Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
NPJ Vaccines ; 9(1): 4, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167505

RESUMO

Here, we assessed the efficacy of a lipid nanoparticle-based mRNA vaccine candidate encoding the receptor-binding domain (LNP-mRNA-RBD) in mice. Mice immunized with LNP-mRNA-RBD based on the ancestral strain (ancestral-type LNP-mRNA-RBD) showed similar cellular responses against the ancestral strain and BA.5, but their neutralizing activity against BA.5 was lower than that against the ancestral strain. The ancestral-type LNP-mRNA-RBD protected mice from the ancestral strain or BA.5 challenge; however, its ability to reduce the viral burdens after BA.5 challenge was limited. In contrast, immunization with bivalent LNP-mRNA-RBD consisting of the ancestral-type and BA.4/5-type LNP-mRNA-RBD or monovalent BA.4/5-type LNP-mRNA-RBD elicited robust cellular responses, as well as high and moderate neutralizing titers against BA.5 and XBB.1.5, respectively. Furthermore, the vaccines containing BA.4/5-type LNP-mRNA-RBD remarkably reduced the viral burdens following BA.5 or XBB.1.5 challenge. Overall, our findings suggest that LNP-mRNA-RBD is effective against SARS-CoV-2 infection.

2.
Int Immunol ; 34(7): 353-364, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35419609

RESUMO

Agonists for TLR9 and stimulator of IFN genes (STING) offer therapeutic applications as both anti-tumor agents and vaccine adjuvants, though their clinical applications are limited; the clinically available TLR9 agonist is a weak IFN inducer and STING agonists induce undesired type 2 immunity. Yet, combining TLR9 and STING agonists overcame these limitations by synergistically inducing innate and adaptive IFNγ to become an advantageous type 1 adjuvant, suppressing type 2 immunity, in addition to exerting robust anti-tumor activities when used as a monotherapeutic agent for cancer immunotherapy. Here, we sought to decipher the immunological mechanisms behind the synergism mediated by TLR9 and STING agonists and found that their potent anti-tumor immunity in a Pan02 peritoneal dissemination model of pancreatic cancer was achieved only when agonists for TLR9 and STING were administered locally, and was via mechanisms involving CD4 and CD8 T cells as well as the co-operative action of IL-12 and type I IFNs. Rechallenge studies of long-term cancer survivors suggested that the elicitation of Pan02-specific memory responses provides protection against the secondary tumor challenge. Mechanistically, we found that TLR9 and STING agonists synergistically induce IL-12 and type I IFN production in murine APCs. The synergistic effect of the TLR9 and STING agonists on IL-12p40 was at protein, mRNA and promoter activation levels, and transcriptional regulation was mediated by a 200 bp region situated 983 bp upstream of the IL-12p40 transcription initiation site. Such intracellular transcriptional synergy may hold a key in successful cancer immunotherapy and provide further insights into dual agonism of innate immune sensors during host homeostasis and diseases.


Assuntos
Proteínas de Membrana , Neoplasias , Receptor Toll-Like 9 , Adjuvantes Imunológicos/farmacologia , Animais , Imunoterapia , Interleucina-12 , Subunidade p40 da Interleucina-12 , Proteínas de Membrana/metabolismo , Camundongos , Receptor Toll-Like 9/metabolismo
3.
J Immunol ; 198(12): 4707-4715, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28507029

RESUMO

DNA vaccines are attractive immunogens for priming humoral and cellular immune responses to the encoded Ag. However, their ability to induce Ag-specific CD8+ T cell responses requires improvement. Among the strategies for improving DNA vaccine immunogenicity are booster vaccinations, alternate vaccine formulations, electroporation, and genetic adjuvants, but few, such as extracellular vesicles (EVs), target natural Ag delivery systems. By focusing on CD63, a tetraspanin protein expressed on various cellular membranes, including EVs, we examined whether a DNA vaccine encoding an Ag fused to CD63 delivered into EVs would improve vaccine immunogenicity. In vitro transfection with plasmid DNA encoding an OVA Ag fused to CD63 (pCD63-OVA) produced OVA-carrying EVs. Immunizations with the purified OVA-carrying EVs primed naive mice to induce OVA-specific CD4+ and CD8+ T cells, whereas immunization with EVs purified from cells transfected with control plasmids encoding OVA protein alone or a calnexin-OVA fusion protein delivered into the endoplasmic reticulum failed to do so. Vaccinating mice with pCD63-OVA induced potent Ag-specific T cell responses, particularly those from CD8+ T cells. CD63 delivery into EVs led to better CD8+ T cell responses than calnexin delivery into the endoplasmic reticulum. When we used a mouse tumor implantation model to evaluate pCD63-OVA as a therapeutic vaccine, the EV-delivered DNA vaccination significantly inhibited tumor growth compared with the control DNA vaccinations. These results indicate that EV Ag delivery via DNA vaccination offers a new strategy for eliciting strong CD8+ T cell responses to the encoded Ag, making it a potentially useful cancer vaccine.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vesículas Extracelulares/imunologia , Ativação Linfocitária , Tetraspanina 30/imunologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/imunologia , Feminino , Imunidade Celular , Imunização Secundária , Imunogenicidade da Vacina , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Tetraspanina 30/genética , Vacinas de DNA/administração & dosagem
4.
EBioMedicine ; 16: 124-135, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28111238

RESUMO

Respiratory syncytial virus (RSV) is one of the most prevalent causative agents of lower respiratory tract infections worldwide, especially in infants around 3 to 4months old. Infants at such a young age have maternally-transferred passive antibodies against RSV but do not have active immune systems efficient enough for the control of RSV infection. In order to elucidate age-specific profiles of immune responses against RSV protection, antibody responses were examined by using blood samples in both acute and convalescent phases obtained from child patients and adult patients. In addition to the serum neutralization activity, antibody responses to the RSV fusion protein (F protein) were dissected by analyzing levels of total IgG, IgG subclasses, the binding stability, and the levels of antibody for the neutralization epitopes. It was suggested that children's antibody responses against RSV are matured over months and years in at least 5 stages based on 1) levels of the neutralization titer and IgG3 for F protein in the convalescent phase, 2) geometric mean ratios of the neutralization titers and levels of IgG1 and IgG2 for F protein in the convalescent phase compared to those levels in the acute phase, 3) the affinity maturation of IgG for F protein and the cross reactivity of IgG for RSV glycoproteins of groups A and B, 4) levels of neutralization epitope-specific IgG, and 5) augmentation of overall antibody responses due to repetitive RSV infection.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/imunologia , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antivirais/sangue , Linhagem Celular Tumoral , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Infecções por Vírus Respiratório Sincicial/sangue , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia
5.
EBioMedicine ; 9: 87-96, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27333035

RESUMO

The innate immune protein Stimulator of interferon genes (STING) promotes the induction of interferon beta (IFN-ß) production via the phosphorylation of its C-terminal tail (CTT) by TANK-binding kinase 1 (TBK1). Potent ligands of STING are, therefore, promising candidates for novel anti-cancer drugs or vaccine adjuvants. However, the intrinsically flexible CTT poses serious problems in in silico drug discovery. Here, we performed molecular dynamics simulations of the STING fragment containing the CTT in ligand-bound and unbound forms and observed that the binding of a potent ligand cyclic GMP-AMP (cGAMP) induced a local structure in the CTT, reminiscent of the known structure of a TBK1 substrate. The subsequent molecular biological experiments confirmed the observed dynamics of the CTT and identified essential residues for the activation of the IFN-ß promoter, leading us to propose a new mechanism of STING activation.


Assuntos
Ligantes , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sítios de Ligação , AMP Cíclico/química , AMP Cíclico/metabolismo , GMP Cíclico/química , GMP Cíclico/metabolismo , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Interferon beta/genética , Interferon beta/metabolismo , Proteínas de Membrana/genética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Domínios Proteicos , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética
6.
J Dermatol Sci ; 82(1): 38-45, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26674124

RESUMO

BACKGROUND: It has been clinically demonstrated that intradermal (ID) vaccines have a potential to confer a superior immunogenic profile compared to intramuscular (IM) or subcutaneous (SC) vaccines. In terms of distribution of a vaccine antigen depending on the administration routes, at least two independent immunogenic pathways of the vaccines have been proposed: (1) the antigen recognition by the immune cells present at the vaccine-administered site and (2) the antigen recognition by the lymph node (LN)-resident immune cells through the lymphatic flow from the vaccine-administered site after the antigen is directly delivered into the draining LNs. OBJECTIVE: In order to clarify the key components for the immunogenic pathway of the ID vaccine, the correlation between the kinetics of the antigen distribution to the draining LNs and antibody responses to the antigen were evaluated. METHODS: We compared the antibody responses in the groups with by surgical removal of the administration site immediately after the ID administration, and by surgical removal of the draining LNs before the ID administration. RESULTS: The results suggested that the efficient and direct antigen delivery to the draining LNs plays an important role in the antibody responses to the ID vaccine. Indeed, it was confirmed that the direct administration into the draining LNs with the antigen elicited comparable levels of the antibody responses with the ID vaccine. At the cellular level, it was shown that the LN-resident immune cells such as B cells, dendritic cells, and macrophages including medullary macrophages and subcapsular sinus macrophages interacting with the antigens following the ID administration. Finally, we demonstrated by immunofluorescence analysis that the lymphatic vessels are more diffusely distributed in the dermis as compared with the subcutaneous area and muscle. CONCLUSION: The results of the present study suggested that the skin is an optimal tissue to facilitate the vaccine antigen access to the draining LNs, which is an important immunogenic pathway of the ID vaccine. Further elucidation of regulatory mechanisms underlying such an immunogenic pathway of the ID vaccine would provide us with elements for the development of novel adjuvants and devices to enhance the immunogenicity of the ID vaccines.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/administração & dosagem , Imunogenicidade da Vacina , Vacinas contra Influenza/administração & dosagem , Linfonodos/imunologia , Pele/imunologia , Vacinação , Animais , Anticorpos Antivirais/sangue , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vacinas contra Influenza/imunologia , Injeções Intradérmicas , Injeções Subcutâneas , Cinética , Linfonodos/metabolismo , Camundongos Endogâmicos BALB C , Pele/metabolismo
7.
Eur J Immunol ; 45(4): 1159-69, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25529558

RESUMO

Agonists for TLR9 and Stimulator of IFN Gene (STING) act as vaccine adjuvants that induce type-1 immune responses. However, currently available CpG oligodeoxynucleotide (ODN) (K-type) induces IFNs only weakly and STING ligands rather induce type-2 immune responses, limiting their potential therapeutic applications. Here, we show a potent synergism between TLR9 and STING agonists. Together, they make an effective type-1 adjuvant and an anticancer agent. The synergistic effect between CpG ODN (K3) and STING-ligand cyclic GMP-AMP (cGAMP), culminating in NK cell IFN-γ (type-II IFN) production, is due to the concurrent effects of IL-12 and type-I IFNs, which are differentially regulated by IRF3/7, STING, and MyD88. The combination of CpG ODN with cGAMP is a potent type-1 adjuvant, capable of inducing strong Th 1-type responses, as demonstrated by enhanced antigen-specific IgG2c and IFN-γ production, as well as cytotoxic CD8(+) T-cell responses. In our murine tumor models, intratumoral injection of CpG ODN and cGAMP together reduced tumor size significantly compared with the singular treatments, acting as an antigen-free anticancer agent. Thus, the combination of CpG ODN and a STING ligand may offer therapeutic application as a potent type-II IFN inducer.


Assuntos
Interferon gama/biossíntese , Proteínas de Membrana/agonistas , Neoplasias/terapia , Nucleotídeos Cíclicos/farmacologia , Oligodesoxirribonucleotídeos/farmacologia , Linfócitos T Citotóxicos/imunologia , Receptor Toll-Like 9/agonistas , Adjuvantes Imunológicos/farmacologia , Animais , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Interferon gama/imunologia , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo
8.
Hum Vaccin Immunother ; 9(10): 2216-21, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23912600

RESUMO

Since the introduction of DNA vaccines two decades ago, this attractive strategy has been hampered by its low immunogenicity in humans. Studies conducted to improve the immunogenicity of DNA vaccines have shown that understanding the mechanism of action of DNA vaccines might be the key to successfully improving their immunogenicity. Our current understanding is that DNA vaccines induce innate and adaptive immune responses in two ways: (1) encoded protein (or polypeptide) antigen(s) by the DNA plasmid can be expressed in stromal cells (i.e., muscle cells) as well as DCs, where these antigens are processed and presented to naïve CD4 or CD8 T cells either by direct or cross presentation, respectively; and (2) the transfected DNA plasmid itself may bind to an un-identified cytosolic DNA sensor and activate the TBK1-STING pathway and the production of type I interferons (IFNs) which function as an adjuvant. Recent studies investigating double-stranded cytosolic DNA sensor(s) have highlighted new mechanisms in which cytosolic DNA may release secondary metabolites, which are in turn recognized by a novel DNA sensing machinery. Here, we discuss these new metabolites and the possibilities of translating this knowledge into improved immunogenicity for DNA vaccines.


Assuntos
Vacinas de DNA/imunologia , Imunidade Adaptativa , Adjuvantes Imunológicos/metabolismo , Animais , DNA/metabolismo , Humanos , Imunidade Inata , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia
9.
Front Genet ; 4: 91, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23734163

RESUMO

Histones are essential components of chromatin structure, and histone modification plays an important role in various cellular functions including transcription, gene silencing, and immunity. Histones also play distinct roles in extrachromosomal settings. Extrachromosomal histone H2B acts as a cytosolic sensor to detect double-stranded DNA (dsDNA) fragments derived from infectious agents or damaged cells to activate innate and acquired immune responses in various cell types. It also physically interacts with interferon (IFN)-ß promoter stimulator 1 (IPS-1), an essential adaptor molecule that activates innate immunity, through COOH-terminal importin 9-related adaptor organizing histone H2B and IPS-1 (CIAO), resulting in a distinct signaling complex that induces dsDNA-induced type I IFN production. Such a molecular platform acts as a cellular sensor to recognize aberrant dsDNA in cases of viral infection and cell damage. This mechanism may also play roles in autoimmunity, transplantation rejection, gene-mediated vaccines, and other therapeutic applications.

10.
PLoS One ; 8(3): e60038, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555875

RESUMO

5,6-Dimethylxanthenone-4-acetic acid (DMXAA), a potent type I interferon (IFN) inducer, was evaluated as a chemotherapeutic agent in mouse cancer models and proved to be well tolerated in human cancer clinical trials. Despite its multiple biological functions, DMXAA has not been fully characterized for the potential application as a vaccine adjuvant. In this report, we show that DMXAA does act as an adjuvant due to its unique property as a soluble innate immune activator. Using OVA as a model antigen, DMXAA was demonstrated to improve on the antigen specific immune responses and induce a preferential Th2 (Type-2) response. The adjuvant effect was directly dependent on the IRF3-mediated production of type-I-interferon, but not IL-33. DMXAA could also enhance the immunogenicity of influenza split vaccine which led to significant increase in protective responses against live influenza virus challenge in mice compared to split vaccine alone. We propose that DMXAA can be used as an adjuvant that targets a specific innate immune signaling pathway via IRF3 for potential applications including vaccines against influenza which requires a high safety profile.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Fator Regulador 3 de Interferon/metabolismo , Xantonas/uso terapêutico , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade Inata/efeitos dos fármacos , Fator Regulador 3 de Interferon/genética , Interleucina-33 , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Camundongos , Camundongos Knockout
11.
Vaccines (Basel) ; 1(3): 278-92, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-26344113

RESUMO

DNA vaccines can induce both humoral and cellular immune responses. Although some DNA vaccines are already licensed for infectious diseases in animals, they are not licensed for human use because the risk and benefit of DNA vaccines is still controversial. Indeed, in humans, the immunogenicity of DNA vaccines is lower than that of other traditional vaccines. To develop the use of DNA vaccines in the clinic, various approaches are in progress to enhance or improve the immunogenicity of DNA vaccines. Recent studies have shown that immunogenicity of DNA vaccines are regulated by innate immune responses via plasmid DNA recognition through the STING-TBK1 signaling cascade. Similarly, molecules that act as dsDNA sensors that activate innate immune responses through STING-TBK1 have been identified and used as genetic adjuvants to enhance DNA vaccine immunogenicity in mouse models. However, the mechanisms that induce innate immune responses by DNA vaccines are still unclear. In this review, we will discuss innate immune signaling upon DNA vaccination and genetic adjuvants of innate immune signaling molecules.

12.
Adv Exp Med Biol ; 738: 60-78, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22399374

RESUMO

In order to survive, all organisms must recognize and eliminate foreign invaders such as infectious pathogens, chemicals, ultraviolet rays, metabolites and damaged or transformed self-tissues, as well as allogenic organs in cases of transplantation. Recent research in innate immunity has elucidated that there are versatile inflammatory sensors on spatiotemporal 'sentry duty' that recognize substances derived from both 'nonself' and 'self', e.g., Toll-like receptors, retinoic acid-inducible gene-I-like receptors, nucleotide oligomerization domain-like receptors and c-type lectin receptors. Having acquired high-level functions through the development of multiple molecules, higher organisms have established both extracellular and intracellular sensors that can discriminate danger-associated molecular patterns from promiscuous, but biologically similar, molecular patterns. In addition, 'loss-of-function' or 'gain-of-function' mutations in these inflammatory sensors have been linked (at least in part) with the etiology and severity of autoimmune diseases, autoinflammatory diseases and immunocompromised diseases in humans. Further studies focusing on the role of these inflammatory sensors in the development of immune disorders would highlight new avenues for the development of novel diagnostic and therapeutic applications with regard to these diseases.


Assuntos
Histocompatibilidade/fisiologia , Imunidade Inata/fisiologia , Receptores Toll-Like/imunologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Mutação , Receptores Toll-Like/genética , Transativadores , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-23316484

RESUMO

All mammalian cells are equipped with large numbers of sensors for protection from various sorts of invaders, who, in turn, are equipped with molecules containing pathogen-associated molecular patterns (PAMPs). Once these sensors recognize non-self antigens containing PAMPs, various physiological responses including inflammation are induced to eliminate the pathogens. However, the host sometimes suffers from chronic infection or continuous injuries, resulting in production of self-molecules containing damage-associated molecular patterns (DAMPs). DAMPs are also responsible for the elimination of pathogens, but promiscuous recognition of DAMPs through sensors against PAMPs has been reported. Accumulation of DAMPs leads to massive inflammation and continuous production of DAMPs; that is, a vicious circle leading to the development of autoimmune disease. From a vaccinological point of view, the accurate recognition of both PAMPs and DAMPs is important for vaccine immunogenicity, because vaccine adjuvants are composed of several PAMPs and/or DAMPs, which are also associated with severe adverse events after vaccination. Here, we review as the roles of PAMPs and DAMPs upon infection with pathogens or inflammation, and the sensors responsible for recognizing them, as well as their relationship with the development of autoimmune disease or the immunogenicity of vaccines.


Assuntos
Imunidade Inata , Infecções/imunologia , Inflamação/imunologia , Ácidos Nucleicos/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais , Vacinas/imunologia , Imunidade Adaptativa , Animais , Doenças Autoimunes , Humanos , Ácidos Nucleicos/metabolismo , Receptores Imunológicos/metabolismo
14.
J Immunol ; 186(3): 1646-55, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21209283

RESUMO

Although more than 20 putative members have been assigned to the nucleotide-binding and oligomerization domain-like receptor (NLR) family, their physiological and biological roles, with the exception of the inflammasome, are not fully understood. In this article, we show that NLR members, such as NLRC4, NLRP3, NLRP4, and NLRP10 interact with Beclin1, an important regulator of autophagy, through their neuronal apoptosis inhibitory protein, MHC class II transcription activator, incompatibility locus protein from Podospora anserina, and telomerase-associated protein domain. Among such NLRs, NLRP4 had a strong affinity to the Beclin1 evolutionally conserved domain. Compromising NLRP4 via RNA interference resulted in upregulation of the autophagic process under physiological conditions and upon invasive bacterial infections, leading to enhancement of the autophagic bactericidal process of group A streptococcus. NLRP4 recruited to the subplasma membrane phagosomes containing group A streptococcus and transiently dissociated from Beclin1, suggesting that NLRP4 senses bacterial infection and permits the initiation of Beclin1-mediated autophagic responses. In addition to a role as a negative regulator of the autophagic process, NLRP4 physically associates with the class C vacuolar protein-sorting complex, thereby negatively regulating maturation of the autophagosome and endosome. Collectively, these results provide novel evidence that NLRP4, and possibly other members of the NLR family, plays a crucial role in biogenesis of the autophagosome and its maturation by the association with regulatory molecules, such as Beclin1 and the class C vacuolar protein-sorting complex.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Autofagia/imunologia , Regulação para Baixo/imunologia , Farmacorresistência Bacteriana/imunologia , Proteínas de Membrana/fisiologia , Proteínas Repressoras/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Atividade Bactericida do Sangue/imunologia , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Lisossomos/imunologia , Lisossomos/microbiologia , Lisossomos/patologia , Proteínas de Membrana/metabolismo , Camundongos , Fagossomos/imunologia , Fagossomos/microbiologia , Fagossomos/patologia , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/imunologia , Vacúolos/imunologia , Vacúolos/microbiologia , Vacúolos/patologia
15.
Crit Rev Immunol ; 30(5): 395-421, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21083523

RESUMO

Protein-transduction technology is one of the most promising therapeutic tools for the control of intracellular events. A number of studies have demonstrated that minimal and efficient protein-transduction domains (PTDs) can act as a peptide vector to transfer bioactive cargo molecules from outside to inside the cell. PTD-mediated transduction has the ability to cross the blood-brain barrier, with transduction taking place in most tissues and cell types in vivo. Thus, recombinant proteins fused to or conjugated with PTDs have the potential to be harnessed as supplementary and/or intervention agents directly modulating cell signaling and/or metabolism, or to be applied to vaccine antigens/adjuvants that are efficiently delivered to the optimal site of action to enhance vaccine immunogenicity. This review introduces the mechanism of action, recent applications, and future perspectives of protein-transduction technology as an alternative therapeutic in the post-genome era.


Assuntos
Proteínas/metabolismo , Transdução de Sinais , Animais , Engenharia Genética , Humanos , Espaço Intracelular/metabolismo , Proteínas/química
16.
Vaccine ; 28(31): 4920-7, 2010 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-20566393

RESUMO

In this study, we explored the possibility of augmenting human immunodeficiency virus (HIV) gp120-specific cell-mediated immune responses in mice by means of a DNA vaccine encoding a mouse Ig Fcgamma2a fragment fused with gp120 (gp120-Ig, Ig-gp120). Western blotting analysis revealed that the HIV gp120 protein expression efficiency was higher in cells transfected with the gp120-Ig-coding plasmid (pGp120Ig) than in those transfected with the gp120 and Ig-gp120 expression plasmids (pGp120 and pIgGp120, respectively). pGp120Ig elicited more HIV-specific CD8 T cells and effector memory CD8 T cells than pGp120 in immunized mice. Furthermore, pGp120Ig significantly reduced the viral load after challenge with an HIV Env gp160-expressing vaccinia virus. These results demonstrate that covalent antigen modification with an Ig sequence can modulate antigen-specific cellular immune responses. The approach may be useful for vaccine development.


Assuntos
Vacinas contra a AIDS/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Imunidade Celular , Vacinas de DNA/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Eletroporação , Feminino , Proteína gp120 do Envelope de HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Humanos , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Carga Viral
17.
J Virol ; 84(2): 822-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19906922

RESUMO

Fragments of double-stranded DNA (dsDNA) forming a right-handed helical structure (B-DNA) stimulate cells to produce type I interferons (IFNs). While an adaptor molecule, IFN-beta promoter stimulator 1 (IPS-1), mediates dsDNA-induced cellular signaling in human cells, the underlying molecular mechanism is not fully understood. Here, we demonstrate that the extrachromosomal histone H2B mediates innate antiviral immune responses in human cells. H2B physically interacts with IPS-1 through the association with a newly identified adaptor, CIAO (COOH-terminal importin 9-related adaptor organizing histone H2B and IPS-1), to transmit the cellular signaling for dsDNA but not immunostimulatory RNA. Extrachromosomal histone H2B was biologically crucial for cell-autonomous responses to protect against multiplication of DNA viruses but not an RNA virus. Thus, the present findings provide evidence indicating that the extrachromosomal histone H2B is engaged in the signaling pathway initiated by dsDNA to trigger antiviral innate immune responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antivirais/metabolismo , DNA/imunologia , Histonas/metabolismo , Imunidade Inata/imunologia , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Chlorocebus aethiops , DNA/metabolismo , Vírus de DNA/fisiologia , Regulação da Expressão Gênica , Células HeLa , Histonas/genética , Humanos , Interferon beta/metabolismo , Camundongos , Células NIH 3T3 , Regiões Promotoras Genéticas , Células Vero
18.
Autophagy ; 4(1): 67-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17921696

RESUMO

Recent research on autophagy clearly demonstrates that the autophagosome-lysosome pathway plays essential roles in elimination of certain pathogens such as group A Streptococcus, Mycobacterium tuberculosis, Listeria monocytogenes, and herpesvirus. (1-4) We have recently found that a key regulator of the autophagic process, the Atg12-Atg5 conjugate, associates with the signaling molecules retinoic acid-inducible gene I (RIG-I) and interferon-beta promoter stimulator 1 (IPS-1), which are essential for recognition of RNA virus infection and which transmit signals to upregulate type I interferons (IFNs). Interestingly, the Atg12-Atg5 conjugate seemed to negatively regulate the type I IFN modulating pathway through direct interaction with caspase recruitment domains (CARDs) presented by RIG-1 and IPS-1.(5) Thus, in contrast to the bactericidal properties of autophagic processes, the autophagy regulator (the Atg12-Atg5 conjugate) appeared to promote RNA virus replication by inhibiting innate anti-virus immune responses. In this addendum, we discuss the non-canonical role of the Atg12-Atg5 conjugate as a suppressor of innate immune responses.


Assuntos
Proteínas de Bactérias/metabolismo , Imunidade Inata , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia/fisiologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Humanos , Fagossomos/metabolismo , Vírus de RNA/genética , Vírus de RNA/metabolismo , Vírus de RNA/patogenicidade , Receptores Imunológicos , Viroses/imunologia , Replicação Viral
19.
Proc Natl Acad Sci U S A ; 104(35): 14050-5, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17709747

RESUMO

Autophagy is an essential process for physiological homeostasis, but its role in viral infection is only beginning to be elucidated. We show here that the Atg5-Atg12 conjugate, a key regulator of the autophagic process, plays an important role in innate antiviral immune responses. Atg5-deficient mouse embryonic fibroblasts (MEFs) were resistant to vesicular stomatitis virus replication, which was largely due to hyperproduction of type I interferons in response to immunostimulatory RNA (isRNA), such as virus-derived, double-stranded, or 5'-phosphorylated RNA. Similar hyperresponse to isRNA was also observed in Atg7-deficient MEFs, in which Atg5-Atg12 conjugation is impaired. Overexpression of Atg5 or Atg12 resulted in Atg5-Atg12 conjugate formation and suppression of isRNA-mediated signaling. Molecular interaction studies indicated that the Atg5-Atg12 conjugate negatively regulates the type I IFN production pathway by direct association with the retinoic acid-inducible gene I (RIG-I) and IFN-beta promoter stimulator 1 (IPS-1) through the caspase recruitment domains (CARDs). Thus, in contrast to its role in promoting the bactericidal process, a component of the autophagic machinery appears to block innate antiviral immune responses, thereby contributing to RNA virus replication in host cells.


Assuntos
Proteínas Associadas aos Microtúbulos/imunologia , Proteínas/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Autofagia , Proteína 12 Relacionada à Autofagia , Proteína 5 Relacionada à Autofagia , Células Cultivadas , Fibroblastos/fisiologia , Genes Reporter , Imunidade Inata , Interferon Tipo I/genética , Camundongos , NF-kappa B/genética , Regiões Promotoras Genéticas , RNA de Cadeia Dupla/genética , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral
20.
J Immunol ; 179(2): 1147-54, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17617608

RESUMO

Flagellin is a key component of the flagella of many pathogens, including Pseudomonas aeruginosa. Flagellin is an attractive vaccine candidate because it is readily produced and manipulated as a recombinant protein and has intrinsic adjuvant activity mediated through TLR5. Although DNA vaccines encoding native Pseudomonas B-type (FliC) or A-type (FlaA) flagellin are strongly immunogenic, the resultant Ab response interferes with the interaction of homologous flagellin with TLR5. This reduces the ability of the host to clear homologous, but not heterologous, flagellin-expressing P. aeruginosa. To circumvent this problem, a DNA vaccine encoding a mutant FliC R90A flagellin was developed. The mutant Ag encoded by this vaccine was highly immunogenic, but its ability to interact with TLR5 was reduced by >100-fold. Vaccination with this flagellin mutant DNA vaccine induced cross-reactive Abs against both FliC and FlaA, but few Abs capable of interfering with TLR5 activation. The flagellin mutant DNA vaccine provided excellent protection against both FliC- and FlaA-expressing P. aeruginosa. These findings suggest that vaccines against flagellated pathogens should avoid inducing Abs against TLR5 and raise the possibility that flagellated bacteria evade host elimination by facilitating the production of Abs that reduce the host's ability to mount an innate immune response.


Assuntos
Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Flagelina/imunologia , Infecções por Pseudomonas/prevenção & controle , Receptor 5 Toll-Like/metabolismo , Vacinas de DNA/imunologia , Sequência de Aminoácidos , Animais , Western Blotting , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Feminino , Flagelina/genética , Camundongos , Dados de Sequência Molecular , Mutação , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Receptor 5 Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...