Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Transl Sci ; 17(5): e13789, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38761014

RESUMO

This first-in-human study evaluated the safety, tolerability, single- and multiple-dose pharmacokinetic profiles with dietary influence, and pharmacodynamics (PD) of DFV890, an oral NLRP3 inhibitor, in healthy participants. In total, 122 participants were enrolled into a three-part trial including single and 2-week multiple ascending oral doses (SAD and MAD, respectively) of DFV890, and were randomized (3:1) to DFV890 or placebo (SAD [3-600 mg] and MAD [fasted: 10-200 mg, once-daily or fed: 25 and 50 mg, twice-daily]). DFV890 was generally well-tolerated. Neither deaths nor serious adverse events were reported. A less than dose-proportional increase in exposure was observed with the initially used crystalline suspension (3-300 mg); however, an adjusted suspension formulation using spray-dried dispersion (SDD; 100-600 mg) confirmed dose-proportional increase in exposure. Relative bioavailability between crystalline suspension and tablets, and food effect were evaluated at 100 mg. Under fasting conditions, Cmax of the tablet yielded 78% compared with the crystalline suspension, and both formulations showed comparable AUC. The fed condition led to a 2.05- and 1.49-fold increase in Cmax and AUC0-last compared with the fasting condition. The median IC50 and IC90 for ex-vivo lipopolysaccharide-stimulated interleukin IL-1ß release inhibition (PD) were 61 (90% CI: 50, 70) and 1340 ng/mL (90% CI: 1190, 1490). Crystalline tablets of 100 mg once-daily or 25 mg twice-daily were sufficient to maintain ~90% of the IL-1ß release inhibition over 24 h at steady state. Data support dose and formulation selection for further development in diseases, in which an overactivated NLRP3 represents the underlying pathophysiology.


Assuntos
Relação Dose-Resposta a Droga , Interleucina-1beta , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Adulto , Feminino , Administração Oral , Pessoa de Meia-Idade , Adulto Jovem , Interleucina-1beta/metabolismo , Voluntários Saudáveis , Interações Alimento-Droga , Método Duplo-Cego , Disponibilidade Biológica , Adolescente , Esquema de Medicação
2.
Toxicol Pathol ; 51(3): 135-147, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37439009

RESUMO

Branaplam is a splicing modulator previously under development as a therapeutic agent for Spinal Muscular Atrophy Type 1 and Huntington's disease. Branaplam increased the levels of survival motor neuron protein in preclinical studies and was well tolerated in early clinical studies; however, peripheral neurotoxicity was observed in a preclinical safety study in juvenile dogs. The aim of this study was to determine whether serum neurofilament light chain (NfL) concentrations in dogs could serve as a monitoring biomarker for branaplam-induced peripheral neurotoxicity. A 30-week time-course investigative study in dogs treated with vehicle control (negative control), neurotoxic pyridoxine (positive control), or branaplam was conducted to assess neuropathology, nerve morphometry, electrophysiological measurements, gene expression profiles, and correlation to NfL serum concentrations. In branaplam-treated animals, a mild to moderate nerve fiber degeneration was observed in peripheral nerves correlating with increased serum NfL concentrations, but there were no observed signs or changes in electrophysiological parameters. Dogs with pyridoxine-induced peripheral axonal degeneration displayed clinical signs and electrophysiological changes in addition to elevated serum NfL. This study suggests that NfL may be useful as an exploratory biomarker to assist in detecting and monitoring treatment-related peripheral nerve injury, with or without clinical signs, associated with administration of branaplam and other compounds bearing a neurotoxic risk.


Assuntos
Filamentos Intermediários , Síndromes Neurotóxicas , Animais , Cães , Piridoxina , Biomarcadores , Síndromes Neurotóxicas/etiologia , Degeneração Neural
3.
Front Immunol ; 11: 745, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32425939

