Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(5): 1941-6, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23322736

RESUMO

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.


Assuntos
Doença de Alzheimer/prevenção & controle , Encéfalo/efeitos dos fármacos , Lipídeo A/análogos & derivados , Receptor 4 Toll-Like/agonistas , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Imunidade Inata/efeitos dos fármacos , Ligantes , Lipídeo A/administração & dosagem , Lipídeo A/uso terapêutico , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microscopia de Fluorescência , Fagocitose/efeitos dos fármacos , Presenilina-1/genética , Presenilina-1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 4 Toll-Like/metabolismo
2.
Proc Natl Acad Sci U S A ; 107(32): 14443-8, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660723

RESUMO

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.


Assuntos
Encéfalo/imunologia , Sistema Imunitário/imunologia , Tirosina Quinase 3 Semelhante a fms/imunologia , Adjuvantes Imunológicos , Animais , Antígenos/imunologia , Células Dendríticas/imunologia , Hemaglutininas/imunologia , Imunidade , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Prosencéfalo/imunologia , Medula Espinal/imunologia , Linfócitos T Reguladores/imunologia
3.
Mol Ther ; 19(10): 1793-801, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21505426

RESUMO

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.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Timidina Quinase/genética , Tirosina Quinase 3 Semelhante a fms/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Memória Imunológica , Interferon gama/metabolismo , Linfócitos T/imunologia
4.
Clin Transl Immunology ; 11(1): e1360, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35035955

RESUMO

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.

5.
J Immunol ; 183(10): 6186-97, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864596

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Hidróxido de Alumínio/farmacologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Lipídeo A/análogos & derivados , Infecções por Papillomavirus/imunologia , Vacinas contra Papillomavirus/imunologia , Receptor 4 Toll-Like/agonistas , Animais , Antígeno B7-2/imunologia , Antígeno B7-2/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Linhagem Celular , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Feminino , Papillomavirus Humano 16/imunologia , Papillomavirus Humano 18/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Lipídeo A/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , NF-kappa B/agonistas , NF-kappa B/imunologia , NF-kappa B/metabolismo , Ovalbumina/imunologia , Infecções por Papillomavirus/virologia , Receptor 4 Toll-Like/imunologia , Transfecção
6.
Front Immunol ; 12: 738955, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603321

RESUMO

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.


Assuntos
Infecções Bacterianas/prevenção & controle , Memória Imunológica , Pulmão/imunologia , Células B de Memória/imunologia , Células T de Memória/imunologia , Infecções Respiratórias/imunologia , Viroses/prevenção & controle , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Interações Hospedeiro-Patógeno , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/virologia , Células B de Memória/metabolismo , Células B de Memória/microbiologia , Células B de Memória/virologia , Células T de Memória/metabolismo , Células T de Memória/microbiologia , Células T de Memória/virologia , Fenótipo , Infecções Respiratórias/microbiologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/virologia , Vacinação , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Viroses/imunologia , Viroses/metabolismo , Viroses/microbiologia
7.
iScience ; 24(9): 102970, 2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34471863

RESUMO

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.

8.
J Virol ; 82(9): 4680-4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18287240

RESUMO

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.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Glioblastoma/terapia , Timidina Quinase/uso terapêutico , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/farmacologia , Encéfalo/patologia , Neoplasias Encefálicas/terapia , Modelos Animais de Doenças , Herpesvirus Humano 1/enzimologia , Humanos , Ratos , Ratos Endogâmicos Lew , Taxa de Sobrevida , Timidina Quinase/genética , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos
9.
Mol Ther ; 16(2): 343-51, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18180781

RESUMO

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.


Assuntos
Adenoviridae/genética , Encéfalo/metabolismo , Imunização/métodos , Transgenes/genética , Animais , Western Blotting , Encéfalo/imunologia , Feminino , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Imuno-Histoquímica , Inflamação/imunologia , Camundongos , Plasmídeos/genética , Tetraciclina/farmacologia , beta-Galactosidase/metabolismo
10.
Nat Neurosci ; 8(1): 27-33, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15568022

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/genética , Genes Recessivos , Mutação , Oligodendroglia/citologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Supressoras de Tumor/genética , Animais , Ciclo Celular , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Corpo Caloso/citologia , Inibidor de Quinase Dependente de Ciclina p27 , Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia , Camundongos , Camundongos Quaking , Mutação/fisiologia , Proteína Básica da Mielina/metabolismo , Neurônios/fisiologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estabilidade de RNA , Ratos , Células-Tronco/citologia , Células-Tronco/fisiologia , Telencéfalo/embriologia
11.
Neuron ; 36(5): 815-29, 2002 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-12467586

RESUMO

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.


Assuntos
Proteína Básica da Mielina/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Processamento Alternativo , Animais , Sítios de Ligação , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Doenças Desmielinizantes/fisiopatologia , Éxons/genética , Humanos , Camundongos , Camundongos Quaking , Proteína Básica da Mielina/genética , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Mutação Puntual , Ligação Proteica , Isoformas de Proteínas/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
Vaccine ; 34(36): 4304-12, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27402566

RESUMO

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.


Assuntos
Imunidade Adaptativa , Adjuvantes Imunológicos , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Vacinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/síntese química , Adjuvantes Imunológicos/química , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/biossíntese , Células Dendríticas/imunologia , Humanos , Imidazóis/síntese química , Imidazóis/química , Imidazóis/imunologia , Imidazóis/farmacologia , Piridinas/síntese química , Piridinas/imunologia , Piridinas/farmacologia , Quinolinas/síntese química , Quinolinas/química , Quinolinas/imunologia , Quinolinas/farmacologia , Suínos , Vacinas/administração & dosagem
14.
RNA Biol ; 2(2): 37-40, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17132940

RESUMO

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.


Assuntos
Bainha de Mielina/fisiologia , Neuroglia/citologia , Proteínas de Ligação a RNA/fisiologia , Animais , Diferenciação Celular , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Viabilidade Fetal , Camundongos , Camundongos Quaking , Modelos Biológicos , Proteína Básica da Mielina/metabolismo , Oligodendroglia/citologia , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/metabolismo
15.
J Immunol Methods ; 424: 64-79, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26002154

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fagocitose/imunologia , Actinas/metabolismo , Adjuvantes Imunológicos , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Modelos Animais de Doenças , Combinação de Medicamentos , Imunofenotipagem , Imunoterapia , Lipídeo A/administração & dosagem , Lipídeo A/análogos & derivados , Lipídeo A/imunologia , Camundongos , Camundongos Transgênicos , Multimerização Proteica , Proteólise , Saponinas/administração & dosagem , Saponinas/imunologia , Vacinação
16.
PLoS One ; 6(6): e21065, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21731648

RESUMO

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.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Poliubiquitina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Linhagem Celular Tumoral , Humanos , Proteína 1 Inibidora do Crescimento , Peptídeos e Proteínas de Sinalização Intracelular/química , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Fosfolipídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Supressoras de Tumor/química , Ubiquitina Tiolesterase/metabolismo , Peptidase 7 Específica de Ubiquitina
17.
PLoS One ; 4(6): e5867, 2009 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-19517016

RESUMO

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.


Assuntos
Bainha de Mielina/química , Proteínas de Ligação a RNA/genética , Células de Schwann/metabolismo , Processamento Alternativo , Animais , Núcleo Celular/metabolismo , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citoplasma/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Gânglios Espinais/metabolismo , Camundongos , Proteína Básica da Mielina/metabolismo , Sistema Nervoso Periférico/fisiologia , Isoformas de Proteínas , Proteínas de Ligação a RNA/química , Ratos
18.
Mol Ther ; 15(12): 2154-63, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17895861

RESUMO

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.


Assuntos
Adenoviridae/imunologia , Encéfalo/metabolismo , Vetores Genéticos , Adenoviridae/genética , Animais , Sequência de Bases , Encéfalo/imunologia , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Testes de Neutralização , Reação em Cadeia da Polimerase
19.
J Biol Chem ; 280(46): 38639-47, 2005 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-16179349

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Neoplasias da Mama/patologia , Proteínas de Ligação a DNA/química , Proteínas de Neoplasias/metabolismo , Neoplasias/patologia , Fosfoproteínas/química , Proteínas Tirosina Quinases/metabolismo , Proteínas de Ligação a RNA/química , Tirosina/química , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Encéfalo/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Fator de Crescimento Epidérmico/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Vetores Genéticos , Células HeLa , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Dados de Sequência Molecular , Peptídeos/química , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/metabolismo , Interferência de RNA , Ratos , Homologia de Sequência de Aminoácidos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA