Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Vaccine ; 34(34): 4017-24, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27317451

RESUMO

TH2-biased immune responses are associated with inadequate protection against some pathogens and with cancer, colitis, asthma and allergy. Since most currently used vaccine adjuvants induce a TH2-biased response, this has led to interest in developing adjuvants capable of activating TH1 immunity and modulating existing TH2 responses. Immunotherapies to shift immune responses from TH2 to TH1 have generally required prolonged immunization protocols and have not induced effective TH1 responses. We have demonstrated that nanoscale emulsions (NE), a novel mucosal adjuvant, induce robust IgA and IgG antibody responses and TH1/TH17 cellular immunity resulting in protection against a variety of respiratory and mucosal infections. Because intranasal (i.n.) delivery of NE adjuvant consistently induces TH1/TH17 biased responses, we hypothesized that NE could be used as a therapeutic vaccine to redirect existing TH2 polarized immunity towards a more balanced TH1/TH2 profile. To test this, a TH2 immune response was established by intramuscular immunization of mice with alum-adjuvanted hepatitis B surface antigen (HBs), followed by a single subsequent i.n. immunization with NE-HBs. These animals exhibited increased TH1 associated immune responses and IL-17, and decreased TH2 cytokines (IL-4 and IL-5) and IgG1. NE immunization induced regulatory T cells and IL-10, and IL-10 was required for the suppression of TH2 immunity. These data demonstrate that NE-based vaccines can modulate existing TH2 immune responses to promote TH1/TH17 immunity and suggest the potential therapeutic use of NE vaccines for diseases associated with TH2 immunity.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Imunidade nas Mucosas , Nanoestruturas/administração & dosagem , Células Th2/imunologia , Administração Intranasal , Animais , Citocinas/imunologia , Emulsões/administração & dosagem , Feminino , Antígenos de Superfície da Hepatite B/imunologia , Imunidade Celular , Imunidade Humoral , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Camundongos , Células Th1/imunologia , Células Th17/imunologia
2.
PLoS One ; 10(5): e0126120, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25962136

RESUMO

Vaccine adjuvants have been reported to induce both mucosal and systemic immunity when applied to mucosal surfaces and this dual response appears important for protection against certain pathogens. Despite the potential advantages, however, no mucosal adjuvants are currently approved for human use. Evaluating compounds as mucosal adjuvants is a slow and costly process due to the need for lengthy animal immunogenicity studies. We have constructed a library of 112 intranasal adjuvant candidate formulations consisting of oil-in-water nanoemulsions that contain various cationic and nonionic surfactants. To facilitate adjuvant development we first evaluated this library in a series of high-throughput, in vitro assays for activities associated with innate and adaptive immune activation in vivo. These in vitro assays screened for the ability of the adjuvant to bind to mucin, induce cytotoxicity, facilitate antigen uptake in epithelial and dendritic cells, and activate cellular pathways. We then sought to determine how these parameters related to adjuvant activity in vivo. While the in vitro assays alone were not enough to predict the in vivo adjuvant activity completely, several interesting relationships were found with immune responses in mice. Furthermore, by varying the physicochemical properties of the surfactant components (charge, surfactant polar head size and hydrophobicity) and the surfactant blend ratio of the formulations, the strength and type of the immune response generated (TH1, TH2, TH17) could be modulated. These findings suggest the possibility of using high-throughput screens to aid in the design of custom adjuvants with unique immunological profiles to match specific mucosal vaccine applications.


Assuntos
Adjuvantes Imunológicos/química , Vacinas/administração & dosagem , Vacinas/química , Adjuvantes Imunológicos/toxicidade , Administração Intranasal , Animais , Linhagem Celular , Química Farmacêutica , Citocinas/biossíntese , Emulsões , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Ensaios de Triagem em Larga Escala , Imunidade Celular , Imunidade Humoral , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Nanotecnologia
3.
J Gene Med ; 16(3-4): 75-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24700644

RESUMO

BACKGROUND: Although cytosolic DNA degradation plays an important role in decreasing transgene expression, the plasmid degradation pattern remains largely unexplored. METHODS: Illumina dye sequencing was employed to provide degradation site information for S1 and cytosolic nucleases. S1 nuclease provided a positive control for a comparison between the agarose gel method and sequencing approaches. RESULTS: The poly(A) region between the ß-lactamase gene and the cytomegalovirus (CMV) promoter was identified as the most likely cut site for polyplex-treated cytosol. The second most likely site, at the 5' end of the ß-lactamase gene, was identified by gel electrophoresis and sequencing. Additional sites were detected in the OriC region, the SV40/poly(A) region, the luciferase gene and the CMV promoter. Sequence analysis of plasmid treated with cytosol from control cells showed the greatest cut activity in the OriC region, the ß-lactamase gene and the poly(A) region following the luciferase gene. Additional regions of cut activity include the SV40 promoter and the ß-lactamase poly(A) termination sequence. Both cytosolic nucleases and the S1 nuclease showed substantial activity at the bacterial origin of replication (OriC). CONCLUSIONS: High-throughput plasmid sequencing revealed regions of the luciferase plasmid DNA sequence that are sensitive to cytosolic nuclease degradation. This provides new targets for improving plasmid and/or polymer design to optimize the likelihood of protein expression.


Assuntos
DNA/metabolismo , Técnicas de Transferência de Genes , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Plasmídeos/metabolismo , Citomegalovirus/genética , Eletroforese em Gel de Ágar , Endonucleases/genética , Endonucleases/metabolismo , Terapia Genética , Vetores Genéticos , Luciferases/genética , Luciferases/metabolismo , Complexo de Reconhecimento de Origem/genética , Proteínas de Ligação a Poli(A)/genética , Proteínas de Ligação a Poli(A)/metabolismo , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Transgenes , beta-Lactamases/genética
4.
J Immunol ; 192(6): 2722-33, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24532579

RESUMO

Nasal administration of an oil-in-water nanoemulsion (NE) adjuvant W805EC produces potent systemic and mucosal, Th-1- and Th-17-balanced cellular responses. However, its molecular mechanism of action has not been fully characterized and is of particular interest because NE does not contain specific ligands for innate immune receptors. In these studies, we demonstrate that W805EC NE adjuvant activates innate immunity, induces specific gene transcription, and modulates NF-κB activity via TLR2 and TLR4 by a mechanism that appears to be distinct from typical TLR agonists. Nasal immunization with NE-based vaccine showed that the TLR2, TLR4, and MyD88 pathways and IL-12 and IL-12Rß1 expression are not required for an Ab response, but they are essential for the induction of balanced Th-1 polarization and Th-17 cellular immunity. NE adjuvant induces MHC class II, CD80, and CD86 costimulatory molecule expression and dendritic cell maturation. Further, upon immunization with NE, adjuvant mice deficient in the CD86 receptor had normal Ab responses but significantly reduced Th-1 cellular responses, whereas animals deficient in both CD80 and CD86 or lacking CD40 failed to produce either humoral or cellular immunity. Overall, our data show that intranasal administration of Ag with NE induces TLR2 and TLR4 activation along with a MyD88-independent Ab response and a MyD88-dependent Th-1 and Th-17 cell-mediated immune response. These findings suggest that the unique properties of NE adjuvant may offer novel opportunities for understanding previously unrecognized mechanisms of immune activation important for generating effective mucosal and systemic immune responses.


Assuntos
Adjuvantes Imunológicos/farmacologia , Emulsões/farmacologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Linhagem Celular , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Emulsões/administração & dosagem , Feminino , Células HEK293 , Humanos , Imunidade Celular/genética , Imunidade Celular/imunologia , Imunidade Humoral/genética , Imunidade Humoral/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-12/metabolismo , Subunidade beta 1 de Receptor de Interleucina-12/genética , Subunidade beta 1 de Receptor de Interleucina-12/imunologia , Subunidade beta 1 de Receptor de Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Transcriptoma/imunologia
5.
Vaccine ; 31(7): 1072-9, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23273511

RESUMO

Nanoemulsions are adjuvants that enhance antigen penetration in the nasal mucosa, increase cellular uptake of antigens by both epithelial dendritic cells, and promote migration of antigen-loaded dendritic cells to regional lymph nodes within a day of vaccine administration. The objective of this study was to determine whether the W(80)5EC nanoemulsion adjuvant enhances immune response not only by direct uptake of antigen by dendritic cells, but also indirectly, by phagocytosis of antigen-primed, apoptotic, epithelial cells. Consistent with this, we show that exposure of both epithelial cells (TC-1s) and dendritic cells (JAWS II or bone marrow derived dendritic cells (BMDCs)) to nanoemulsion exhibited augmented antigen uptake in cell culture. TC-1 cells subsequently underwent G(2)/M cell cycle arrest and apoptosis, and when co-cultured with JAWS II or BMDCs were rapidly engulfed by the dendritic cells, which responded by up-regulating dendritic cell maturation marker CD86. Altogether these results suggest that the effectiveness of nanoemulsions as adjuvants stems, at least in part, from the engulfment of antigen-loaded epithelial cells, leading to enhanced antigen processing and a strong and balanced mucosal and systemic immune response.


Assuntos
Adjuvantes Imunológicos/metabolismo , Antígenos/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Emulsões/metabolismo , Células Epiteliais/imunologia , Fagocitose/efeitos dos fármacos , Animais , Antígenos/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL
6.
Eur J Immunol ; 42(8): 2073-86, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22653620

RESUMO

While the nasal mucosa is a potentially useful site for human immunization, toxin-based nasal adjuvants are generally unsafe and less effective in humans. Safe mucosal adjuvants that activate protective immunity via mucosal administration are highly dependent on barrier antigen sampling by epithelial and DCs. Here, we demonstrate that protein antigens formulated in unique oil-in-water nanoemulsions (NEs) result in distinctive transcellular antigen uptake in ciliated nasal epithelial cells, leading to delivery into nasal associated lymphoid tissue. NE formulation also enhances MHC class II expression in epithelial cells and DC activation/trafficking to regional lymphoid tissues in mice. These materials appear to induce local epithelial cell apoptosis and heterogeneous cytokine production by mucosal epithelial cells and mixed nasal tissues, including G-CSF, GM-CSF, IL-1a, IL-1b, IL-5, IL-6, IL-12, IP-10, KC, MIP-1a, TGF-ß, and TSLP. This is the first observation of a nasal adjuvant that activates calreticulin-associated apoptosis of ciliated nasal epithelial cells to generate broad cytokine/chemokine responses in mucosal tissue.


Assuntos
Adjuvantes Imunológicos , Apoptose , Citocinas/biossíntese , Células Dendríticas/imunologia , Mucosa Nasal/imunologia , Animais , Transporte Biológico/imunologia , Calreticulina , Movimento Celular , Células Cultivadas , Células Dendríticas/metabolismo , Emulsões , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Genes MHC da Classe II , Interleucina-6/imunologia , Interleucina-6/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Nasal/metabolismo
7.
Crit Rev Immunol ; 30(2): 189-99, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20370629

RESUMO

T17 (T-helper-17) cytokine responses have been recently recognized as an important component for the protective immunity produced by vaccination. However, the mechanism by which immune adjuvants induce T17 immunity has not been defined. We have developed a novel mucosal nanoemulsion (NE) adjuvant that produces a robust humoral and T1 cellular immunity. Herein, we demonstrate that immunization with NE adjuvant induces a T17 response to diverse antigens in both outbred and inbred mice. CD86 deficiency had a limited effect on the induction of IL-17, however, double CD80/CD86, CD40, and IL-6 (interleukin 6) mutant mice failed to produce T17 immunity in response to NE adjuvant. Mice deficient in TLR2 and TLR4 (Toll-like receptors 2 and 4) had a diminished IL-17 response. Our data indicate that nasal mucosal immunization with NE adjuvant produces T1 and T17 immunity; that this process requires IL-6, CD40, and at least one of the CD80/CD86 molecules; and that the induction of TH17 is enhanced by the presence of TLR2 and TLR4 receptors. This unique approach to vaccination may have a significant role in protection against mucosal and intracellular pathogens.


Assuntos
Adjuvantes Imunológicos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Interleucina-17/imunologia , Nanoestruturas , Animais , Emulsões , Vacinação
8.
J Aerosol Med Pulm Drug Deliv ; 23(2): 77-89, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19778268

RESUMO

BACKGROUND: Many infectious diseases that cause significant morbidity and mortality, especially in the developing world, could be preventable through vaccination. The effort to produce safe, thermally stable, and needle-free mucosal vaccines has become increasingly important for global health considerations. We have previously demonstrated that a thermally stable nanoemulsion, a mucosal adjuvant for needle-free nasal immunization, is safe and induces protective immunity with a variety of antigens, including recombinant protein. The successful use of nanoemulsion-based vaccines, however, poses numerous challenges. Among the challenges is optimization of the formulation to maintain thermal stability and potency and another is accuracy and efficiency of dispensing the vaccines to the nasal mucosa in the anterior and turbinate region of the nasal cavity or potentially to the nasopharynx-associated lymphoid tissue. METHODS: We have examined the effects of different diluents [phosphate-buffered saline (PBS) and 0.9% NaCl] on the stability and potency of nanoemulsion-based vaccines. In addition, we have determined the efficiency of delivering them using commercially available nasal spray devices (Pfeiffer SAP-62602 multidose pump and the BD Hypak SCF 0.5 ml unit dose Accuspray(TM)). RESULTS: We report the stability and potency of PBS-diluted ovalbumin-nanomeulsion mixtures for up to 8 months and NaCl-diluted mixtures up to 6 months when stored at room temperature. Significant differences in spray characteristics including droplet size, spray angle, plume width, and ovality ratios were observed between the two pumps. Further, we have demonstrated that the nanoemulsion-based vaccines are not physically or chemically altered and retain potency following actuation with nasal spray devices. Using either device, the measured spray characteristics suggest deposition of nanoemulsion-based vaccines in inductive tissues located in the anterior region of the nasal cavity. CONCLUSIONS: The results of this study suggest that nanoemulsion-based vaccines do not require specially engineered delivery devices and support their potential use as nasopharyngeal vaccine adjuvants.


Assuntos
Nanopartículas , Nebulizadores e Vaporizadores , Ovalbumina/administração & dosagem , Vacinas/administração & dosagem , Administração Intranasal , Aerossóis , Fosfatase Alcalina/administração & dosagem , Fosfatase Alcalina/química , Fosfatase Alcalina/imunologia , Animais , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Emulsões , Excipientes/química , Feminino , Antígenos de Superfície da Hepatite B/administração & dosagem , Antígenos de Superfície da Hepatite B/química , Antígenos de Superfície da Hepatite B/imunologia , Camundongos , Ovalbumina/química , Ovalbumina/imunologia , Tamanho da Partícula , Cloreto de Sódio/química , Suínos , Vacinas/imunologia
9.
Bioconjug Chem ; 20(8): 1503-13, 2009 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-19583240

RESUMO

Generation 7 (G7) poly(amidoamine) (PAMAM) dendrimers with amine, acetamide, and carboxylate end groups were prepared to investigate polymer/cell membrane interactions in vitro. G7 PAMAM dendrimers were used in this study because higher-generation of dendrimers are more effective in permeabilization of cell plasma membranes and in the formation of nanoscale holes in supported lipid bilayers than smaller, lower-generation dendrimers. Dendrimer-based conjugates were characterized by (1)H NMR, UV/vis spectroscopy, GPC, HPLC, and CE. Positively charged amine-terminated G7 dendrimers (G7-NH(2)) were observed to internalize into KB, Rat2, and C6 cells at a 200 nM concentration. By way of contrast, neither negatively charged G7 carboxylate-terminated dendrimers (G7-COOH) nor neutral acetamide-terminated G7 dendrimers (G7-Ac) associated with the cell plasma membrane or internalized under similar conditions. A series of in vitro experiments employing endocytic markers cholera toxin subunit B (CTB), transferrin, and GM(1)-pyrene were performed to further investigate mechanisms of dendrimer internalization into cells. G7-NH(2) dendrimers colocalized with CTB; however, experiments with C6 cells indicated that internalization of G7-NH(2) was not ganglioside GM(1) dependent. The G7/CTB colocalization was thus ascribed to an artifact of direct interaction between the two species. The presence of GM(1) in the membrane also had no effect upon XTT assays of cell viability or lactate dehydrogenase (LDH) assays of membrane permeability.


Assuntos
Membrana Celular/metabolismo , Dendrímeros/metabolismo , Gangliosídeo G(M1)/metabolismo , Bicamadas Lipídicas/metabolismo , Poliaminas/metabolismo , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dendrímeros/química , Relação Dose-Resposta a Droga , Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/farmacologia , Humanos , Células KB , Modelos Biológicos , Estrutura Molecular , Poliaminas/química , Ratos , Propriedades de Superfície
10.
PLoS One ; 3(8): e2954, 2008 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-18698426

RESUMO

BACKGROUND: Hepatitis B virus infection remains an important global health concern despite the availability of safe and effective prophylactic vaccines. Limitations to these vaccines include requirement for refrigeration and three immunizations thereby restricting use in the developing world. A new nasal hepatitis B vaccine composed of recombinant hepatitis B surface antigen (HBsAg) in a novel nanoemulsion (NE) adjuvant (HBsAg-NE) could be effective with fewer administrations. METHODOLOGY AND PRINCIPAL FINDINGS: Physical characterization indicated that HBsAg-NE consists of uniform lipid droplets (349+/-17 nm) associated with HBsAg through electrostatic and hydrophobic interactions. Immunogenicity of HBsAg-NE vaccine was evaluated in mice, rats and guinea pigs. Animals immunized intranasally developed robust and sustained systemic IgG, mucosal IgA and strong antigen-specific cellular immune responses. Serum IgG reached > or = 10(6) titers and was comparable to intramuscular vaccination with alum-adjuvanted vaccine (HBsAg-Alu). Normalization showed that HBsAg-NE vaccination correlates with a protective immunity equivalent or greater than 1000 IU/ml. Th1 polarized immune response was indicated by IFN-gamma and TNF-alpha cytokine production and elevated levels of IgG(2) subclass of HBsAg-specific antibodies. The vaccine retains full immunogenicity for a year at 4 degrees C, 6 months at 25 degrees C and 6 weeks at 40 degrees C. Comprehensive pre-clinical toxicology evaluation demonstrated that HBsAg-NE vaccine is safe and well tolerated in multiple animal models. CONCLUSIONS: Our results suggest that needle-free nasal immunization with HBsAg-NE could be a safe and effective hepatitis B vaccine, or provide an alternative booster administration for the parenteral hepatitis B vaccines. This vaccine induces a Th1 associated cellular immunity and also may provide therapeutic benefit to patients with chronic hepatitis B infection who lack cellular immune responses to adequately control viral replication. Long-term stability of this vaccine formulation at elevated temperatures suggests a direct advantage in the field, since potential excursions from cold chain maintenance could be tolerated without a loss in therapeutic efficacy.


Assuntos
Emulsões , Antígenos de Superfície da Hepatite B/administração & dosagem , Vacinas contra Hepatite B/administração & dosagem , Adjuvantes Imunológicos , Animais , Formação de Anticorpos , Química Farmacêutica , Formas de Dosagem , Antígenos de Superfície da Hepatite B/imunologia , Humanos , Imunoglobulina G/sangue , Camundongos , Tamanho da Partícula , Proteínas Recombinantes/administração & dosagem , Vacinas de DNA/imunologia
11.
J Biomed Opt ; 13(3): 034008, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18601553

RESUMO

Flow cytometry is a powerful technique for quantitative characterization of fluorescence in cells. Quantitation is achieved by ensuring a high degree of uniformity in the optical excitation and detection, generally by using a highly controlled flow. Two-photon excitation has the advantages that it enables simultaneous excitation of multiple dyes and achieves a very high SNR through simplified filtering and fluorescence background reduction. We demonstrate that two-photon excitation in conjunction with a targeted multidye labeling strategy enables quantitative flow cytometry even under conditions of nonuniform flow, such as may be encountered in simple capillary flow or in vivo. By matching the excitation volume to the size of a cell, single-cell detection is ensured. Labeling cells with targeted nanoparticles containing multiple fluorophores enables normalization of the fluorescence signal and thus quantitative measurements under nonuniform excitation. Flow cytometry using two-photon excitation is demonstrated for detection and differentiation of particles and cells both in vitro in a glass capillary and in vivo in the blood stream of live mice. The technique also enables us to monitor the fluorescent dye labeling dynamics in vivo. In addition, we present a unique two-beam scanning method to conduct cell size measurement in nonuniform flow.


Assuntos
Separação Celular/instrumentação , Citometria de Fluxo/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Animais , Separação Celular/métodos , Desenho de Equipamento , Análise de Falha de Equipamento , Feminino , Citometria de Fluxo/métodos , Humanos , Células Jurkat , Camundongos , Camundongos Nus , Microscopia de Fluorescência por Excitação Multifotônica/métodos
12.
AIDS Res Hum Retroviruses ; 24(2): 271-81, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18260780

RESUMO

Epidemiological and experimental data suggest that both robust neutralizing antibodies and potent cellular responses play important roles in controlling primary HIV-1 infection. In this study we have investigated the induction of systemic and mucosal immune responses to HIV gp120 monomer immunogen administered intranasally in a novel, oil-in-water nanoemulsion (NE) adjuvant. Mice and guinea pigs intranasally immunized by the application of recombinant HIV gp120 antigen mixed in NE demonstrated robust serum anti-gp120 IgG, as well as bronchial, vaginal, and serum anti-gp120 IgA in mice. The serum of these animals demonstrated antibodies that cross-reacted with heterologous serotypes of gp120 and had significant neutralizing activity against two clade-B laboratory strains of HIV (HIVBaL and HIVSF162) and five primary HIV-1 isolates. The analysis of gp120-specific CTL proliferation, INF-gamma induction, and prevalence of anti-gp120 IgG2 subclass antibodies indicated that nasal vaccination in NE also induced systemic, Th1-polarized cellular immune responses. This study suggests that NE should be evaluated as a mucosal adjuvant for multivalent HIV vaccines.


Assuntos
Adjuvantes Imunológicos , Emulsões/administração & dosagem , Anticorpos Anti-HIV/sangue , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Linfócitos T Citotóxicos/imunologia , Administração Intranasal , Animais , Proliferação de Células , Reações Cruzadas , Feminino , Cobaias , Proteína gp120 do Envelope de HIV/administração & dosagem , Imunoglobulina A/análise , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Interferon gama/biossíntese , Pulmão/química , Pulmão/imunologia , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vagina/química , Vagina/imunologia
13.
Clin Vaccine Immunol ; 15(2): 348-58, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18057181

RESUMO

Live-virus vaccines for smallpox are effective but have risks that are no longer acceptable for routine use in populations at minimal risk of infection. We have developed a mucosal, killed-vaccinia virus (VV) vaccine based on antimicrobial nanoemulsion (NE) of soybean oil and detergent. Incubation of VV with 10% NE for at least 60 min causes the complete disruption and inactivation of VV. Simple mixtures of NE and VV (Western Reserve serotype) (VV/NE) applied to the nares of mice resulted in both systemic and mucosal anti-VV immunity, virus-neutralizing antibodies, and Th1-biased cellular responses. Nasal vaccination with VV/NE vaccine produced protection against lethal infection equal to vaccination by scarification, with 100% survival after challenge with 77 times the 50% lethal dose of live VV. However, animals protected with VV/NE immunization did after virus challenge have clinical symptoms more extensive than animals vaccinated by scarification. VV/NE-based vaccines are highly immunogenic and induce protective mucosal and systemic immunity without the need for an inflammatory adjuvant or infection with live virus.


Assuntos
Vacina Antivariólica/administração & dosagem , Vacina Antivariólica/imunologia , Varíola/prevenção & controle , Vaccinia virus/imunologia , Inativação de Vírus , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Líquido da Lavagem Broncoalveolar/imunologia , Feminino , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina A/sangue , Imunoglobulina G/análise , Imunoglobulina G/sangue , Interferon gama/biossíntese , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Varíola/imunologia , Varíola/fisiopatologia , Baço/imunologia , Análise de Sobrevida , Vacinas de Produtos Inativados/imunologia
14.
Infect Immun ; 75(8): 4020-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17502384

RESUMO

The currently available commercial human anthrax vaccine requires multiple injections for efficacy and has side effects due to its alum adjuvant. These factors limit its utility when immunizing exposed populations in emergent situations. We evaluated a novel mucosal adjuvant that consists of a nontoxic, water-in-oil nanoemulsion (NE). This material does not contain a proinflammatory component but penetrates mucosal surfaces to load antigens into dendritic cells. Mice and guinea pigs were intranasally immunized with recombinant Bacillus anthracis protective antigen (rPA) mixed in NE as an adjuvant. rPA-NE immunization was effective in inducing both serum anti-PA immunoglobulin G (IgG) and bronchial anti-PA IgA and IgG antibodies after either one or two mucosal administrations. Serum anti-PA IgG2a and IgG2b antibodies and PA-specific cytokine induction after immunization indicate a Th1-polarized immune response. rPA-NE immunization also produced high titers of lethal-toxin-neutralizing serum antibodies in both mice and guinea pigs. Guinea pigs nasally immunized with rPA-NE vaccine were protected against an intradermal challenge with approximately 1,000 times the 50% lethal dose ( approximately 1,000x LD(50)) of B. anthracis Ames strain spores (1.38 x 10(3) spores), which killed control animals within 96 h. Nasal immunization also resulted in 70% and 40% survival rates against intranasal challenge with 10x LD(50) and 100x LD(50) (1.2 x 10(6) and 1.2 x 10(7)) Ames strain spores. Our results indicate that NE can effectively adjuvant rPA for intranasal immunization. This potentially could lead to a needle-free anthrax vaccine requiring fewer doses and having fewer side effects than the currently available human vaccine.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Imunidade nas Mucosas , Nanopartículas , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antibacterianos/sangue , Antitoxinas/sangue , Bacillus anthracis/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Modelos Animais de Doenças , Emulsões , Feminino , Cobaias , Humanos , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Análise de Sobrevida , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/imunologia
15.
Nano Lett ; 5(11): 2123-30, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16277438

RESUMO

We have synthesized water-soluble, biocompatible, fluorescent, and stable silver/dendrimer nanocomposites that exhibit a potential for in vitro cell labeling. Amino-, hydroxyl-, and carboxyl-terminated ethylenediamine core generation 5 poly(amidoamine) dendrimers were utilized to prepare aqueous silver(I)-dendrimer complexes (with the molar ratio of 25 Ag+ per dendrimer) at the biologic pH of 7.4. Conversion of silver(I)-dendrimer complexes into dendrimer nanocomposites was achieved by irradiating the solutions with UV light to reduce the bound Ag+ cations to zerovalent Ag0 atoms, which were simultaneously trapped in the dendrimer network, resulting in the formation of {(Ag0)25-PAMAM_E5.NH2}, {(Ag0)25-PAMAM_E5.NGly}, and {(Ag0)25-PAMAM_E5.NSAH} dendrimer nanocomposites (DNC), respectively. The silver-DNCs were characterized by means of UV-vis, fluorescence spectroscopy, dynamic light-scattering, zeta potential measurements, high-resolution transmission electron microscopy, X-ray energy dispersive spectroscopy, and selected area electron diffraction. The cytotoxicity of dendrimers and related silver nanocomposites was evaluated using an XTT colorimetric assay of cellular viability. The cellular uptake of nanoparticles was examined by transmission electron and confocal microscopy. Results indicate that {(Ag0)25-PAMAM_E5.NH2}, {(Ag0-)25-PAMAM_E5.NGly}, and {(Ag0)25-PAMAM_E5.NSAH} form primarily single particles with diameters between 3 and 7 nm. The dendrimer nanocomposites are fluorescent, and their surface charge, cellular internalization, toxicity, and cell labeling capabilities are determined by the surface functionalities of dendrimer templates. The {(Ag0)25-PAMAM_E5.NH2} and {(Ag0)25-PAMAM_E5.NSAH} nanocomposites exhibit potential application as cell biomarkers.


Assuntos
Nanoestruturas , Animais , Materiais Biocompatíveis , Biomarcadores , Dendrímeros/análise , Dendrímeros/síntese química , Dendrímeros/toxicidade , Humanos , Camundongos , Microscopia Eletrônica , Células NIH 3T3 , Nanoestruturas/análise , Nanoestruturas/química , Nanoestruturas/toxicidade , Nanotecnologia , Poliaminas/análise , Poliaminas/síntese química , Poliaminas/toxicidade , Prata/análise , Prata/química , Prata/toxicidade , Células U937
16.
Bioconjug Chem ; 15(4): 774-82, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15264864

RESUMO

We have investigated poly(amidoamine) (PAMAM) dendrimer interactions with supported 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) lipid bilayers and KB and Rat2 cell membranes using atomic force microscopy (AFM), enzyme assays, flow cell cytometry, and fluorescence microscopy. Amine-terminated generation 7 (G7) PAMAM dendrimers (10-100 nM) were observed to form holes of 15-40 nm in diameter in aqueous, supported lipid bilayers. G5 amine-terminated dendrimers did not initiate hole formation but expanded holes at existing defects. Acetamide-terminated G5 PAMAM dendrimers did not cause hole formation in this concentration range. The interactions between PAMAM dendrimers and cell membranes were studied in vitro using KB and Rat 2 cell lines. Neither G5 amine- nor acetamide-terminated PAMAM dendrimers were cytotoxic up to a 500 nM concentration. However, the dose dependent release of the cytoplasmic proteins lactate dehydrogenase (LDH) and luciferase (Luc) indicated that the presence of the amine-terminated G5 PAMAM dendrimer decreased the integrity of the cell membrane. In contrast, the presence of acetamide-terminated G5 PAMAM dendrimer had little effect on membrane integrity up to a 500 nM concentration. The induction of permeability caused by the amine-terminated dendrimers was not permanent, and leaking of cytosolic enzymes returned to normal levels upon removal of the dendrimers. The mechanism of how PAMAM dendrimers altered cells was investigated using fluorescence microscopy, LDH and Luc assays, and flow cytometry. This study revealed that (1) a hole formation mechanism is consistent with the observations of dendrimer internalization, (2) cytosolic proteins can diffuse out of the cell via these holes, and (3) dye molecules can be detected diffusing into the cell or out of the cell through the same membrane holes. Diffusion of dendrimers through holes is sufficient to explain the uptake of G5 amine-terminated PAMAM dendrimers into cells and is consistent with the lack of uptake of G5 acetamide-terminated PAMAM dendrimers.


Assuntos
Amidas/química , Amidas/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Poliaminas/química , Poliaminas/metabolismo , Polímeros/química , Polímeros/metabolismo , Acetamidas/química , Acetamidas/metabolismo , Aminas/química , Aminas/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Permeabilidade da Membrana Celular , Corantes/química , Corantes/metabolismo , Difusão , Dimiristoilfosfatidilcolina/química , Dimiristoilfosfatidilcolina/metabolismo , Sistemas de Liberação de Medicamentos , Citometria de Fluxo , Luciferases/metabolismo , Microscopia de Força Atômica , Estrutura Molecular , Ratos
18.
Vaccine ; 21(25-26): 3801-14, 2003 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-12922114

RESUMO

Nanoemulsion, a water-in-oil formulation stabilized by small amounts of surfactant, is non-toxic to mucous membranes and produces biocidal activity against enveloped viruses. We evaluated nanoemulsion as an adjuvant for mucosal influenza vaccines. Mice (C3H/HeNHsd strain) were vaccinated intranasally with 5 x 10(5) plaque forming units (pfu) of influenza A virus (Ann Arbor/6/60 strain) and a nanoemulsion mixture. The mice were challenged on day 21 after immunization with an intranasal lethal dose of 2 x 10(5) pfu of virus. Animals vaccinated with the influenza A/nanoemulsion mixture were completely protected against infection, while animals vaccinated with either formaldehyde-killed virus or nanoemulsion alone developed viral pneumonitis and died by day 6 after the challenge. Mice vaccinated with virus/nanoemulsion mixture had rapid cytokine responses followed by high levels of specific anti-influenza immunoglobulin G (IgG) and immunoglobulin A (IgA) antibodies. Specificity of the immune response was confirmed by assessment of the proliferation and cytokine production in splenocytes. This paper demonstrates that nanoemulsion can be employed as a non-toxic mucosal adjuvant for influenza virus vaccine.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Pneumonia/prevenção & controle , Administração Intranasal , Animais , Especificidade de Anticorpos , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/virologia , Divisão Celular/fisiologia , Citocinas/biossíntese , Emulsões , Feminino , Citometria de Fluxo , Imunidade nas Mucosas/efeitos dos fármacos , Imunidade nas Mucosas/imunologia , Imunização , Imunoglobulina A/biossíntese , Imunoglobulina G/biossíntese , Vacinas contra Influenza/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Pneumonia/imunologia , RNA Viral/análise , RNA Viral/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA