Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 21(1): 479, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38093320

RESUMO

Vaccination is still the most promising strategy for combating influenza virus pandemics. However, the highly variable characteristics of influenza virus make it difficult to develop antibody-based universal vaccines, until now. Lung tissue-resident memory T cells (TRM), which actively survey tissues for signs of infection and react rapidly to eliminate infected cells without the need for a systemic immune reaction, have recently drawn increasing attention towards the development of a universal influenza vaccine. We previously designed a sequential immunization strategy based on orally administered Salmonella vectored vaccine candidates. To further improve our vaccine design, in this study, we used two different dendritic cell (DC)-targeting strategies, including a single chain variable fragment (scFv) targeting the surface marker DC-CD11c and DC targeting peptide 3 (DCpep3). Oral immunization with Salmonella harboring plasmid pYL230 (S230), which displayed scFv-CD11c on the bacterial surface, induced dramatic production of spleen effector memory T cells (TEM). On the other hand, intranasal boost immunization using purified DCpep3-decorated 3M2e-ferritin nanoparticles in mice orally immunized twice with S230 (S230inDC) significantly stimulated the differentiation of lung CD11b+ DCs, increased intracellular IL-17 production in lung CD4+ T cells and elevated chemokine production in lung sections, such as CXCL13 and CXCL15, as determined by RNAseq and qRT‒PCR assays, resulting in significantly increased percentages of lung TRMs, which could provide efficient protection against influenza virus challenge. The dual DC targeting strategy, together with the sequential immunization approach described in this study, provides us with a novel "prime and pull" strategy for addressing the production of protective TRM cells in vaccine design.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Camundongos , Animais , Células T de Memória , Pulmão , Células Dendríticas , Infecções por Orthomyxoviridae/prevenção & controle
2.
Int J Mol Sci ; 24(15)2023 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-37569548

RESUMO

Finding a long-term cure for tumor patients still represents a major challenge. Immunotherapies offer promising therapy options, since they are designed to specifically prime the immune system against the tumor and modulate the immunosuppressive tumor microenvironment. Using nucleic-acid-based vaccines or cellular vaccines often does not achieve sufficient activation of the immune system in clinical trials. Additionally, the rapid degradation of drugs and their non-specific uptake into tissues and cells as well as their severe side effects pose a challenge. The encapsulation of immunomodulatory molecules into nanocarriers provides the opportunity of protected cargo transport and targeted uptake by antigen-presenting cells. In addition, different immunomodulatory cargos can be co-delivered, which enables versatile stimulation of the immune system, enhances anti-tumor immune responses and improves the toxicity profile of conventional chemotherapeutic agents.

3.
Nano Lett ; 23(7): 2733-2742, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-36930562

RESUMO

Dendritic cells (DCs) play an essential role in both the induction of the immune response and the maintenance of immune tolerance, with any malfunction of DCs potentially causing several diseases. While gene-based therapy for DC manipulation is a promising approach, it remains challenging due to the lack of efficient delivery systems for DC targeting. Herein, we describe a novel bacterial nanomedicine (BNM) system for pathogen recognition-mediated DCs-specific gene silencing and gene editing. BNMs contain components from bacterial outer membranes and achieve efficient DC targeting through the recognition of pathogen-associated molecular patterns by pattern recognition receptors on DCs. The targeting efficiency of BNMs is reduced in DCs lacking toll-like receptor 4, which is responsible for recognizing lipopolysaccharide, a major component of the bacterial outer membrane. As a proof-of-concept demonstration, we present gene-based therapy mediated by BNMs for enhancing antigen cross-presentation in DCs, which generates a remarkable antitumor effect.


Assuntos
Apresentação de Antígeno , Lipopolissacarídeos , Células Dendríticas , Inativação Gênica
4.
Front Microbiol ; 13: 937192, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36003947

RESUMO

A universal influenza vaccine is required for broad protection against influenza infection. Here, we revealed the efficacy of novel influenza vaccine candidates based on Ebola glycoprotein dendritic cell (DC)-targeting domain (EΔM) fusion protein technology. The four copies of ectodomain matrix protein of influenza (tM2e) or M2e hemagglutinin stalk (HA stalk) peptides (HM2e) were fused with EΔM to generate EΔM-tM2e or EΔM-HM2e, respectively. We demonstrated that EΔM-HM2e- or EΔM-tM2e-pseudotyped viral particles can efficiently target DC/macrophages in vitro and induced significantly high titers of anti-HA and/or anti-M2e antibodies in mice. Significantly, the recombinant vesicular stomatitis virus (rVSV)-EΔM-tM2e and rVSV-EΔM-HM2e vaccines mediated rapid and potent induction of M2 or/and HA antibodies in mice sera and mucosa. Importantly, vaccination of rVSV-EΔM-tM2e or rVSV-EΔM-HM2e protected mice from influenza H1N1 and H3N2 challenges. Taken together, our study suggests that rVSV-EΔM-tM2e and rVSV-EΔM-HM2e are promising candidates that may lead to the development of a universal vaccine against different influenza strains.

5.
Cell Rep ; 39(9): 110901, 2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35649357

RESUMO

Antibodies are important for vaccine efficacy. Targeting antigens to antigen-presenting cells (APCs) increases antibody levels. Here, we explore the role of antigen valency in MHC class II (MHCII)-targeted vaccines delivered as DNA. We design heterodimeric proteins that carry either two identical (bivalent vaccines), or two different antigens (monovalent vaccines). Bivalent vaccines with two identical influenza hemagglutinins (HA) elicit higher amounts of anti-HA antibodies in mice than monovalent versions with two different HAs. Bivalent vaccines increase the levels of germinal center (GC) B cells and long-lived plasma cells. Only HA-bivalent vaccines completely protect mice against challenge with homologous influenza virus. Similar results are obtained with other antigens by targeting CD11c and Xcr1 on dendritic cells (DCs) or when administering the vaccine as protein with adjuvant. Bivalency probably increases B cell responses by cross-linking BCRs in readily observable DC-B cell synapses. These results are important for generating potent APC-targeted vaccines.


Assuntos
Vacinas Anticâncer , Vacinas contra Influenza , Vacinas de DNA , Animais , Anticorpos Antivirais , Células Apresentadoras de Antígenos , Hemaglutininas , Camundongos , Vacinas Combinadas , Vacinas de DNA/genética
6.
Biomaterials ; 286: 121542, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35594837

RESUMO

Therapeutic cancer vaccines (TCVs) should induce robust tumor-specific T cell responses. To achieve this, TCVs incorporate T cell epitopes and strong adjuvants. Here, we report an all-in-one adjuvanted cancer vaccine platform that targets the intracellular compartment of dentritic cells and subsequently induces effective cytotoxic T cell responses. We screened a novel peptide (DCpep6) that specifically binds and transmits into CD11c+ cells through a novel in vivo phage biopanning. We then engineered a protein-based TCV (DEF) consisting of DCpep6 (D), an optimized HPV E7 tumor antigen (E), and a built-in flagellin adjuvant (F) as a single molecule. DEF was stably expressed, and each component was functional. In vivo-administered DEF rapidly biodistributed in draining LNs and internalized into CD11c+ cells. DEF immunization elicited strong antitumor T cell responses and provided long-term survival of TC-1 tumor-implanted mice. The DEF-mediated antitumor effect was abolished in NLRC4-/- mice. Taken together, we propose a protein-based all-in-one TCV platform that intracellularly codelivers tumor antigen and inflammasome activator to DCs to induce long-lasting antitumor T cell responses.


Assuntos
Vacinas Anticâncer , Neoplasias , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos , Citosol , Células Dendríticas , Inflamassomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo
7.
Vaccine ; 40(16): 2409-2419, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35305824

RESUMO

Breast cancer was ranked first in global cancer incidence in 2020, and HER2 overexpression in breast cancer accounts for 20-30% of breast cancer patients. Current therapeutic strategies increase the survival rate, but resistance to them occurs frequently, and there is an urgent need to develop novel treatments such as DNA vaccines which can induce a specific and long-lasting immune response against HER2 antigens. To enhance the immunogenicity of DNA vaccines, dendritic cells (DCs) can be targeted using multi-epitope proteins that provide accurate immune focusing. For this purpose, we generated a DNA vaccine encoding a fusion protein composed of 1) in silico discovered antigenic epitopes of human and rat HER2 proteins (MeHer2) and 2) a single-chain antibody fragment (ScFv) specific for the DC-restricted antigen-uptake receptor DEC205 (ScFvDEC). The xenogeneic multi-epitope DNA vaccine (pMeHer2) encodes three only T-cell epitopes, two only B-cell epitopes, and two T and B cell epitopes, and pScFvDEC-MeHer2 vaccine additionally encodes ScFvDEC introduced at the N terminus of the MeHer2. Then, mouse groups were immunized with pScFvDEC-MeHer2, pMeHer2, pScFvDEC, pEmpty, and PBS to determine the elicited immune response. pScFvDEC-MeHer2 vaccinated mice showed a strong IgG response (P < 0.0001) and pScFvDEC-MeHer2 induced a significant IgG2a increase (P < 0.01). The percentages of both IFN-γ secreting CD4 and CD8 T cells were higher in mice immunized with pScFvDEC-MeHer2 compared with the pMeHer2. pScFvDEC-MeHer2 and pMeHer2 secreted significantly higher levels of extracellular IFN-γ compared with to control groups (P < 0.0001). In addition, the IFN-γ level of the pScFvDEC-MeHer2 vaccine group was approximately two times higher than the pMeHer2 group (P < 0.0001). Overall, this study identified the pScFvDECMeHer2 construct as a potential DNA vaccine candidate, supporting further studies to be conducted on HER2+ animal models.


Assuntos
Neoplasias da Mama , Vacinas de DNA , Animais , Neoplasias da Mama/prevenção & controle , Células Dendríticas , Epitopos de Linfócito T/genética , Feminino , Humanos , Camundongos , Ratos , Receptor ErbB-2/genética
8.
Transbound Emerg Dis ; 68(2): 587-597, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32643286

RESUMO

New technologies in the field of vaccinology arise as a necessity for the treatment and control of many diseases. Whole virus inactivated vaccines and modified live virus ones used against Bovine Herpesvirus-1 (BoHV-1) infection have several disadvantages. Previous works on DNA vaccines against BoHV-1 have demonstrated the capability to induce humoral and cellular immune responses. Nevertheless, 'naked' DNA induces low immunogenic response. Thus, loading of antigen encoding DNA sequences in liposomal formulations targeting dendritic cell receptors could be a promising strategy to better activate these antigen-presenting cells (APC). In this work, a DNA-based vaccine encoding the truncated version of BoHV-1 glycoprotein D (pCIgD) was evaluated alone and encapsulated in a liposomal formulation containing LPS and decorated with MANα1-2MAN-PEG-DOPE (pCIgD-Man-L). The vaccinations were performed in mice and bovines. The results showed that the use of pCIgD-Man-L enhanced the immune response in both animal models. For humoral immunity, significant differences were achieved when total antibody titres and isotypes were assayed in sera. Regarding cellular immunity, a significant increase in the proliferative response against BoHV-1 was detected in animals vaccinated with pCIgD-Man-L when compared to the response induced in animals vaccinated with pCIgD. In addition, upregulation of CD40 molecules on the surface of bovine dendritic cells (DCs) was observed when cells were stimulated and activated with the vaccine formulations. When viral challenge was performed, bovines vaccinated with MANα1-2MAN-PEG-DOPE elicited better protection which was evidenced by a lower viral excretion. These results demonstrate that the dendritic cell targeting using MANα1-2MAN decorated liposomes can boost the immunogenicity resulting in a long-lasting immunity. Liposomes decorated with MANα1-2MAN-PEG-DOPE were tested for the first time as a DNA vaccine nanovehicle in cattle as a preventive treatment against BoHV-1. These results open new perspectives for the design of vaccines for the control of bovine rhinotracheitis.


Assuntos
Doenças dos Bovinos/prevenção & controle , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1/imunologia , Vacinas contra Herpesvirus/administração & dosagem , Vacinação/veterinária , Animais , Bovinos , Infecções por Herpesviridae/prevenção & controle , Masculino , Camundongos , Vacinas de DNA/administração & dosagem
9.
Vaccine ; 37(35): 4963-4974, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31320219

RESUMO

Vaccination is the most efficient strategy to protect from infectious diseases and the induction of a protective immune response not only depends on the nature of the antigen, but is also influenced by the vaccination strategy and the co-administration of adjuvants. Therefore, the precise monitoring of adjuvant candidates and their immune modulatory properties is a crucial step in vaccine development. Here, one central aspect is the induction of appropriate humoral and cellular effector mechanisms. In our study we performed a direct comparison of two promising candidates in adjuvant development, the STING activator bis-(3,5)-cyclic dimeric adenosine monophosphate (c-di-AMP) and the Toll-like receptor ligand formulation poly(I:C)/CpG. These were evaluated in C57BL/6 mice using the model antigen ovalbumin (OVA) in subcutaneous vaccination with soluble protein as well as in a dendritic cell (DC) targeting approach (αDEC-OVA). Strikingly, c-di-AMP as compared to poly(I:C)/CpG resulted in significantly higher antigen-specific IgG antibody levels when used in immunization with soluble OVA as well as in antigen targeting to DC. In vaccination with soluble OVA, c-di-AMP induced a significantly stronger CTL, Th1 and IFNγ-producing CD8+ memory T cell response than poly(I:C)/CpG. The response was CTL and Th1 cell dominated, a profile shared by both adjuvants. In the context of targeting OVA to DC, c-di-AMP induced significantly increased Th1 and Th2 cell responses as compared to poly(I:C)/CpG. Interestingly, the Th1 response dominated the overall T cell response only when c-di-AMP was used, indicating a distinct modulatory property of c-di-AMP when the DC targeting immunization approach was exploited. Taken together, we describe superior properties of c-di-AMP as compared to poly(I:C)/CpG in subcutaneous vaccination with soluble antigen as well as antigen targeting to DC. This indicates exceptionally effective adjuvant properties for c-di-AMP and provides compelling evidence of its potential for further adjuvant development, especially also when using DC targeting approaches.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos CD/imunologia , Células Dendríticas/imunologia , Fosfatos de Dinucleosídeos/imunologia , Lectinas Tipo C/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Receptores de Superfície Celular/imunologia , Animais , Vacinas Anticâncer , Fosfatos de Dinucleosídeos/administração & dosagem , Feminino , Imunoglobulina G/imunologia , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Poli I-C/administração & dosagem , Poli I-C/imunologia , Organismos Livres de Patógenos Específicos , Vacinação
10.
Front Oncol ; 8: 404, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319969

RESUMO

Dendritic cells (DCs) are the primary antigen-presenting cells and play key roles in the orchestration of the innate and adaptive immune system. Targeting DCs by nanotechnology stands as a promising strategy for cancer immunotherapy. The physicochemical properties of nanoparticles (NPs) influence their interactions with DCs, thus altering the immune outcome of DCs by changing their functions in the processes of maturation, homing, antigen processing and antigen presentation. In this review, we summarize the recent progress in targeting DCs using NPs as a drug delivery carrier in cancer immunotherapy, the recognition of NPs by DCs, and the ways the physicochemical properties of NPs affect DCs' functions. Finally, the molecular pathways in DCs that are affected by NPs are also discussed.

11.
Cell Cycle ; 17(10): 1220-1234, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30019620

RESUMO

Dendritic cells (DCs) play a predominant role in initiating cell immune responses. Here we generated a DC-targeting lentiviral vector (LVDC-UbHBcAg-LIGHT) and evaluated its capacity to elicit HBV-specific cytotoxic T lymphocyte (CTL) responses. DC-SIGN-mediated specific transduction using this construct was confirmed in DC-SIGN-expressing 293T cells and ex vivo-cultured bone marrow cells. LVDC-UbHBcAg-LIGHT-loaded DCs were highly effective in inducing HBV-specific CTLs. Mechanistic studies demonstrated autophagy blocking led to a significant increase in apoptosis and obvious inhibition of CD8 + T cells entry into S-phase, correspondingly attenuated LVDC-UbHBcAg-LIGHT-loaded DC-induced T cell responses. This observation was supported by accumulation of pro-apoptotic proteins and the main negative cell cycle regulator-CDKN1B that otherwise would be degraded in activated T cells where autophagy preferentially occured. Our findings revealed an important role of autophagy in the activation of T cells and suggested LVDC-UbHBcAg-LIGHT may potentially be used as a therapeutic strategy to combat persistent HBV infection with higher security.


Assuntos
Autofagia , Células Dendríticas/metabolismo , Vetores Genéticos/metabolismo , Vírus da Hepatite B/imunologia , Lentivirus/genética , Linfócitos T Citotóxicos/imunologia , Transdução Genética , Regulação para Cima , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia/metabolismo , Linfócitos T CD8-Positivos/imunologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/efeitos dos fármacos , Engenharia Genética , Antígenos do Núcleo do Vírus da Hepatite B/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos Endogâmicos BALB C , Receptores de Superfície Celular/metabolismo , Fase S/efeitos dos fármacos , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/ultraestrutura , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Ubiquitina/metabolismo , Regulação para Cima/efeitos dos fármacos
12.
Cell Physiol Biochem ; 48(3): 1041-1059, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30041239

RESUMO

BACKGROUND/AIMS: Developing engineered dendritic cell (DC)-targeting lentivectors (LVs) have been the target of intense research for their potential to create antigen-directed immunotherapeutics which can be safely administered to patients. In this study, we constructed a DC-directed LV (LVDC-UbHBcAg-LIGHT) as a potential vaccine to induce anti-HBV immune responses. METHODS: Specificity of LVDC-UbHBcAg-LIGHT for DCs in vivo was confirmed through live animal imaging studies. The levels of cytokine production in T cells were assessed by flow cytometry. The HBcAg-specific cytotoxic T lymphocyte (CTL) responses and antibody responses induced by direct administration of the LVs were detected by LDH release assay and ELISA respectively. The levels of serum HBsAg and HBV DNA were evaluated by Abbott kits and quantitative PCR respectively. The expression levels of HBsAg and HBcAg in liver tissues of HBV transgenic mice were examined by immunohistochemistry. In addition, molecular mechanism underlying the activation of CD8+ T cells was explored. RESULTS: Live animal imaging studies showed that following subcutaneous administration of LVDC-UbHBcAg-LIGHT, no obvious luminescence signal was detected at the injection site. Immunization with LVDC-UbHBcAg-LIGHT elicited potent T cell responses in HBV transgenic mice evidenced by increased percentages of IFN-γ, TNF-α and GzmB producing CD8+ T cells as well as IFN-γ producing CD4+ T cells, improved HBcAg-specific CTL activities and antibody responses. Additionally, vaccination with LVDC-UbHBcAg-LIGHT efficiently reduced serum HBsAg, HBV DNA levels and the expression of HBsAg and HBcAg in liver tissues of HBV transgenic mice. More importantly, autophagy was induced in the activated CD8+ T cells, and the induced autophagy noticeably promoted the proliferation of T cells and decreased the frequencies of apoptotic CD8+ T cells by selectively degrading ubiquitinated apoptosis and cell cycle-associated protein aggregates. Futhermore, we confirmed the interaction between autophagosomes and ubiquitinated aggregates by confocal microscopy and immunoprecipitation analysis. CONCLUSIONS: These results demonstrated that LVDC-UbHBcAg-LIGHT provided a simple method of eliciting effective antiviral immune responses in HBV transgenic mice and might potentially be used as a therapeutic strategy to eradicate HBV with more safety and efficiency. Moreover, our results revealed a direct role of autophagy in promoting the survival and proliferation of activated CD8+ T cells.


Assuntos
Autofagia , Vetores Genéticos/metabolismo , Antígenos do Núcleo do Vírus da Hepatite B/metabolismo , Vírus da Hepatite B/fisiologia , Linfócitos T Citotóxicos/metabolismo , Animais , Anticorpos Antivirais/sangue , Proteína 11 Semelhante a Bcl-2/genética , Proteína 11 Semelhante a Bcl-2/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Vetores Genéticos/genética , Granzimas/metabolismo , Antígenos do Núcleo do Vírus da Hepatite B/genética , Antígenos do Núcleo do Vírus da Hepatite B/imunologia , Humanos , Interferon gama/análise , Lentivirus/genética , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Replicação Viral
13.
Front Immunol ; 9: 3123, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687326

RESUMO

Increased susceptibility to infectious diseases is a hallmark of the neonatal period of life that is generally attributed to a relative immaturity of the immune system. Dendritic cells (DCs) are innate immune sentinels with vital roles in the initiation and orchestration of immune responses, thus, constituting a promising target for promoting neonatal immunity. However, as is the case for other immune cells, neonatal DCs have been suggested to be functionally immature compared to their adult counterparts. Here we review some of the unique aspects of neonatal DCs that shape immune responses in early life and speculate whether the functional properties of neonatal DCs could be exploited or manipulated to promote more effective vaccination in early life.


Assuntos
Células Dendríticas/imunologia , Vacinação/métodos , Vacinas/imunologia , Adulto , Fatores Etários , Animais , Animais Recém-Nascidos , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Infecções Bacterianas/prevenção & controle , Humanos , Imunogenicidade da Vacina , Recém-Nascido , Camundongos , Vacinas/administração & dosagem , Viroses/imunologia , Viroses/microbiologia , Viroses/prevenção & controle
14.
Expert Rev Vaccines ; 16(10): 955-972, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28879788

RESUMO

INTRODUCTION: Development of a safe, effective and globally affordable Human Immunodeficiency Virus strain 1 (HIV-1) vaccine offers the best hope for future control of the HIV-1 pandemic. However, with the exception of the recent RV144 trial, which elicited a modest level of protection against infection, no vaccine candidate has shown efficacy in preventing HIV-1 infection or in controlling virus replication in humans. There is also a great need for a successful immunotherapeutic vaccine since combination antiretroviral therapy (cART) does not eliminate the reservoir of HIV-infected cells. But to date, no vaccine candidate has proven to significantly alter the natural history of an individual with HIV-1 infection. Areas covered: For over 25 years, the ANRS (France Recherche Nord&Sud Sida-HIV hépatites) has been committed to an original program combining basic science and clinical research developing an epitope-based vaccine strategy to induce a multiepitopic cellular response against HIV-1. This review describes the evolution of concepts, based on strategies using HIV-1 lipopeptides towards the use of dendritic cell (DC) manipulation. Expert commentary: Understanding the crucial role of DCs in immune responses allowed moving from the non-specific administration of HIV-1 sequences with lipopeptides to DC-based vaccines. These DC-targeting strategies should improve HIV-1 vaccine efficacy.


Assuntos
Vacinas contra a AIDS/biossíntese , Células Dendríticas/imunologia , Epitopos/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Lipopeptídeos/imunologia , Vacinas contra a AIDS/genética , Vacinas contra a AIDS/farmacologia , Sequência de Aminoácidos , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Ensaios Clínicos como Assunto , Células Dendríticas/efeitos dos fármacos , Epitopos/química , Epitopos/genética , França , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Lipopeptídeos/química , Lipopeptídeos/genética , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas
15.
Benef Microbes ; 8(3): 379-391, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28504575

RESUMO

FaeG is the major subunit of K88 fimbriae. These cell surface attachments are considered to be the major virulence factor of enterotoxigenic Escherichia coli (ETEC), which causes diarrhoea in piglets. The use of dendritic cell-targeting peptide (DCpep) has been demonstrated to be an effective approach to enhance the immunity of vaccines. Lactobacillus plantarum is an attractive candidate for oral vaccination owing to its beneficial effects and safety. In this study, L. plantarum was employed to deliver a FaeG-DCpep fusion antigen, and the immune response in mice was evaluated. The synthesis of FaeG-DCpep dramatically increased the adhesion of recombinant L. plantarum (RLP) to IPEC-J2 cell surfaces, resulting in direct competition between L. plantarum and ETEC during adhesion assays. Significantly higher levels of body weight gain, sera immunoglobulin G and intestinal immunoglobulin A were observed in BALB/c mice immunised with RLP. In addition, the number of CD19+ B cells and CD11c+DC cells and the expression levels of several cytokines in the spleen and lymph nodes increased significantly compared to non-immunised mice. The oral administration of RLP also alleviated the symptoms of ETEC challenge, as shown by haematoxylin-eosin staining, indicating that RLP may be an efficient vaccine candidate.


Assuntos
Adesinas de Escherichia coli/imunologia , Células Dendríticas/imunologia , Escherichia coli Enterotoxigênica/efeitos dos fármacos , Lactobacillus plantarum/metabolismo , Probióticos/farmacologia , Proteínas Recombinantes de Fusão/imunologia , Adesinas de Escherichia coli/genética , Animais , Linfócitos B/imunologia , Aderência Bacteriana/genética , Escherichia coli Enterotoxigênica/imunologia , Escherichia coli Enterotoxigênica/patogenicidade , Feminino , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética
16.
J Control Release ; 256: 170-181, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28414151

RESUMO

The purpose of the present study was to identify an "easy-to-adopt" strategy to enhance immune responses using functionalized alginate (ALG) nanoparticles (MAN-ALG/ALG=OVA NPs), which were prepared by CaCl2 cross-linking of two different types of ALG. The mannose (MAN) modified ALG (MAN-ALG) was used for dendritic cell targeting. The other component, composed of ovalbumin (OVA), a model antigen, is conjugated to ALG (ALG=OVA) via pH sensitive Schiff base bond. Grafting of alginate was demonstrated by FT-IR and 1H NMR, while the morphological structure, particle size, Zeta potential of MAN-ALG/ALG=OVA NPs were measured using TEM and DLS. The OVA releasing behavior of MAN-ALG/ALG=OVA NPs was determined as a function of pH. Antigen uptake was examined by flow cytometry and confocal laser scanning microscopy in vitro using mouse bone marrow dendritic cells (BMDCs). The results showed that MAN-ALG/ALG=OVA NPs facilitated antigen uptake of BMDCs and cytosolic release of the antigen. Significant up-regulation of cytokine secretion and expression levels of the surface co-stimulatory molecules were also observed in MAN-ALG/ALG=OVA NPs-treated BMDCs, compared to free OVA. In vivo bio-distribution study using Cy7 (a near-infrared fluorescence dye) labeled MAN-ALG/ALG=OVA NPs showed efficient in vivo trafficking of the nanoparticles from the injection site to the draining lymph nodes. Moreover, MAN-ALG/ALG=OVA NPs were found to enhance cross-presentation of OVA to B3Z T cell hybridoma in vitro. Subcutaneous administration of MAN-ALG/ALG=OVA NPs also induced major cytotoxic T lymphocytes (CTL) response and inhibition of E.G7 tumor growth in C57BL/6 mice. In summary, we report here that the MAN-ALG/ALG=OVA NPs have the potential as a potent nanovaccine for cancer immunotherapy.


Assuntos
Antígenos/administração & dosagem , Células Dendríticas/imunologia , Imunoterapia , Nanopartículas/administração & dosagem , Neoplasias/terapia , Ovalbumina/administração & dosagem , Alginatos/administração & dosagem , Alginatos/química , Animais , Antígenos/química , Linhagem Celular Tumoral , Citocinas/imunologia , Feminino , Ácido Glucurônico/administração & dosagem , Ácido Glucurônico/química , Ácidos Hexurônicos/administração & dosagem , Ácidos Hexurônicos/química , Manose/administração & dosagem , Manose/química , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/química , Neoplasias/imunologia , Ovalbumina/química , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
17.
Int Immunopharmacol ; 46: 62-69, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28259002

RESUMO

Human respiratory syncytial virus (RSV) is the most important cause of serious lower respiratory tract infection in infants, the elderly, and the immunocompromised population. There is no licensed vaccine against RSV until now. It has been reported that targeting antigen to DEC205, a phagocytosis receptor on dendritic cells (DCs), could induce enhanced CD4+ and CD8+ T cell responses in mice. To develop RSV DNA vaccine and target the encoded antigen protein to DCs, the ectodomain of fusion glycoprotein (sF, amino acids: 23-524) of RSV was fused with anti-DEC205 single-chain Fv fragment (scDEC) and designated scDECF. Following successful expression from the recombinant plasmid of pVAX1/scDECF, the recombinant protein of scDECF was found capable of specifically binding to DEC205 receptor on CHOmDEC205 cells, and facilitating uptake of RSV F by DC2.4 cells in vitro. Furthermore, the higher levels of RSV-specific IgG antibody responses and neutralization antibody titers, as well as RSV F-specific CD8+ T cell responses were induced in mice immunized intramuscularly by pVAX1/scDECF than by the control plasmid of pVAX1/scISOF encoding sF protein fused with isotype matched control single-chain Fv fragment (scISO). Compared with pVAX1/scISOF, both the ratio of IgG2a/IgG1, >1, and the enhanced IFN-γ cytokine were induced in mice following pVAX1/scDECF immunization, which exhibited a Th1 dominant response in pVAX1/scDECF vaccinated mice. Notably, the elevated efficiency of RSV F protein bound by DCs in vivo could also be observed in mice inoculated by pVAX1/scDECF. Collectively, these results demonstrate the enhanced IgG and CD8+ T cell immune responses have been induced successfully by DNA vaccine against RSV by targeting F antigen to DCs via the DEC205 receptor, and this DC-targeting vaccine strategy merits further investigation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vacinas Virais/imunologia , Idoso , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , ELISPOT , Humanos , Imunidade Celular , Hospedeiro Imunocomprometido , Lactente , Recém-Nascido , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Anticorpos de Cadeia Única/genética , Vacinação , Vacinas de DNA , Vacinas Virais/genética
18.
J Control Release ; 237: 14-22, 2016 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-27374625

RESUMO

Considering the rising incidence of allergic asthma, the symptomatic treatments that are currently applied in most cases are less than ideal. Specific immunotherapy is currently the only treatment that is able to change the course of the disease, but suffers from a long treatment duration. A gene based immunization that elicits the targeting of allergens towards dendritic cells in a steady-state environment might have the potential to amend these difficulties. Here we used a replication deficient adenovirus to induce the mucosal expression of OVA coupled to a single-chain antibody against DEC-205. A single intranasal vaccination was sufficient to mitigate an OVA-dependent asthmatic phenotype in a murine model. Invasive airway measurements demonstrated improved lung function after Ad-Dec-OVA treatment, which was in line with a marked reduction of goblet cell hyperplasia and lung eosinophilia. Furthermore OVA-specific IgE titers and production of type 2 cytokines were significantly reduced. Together, the here presented data demonstrate the feasibility of mucosal expression of DEC-targeted allergens as a treatment of allergic asthma.


Assuntos
Adenoviridae/genética , Alérgenos/imunologia , Antígenos CD/imunologia , Asma/prevenção & controle , Imunização/métodos , Lectinas Tipo C/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Ovalbumina/imunologia , Receptores de Superfície Celular/imunologia , Anticorpos de Cadeia Única/imunologia , Alérgenos/genética , Animais , Asma/imunologia , Citocinas/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Imunoglobulina E/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/genética , Anticorpos de Cadeia Única/genética
19.
J Viral Hepat ; 23(10): 798-804, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27126208

RESUMO

DNA vaccine targeting delivery to DC represents one effective strategy to improve the immunogenicity of the vaccine. In a previous study, we developed a novel DC-targeting recombinant protein that can deliver plasmid DNA to DCs by an electrostatic coupling effect and can thus improve the uptake efficiency of DCs, improving the expression of plasmid DNA in DCs. In this study, we coupled the protein with the HBV DNA vaccine pSVK-HBVA and investigated whether the immunogenicity and antiviral ability of the vaccine can be improved in HBV transgenic mice. The results show that a stronger specific immune response can be induced in mice after immunization with the coupling vaccine. The HBV DNA copy number and circulating antigen HBsAg in the serum of HBV transgenic mice were significantly decreased. Therefore, this study has demonstrated that the DC-targeting protein has the ability to improve the immunogenicity and the antiviral activity of the HBV DNA vaccine pSVK-HBVA. These findings indicate that this DC-targeting protein can be a potential method for the delivery of DNA vaccines directly to DCs.


Assuntos
Células Dendríticas/imunologia , Vacinas contra Hepatite B/administração & dosagem , Vacinas contra Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Hepatite B/prevenção & controle , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Animais , Feminino , Vírus da Hepatite B/genética , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
20.
Immunol Lett ; 168(2): 293-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26475398

RESUMO

Strategies for inducing an effective immune response following vaccination have focused on targeting antigens to dendritic cells (DCs) through the DC-specific surface molecule DEC-205. The immunogenicity and efficacy of DNA vaccination can also be enhanced by fusing the encoded antigen to single-chain antibodies directed against DEC-205. Here, we investigated this promising approach for its enhancement of hepatitis B virus (HBV)-specific cellular and humoral immune responses and its antiviral effects in HBV transgenic mice. A plasmid DNA vaccine encoding mouse DEC-205 single-chain fragment variable (mDEC-205-scFv) linked with the hepatitis B surface antigen (HBsAg) was constructed. Vaccination with this fusion DNA vaccine in HBV transgenic mice induced robust antiviral T cell and antibody immunity against HBsAg. The levels of serum-circulating HBsAg and the HBV DNA copy number were downregulated by the induction of a higher HBsAg-specific response. Thus, in this study, we demonstrated the therapeutic efficacy of the novel mDEC-205-scFv-fused DNA vaccine in a mouse model of immune-tolerant, chronic HBV infection.


Assuntos
Células Dendríticas/imunologia , Vacinas contra Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Hepatite B/imunologia , Imunidade Humoral/imunologia , Linfócitos T/imunologia , Vacinas de DNA/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Células HEK293 , Hepatite B/prevenção & controle , Hepatite B/virologia , Antígenos de Superfície da Hepatite B/sangue , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/imunologia , Vacinas contra Hepatite B/administração & dosagem , Vacinas contra Hepatite B/genética , Vírus da Hepatite B/genética , Vírus da Hepatite B/fisiologia , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Linfócitos T/metabolismo , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...