RESUMO

Genetic disruption or short-term pharmacological inhibition of MALT1 protease is effective in several preclinical models of autoimmunity and B cell malignancies. Despite these protective effects, the severe reduction in regulatory T cells (Tregs) and the associated IPEX-like pathology occurring upon congenital disruption of the MALT1 protease in mice has raised concerns about the long-term safety of MALT1 inhibition. Here we describe the results of a series of toxicology studies in rat and dog species using MLT-943, a novel potent and selective MALT1 protease inhibitor. While MLT-943 effectively prevented T cell-dependent B cell immune responses and reduced joint inflammation in the collagen-induced arthritis rat pharmacology model, in both preclinical species, pharmacological inhibition of MALT1 was associated with a rapid and dose-dependent reduction in Tregs and resulted in the progressive appearance of immune abnormalities and clinical signs of an IPEX-like pathology. At the 13-week time point, rats displayed severe intestinal inflammation associated with mast cell activation, high serum IgE levels, systemic T cell activation and mononuclear cell infiltration in multiple tissues. Importantly, using thymectomized rats we demonstrated that MALT1 protease inhibition affects peripheral Treg frequency independently of effects on thymic Treg output and development. Our data confirm the therapeutic potential of MALT1 protease inhibitors but highlight the safety risks and challenges to consider before potential application of such inhibitors into the clinic.


Assuntos
Diabetes Mellitus Tipo 1/congênito , Diarreia/etiologia , Doenças Genéticas Ligadas ao Cromossomo X/etiologia , Doenças do Sistema Imunitário/congênito , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/antagonistas & inibidores , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Diabetes Mellitus Tipo 1/etiologia , Cães , Feminino , Humanos , Doenças do Sistema Imunitário/etiologia , Inflamação/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Linfócitos T Reguladores/imunologia
4.
ILAR J ; 58(1): 69-79, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575330

RESUMO

Originally conceptualized as an integrated approach combining conventional toxicology methods with genome-wide expression profiling, toxicogenomics has promised to provide unequivocal relationships between the molecular changes elicited by a compound or a target pathway and the lesions that appear subsequently in the tissues. However, the discipline has only partially delivered on this promise, and the number of publications and submissions related to toxicogenomics is stagnating. The purpose of this article is to outline key factors contributing to a successful implementation of toxicogenomics in the drug discovery and development process. Paradigms and methods of toxicogenomics are briefly reviewed, and the prominence of biostatistics and its limitations in the particular context of nonclinical toxicology studies are discussed. We present specific approaches for pathophysiological contextualization of gene expression data derived from tissues with lesions at variable incidence and severity: "unmixing" (deconvolution) of molecular expression profiles from complex tissues, the invaluable contribution of reference data, the role of establishing causation between expression signals and pathologic changes (phenotypic anchoring), and especially molecular localization. These approaches compensate for the limitations of biostatistical analysis, which in turn, derive from tissue heterogeneity. Finally, impactful applications of toxicogenomics along the drug discovery and development process are exemplified, from the evaluation of potential target toxicities to the selection of candidate compounds and elucidation of the molecular and cellular mechanisms leading to chronic toxicity.


Assuntos
Perfilação da Expressão Gênica , Toxicogenética , Descoberta de Drogas , Medição de Risco
5.
Toxicol Sci ; 155(1): 283-297, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27742868

RESUMO

The aim of this study was to determine the relative safety of 4 antiviral drugs (telbivudine, tenofovir, adefovir, and entecavir) against hepatitis B virus with respect to kidney function and toxicity in male Sprague Dawley rats. The antiviral drugs were administered once daily for 4 weeks by oral gavage at ∼10 and 25-40 times the human equivalent dose. Main assessments included markers of renal toxicity in urine, magnetic resonance imaging (MRI) of kidney function, histopathology, and electron microscopic examination. Administration of adefovir at 11 and 28 mg/kg for 4 weeks caused functional and morphological kidney alterations in a time- and dose-dependent manner, affecting mainly the proximal tubules and suggesting a mechanism of toxicity related to mitochondrial degeneration/depletion. Of note, the observed adefovir-induced reduction of kidney function was not detected by the standard method of glomerular filtration rate (GFR) measurements (clearance rate of the endogenous marker, creatinine), thereby emphasizing the superiority of MRI in terms of sensitive detection of GFR in rats. For the low dose of 300 mg/kg of tenofovir, minor kidney effects such as nuclear enlargement in the tubular epithelium, and hyaline droplets accumulation were detected, which was also observed for the low dose (11 mg/kg) of adefovir. No assessments could be done at the higher dose of 600/1000 mg/kg tenofovir due to gastrointestinal tract toxicity which prevented treatment of the animals for longer than 1 week. Entecavir at 1 and 3 mg/kg and telbivudine at 600 and 1600 mg/kg caused no toxicologically relevant effects on the kidney.


Assuntos
Antivirais/efeitos adversos , Hepatite B/tratamento farmacológico , Rim/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Rim/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Sprague-Dawley
6.
Sci Rep ; 6: 36923, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27853279

RESUMO

Antibodies targeting IL-17A or its receptor IL-17RA show unprecedented efficacy in the treatment of autoimmune diseases such as psoriasis. These therapies, by neutralizing critical mediators of immunity, may increase susceptibility to infections. Here, we compared the effect of antibodies neutralizing IL-17A, IL-17F or TNFα on murine host responses to Mycobacterium tuberculosis infection by evaluating lung transcriptomic, microbiological and histological analyses. Coinciding with a significant increase of mycobacterial burden and pathological changes following TNFα blockade, gene array analyses of infected lungs revealed major changes of inflammatory and immune gene expression signatures 4 weeks post-infection. Specifically, gene expression associated with host-pathogen interactions, macrophage recruitment, activation and polarization, host-antimycobacterial activities, immunomodulatory responses, as well as extracellular matrix metallopeptidases, were markedly modulated by TNFα blockade. IL-17A or IL-17F neutralization elicited only mild changes of few genes without impaired host resistance four weeks after M. tuberculosis infection. Further, the absence of both IL-17RA and IL-22 pathways in genetically deficient mice did not profoundly compromise host control of M. tuberculosis over a 6-months period, ruling out potential compensation between these two pathways, while TNFα-deficient mice succumbed rapidly. These data provide experimental confirmation of the low clinical risk of mycobacterial infection under anti-IL-17A therapy, in contrast to anti-TNFα treatment.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos/imunologia , Interleucina-17/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Interações Hospedeiro-Patógeno/imunologia , Imunidade/imunologia , Interleucinas/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-17/imunologia , Interleucina 22
7.
Toxicol Sci ; 153(1): 39-54, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27255383

RESUMO

Histone deacetylase (HDAC) inhibitors are an emerging class of anticancer agents that modify gene expression by altering the acetylation status of lysine residues of histone proteins, thereby inducing transcription, cell cycle arrest, differentiation, and cell death or apoptosis of cancer cells. In the clinical setting, treatment with HDAC inhibitors has been associated with delayed cardiac repolarization and in rare instances a lethal ventricular tachyarrhythmia known as torsades de pointes. The mechanism(s) of HDAC inhibitor-induced effects on cardiac repolarization is unknown. We demonstrate that administration of structurally diverse HDAC inhibitors to dogs causes delayed but persistent increases in the heart rate corrected QT interval (QTc), an in vivo measure of cardiac repolarization, at timepoints far removed from the Tmax for parent drug and metabolites. Transcriptional profiling of ventricular myocardium from dogs treated with various HDAC inhibitors demonstrated effects on genes involved in protein trafficking, scaffolding and insertion of various ion channels into the cell membrane as well as genes for specific ion channel subunits involved in cardiac repolarization. Extensive in vitro ion channel profiling of various structural classes of HDAC inhibitors (and their major metabolites) by binding and acute patch clamp assays failed to show any consistent correlations with direct ion channel blockade. Drug-induced rescue of an intracellular trafficking-deficient mutant potassium ion channel, hERG (G601S), and decreased maturation (glycosylation) of wild-type hERG expressed by CHO cells in vitro correlated with prolongation of QTc intervals observed in vivo The results suggest that HDAC inhibitor-induced prolongation of cardiac repolarization may be mediated in part by transcriptional changes of genes required for ion channel trafficking and localization to the sarcolemma. These data have broad implications for the development of these drug classes and suggest that the optimal time to assess potentially transcriptionally mediated physiologic effects will be delayed relative to an epigenetic drug's Tmax/Cmax.


Assuntos
Coração/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Transcrição Gênica , Animais , Transporte Biológico , Cães , Coração/fisiologia , Inibidores de Histona Desacetilases/farmacocinética , Masculino
8.
J Leukoc Biol ; 99(6): 1153-64, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26729813

RESUMO

Antibodies targeting IL-17A or its receptor, IL-17RA, are approved to treat psoriasis and are being evaluated for other autoimmune conditions. Conversely, IL-17 signaling is critical for immunity to opportunistic mucosal infections caused by the commensal fungus Candida albicans, as mice and humans lacking the IL-17R experience chronic mucosal candidiasis. IL-17A, IL-17F, and IL-17AF bind the IL-17RA-IL-17RC heterodimeric complex and deliver qualitatively similar signals through the adaptor Act1. Here, we used a mouse model of acute oropharyngeal candidiasis to assess the impact of blocking IL-17 family cytokines compared with specific IL-17 cytokine gene knockout mice. Anti-IL-17A antibodies, which neutralize IL-17A and IL-17AF, caused elevated oral fungal loads, whereas anti-IL-17AF and anti-IL-17F antibodies did not. Notably, there was a cooperative effect of blocking IL-17A, IL-17AF, and IL-17F together. Termination of anti-IL-17A treatment was associated with rapid C. albicans clearance. IL-17F-deficient mice were fully resistant to oropharyngeal candidiasis, consistent with antibody blockade. However, IL-17A-deficient mice had lower fungal burdens than anti-IL-17A-treated mice. Act1-deficient mice were much more susceptible to oropharyngeal candidiasis than anti-IL-17A antibody-treated mice, yet anti-IL-17A and anti-IL-17RA treatment caused equivalent susceptibilities. Based on microarray analyses of the oral mucosa during infection, only a limited number of genes were associated with oropharyngeal candidiasis susceptibility. In sum, we conclude that IL-17A is the main cytokine mediator of immunity in murine oropharyngeal candidiasis, but a cooperative relationship among IL-17A, IL-17AF, and IL-17F exists in vivo. Susceptibility displays the following hierarchy: IL-17RA- or Act1-deficiency > anti-IL-17A + anti-IL-17F antibodies > anti-IL-17A or anti-IL-17RA antibodies > IL-17A deficiency.


Assuntos
Anticorpos Bloqueadores/farmacologia , Candidíase Bucal/imunologia , Candidíase Bucal/patologia , Imunidade nas Mucosas/efeitos dos fármacos , Interleucina-17/antagonistas & inibidores , Mucosa Bucal/imunologia , Mucosa Bucal/patologia , Doença Aguda , Animais , Anticorpos Antifúngicos/farmacologia , Anticorpos Neutralizantes/farmacologia , Candida/efeitos dos fármacos , Candidíase Bucal/microbiologia , Suscetibilidade a Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Predisposição Genética para Doença , Interleucina-17/metabolismo , Camundongos Endogâmicos C57BL , Mucosa Bucal/efeitos dos fármacos
9.
Sci Transl Med ; 6(263): 263ra160, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25411473

RESUMO

Adjuvants increase vaccine potency largely by activating innate immunity and promoting inflammation. Limiting the side effects of this inflammation is a major hurdle for adjuvant use in vaccines for humans. It has been difficult to improve on adjuvant safety because of a poor understanding of adjuvant mechanism and the empirical nature of adjuvant discovery and development historically. We describe new principles for the rational optimization of small-molecule immune potentiators (SMIPs) targeting Toll-like receptor 7 as adjuvants with a predicted increase in their therapeutic indices. Unlike traditional drugs, SMIP-based adjuvants need to have limited bioavailability and remain localized for optimal efficacy. These features also lead to temporally and spatially restricted inflammation that should decrease side effects. Through medicinal and formulation chemistry and extensive immunopharmacology, we show that in vivo potency can be increased with little to no systemic exposure, localized innate immune activation and short in vivo residence times of SMIP-based adjuvants. This work provides a systematic and generalizable approach to engineering small molecules for use as vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Desenho de Fármacos , Vacinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacocinética , Disponibilidade Biológica
10.
Vaccine ; 31(33): 3363-9, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23684834

RESUMO

MF59 is a safe and effective vaccine adjuvant that has been used in a licensed seasonal influenza vaccine for 15 years. The purpose of the present studies was to directly address a question that has been asked of us on many occasions: "which is the adjuvant active component of MF59?". Since we have recently gained a number of insights on how MF59 works as an adjuvant, we were able to use these approaches to evaluate if the individual components of MF59 (squalene oil, the surfactants Span 85 and Tween 80 or the citrate buffer) showed any direct immunostimulatory activity. We assessed the ability of the individual components to stimulate the innate and adaptive immune responses that we have shown to be indicative of MF59-mediated adjuvanticity. No immune stimulatory capacities could be attributed to squalene, Tween 80 or the citrate buffer alone. Instead, we found that the lipophilic surfactant Span 85 contributes to activation of the muscle transcriptome. However, despite this local activation, Span 85 alone - like the other single components of MF59 - is not sufficient to induce an adjuvant effect. Only the fully formulated MF59 emulsion induces all the established hallmarks of innate and adaptive immune activation, which includes activation of genes indicative of transendothelial cell migration, strong influx of immune cells into the injection site and their enhanced antigen uptake and transport to the lymph nodes. These observations may have important implications in the design of optimal emulsion-based vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Emulsões/farmacologia , Polissorbatos/farmacologia , Esqualeno/farmacologia , Imunidade Adaptativa , Adjuvantes Imunológicos/química , Animais , Anticorpos Antivirais/sangue , Apresentação de Antígeno/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Emulsões/química , Feminino , Testes de Inibição da Hemaglutinação , Hexoses/farmacologia , Humanos , Imunidade Inata , Vacinas contra Influenza/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Ativação Linfocitária , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Polissorbatos/química , Esqualeno/química , Transcriptoma/efeitos dos fármacos
11.
J Immunol ; 188(7): 3088-98, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22351935

RESUMO

The innate immune pathways induced by adjuvants required to increase adaptive responses to influenza subunit vaccines are not well characterized. We profiled different TLR-independent (MF59 and alum) and TLR-dependent (CpG, resiquimod, and Pam3CSK4) adjuvants for the ability to increase the immunogenicity to a trivalent influenza seasonal subunit vaccine and to tetanus toxoid (TT) in mouse. Although all adjuvants boosted the Ab responses to TT, only MF59 and Pam3CSK4 were able to enhance hemagglutinin Ab responses. To identify innate immune correlates of adjuvanticity to influenza subunit vaccine, we investigated the gene signatures induced by each adjuvant in vitro in splenocytes and in vivo in muscle and lymph nodes using DNA microarrays. We found that flu adjuvanticity correlates with the upregulation of proinflammatory genes and other genes involved in leukocyte transendothelial migration at the vaccine injection site. Confocal and FACS analysis confirmed that MF59 and Pam3CSK4 were the strongest inducers of blood cell recruitment in the muscle compared with the other adjuvants tested. Even though it has been proposed that IFN type I is required for adjuvanticity to influenza vaccines, we found that MF59 and Pam3CSK4 were not good inducers of IFN-related innate immunity pathways. By contrast, resiquimod failed to enhance the adaptive response to flu despite a strong activation of the IFN pathway in muscle and lymph nodes. By blocking IFN type I receptor through a mAb, we confirmed that the adjuvanticity of MF59 and Pam3CSK4 to a trivalent influenza vaccine and to TT is IFN independent.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas contra Influenza/imunologia , Interferon-alfa/imunologia , Lipopeptídeos/farmacologia , Polissorbatos/farmacologia , Esqualeno/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Quimiotaxia/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Imunidade Inata/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza B/imunologia , Linfonodos/imunologia , Linfonodos/metabolismo , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Músculos/imunologia , Músculos/metabolismo , Organismos Livres de Patógenos Específicos , Baço/citologia , Baço/imunologia , Baço/metabolismo , Toxoide Tetânico/imunologia , Vacinas de Subunidades Antigênicas/imunologia
12.
J Innate Immun ; 2(2): 144-59, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20375632

RESUMO

CpG-containing oligodeoxynucleotides are potent mucosal adjuvants and effective as stand-alone treatment of respiratory infections in mice. Although CpG is also used as a type 1 helper immunomodulator in the treatment of asthma and allergic disease, immune modulation following intranasal application has not been fully characterized yet. Using a B-type CpG, we monitored RNA expression profiles, cytokine production and cellular activation in lung tissue and bronchoalveolar lavages ex vivo and cytokine production of purified cell populations in vitro. CpG triggered the upregulation of many transcripts, including interferon response genes and proinflammatory cytokine genes, between 3 h and 4 days. Overlapping subsets of these cytokine proteins were induced in vitro in purified CD11c+ cells, B cells and alveolar macrophages from the lung, thus identifying these cells as direct targets of CpG. While lung B cells strongly respond to CpG in vitro, less activation is found ex vivo, suggesting efficient CpG sequestering or rapid B cell migration after activation. In contrast, a type II alveolar epithelial cell line did not respond to CpG in vitro. We noted selective recruitment of plasmacytoid dendritic cells (DCs) into the lung tissue, and of conventional DCs and natural killer (NK) cells into the lung tissue and bronchoalveolar space. Furthermore, CpG induced activation of intrapulmonary DCs, NK and T cells. We hypothesize that CpG-linked adjuvanticity and clearance of respiratory pathogens are mediated by two major mechanisms: transient induction of the interferon pathway limiting microbial survival and selective recruitment of DCs and NK cells, which allows for better adaptive responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Pulmão/imunologia , Oligodesoxirribonucleotídeos/administração & dosagem , Administração Intranasal , Animais , Linfócitos B/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Antígeno CD11c/metabolismo , Feminino , Pulmão/citologia , Ativação Linfocitária/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/imunologia
13.
Vaccine ; 27(25-26): 3331-4, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19200813

RESUMO

Despite the fact that alum and oil-in-water emulsions have been used for decades as human vaccine adjuvants in a large number of individuals, their mechanism of action is not completely understood. It has been reported that these particulate adjuvants act by increasing antigen availability and uptake by immune cells. However, recent work on alum and on the squalene-based emulsion MF59, has demonstrated that besides antigen delivery functions, these classes of adjuvants can also activate innate immunity pathways in vivo, generating an immunocompetent environment at injection site. Interestingly, it has been demonstrated that alum adjuvanticity depends on the activation of a protein complex called NLPR3/inflammasome, which is required for the correct processing of a number of pro-inflammatory cytokines, including IL1beta. More work needs to be performed to investigate if the inflammasome is also required for the activity of MF59 and of other particulate vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Polissorbatos/farmacologia , Esqualeno/farmacologia , Animais , Proteínas de Transporte/fisiologia , Células Dendríticas/fisiologia , Humanos , Imunidade Inata/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores Toll-Like/fisiologia
14.
Eur J Immunol ; 38(8): 2068-71, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18651701

RESUMO

The development of vaccine adjuvants for human use has been one of the slowest processes in the history of medicine. For almost one century, aluminium hydroxide (alum) has been the only vaccine adjuvant approved worldwide. Only in the past decade have two oil-in-water emulsions and one TLR agonist been approved by the European authorities as new vaccine adjuvants. Despite the fact that alum has been injected into billions of people, its mechanism of action is not fully understood. Recently, several reports have greatly increased our knowledge of the molecular and cellular events triggered by alum; however, the contribution of each of these processes to alum adjuvanticity is still unclear. A study published in this issue of the European Journal of Immunology, together with two recent publications, have demonstrated that the NOD-like receptor, pyrin domain containing 3 (Nlrp3)-inflammasome is the molecular target of alum immunostimulatory activity in vitro. Surprisingly, these three studies reported conflicting results on the requirement of the Nlrp3 inflammasome complex for alum adjuvant effects in vivo. This commentary attempts to resolve some of these discrepancies.


Assuntos
Adjuvantes Imunológicos/farmacologia , Hidróxido de Alumínio/farmacologia , Animais , Proteínas de Transporte/fisiologia , Humanos , Inflamação/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores Toll-Like/fisiologia , Vacinação
15.
J Immunol ; 179(8): 5346-57, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17911621

RESUMO

LTK63, a nontoxic mutant of Escherichia coli heat labile enterotoxin (LT), is a potent and safe mucosal adjuvant that has also been shown to confer generic protection to several respiratory pathogens. To understand the mechanisms of action underlying the LTK63 protective effect, we analyzed the molecular and cellular events triggered by its administration in vivo. We show here that LTK63 intrapulmonary administration induced in the mouse lung a specific gene expression signature characterized by the up-regulation of cell cycle genes, several host defense genes, chemokines, chemokine receptors, and immune cell-associated genes. Such a transcriptional profile reflected the activation of alveolar macrophages and the recruitment to the lung of T and B cells and innate immune cells such as granulocytes, NK, and dendritic cells. All of these events were T cell dependent and specific for LTK63 because they were absent in SCID and nude mice. Additionally, we showed that LTK63 induces a potent adaptive immune response against itself directed to the lung. We propose that acquired response to LTK63 is the driving force for the local recruitment of both adaptive and innate immune cells. Our data suggest that LTK63 acts as an airway infection mimic that establishes a generic protective environment limiting respiratory infection by innate immune mechanisms and by improving adaptive responses to invading pathogens.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/imunologia , Enterotoxinas/administração & dosagem , Enterotoxinas/imunologia , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Escherichia coli/imunologia , Imunidade nas Mucosas , Pulmão/imunologia , Pulmão/metabolismo , Animais , Subpopulações de Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/imunologia , Quimiocinas/biossíntese , Quimiocinas/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Imunidade Celular , Imunidade Inata , Intubação Intratraqueal , Pulmão/citologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Transcrição Gênica/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA