Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Clin Transl Immunology ; 11(1): e1360, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35035955

RESUMEN

OBJECTIVES: We previously described the Phase I-II evaluation of SARS-CoV-2 recombinant protein candidate vaccine, CoV2-PreS-dTM, with AF03- or AS03-adjuvant systems (ClinicalTrials.gov, NCT04537208). Here, we further characterise the cellular immunogenicity profile of this vaccine candidate using a whole-blood secretion assay in parallel to intracellular cytokine staining (ICS) of cryopreserved peripheral blood mononuclear cells (PBMCs). METHODS: A randomly allocated subset of 90 healthy, SARS-CoV-2-seronegative adults aged ≥ 18 years who had received (random allocation) one or two separate injections (on study day [D]1 and D22) of saline placebo or CoV2-PreS-dTM formulated with AS03 or AF03 were included. Cytokine secretion was assessed using a TruCulture® whole-blood stimulation system in combination with multiplex bead array, and intracellular cytokine profiles were evaluated on thawed PBMCs following ex vivo stimulation with recombinant S protein at pre-vaccination (D1), post-dose 1 (D22) and post-dose 2 (D36). RESULTS: Both methods detected similar vaccine-induced responses after the first and second doses. We observed a Th1 bias (Th1/Th2 ratio > 1.0) for most treatment groups when analysed in whole blood, mainly characterised by increased IFN-γ, IL-2 and TNF-α secretion. Among participants aged ≥ 50 years, the Th1/Th2 ratio was higher for those who received vaccine candidate with AS03 versus AF03 adjuvant. ICS revealed that this higher Th1/Th2 ratio resulted from higher levels of IFN-γ expression and that the vaccine induced polyfunctional CD4+ T cells. CONCLUSIONS: The whole-blood cytokine secretion assay is a high-throughput alternative for assessing the quantity and character of vaccine-induced cellular responses.

2.
Front Immunol ; 12: 738955, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603321

RESUMEN

There is increasing evidence that lung-resident memory T and B cells play a critical role in protecting against respiratory reinfection. With a unique transcriptional and phenotypic profile, resident memory lymphocytes are maintained in a quiescent state, constantly surveying the lung for microbial intruders. Upon reactivation with cognate antigen, these cells provide rapid effector function to enhance immunity and prevent infection. Immunization strategies designed to induce their formation, alongside novel techniques enabling their detection, have the potential to accelerate and transform vaccine development. Despite most data originating from murine studies, this review will discuss recent insights into the generation, maintenance and characterisation of pulmonary resident memory lymphocytes in the context of respiratory infection and vaccination using recent findings from human and non-human primate studies.


Asunto(s)
Infecciones Bacterianas/prevención & control , Memoria Inmunológica , Pulmón/inmunología , Células B de Memoria/inmunología , Células T de Memoria/inmunología , Infecciones del Sistema Respiratorio/inmunología , Virosis/prevención & control , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Interacciones Huésped-Patógeno , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/virología , Células B de Memoria/metabolismo , Células B de Memoria/microbiología , Células B de Memoria/virología , Células T de Memoria/metabolismo , Células T de Memoria/microbiología , Células T de Memoria/virología , Fenotipo , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/prevención & control , Infecciones del Sistema Respiratorio/virología , Vacunación , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Virosis/inmunología , Virosis/metabolismo , Virosis/microbiología
3.
iScience ; 24(9): 102970, 2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34471863

RESUMEN

The elderly are an important target for influenza vaccination, and the determination of factors that underlie immune responsiveness is clinically valuable. We evaluated the immune and metabolic profiles of 205 elderly Singaporeans administered with Vaxigrip. Despite high seroprotection rates, we observed heterogeneity in the response. We stratified the cohort into complete (CR) or incomplete responders (IR), where IR exhibited signs of accelerated T cell aging. We found a higher upregulation of genes associated with the B-cell endoplasmic-reticulum stress response in CR, where XBP-1 acts as a key upstream regulator. B-cells from IR were incapable of matching the level of XBP-1 upregulation observed in CR after inducing ER stress with tunicamycin in vitro. Metabolic signatures also distinguished CR and IR - as CR presented with a greater diversity of bile acids. Our findings suggest that the ER-stress pathway activation could improve influenza vaccination in the elderly.

4.
Vaccine ; 34(36): 4304-12, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27402566

RESUMEN

Small-molecule adjuvants that boost and direct adaptive immunity provide a powerful means to increase the effectiveness of vaccines. Through rational design several novel imidazoquinoline and oxoadenine TLR7/8 agonists, each with unique molecular modifications, were synthesized and assessed for their ability to augment adaptive immunity. All agonists bound human TLR7 and TLR8 and induced maturation of both human mDCs and pDCs. All agonists prompted production of type I interferon and/or proinflammatory cytokines, albeit with varying potencies. In most in vitro assays, the oxoadenine class of agonists proved more potent than the imidazoquinolines. Therefore, an optimized oxoadenine TLR7/8 agonist that demonstrated maximal activity in the in vitro assays was further assessed in a vaccine study with the CRM197 antigen in a porcine model. Antigen-specific antibody production was greatly enhanced in a dose dependent manner, with antibody titers increased 800-fold compared to titers from pigs vaccinated with the non-adjuvanted vaccine. Moreover, pigs vaccinated with antigen containing the highest dose of adjuvant promoted a 13-fold increase in the percentage of antigen-specific CD3(+)/CD8(+) T cells over pigs vaccinated with antigen alone. Together this work demonstrates the promise of these novel TLR7/8 agonists as effective human vaccine adjuvants.


Asunto(s)
Inmunidad Adaptativa , Adyuvantes Inmunológicos , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Vacunas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/síntesis química , Adyuvantes Inmunológicos/química , Animales , Linfocitos T CD8-positivos/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Humanos , Imidazoles/síntesis química , Imidazoles/química , Imidazoles/inmunología , Imidazoles/farmacología , Piridinas/síntesis química , Piridinas/inmunología , Piridinas/farmacología , Quinolinas/síntesis química , Quinolinas/química , Quinolinas/inmunología , Quinolinas/farmacología , Porcinos , Vacunas/administración & dosificación
5.
J Immunol Methods ; 424: 64-79, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26002154

RESUMEN

Antibody-mediated capture of amyloid-beta (Aß) in peripheral blood was identified as an attractive strategy to eliminate cerebral toxic amyloid in Alzheimer's disease (AD) patients and murine models. Alternatively, defective capacity of peripheral monocytes to engulf Aß was reported in individuals with AD. In this report, we developed different approaches to investigate cellular uptake and phagocytosis of Aß, and to examine how two immunological devices--an immunostimulatory Adjuvant System and different amyloid specific antibodies--may affect these biological events. Between one and thirteen months of age, APPswe X PS1.M146V (TASTPM) AD model mice had decreasing concentrations of Aß in their plasma. In contrast, the proportion of blood monocytes containing Aß tended to increase with age. Importantly, the TLR-agonist containing Adjuvant System AS01B primed monocytes to promote de novo Aß uptake capacity, particularly in the presence of anti-Aß antibodies. Biochemical experiments demonstrated that cells achieved Aß uptake and internalization followed by Aß degradation via mechanisms that required effective actin polymerization and proteolytic enzymes such as insulin-degrading enzyme. We further demonstrated that both Aß-specific monoclonal antibodies and plasma from Aß-immunized mice enhanced the phagocytosis of 1 µm Aß-coated particles. Together, our data highlight a new biomarker testing to follow amyloid clearance within the blood and a mechanism of Aß uptake by peripheral monocytes in the context of active or passive immunization, and emphasize on novel approaches to investigate this phenomenon.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Fagocitosis/inmunología , Actinas/metabolismo , Adyuvantes Inmunológicos , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/sangre , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Combinación de Medicamentos , Inmunofenotipificación , Inmunoterapia , Lípido A/administración & dosificación , Lípido A/análogos & derivados , Lípido A/inmunología , Ratones , Ratones Transgénicos , Multimerización de Proteína , Proteolisis , Saponinas/administración & dosificación , Saponinas/inmunología , Vacunación
6.
Proc Natl Acad Sci U S A ; 110(5): 1941-6, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23322736

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia worldwide. The pathogenesis of this neurodegenerative disease, currently without curative treatment, is associated with the accumulation of amyloid ß (Aß) in brain parenchyma and cerebral vasculature. AD patients are unable to clear this toxic peptide, leading to Aß accumulation in their brains and, presumably, the pathology associated with this devastating disease. Compounds that stimulate the immune system to clear Aß may therefore have great therapeutic potential in AD patients. Monophosphoryl lipid A (MPL) is an LPS-derived Toll-like receptor 4 agonist that exhibits unique immunomodulatory properties at doses that are nonpyrogenic. We show here that repeated systemic injections of MPL, but not LPS, significantly improved AD-related pathology in APP(swe)/PS1 mice. MPL treatment led to a significant reduction in Aß load in the brain of these mice, as well as enhanced cognitive function. MPL induced a potent phagocytic response by microglia while triggering a moderate inflammatory reaction. Our data suggest that the Toll-like receptor 4 agonist MPL may be a treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Encéfalo/efectos de los fármacos , Lípido A/análogos & derivados , Receptor Toll-Like 4/agonistas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inmunidad Innata/efectos de los fármacos , Ligandos , Lípido A/administración & dosificación , Lípido A/uso terapéutico , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microscopía Fluorescente , Fagocitosis/efectos de los fármacos , Presenilina-1/genética , Presenilina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 4/metabolismo
8.
PLoS One ; 6(6): e21065, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731648

RESUMEN

The INhibitor of Growth tumor suppressors (ING1-ING5) affect aging, apoptosis, DNA repair and tumorigenesis. Plant homeodomains (PHD) of ING proteins bind histones in a methylation-sensitive manner to regulate chromatin structure. ING1 and ING2 contain a polybasic region (PBR) adjacent to their PHDs that binds stress-inducible phosphatidylinositol monophosphate (PtIn-MP) signaling lipids to activate these INGs. ING1 induces apoptosis independently of p53 but other studies suggest proapoptotic interdependence of ING1 and p53 leaving their functional relationship unclear. Here we identify a novel ubiquitin-binding domain (UBD) that overlaps with the PBR of ING1 and shows similarity to previously described UBDs involved in DNA damage responses. The ING1 UBD binds ubiquitin with high affinity (K(d)∼100 nM) and ubiquitin competes with PtIn-MPs for ING1 binding. ING1 expression stabilized wild-type, but not mutant p53 in an MDM2-independent manner and knockdown of endogenous ING1 depressed p53 levels in a transcription-independent manner. ING1 stabilized unmodified and six multimonoubiquitinated forms of wild-type p53 that were also seen upon DNA damage, but not p53 mutants lacking the six known sites of ubiquitination. We also find that ING1 physically interacts with herpesvirus-associated ubiquitin-specific protease (HAUSP), a p53 and MDM2 deubiquitinase (DUB), and knockdown of HAUSP blocks the ability of ING1 to stabilize p53. These data link lipid stress signaling to ubiquitin-mediated proteasomal degradation through the PBR/UBD of ING1 and further indicate that ING1 stabilizes p53 by inhibiting polyubiquitination of multimonoubiquitinated forms via interaction with and colocalization of the HAUSP-deubiquitinase with p53.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Poliubiquitina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación , Línea Celular Tumoral , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular/química , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Fosfolípidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Supresoras de Tumor/química , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
9.
Mol Ther ; 19(10): 1793-801, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21505426

RESUMEN

Glioblastoma multiforme (GBM) is a primary brain tumor with a median survival of 14.6 months postdiagnosis. The infiltrative nature of GBM prevents complete resection and residual brain tumor cells give rise to recurrent GBM, a hallmark of this disease. Recurrent GBMs are known to harbor numerous mutations/gene rearrangements when compared to the primary tumor, which leads to the potential expression of novel proteins that could serve as tumor neoantigens. We have developed a combined immune-based gene therapeutic approach for GBM using adenoviral (Ads) mediated gene delivery of Herpes Simplex Virus Type 1-thymidine kinase (TK) into the tumor mass to induce tumor cells' death combined with an adenovirus expressing fms-like tyrosine kinase 3 ligand (Flt3L) to recruit dendritic cells (DCs) into the tumor microenvironment. This leads to the induction of specific anti-brain tumor immunity and immunological memory. In a model of GBM recurrence, we demonstrate that Flt3L/TK mediated immunological memory is capable of recognizing brain tumor neoantigens absent from the original treated tumor. These data demonstrate that the Flt3L/TK gene therapeutic approach can induce systemic immunological memory capable of recognizing a brain tumor neoantigen in a model of recurrent GBM.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Timidina Quinasa/genética , Tirosina Quinasa 3 Similar a fms/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Linfocitos T/inmunología
10.
Proc Natl Acad Sci U S A ; 107(32): 14443-8, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660723

RESUMEN

Soluble antigens diffuse out of the brain and can thus stimulate a systemic immune response, whereas particulate antigens (from infectious agents or tumor cells) remain within brain tissue, thus failing to stimulate a systemic immune response. Immune privilege describes how the immune system responds to particulate antigens localized selectively within the brain parenchyma. We believe this immune privilege is caused by the absence of antigen presenting dendritic cells from the brain. We tested the prediction that expression of fms-like tyrosine kinase ligand 3 (Flt3L) in the brain will recruit dendritic cells and induce a systemic immune response against exogenous influenza hemagglutinin in BALB/c mice. Coexpression of Flt3L with HA in the brain parenchyma induced a robust systemic anti-HA immune response, and a small response against myelin basic protein and proteolipid protein epitopes. Depletion of CD4(+)CD25+ regulatory T cells (Tregs) enhanced both responses. To investigate the autoimmune impact of these immune responses, we characterized the neuropathological and behavioral consequences of intraparenchymal injections of Flt3L and HA in BALB/c and C57BL/6 mice. T cell infiltration in the forebrain was time and strain dependent, and increased in animals treated with Flt3L and depleted of Tregs; however, we failed to detect widespread defects in myelination throughout the forebrain or spinal cord. Results of behavioral tests were all normal. These results demonstrate that Flt3L overcomes the brain's immune privilege, and supports the clinical development of Flt3L as an adjuvant to stimulate clinically effective immune responses against brain neo-antigens, for example, those associated with brain tumors.


Asunto(s)
Encéfalo/inmunología , Sistema Inmunológico/inmunología , Tirosina Quinasa 3 Similar a fms/inmunología , Adyuvantes Inmunológicos , Animales , Antígenos/inmunología , Células Dendríticas/inmunología , Hemaglutininas/inmunología , Inmunidad , Ligandos , Ratones , Ratones Endogámicos BALB C , Prosencéfalo/inmunología , Médula Espinal/inmunología , Linfocitos T Reguladores/inmunología
11.
J Immunol ; 183(10): 6186-97, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19864596

RESUMEN

Adjuvant System 04 (AS04) combines the TLR4 agonist MPL (3-O-desacyl-4'-monophosphoryl lipid A) and aluminum salt. It is a new generation TLR-based adjuvant licensed for use in human vaccines. One of these vaccines, the human papillomavirus (HPV) vaccine Cervarix, is used in this study to elucidate the mechanism of action of AS04 in human cells and in mice. The adjuvant activity of AS04 was found to be strictly dependent on AS04 and the HPV Ags being injected at the same i.m. site within 24 h of each other. During this period, AS04 transiently induced local NF-kappaB activity and cytokine production. This led to an increased number of activated Ag-loaded dendritic cells and monocytes in the lymph node draining the injection site, which further increased the activation of Ag-specific T cells. AS04 was also found to directly stimulate those APCs in vitro but not directly stimulate CD4(+) T or B lymphocytes. These AS04-induced innate responses were primarily due to MPL. Aluminum salt appeared not to synergize with or inhibit MPL, but rather it prolonged the cytokine responses to MPL at the injection site. Altogether these results support a model in which the addition of MPL to aluminum salt enhances the vaccine response by rapidly triggering a local cytokine response leading to an optimal activation of APCs. The transient and confined nature of these responses provides further supporting evidence for the favorable safety profile of AS04 adjuvanted vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Hidróxido de Aluminio/farmacología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Lípido A/análogos & derivados , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/inmunología , Receptor Toll-Like 4/agonistas , Animales , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Línea Celular , Citocinas/efectos de los fármacos , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 18/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Lípido A/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/agonistas , FN-kappa B/inmunología , FN-kappa B/metabolismo , Ovalbúmina/inmunología , Infecciones por Papillomavirus/virología , Receptor Toll-Like 4/inmunología , Transfección
12.
PLoS One ; 4(6): e5867, 2009 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-19517016

RESUMEN

BACKGROUND: The quaking viable (qk(v)) mice have uncompacted myelin in their central and peripheral nervous system (CNS, PNS). The qk gene encodes 3 major alternatively spliced isoforms that contain unique sequence at their C-terminus dictating their cellular localization. QKI-5 is a nuclear isoform, whereas QKI-6 and QKI-7 are cytoplasmic isoforms. The qk(v) mice harbor an enhancer/promoter deletion that prevents the expression of isoforms QKI-6 and QKI-7 in myelinating cells resulting in a dysmyelination phenotype. It was shown that QKI regulates the differentiation of oligodendrocytes, the myelinating cells of the CNS, however, little is known about the role of the QKI proteins, or RNA binding proteins in PNS myelination. METHODOLOGY/PRINCIPAL FINDINGS: To define the role of the QKI proteins in PNS myelination, we ectopically expressed QKI-6 and QKI-7 in primary rat Schwann cell/neuron from dorsal root ganglia cocultures. We show that the QKI isoforms blocked proliferation and promoted Schwann cell differentiation and myelination. In addition, these events were coordinated with elevated proteins levels of p27(KIP1) and myelin basic protein (MBP), markers of Schwann cell differentiation. QKI-6 and QKI-7 expressing co-cultures contained myelinated fibers that had directionality and contained significantly thicker myelin, as assessed by electron microscopy. Moreover, QKI-deficient Schwann cells had reduced levels of MBP, p27(KIP1) and Krox-20 mRNAs, as assessed by quantitative RT-PCR. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that the QKI-6 and QKI-7 RNA binding proteins are positive regulators of PNS myelination and show that the QKI RNA binding proteins play a key role in Schwann cell differentiation and myelination.


Asunto(s)
Vaina de Mielina/química , Proteínas de Unión al ARN/genética , Células de Schwann/metabolismo , Empalme Alternativo , Animales , Núcleo Celular/metabolismo , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citoplasma/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Ganglios Espinales/metabolismo , Ratones , Proteína Básica de Mielina/metabolismo , Sistema Nervioso Periférico/fisiología , Isoformas de Proteínas , Proteínas de Unión al ARN/química , Ratas
13.
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
14.
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
15.
Mol Ther ; 15(12): 2154-63, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17895861

RESUMEN

The main challenge of gene therapy is to provide long-term, efficient transgene expression. Long-term transgene expression from first generation adenoviral vectors (Advs) delivered to the central nervous system (CNS) is elicited in animals not previously exposed to adenovirus (Ad). However, upon systemic immunization against Ad, transgene expression from a first generation Adv is abolished. High-capacity Advs (HC-Advs) provide sustained very long-term transgene expression in the brain, even in animals pre-immunized against Ad. In this study, we tested the hypothesis that a HC-Adv in the brain would allow for long-term transgene expression, for up to 1 year, in the brain of mice immunized against Ad prior to delivery of the vector to the striatum. In naïve animals, the expression of beta-galactosidase from Adv or HC-Adv was sustained for 1 year. In animals immunized prior to vector delivery, expression from a first generation Adv was abolished. These results point to a very long-term HC-Adv-mediated transgene expression in the brain, even in animals that had been immunized systemically against Ad before the delivery of HC-Adv into the brain. This study therefore indicates the utility of HC-Adv as a powerful gene therapy vector for chronic neurological disorders, even in patients who had been pre-exposed to Ad prior to gene therapy.


Asunto(s)
Adenoviridae/inmunología , Encéfalo/metabolismo , Vectores Genéticos , Adenoviridae/genética , Animales , Secuencia de Bases , Encéfalo/inmunología , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Ratones , Ratones Transgénicos , Pruebas de Neutralización , Reacción en Cadena de la Polimerasa
16.
J Biol Chem ; 280(46): 38639-47, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16179349

RESUMEN

The breast tumor kinase (BRK) is a growth promoting non-receptor tyrosine kinase overexpressed in the majority of human breast tumors. BRK is known to potentiate the epidermal growth factor (EGF) response in these cells. Although BRK is known to phosphorylate the RNA-binding protein Sam68, the specific tyrosines phosphorylated and the exact role of this phosphorylation remains unknown. Herein, we have generated Sam68 phospho-specific antibodies against C-terminal phosphorylated tyrosine residues within the Sam68 nuclear localization signal. We show that BRK phosphorylates Sam68 on all three tyrosines in the nuclear localization signal. By indirect immunofluorescence we observed that BRK and EGF treatment not only phosphorylates Sam68 but also induces its relocalization. Tyrosine 440 was identified as a principal modulator of Sam68 localization and this site was phosphorylated in response to EGF treatment in human breast tumor cell lines. Moreover, this phosphorylation event was inhibited by BRK small interfering RNA treatment, consistent with Sam68 being a physiological substrate of BRK downstream of the EGF receptor in breast cancer cells. Finally, we observed that Sam68 suppressed BRK-induced cell proliferation, suggesting that Sam68 does indeed contain anti-proliferative properties that may be neutralized in breast cancer cells by phosphorylation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Neoplasias de la Mama/patología , Proteínas de Unión al ADN/química , Proteínas de Neoplasias/metabolismo , Neoplasias/patología , Fosfoproteínas/química , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Unión al ARN/química , Tirosina/química , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Astrocitos/citología , Encéfalo/metabolismo , Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Vectores Genéticos , Células HeLa , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Datos de Secuencia Molecular , Péptidos/química , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/metabolismo , Interferencia de ARN , Ratas , Homología de Secuencia de Aminoácido , Transducción de Señal
17.
RNA Biol ; 2(2): 37-40, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17132940

RESUMEN

The quaking viable (qk(v)) mice have attracted attention because of their characteristic tremor caused by their dysmyelination. In the central nervous system, qk(v) mice fail to develop mature myelinating oligodendrocytes and display uncompacted myelin. The genetic defect in the qk(v) mice prevents the proper expression of alternatively spliced KH-type QKI RNA binding proteins. Thus qk(v) mice provide a unique animal model linking RNA binding proteins to defects in oligodendrocyte cell fate and myelination. The fact that QKI proteins are modified post-translationally makes them Signal Transduction Activiators of RNA (STAR) proteins. We have used a gain-of-function approach with the ectopic expression of the separate QKI isoforms using adenoviruses and retroviruses to determine their separate roles in cell fate and myelination. Herein, we discuss the recent advances in characterizing the QKI KH-type proteins as glial cell fate and myelin egulators.


Asunto(s)
Vaina de Mielina/fisiología , Neuroglía/citología , Proteínas de Unión al ARN/fisiología , Animales , Diferenciación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Viabilidad Fetal , Ratones , Ratones Quaking , Modelos Biológicos , Proteína Básica de Mielina/metabolismo , Oligodendroglía/citología , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/metabolismo
18.
Nat Neurosci ; 8(1): 27-33, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15568022

RESUMEN

The quaking (Qk) locus expresses a family of RNA binding proteins, and the expression of several alternatively spliced isoforms coincides with the development of oligodendrocytes and the onset of myelination. Quaking viable (Qk(v)) mice harboring an autosomal recessive mutation in this locus have uncompacted myelin in the central nervous system owing to the inability of oligodendrocytes to properly mature. Here we show that the expression of two QKI isoforms, absent from oligodendrocytes of Qk(v) mice, induces cell cycle arrest of primary rat oligodendrocyte progenitor cells and differentiation into oligodendrocytes. Injection of retroviruses expressing QKI into the telencephalon of mouse embryos induced differentiation and migration of multipotential neural progenitor cells into mature oligodendrocytes localized in the corpus callosum. The mRNA encoding the cyclin-dependent kinase (CDK)-inhibitor p27(Kip1) was bound and stabilized by QKI, leading to an increased accumulation of p27(Kip1) protein in oligodendrocytes. Our findings demonstrate that QKI is upstream of p27(Kip1) during oligodendrocyte differentiation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Genes Recesivos , Mutación , Oligodendroglía/citología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Ciclo Celular , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Cuerpo Calloso/citología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Ratones , Ratones Quaking , Mutación/fisiología , Proteína Básica de Mielina/metabolismo , Neuronas/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estabilidad del ARN , Ratas , Células Madre/citología , Células Madre/fisiología , Telencéfalo/embriología
19.
Neuron ; 36(5): 815-29, 2002 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-12467586

RESUMEN

Quaking viable (qk(v)) mice fail to properly compact myelin in their central nervous systems. Although the defect in the qk(v) mice involves a mutation affecting the expression of the alternatively spliced qk gene products, their roles in myelination are unknown. We show that the QKI RNA binding proteins regulate the nuclear export of MBP mRNAs. Disruption of the QKI nucleocytoplasmic equilibrium in oligodendrocytes results in nuclear and perikaryal retention of the MBP mRNAs and lack of export to cytoplasmic processes, as it occurs in qk(v) mice. MBP mRNA export defect leads to a reduction in the MBP levels and their improper cellular targeting to the periphery. Our findings suggest that QKI participates in myelination by regulating the mRNA export of key protein components.


Asunto(s)
Proteína Básica de Mielina/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3'/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Empalme Alternativo , Animales , Sitios de Unión , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Enfermedades Desmielinizantes/fisiopatología , Exones/genética , Humanos , Ratones , Ratones Quaking , Proteína Básica de Mielina/genética , Oligodendroglía/citología , Oligodendroglía/fisiología , Mutación Puntual , Unión Proteica , Isoformas de Proteínas/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA