas Atenuadas , Vacunas de Productos Inactivados , Proteínas Virales/genética , Vacunas Virales/efectos adversos
5.
Toxicol Appl Pharmacol ; 268(3): 318-30, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23403069

RESUMEN

Adenoviral vectors (Ads) are promising gene delivery vehicles due to their high transduction efficiency; however, their clinical usefulness has been hampered by their immunogenicity and the presence of anti-Ad immunity in humans. We reported the efficacy of a gene therapy approach for glioma consisting of intratumoral injection of Ads encoding conditionally cytotoxic herpes simplex type 1 thymidine kinase (Ad-TK) and the immunostimulatory cytokine fms-like tyrosine kinase ligand 3 (Ad-Flt3L). Herein, we report the biodistribution, efficacy, and neurological and systemic effects of a bicistronic high-capacity Ad, i.e., HC-Ad-TK/TetOn-Flt3L. HC-Ads elicit sustained transgene expression, even in the presence of anti-Ad immunity, and can encode large therapeutic cassettes, including regulatory elements to enable turning gene expression "on" or "off" according to clinical need. The inclusion of two therapeutic transgenes within a single vector enables a reduction of the total vector load without adversely impacting efficacy. Because clinically the vectors will be delivered into the surgical cavity, normal regions of the brain parenchyma are likely to be transduced. Thus, we assessed any potential toxicities elicited by escalating doses of HC-Ad-TK/TetOn-Flt3L (1×10(8), 1×10(9), or 1×10(10) viral particles [vp]) delivered into the rat brain parenchyma. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points. The results indicate that doses up to 1×10(9) vp of HC-Ad-TK/TetOn-Flt3L can be safely delivered into the normal rat brain and underpin further developments for its implementation in a phase I clinical trial for glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Ensayos Clínicos Fase I como Asunto/métodos , Citotoxinas/administración & dosificación , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioblastoma/tratamiento farmacológico , Inmunización/métodos , Adenoviridae/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citotoxinas/efectos adversos , Citotoxinas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Ratas , Ratas Endogámicas Lew , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Resultado del Tratamiento
6.
Mol Ther ; 20(4): 808-19, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22233583

RESUMEN

The adaptive immune response to viral vectors reduces vector-mediated transgene expression from the brain. It is unknown, however, whether this loss is caused by functional downregulation of transgene expression or death of transduced cells. Herein, we demonstrate that during the elimination of transgene expression, the brain becomes infiltrated with CD4(+) and CD8(+) T cells and that these T cells are necessary for transgene elimination. Further, the loss of transgene-expressing brain cells fails to occur in the absence of IFNγ, perforin, and TNFα receptor. Two methods to induce severe immune suppression in immunized animals also fail to restitute transgene expression, demonstrating the irreversibility of this process. The need for cytotoxic molecules and the irreversibility of the reduction in transgene expression suggested to us that elimination of transduced cells is responsible for the loss of transgene expression. A new experimental paradigm that discriminates between downregulation of transgene expression and the elimination of transduced cells demonstrates that transduced cells are lost from the brain upon the induction of a specific antiviral immune response. We conclude that the anti-adenoviral immune response reduces transgene expression in the brain through loss of transduced cells.


Asunto(s)
Encéfalo/citología , Interferón gamma/metabolismo , Perforina/metabolismo , Transducción Genética/métodos , Transgenes/genética , Factor de Necrosis Tumoral alfa/metabolismo , Adenoviridae/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos
7.
Proc Natl Acad Sci U S A ; 107(46): 20021-6, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21030678

RESUMEN

Restricting the cytotoxicity of anticancer agents by targeting receptors exclusively expressed on tumor cells is critical when treating infiltrative brain tumors such as glioblastoma multiforme (GBM). GBMs express an IL-13 receptor (IL13Rα2) that differs from the physiological IL4R/IL13R receptor. We developed a regulatable adenoviral vector (Ad.mhIL-4.TRE.mhIL-13-PE) encoding a mutated human IL-13 fused to Pseudomonas exotoxin (mhIL-13-PE) that specifically binds to IL13Rα2 to provide sustained expression, effective anti-GBM cytotoxicity, and minimal neurotoxicity. The therapeutic Ad also encodes mutated human IL-4 that binds to the physiological IL4R/IL13R without interacting with IL13Rα2, thus inhibiting potential binding of mhIL-13-PE to normal brain cells. Using intracranial GBM xenografts and syngeneic mouse models, we tested the Ad.mhIL-4.TRE.mhIL-13-PE and two protein formulations, hIL-13-PE used in clinical trials (Cintredekin Besudotox) and a second-generation mhIL-13-PE. Cintredekin Besudotox doubled median survival without eliciting long-term survival and caused severe neurotoxicity; mhIL-13-PE led to ∼40% long-term survival, eliciting severe neurological toxicity at the high dose tested. In contrast, Ad-mediated delivery of mhIL-13-PE led to tumor regression and long-term survival in over 70% of the animals, without causing apparent neurotoxicity. Although Cintredekin Besudotox was originally developed to target GBM, when tested in a phase III trial it failed to achieve clinical endpoints and revealed neurotoxicity. Limitations of Cintredekin Besudotox include its short half-life, which demanded frequent or continued administration, and binding to IL4R/IL13R, present in normal brain cells. These shortcomings were overcome by our therapeutic Ad, thus representing a significant advance in the development of targeted therapeutics for GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Citotoxinas/genética , Citotoxinas/uso terapéutico , Técnicas de Transferencia de Gen , Terapia Genética , Glioma/tratamiento farmacológico , Adenoviridae/genética , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Exotoxinas/genética , Exotoxinas/uso terapéutico , Vectores Genéticos/genética , Glioma/patología , Humanos , Inmunocompetencia/inmunología , Interleucina-13/genética , Interleucina-13/uso terapéutico , Ratones , Ratones Desnudos , Mutación/genética , Neurotoxinas/toxicidad , Pseudomonas/metabolismo , Transgenes/genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Virol ; 84(12): 6007-17, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20375153

RESUMEN

Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Conditional cytotoxic/immune-stimulatory gene therapy (Ad-TK and Ad-Flt3L) elicits tumor regression and immunological memory in rodent GBM models. Since the majority of patients enrolled in clinical trials would exhibit adenovirus immunity, which could curtail transgene expression and therapeutic efficacy, we used high-capacity adenovirus vectors (HC-Ads) as a gene delivery platform. Herein, we describe for the first time a novel bicistronic HC-Ad driving constitutive expression of herpes simplex virus type 1 thymidine kinase (HSV1-TK) and inducible Tet-mediated expression of Flt3L within a single-vector platform. We achieved anti-GBM therapeutic efficacy with no overt toxicities using this bicistronic HC-Ad even in the presence of systemic Ad immunity. The bicistronic HC-Ad-TK/TetOn-Flt3L was delivered into intracranial gliomas in rats. Survival, vector biodistribution, neuropathology, systemic toxicity, and neurobehavioral deficits were assessed for up to 1 year posttreatment. Therapeutic efficacy was also assessed in animals preimmunized against Ads. We demonstrate therapeutic efficacy, with vector genomes being restricted to the brain injection site and an absence of overt toxicities. Importantly, antiadenoviral immunity did not inhibit therapeutic efficacy. These data represent the first report of a bicistronic vector platform driving the expression of two therapeutic transgenes, i.e., constitutive HSV1-TK and inducible Flt3L genes. Further, our data demonstrate no promoter interference and optimum gene delivery and expression from within this single-vector platform. Analysis of the efficacy, safety, and toxicity of this bicistronic HC-Ad vector in an animal model of GBM strongly supports further preclinical testing and downstream process development of HC-Ad-TK/TetOn-Flt3L for a future phase I clinical trial for GBM.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Glioma/genética , Glioma/terapia , Herpesvirus Humano 1/enzimología , Timidina Quinasa/uso terapéutico , Proteínas Virales/uso terapéutico , Tirosina Quinasa 3 Similar a fms/uso terapéutico , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glioma/metabolismo , Herpesvirus Humano 1/genética , Humanos , Ratas , Ratas Endogámicas Lew , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
9.
Mol Pain ; 5: 42, 2009 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-19656360

RESUMEN

BACKGROUND: Our goal is to use gene therapy to alleviate pain by targeting glial cells. In an animal model of facial pain we tested the effect of transfecting the glutamic acid decarboxylase (GAD) gene into satellite glial cells (SGCs) of the trigeminal ganglion by using a serotype 5 adenovector with high tropisms for glial cells. We postulated that GABA produced from the expression of GAD would reduce pain behavior by acting on GABA receptors on neurons within the ganglion. RESULTS: Injection of adenoviral vectors (AdGAD65) directly into the trigeminal ganglion leads to sustained expression of the GAD65 isoform over the 4 weeks observation period. Immunohistochemical analysis showed that adenovirus-mediated GAD65 expression and GABA synthesis were mainly in SGCs. GABAA and GABAB receptors were both seen in sensory neurons, yet only GABAA receptors decorated the neuronal surface. GABA receptors were not found on SGCs. Six days after injection of AdGAD65 into the trigeminal ganglion, there was a statistically significant decrease of pain behavior in the orofacial formalin test, a model of inflammatory pain. Rats injected with control virus (AdGFP or AdLacZ) had no reduction in their pain behavior. AdGAD65-dependent analgesia was blocked by bicuculline, a selective GABAA receptor antagonist, but not by CGP46381, a selective GABAB receptor antagonist. CONCLUSION: Transfection of glial cells in the trigeminal ganglion with the GAD gene blocks pain behavior by acting on GABAA receptors on neuronal perikarya.


Asunto(s)
Adenoviridae/genética , Dolor Facial/terapia , Terapia Genética , Vectores Genéticos/genética , Glutamato Descarboxilasa/fisiología , Ganglio del Trigémino/metabolismo , Analgesia/métodos , Animales , Pollos , Glutamato Descarboxilasa/genética , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
10.
J Virol ; 82(9): 4680-4, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18287240

RESUMEN

Gene therapy is proposed as a novel therapeutic strategy for treating glioblastoma multiforme (GBM), a devastating brain cancer. In the clinic, antivector immune responses pose formidable challenges. Herein we demonstrate that high-capacity adenovirus vectors (HC-Ads) carrying the conditional cytotoxic gene herpes simplex virus type 1-thymidine kinase (TK) induce tumor regression and long-term survival in an intracranial glioma model, even in the presence of systemic antiadenovirus immunity, as could be encountered in patients. First-generation Ad-TK failed to elicit tumor regression in this model. These results pave the way for implementing HC-Ad-TK-mediated gene therapy as a powerful adjuvant for treating GBM.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Glioblastoma/terapia , Timidina Quinasa/uso terapéutico , Adenoviridae/inmunología , Animales , Anticuerpos Antivirales/farmacología , Encéfalo/patología , Neoplasias Encefálicas/terapia , Modelos Animales de Enfermedad , Herpesvirus Humano 1/enzimología , Humanos , Ratas , Ratas Endogámicas Lew , Tasa de Supervivencia , Timidina Quinasa/genética , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
11.
Mol Ther ; 16(2): 343-51, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18180781

RESUMEN

Immune responses against vectors or encoded transgenes can impose limitations on gene therapy. We demonstrated that tetracycline-regulated high-capacity adenoviral vectors (HC-Ads) sustain regulated transgene expression in the brain even in the presence of systemic pre-existing immune responses against adenoviruses. In this study we assessed whether systemic pre-existing immune responses against the transgene products, i.e., beta-Gal or the tetracycline-dependent (TetON) regulatory transcription factors (rtTA2(S)M2 and the tTS(Kid)), affect transgene expression levels and the safety profile of HC-Ads in the brain. We pre-immunized mice with plasmids encoding the TetON switch expressing rtTA2(S)M2 and the tTS(Kid) or beta-Gal. HC-Ads expressing beta-Gal under the control of the TetON switch were then injected into the striatum. We assessed levels and distribution of beta-Gal expression, and evaluated local inflammation and neuropathological changes. We found that systemic immunity against beta-Gal, but not against the TetON switch, led to inflammation and reduction of transgene expression in the striatum. Therefore, the regulatory TetON switch appears to be safe to use, and capable of sustaining transgene expression in the brain even in the presence of an immune response against its components. Systemic immunity against the transgene had the effect of curtailing its expression, thereby affecting the efficacy and safety of gene delivery to the brain. This factor should be considered when developing gene therapies for neurological use.


Asunto(s)
Adenoviridae/genética , Encéfalo/metabolismo , Inmunización/métodos , Transgenes/genética , Animales , Western Blotting , Encéfalo/inmunología , Femenino , Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Inmunohistoquímica , Inflamación/inmunología , Ratones , Plásmidos/genética , Tetraciclina/farmacología , beta-Galactosidasa/metabolismo
12.
Neuro Oncol ; 10(1): 19-31, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18079358

RESUMEN

The disseminated characteristics of human glioblastoma multiforme (GBM) make it a particularly difficult tumor to treat with long-term efficacy. Most preclinical models of GBM involve treatment of a single tumor mass. For therapeutic outcomes to translate from the preclinical to the clinical setting, induction of an antitumor response capable of eliminating multifocal disease is essential. We tested the hypothesis that expression of Flt3L (human soluble FMS-like tyrosine kinase 3 ligand) and TK (herpes simplex virus type 1-thymidine kinase) within brain gliomas would mediate regression of the primary, treated tumor mass and a secondary, untreated tumor growing at a distant site from the primary tumor and the site of therapeutic vector injection. In both the single-GBM and multifocal-GBM models used, all saline-treated control animals succumbed to tumors by day 22. Around 70% of the animals bearing a single GBM mass treated with an adenovirus expressing Flt3L (AdFlt3L) and an adenovirus expressing TK (AdTK + GCV) survived long term. Approximately 50% of animals bearing a large primary GBM that were implanted with a second GBM in the contralateral hemisphere at the same time the primary tumors were being treated with AdFlt3L and AdTK also survived long term. A second multifocal GBM model, in which bilateral GBMs were implanted simultaneously and only the right tumor mass was treated with AdFlt3L and AdTK, also demonstrated long-term survival. While no significant difference in survival was found between unifocal and multifocal GBM-bearing animals treated with AdFlt3L and AdTK, both treatments were statistically different from the saline-treated control group (p < 0.05). Our results demonstrate that combination therapy with AdFlt3L and AdTK can eradicate multifocal brain tumor disease in a syngeneic, intracranial GBM model.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Proteínas de la Membrana/metabolismo , Timidina Quinasa/metabolismo , Adenoviridae/genética , Animales , Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Glioblastoma/patología , Humanos , Masculino , Microscopía Confocal , Ratas , Ratas Endogámicas Lew , Trasplante Isogénico
13.
Methods Mol Biol ; 434: 239-66, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18470649

RESUMEN

Regulatable promoter systems allow gene expression to be tightly controlled in vivo. This is highly desirable for the development of safe, efficacious adenoviral vectors that can be used to treat human diseases in the clinic. Ideally, regulatable cassettes should have minimal gene expression in the "OFF" state, and expression should quickly reach therapeutic levels in the "ON" state. In addition, the components of regulatable cassettes should be non-toxic at physiological concentrations and should not be immunogenic, especially when treating chronic illness that requires long-lasting gene expression. In this chapter, we will describe in detail protocols to develop and validate first generation (Ad) and high-capacity adenoviral (HC-Ad) vectors that express therapeutic genes under the control of the TetON regulatable system. Our laboratory has successfully used these protocols to regulate the expression of marker genes, immune stimulatory genes, and toxins for cancer gene therapeutics, i.e., glioma that is a deadly form of brain cancer. We have shown that this third generation TetON regulatable system, incorporating a doxycycline (DOX)-sensitive rtTA(2)S-M2 inducer and tTS(Kid) silencer, is non-toxic, relatively non-immunogenic, and can tightly regulate reporter transgene expression downstream of a TRE promoter from adenoviral vectors in vitro and also in vivo.


Asunto(s)
Adenoviridae/genética , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Glioma/metabolismo , Glioma/terapia , Transgenes/fisiología , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/metabolismo , Antibacterianos/uso terapéutico , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Southern Blotting , Células Cultivadas , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Doxiciclina/uso terapéutico , Exotoxinas/genética , Exotoxinas/metabolismo , Glioma/genética , Humanos , Tetraciclina/uso terapéutico , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Exotoxina A de Pseudomonas aeruginosa
14.
Mol Neurobiol ; 55(2): 1068-1081, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28092084

RESUMEN

When disrupted, iron homeostasis negatively impacts oligodendrocyte (OLG) differentiation and impairs myelination. To better understand myelin formation and OLG maturation, in vivo and in vitro studies were conducted to evaluate the effect of iron deficiency (ID) not only on OLG maturation but also on astrocytes (AST) and microglial cells (MG). In vivo experiments in an ID model were carried out to describe maturational events during OLG and AST development and the reactive profile of MG during myelination when iron availability is lower than normal. In turn, in vitro assays were conducted to explore proliferating and maturational states of each glial cell type derived from control or ID conditions. Studies targeted NG2, PDGFRα, CNPAse, CC1, and MBP expression in OLG, GFAP and S100 expression in AST, and CD11b, ED1, and cytokine expression in MG, as well as BrDU incorporation in the three cell types. Our results show that ID affected OLG development at early stages, not only reducing their maturation capacity but also increasing their proliferation and affecting their morphological complexity. AST ID proliferated more than control ones and were more immature, much like OLG. Cytokine expression in ID animals reflected an anti-inflammatory state which probably influenced OLG maturation. These results show that ID conditions alter all glial cells and may impact myelin formation, which could be regulated by a mechanism involving a cross talk between AST, MG, and oligodendrocyte progenitors (OPC).


Asunto(s)
Anemia Ferropénica/metabolismo , Astrocitos/metabolismo , Hierro/metabolismo , Microglía/metabolismo , Oligodendroglía/metabolismo , Animales , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Homeostasis/fisiología , Vaina de Mielina/metabolismo , Ratas , Ratas Wistar
15.
Mol Neurobiol ; 55(11): 8637-8650, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29582397

RESUMEN

The specific roles of Notch in progressive adulthood neurodegenerative disorders have begun to be unraveled in recent years. A number of independent studies have shown significant increases of Notch expression in brains from patients at later stages of sporadic Alzheimer's disease (AD). However, the impact of Notch canonical signaling activation in the pathophysiology of AD is still elusive. To further investigate this issue, 2-month-old wild-type (WT) and hemizygous McGill-R-Thy1-APP rats (Tg(+/-)) were injected in CA1 with lentiviral particles (LVP) expressing the transcriptionally active fragment of Notch, known as Notch Intracellular Domain (NICD), (LVP-NICD), or control lentivirus particles (LVP-C). The Tg(+/-) rat model captures presymptomatic aspects of the AD pathology, including intraneuronal amyloid beta (Aß) accumulation and early cognitive deficits. Seven months after LVP administration, Morris water maze test was performed, and brains isolated for biochemical and histological analysis. Our results showed a learning impairment and a worsening of spatial memory in LVP-NICD- as compared to LVP-C-injected Tg(+/-) rats. In addition, immuno histochemistry, ELISA multiplex, Western blot, RT-qPCR, and 1H-NMR spectrometry of cerebrospinal fluid (CSF) indicated that chronic expression of NICD promoted hippocampal vessel thickening with accumulation of Aß in brain microvasculature, alteration of blood-brain barrier (BBB) permeability, and a decrease of CSF glucose levels. These findings suggest that, in the presence of early Aß pathology, expression of NICD may contribute to the development of microvascular abnormalities, altering glucose transport at the BBB with impact on early decline of spatial learning and memory.


Asunto(s)
Enfermedad de Alzheimer/patología , Vasos Sanguíneos/patología , Glucosa/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/patología , Receptores Notch/química , Receptores Notch/metabolismo , Memoria Espacial , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Vectores Genéticos/metabolismo , Células HEK293 , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Inflamación/patología , Lentivirus/genética , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Microvasos/patología , Dominios Proteicos , Espectroscopía de Protones por Resonancia Magnética , Ratas Transgénicas , Ratas Wistar
16.
Neuro Oncol ; 9(3): 245-58, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17522335

RESUMEN

Expression of the immune-stimulatory molecule Fms-like tyrosine kinase 3 ligand (Flt3L) and the conditional cytotoxic enzyme herpes simplex virus type 1 thymidine kinase (HSV1-TK) provides long-term immune-mediated survival of large glioblastoma multiforme (GBM) models in rodents. A limitation for predictive testing of novel antiglioma therapies has been the lack of a glioma model in a large animal. Dogs bearing spontaneous GBM may constitute an attractive large-animal model for GBM, which so far has remained underappreciated. In preparation for a clinical trial in dogs bearing spontaneous GBMs, we tested and optimized adenovirus-mediated transgene expression with negligible toxicity in the dog brain in vivo and in canine J3T glioma cells. Expression of the marker gene beta-galactosidase (beta-Gal) was higher when driven by the murine (m) than the human (h) cytomegalovirus (CMV) promoter in the dog brain in vivo, without enhanced inflammation. In the canine brain, beta-Gal was expressed mostly in astrocytes. beta-Gal activity in J3T cells was also higher with the mCMV than the hCMV promoter driving tetracycline-dependent (TetON) transgene expression within high-capacity adenovirus vectors (HC-Ads). Dog glioma cells were efficiently transduced by HC-Ads expressing mCMV-driven HSV1-TK, which induced 90% reduction in cell viability in the presence of ganciclovir. J3T cells were also effectively transduced with HC-Ads expressing Flt3L under the control of the regulatable TetON promoter system, and as predicted, Flt3L release was stringently inducer dependent. HC-Ads encoding therapeutic transgenes under the control of regulatory sequences driven by the mCMV promoter are excellent vectors for the treatment of spontaneous GBM in dogs, which constitute an ideal preclinical animal model.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/genética , Encéfalo/fisiología , Terapia Genética/métodos , Glioma/genética , Regiones Promotoras Genéticas , Transgenes/fisiología , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Citomegalovirus/genética , Perros , Ensayo de Inmunoadsorción Enzimática , Ingeniería Genética/métodos , Vectores Genéticos , Glioma/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Confocal , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transducción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
17.
Cancer Res ; 65(16): 7194-204, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16103070

RESUMEN

In spite of preclinical efficacy and recent randomized, controlled studies with adenoviral vectors expressing herpes simplex virus-1 thymidine kinase (HSV1-TK) showing statistically significant increases in survival, most clinical trials using single therapies have failed to provide major therapeutic breakthroughs. Because glioma is a disease with dismal prognosis and rapid progression, it is an attractive target for gene therapy. Preclinical models using microscopic brain tumor models (e.g., < or =0.3 mm3) may not reflect the pathophysiology and progression of large human tumors. To overcome some of these limitations, we developed a syngeneic large brain tumor model. In this model, administration of single therapeutic modalities, either conditional cytotoxicity or immunostimulation, fail. However, when various immunostimulatory therapies were delivered in combination with conditional cytotoxicity (HSV1-TK), only the combined delivery of fms-like tyrosine kinase ligand (Flt3L) and HSV1-TK significantly prolonged the survival of large tumor-bearing animals (> or =80%; P < or = 0.005). When either macrophages or CD4+ cells were depleted before administration of viral therapy, TK + Flt3L therapy failed to prolong survival. Meanwhile, depletion of CD8+ cells or natural killer cells did not affect TK + Flt3L efficacy. Spinal cord of animals surviving 6 months after TK + Flt3L were evaluated for the presence of autoimmune lesions. Whereas macrophages were present within the corticospinal tract and low levels of T-cell infiltration were detected, these effects are not indicative of an overt autoimmune disorder. We propose that combined Flt3L and HSV1-TK adenoviral-mediated gene therapy may provide an effective antiglioma treatment with increased efficacy in clinical trials of glioma.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Neoplasias Encefálicas/terapia , Ciclosporina/farmacología , Terapia Genética/métodos , Glioma/terapia , Adenovirus Humanos/genética , Secuencia de Aminoácidos , Animales , Autoinmunidad , Encéfalo/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Terapia Combinada , Vectores Genéticos/genética , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/genética , Humanos , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Ratas , Ratas Endogámicas Lew , Médula Espinal/inmunología , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
18.
Curr Gene Ther ; 5(6): 559-72, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16457646

RESUMEN

Pituitary tumors are the most common primary intracranial neoplasms. Although most pituitary tumors are considered typically benign, others can cause severe and progressive disease. The principal aims of pituitary tumor treatment are the elimination or reduction of the tumor mass, normalization of hormone secretion and preservation of remaining pituitary function. In spite of major advances in the therapy of pituitary tumors, for some of the most difficult tumors, current therapies that include medical, surgical and radiotherapeutic methods are often unsatisfactory and there is a need to develop new treatment strategies. Gene therapy, which uses nucleic acids as drugs, has emerged as an attractive therapeutic option for the treatment of pituitary tumors that do not respond to classical treatment strategies if the patients become intolerant to the therapy. The development of animal models for pituitary tumors and hormone hypersecretion has proven to be critical for the implementation of novel treatment strategies and gene therapy approaches. Preclinical trials using several gene therapy approaches for the treatment of anterior pituitary diseases have been successfully implemented. Several issues need to be addressed before clinical implementation becomes a reality, including the development of more effective and safer viral vectors, uncovering novel therapeutic targets and development of targeted expression of therapeutic transgenes. With the development of efficient gene delivery vectors allowing long-term transgene expression with minimal toxicity, gene therapy will become one of the most promising approaches for treating pituitary adenomas.


Asunto(s)
Adenoma/terapia , Terapia Genética/métodos , Neoplasias Hipofisarias/terapia , Adenoma/epidemiología , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Marcación de Gen , Vectores Genéticos/uso terapéutico , Hormonas/metabolismo , Humanos , Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/epidemiología , Neoplasias Hipofisarias/metabolismo
19.
Clin Cancer Res ; 20(6): 1555-1565, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24501391

RESUMEN

PURPOSE: Glioblastoma multiforme is the most common primary brain cancer in adults. Chemotherapy with temozolomide (TMZ) significantly prolongs the survival of patients with glioblastoma multiforme. However, the three-year survival is still approximately 5%. Herein, we combined intratumoral administration of an adenoviral vector expressing Flt3L (Ad-Flt3L) with systemic temozolomide to assess its impact on therapeutic efficacy. EXPERIMENTAL DESIGN: Wild-type or immunodeficient mice bearing intracranial glioblastoma multiforme or metastatic melanoma were treated with an intratumoral injection of Ad-Flt3L alone or in combination with the conditionally cytotoxic enzyme thymidine kinase (Ad-TK), followed by systemic administration of ganciclovir and temozolomide. We monitored survival and measured the tumor-infiltrating immune cells. RESULTS: Although treatment with temozolomide alone led to a small improvement in median survival, when used in combination with gene therapy-mediated immunotherapy, it significantly increased the survival of tumor-bearing mice. The antitumor effect was further enhanced by concomitant intratumoral administration of Ad-TK, leading to 50% to 70% long-term survival in all tumor models. Although temozolomide reduced the content of T cells in the tumor, this did not affect the therapeutic efficacy. The antitumor effect of Ad-Flt3L+Ad-TK+TMZ required an intact immune system because the treatment failed when administered to knock out mice that lacked lymphocytes or dendritic cells. CONCLUSIONS: Our results challenge the notion that chemotherapy leads to a state of immune-suppression which impairs the ability of the immune system to mount an effective antitumor response. Our work indicates that temozolomide does not inhibit antitumor immunity and supports its clinical implementation in combination with immune-mediated therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/patología , Dacarbazina/análogos & derivados , Glioblastoma/patología , Inmunoterapia/métodos , Adenoviridae , Animales , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Temozolomida , Timidina Quinasa/genética , Timidina Quinasa/inmunología
20.
Front Cell Neurosci ; 7: 53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23641196

RESUMEN

Neuroinflammation has received increased attention as a target for putative neuroprotective therapies in Parkinson's Disease (PD). Two prototypic pro-inflammatory cytokines interleukin-1ß (IL-1) and tumor necrosis factor-α (TNF) have been implicated as main effectors of the functional consequences of neuroinflammation on neurodegeneration in PD models. In this review, we describe that the functional interaction between these cytokines in the brain differs from the periphery (e.g., their expression is not induced by each other) and present data showing predominantly a toxic effect of these cytokines when expressed at high doses and for a sustained period of time in the substantia nigra pars compacta (SN). In addition, we highlight opposite evidence showing protective effects of these two main cytokines when conditions of duration, amount of expression or state of activation of the target or neighboring cells are changed. Furthermore, we discuss these results in the frame of previous disappointing results from anti-TNF-α clinical trials against Multiple Sclerosis, another neurodegenerative disease with a clear neuroinflammatory component. In conclusion, we hypothesize that the available evidence suggests that the duration and dose of IL-1ß or TNF-α expression is crucial to predict their functional effect on the SN. Since these parameters are not amenable for measurement in the SN of PD patients, we call for an in-depth analysis to identify downstream mediators that could be common to the toxic (and not the protective) effects of these cytokines in the SN. This strategy could spare the possible neuroprotective effect of these cytokines operative in the patient at the time of treatment, increasing the probability of efficacy in a clinical setting. Alternatively, receptor-specific agonists or antagonists could also provide a way to circumvent undesired effects of general anti-inflammatory or specific anti-IL-1ß or TNF-α therapies against PD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA