Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Virol ; 96(22): e0112522, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36326274

RESUMO

Lyssaviruses cause rabies, which is an acute neurological disease responsible for more than 59,000 human deaths annually and has no available effective treatments. The phosphoprotein (P) of lyssaviruses (lyssavirus-P) plays multiple roles in virus replication and immune evasion. Lyssavirus-P has been identified as the major type I interferon (IFN-I) antagonist, while the precise site and precise molecular mechanism remain unclear. Herein, we found that substitution of site 179 of lyssavirus-P from serine (Ser) to proline (Pro) impairs its antagonism function of IFN-I by sequence alignment and site mutations. Subsequent studies demonstrated that lyssavirus-P containing S179 specifically interacted with I-kappa B kinase ε (IKKε). Specifically, lyssavirus-P containing S179 interacted simultaneously with the kinase domain (KD) and scaffold dimerization domain (SDD) of IKKε, competing with TNF receptor-associated factor 3 (TRAF3) and IFN regulatory factor 3 (IRF3) for binding with IKKε, leading to the inhibition of IFN production. Furthermore, S179 was involved in the viral pathogenicity of the typical lyssavirus rabies virus in a mouse model. Interestingly, we found that S179 is conserved among most lyssavirus-P and functional for IFN antagonism. Collectively, we identified S179 of lyssavirus-P is essential for IFN-I inhibition, which provides deep insight into the immune evasion strategies of lyssaviruses. IMPORTANCE Interferon (IFN) and the IFN-induced cellular antiviral response constitute the first line of defense against viral invasion. Evading host innate immunity, especially IFN signaling, is the key step required for lyssaviruses to establish infection. In this study, S179 of lyssavirus phosphoprotein (lyssavirus-P) was identified as the key site for antagonizing IFN-I production. Mechanistically, lyssavirus-P containing S179 specifically targets the key kinase IKKε and disrupts its interaction with TRAF3 and IRF3. S179P mutation in the P protein of the typical lyssavirus rabies virus (RABV) attenuated its pathogenicity in a mouse model. Our findings provide deep insight into the immune evasion strategies of lyssaviruses, which is helpful for the development of effective antiviral therapeutics.


Assuntos
Interferon Tipo I , Lyssavirus , Vírus da Raiva , Animais , Camundongos , Humanos , Lyssavirus/genética , Quinase I-kappa B/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Interferon Tipo I/metabolismo , Antivirais
2.
J Virol ; 95(24): e0082921, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34613801

RESUMO

Rabies, caused by rabies virus (RABV), is fatal to both humans and animals around the world. Effective clinical therapy for rabies has not been achieved, and vaccination is the most effective means of preventing and controlling rabies. Although different vaccines, such as live attenuated and inactivated vaccines, can induce different immune responses, different expressions of pattern recognition receptors (PRRs) also cause diverse immune responses. Toll-like receptor 4 (TLR4) is a pivotal PRR that induces cytokine production and bridges innate and adaptive immunity. Importantly, TLR4 recognizes various virus-derived pathogen-associated molecular patterns (PAMPs) and virus-induced damage-associated molecular patterns (DAMPs), usually leading to the activation of immune cells. However, the role of TLR4 in the humoral immune response induced by RABV has not yet been revealed. Based on TLR4-deficient (TLR4-/-) and wild-type (WT) mouse models, we report that TLR4-dependent recruitment of the conventional type 2 dendritic cells (CD8α- CD11b+ cDC2) into secondary lymph organs (SLOs) is critical for antigen presentation. cDC2-initiated differentiation of follicular helper T (Tfh) cells promotes the proliferation of germinal center (GC) B cells, the formation of GCs, and the production of plasma cells (PCs), all of which contribute to the production of RABV-specific IgG and virus-neutralizing antibodies (VNAs). Collectively, our work demonstrates that TLR4 is necessary for the recruitment of cDC2 and for the induction of RABV-induced humoral immunity, which is regulated by the cDC2-Tfh-GC B axis. IMPORTANCE Vaccination is the most efficient method to prevent rabies. TLR4, a well-known immune sensor, plays a critical role in initiating innate immune response. Here, we found that TLR4-deficient (TLR4-/-) mice suppressed the induction of humoral immune response after immunization with rabies virus (RABV), including reduced production of VNAs and RABV-specific IgG compared to that occurred in wild-type (WT) mice. As a consequence, TLR4-/- mice exhibited higher mortality than that of WT mice after challenge with virulent RABV. Importantly, further investigation found that TLR4 signaling promoted the recruitment of cDC2 (CD8α+ CD11b-), a subset of cDCs known to induce CD4+ T-cell immunity through their MHC-II presentation machinery. Our results imply that TLR4 is indispensable for an efficient humoral response to rabies vaccine, which provides new insight into the development of novel rabies vaccines.


Assuntos
Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Humoral/imunologia , Tecido Linfoide/imunologia , Vírus da Raiva/imunologia , Receptor 4 Toll-Like/genética , Animais , Anticorpos Antivirais/sangue , Feminino , Imunização , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos C57BL , Raiva/imunologia , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/imunologia , Receptor 4 Toll-Like/imunologia
3.
J Virol ; 95(20): e0079021, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34346765

RESUMO

Rabies is a fatal zoonosis that causes encephalitis in mammals, and vaccination is the most effective method to control and eliminate rabies. Virus-like vesicles (VLVs), which are characterized as infectious, self-propagating membrane-enveloped particles composed of only Semliki Forest virus (SFV) replicase and vesicular stomatitis virus glycoprotein (VSV-G), have been proven safe and efficient as vaccine candidates. However, previous studies showed that VLVs containing rabies virus glycoprotein (RABV-G) grew at relatively low titers in cells, impeding their potential use as a rabies vaccine. In this study, we constructed novel VLVs by transfection of a mutant SFV RNA replicon encoding RABV-G. We found that these VLVs could self-propagate efficiently in cell culture and could evolve to high titers (approximately 108 focus-forming units [FFU]/ml) by extensive passaging 25 times in BHK-21 cells. Furthermore, we found that the evolved amino acid changes in SFV nonstructural protein 1 (nsP1) at positions 470 and 482 was critical for this high-titer phenotype. Remarkably, VLVs could induce robust type I interferon (IFN) expression in BV2 cells and were highly sensitive to IFN-α. We found that direct inoculation of VLVs into the mouse brain caused reduced body weight loss, mortality, and neuroinflammation compared with the RABV vaccine strain. Finally, it could induce increased generation of germinal center (GC) B cells, plasma cells (PCs), and virus-neutralizing antibodies (VNAs), as well as provide protection against virulent RABV challenge in immunized mice. This study demonstrated that VLVs containing RABV-G could proliferate in cells and were highly evolvable, revealing the feasibility of developing an economic, safe, and efficacious rabies vaccine. IMPORTANCE VLVs have been shown to represent a more versatile and superior vaccine platform. In previous studies, VLVs containing the Semliki Forest virus replicase (SFV nsP1 to nsP4) and rabies virus glycoprotein (RABV-G) grew to relatively low titers in cells. In our study, we not only succeeded in generating VLVs that proliferate in cells and stably express RABV-G, but the VLVs that evolved grew to higher titers, reaching 108 FFU/ml. We also found that nucleic acid changes at positions 470 and 482 in nsP1 were vital for this high-titer phenotype. Moreover, the VLVs that evolved in our studies were highly attenuated in mice, induced potent immunity, and protected mice from lethal RABV infection. Collectively, our study showed that high titers of VLVs containing RABV-G were achieved, demonstrating that these VLVs could be an economical, safe, and efficacious rabies vaccine candidate.


Assuntos
Vacina Antirrábica/imunologia , Raiva/imunologia , Vacinação/métodos , Animais , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Modelos Animais de Doenças , Feminino , Engenharia Genética/métodos , Centro Germinativo/imunologia , Glicoproteínas/genética , Imunização/métodos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Raiva/metabolismo , Vacina Antirrábica/metabolismo , Vacina Antirrábica/farmacologia , Vírus da Raiva/imunologia , Vírus da Floresta de Semliki/imunologia , Vesiculovirus/genética , Proteínas Virais/genética
4.
J Virol ; 95(23): e0141421, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495701

RESUMO

Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.


Assuntos
Adjuvantes Imunológicos , Manganês/farmacologia , Vacina Antirrábica/imunologia , Animais , Anticorpos Antivirais/sangue , Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos , Gatos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Cães , Feminino , Centro Germinativo/imunologia , Imunidade Humoral , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Plasmócitos/imunologia , Raiva/imunologia , Vírus da Raiva/imunologia , Vacinação , Desenvolvimento de Vacinas
5.
J Gen Virol ; 102(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34661517

RESUMO

Rabies is a zoonotic disease caused by the rabies virus (RABV). RABV can lead to fatal encephalitis and is still a serious threat in most parts of the world. Interferon regulatory factor 7 (IRF7) is the main transcriptional regulator of type I IFN, and it is crucial for the induction of IFNα/ß and the type I IFN-dependent immune response. In this study, we focused on the role of IRF7 in the pathogenicity and immunogenicity of RABV using an IRF7-/- mouse model. The results showed that the absence of IRF7 made mice more susceptible to RABV, because IRF7 restricted the replication of RABV in the early stage of infection. IRF7 deficiency affected the recruitment of plasmacytoid dendritic cells to the draining lymph nodes (dLNs), reduced the production of type I IFN and expression of IFN-stimulated genes. Furthermore, we found that the ability to produce specific RABV-neutralizing antibody was impaired in IRF7-/- mice. Consistently, IRF7 deficiency affected the recruitment of germinal-centre B cells to dLNs, and the generation of plasma cells and RABV-specific antibody secreting cells. Moreover, the absence of IRF7 downregulated the induction of IFN-γ and reduced type 1 T helper cell (Th1)-dependent antibody production. Collectively, our findings demonstrate that IRF7 promotes humoral immune responses and compromises the pathogenicity of RABV in a mouse model.


Assuntos
Fator Regulador 7 de Interferon/fisiologia , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Raiva/imunologia , Raiva/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Imunidade Humoral , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferons/análise , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vacina Antirrábica/imunologia , Células Th1/imunologia , Carga Viral
6.
Arch Virol ; 164(12): 2963-2974, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31552533

RESUMO

Cholesterol-25-hydroxylase (CH25H) is a reticulum-associated membrane protein that catalyzes the oxidation of cholesterol to 25-hydroxycholesterol (25HC). Recent studies have revealed that CH25H is an interferon-stimulated gene (ISG) that suppresses infection by several viruses. In the present study, we found that overexpression of both human and murine CH25H inhibited rabies virus (RABV) infection in HEK-293T (293T) cells. In contrast, silencing of CH25H enhanced RABV replication in 293T cells, and a catalytic mutant of CH25H lost its ability to inhibit RABV infection. Treatment with the oxysterol 25-hydroxycholesterol (25HC), the product of CH25H, dramatically decreased RABV replication in 293T, BSR and N2a cells by inhibiting viral membrane penetration. These data provide insights into the antiviral function of CH25H against RABV infection, which can potentially be used as a therapeutic agent for rabies.


Assuntos
Vírus da Raiva/fisiologia , Raiva/enzimologia , Esteroide Hidroxilases/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Hidroxicolesteróis/metabolismo , Camundongos , Raiva/genética , Raiva/virologia , Vírus da Raiva/genética , Esteroide Hidroxilases/genética , Replicação Viral
7.
mBio ; 15(1): e0177523, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38078742

RESUMO

IMPORTANCE: messenger RNA (mRNA) vaccines are a key technology in combating existing and emerging infectious diseases. However, the inherent instability of mRNA and the nonspecificity of lipid nanoparticle-encapsulated (LNP) delivery systems result in the need for cold storage and a relatively short-duration immune response to mRNA vaccines. Herein, we develop a novel vaccine in the form of circRNAs encapsulated in LNPs, and the circular structure of the circRNAs enhances their stability. Lyophilization is considered the most effective method for the long-term preservation of RNA vaccines. However, this process may result in irreversible damage to the nanoparticles, particularly the potential disruption of targeting modifications on LNPs. During the selection of lymph node-targeting ligands, we found that LNPs modified with mannose maintained their physical properties almost unchanged after lyophilization. Additionally, the targeting specificity and immunogenicity remained unaffected. In contrast, even with the addition of cryoprotectants such as sucrose, the physical properties of LNPs were impaired, leading to an obvious decrease in immunogenicity. This may be attributed to the protective role of mannose on the surface of LNPs during lyophilization. Freshly prepared and lyophilized mLNP-circRNA vaccines elicited comparable immune responses in both the rabies virus model and the SARS-CoV-2 model. Our data demonstrated that mLNP-circRNA vaccines elicit robust immune responses while improving stability after lyophilization, with no compromise in tissue targeting specificity. Therefore, mannose-modified LNP-circRNA vaccines represent a promising vaccine design strategy.


Assuntos
RNA Circular , Vacinas , Manose/química , Vacinas/genética , Imunidade , Liofilização , RNA Mensageiro/genética
8.
Emerg Microbes Infect ; 13(1): 2300461, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38164714

RESUMO

During the COVID-19 epidemic, the incidence of rabies has increased in several countries, especially in remote and disadvantaged areas, due to inadequate surveillance and declining immunization coverage. Multiple vaccinations with inactivated rabies virus vaccines for pre- or post-exposure prophylaxis are considered inefficient, expensive and impractical in developing countries. Herein, three modified human recombinant adenoviruses type 5 designated Adv-RVG, Adv-E1-RVG, and Adv-RVDG, carrying rabies virus G (RVG) expression cassettes in various combinations within E1 or E3 genomic regions, were constructed to serve as rabies vaccine candidates. Adv-RVDG mediated greater RVG expression both in vitro and in vivo and induced a more robust and durable humoral immune response than the rabies vaccine strain SAD-L16, Adv-RVG, and Adv-E1-RVG by more effectively activating the dendritic cells (DCs) - follicular helper T (Tfh) cells - germinal centre (GC) / memory B cells (MBCs) - long-lived plasma cells (LLPCs) axis with 100% survival after a lethal RABV challenge in mice during the 24-week study period. Similarly, dogs and cats immunized with Adv-RVDG showed stronger and longer-lasting antibody responses than those vaccinated with a commercial inactivated rabies vaccine and showed good tolerance to Adv-RVDG. In conclusion, our study demonstrated that simultaneous insertion of protective antigens into the E1 and E3 genomic regions of adenovirus vector can significantly enhance the immunogenicity of adenoviral-vectored vaccines, providing a theoretical and practical basis for the subsequent development of multivalent and multi-conjugated vaccines using recombinant adenovirus platform. Meanwhile, our data suggest Adv-RVDG is a safe, efficient, and economical vaccine for mass-coverage immunization.


Assuntos
Doenças do Gato , Doenças do Cão , Vacina Antirrábica , Vírus da Raiva , Gatos , Cães , Humanos , Animais , Camundongos , Vírus da Raiva/genética , Vacina Antirrábica/genética , Imunidade Humoral , Anticorpos Antivirais , Adenoviridae/genética
9.
Redox Biol ; 64: 102769, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37285742

RESUMO

Cholesterol-24-hydroxylase (CH24H or Cyp46a1) is a reticulum-associated membrane protein that plays an irreplaceable role in cholesterol metabolism in the brain and has been well-studied in several neuro-associated diseases in recent years. In the present study, we found that CH24H expression can be induced by several neuroinvasive viruses, including vesicular stomatitis virus (VSV), rabies virus (RABV), Semliki Forest virus (SFV) and murine hepatitis virus (MHV). The CH24H metabolite, 24-hydroxycholesterol (24HC), also shows competence in inhibiting the replication of multiple viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). 24HC can increase the cholesterol concentration in multivesicular body (MVB)/late endosome (LE) by disrupting the interaction between OSBP and VAPA, resulting in viral particles being trapped in MVB/LE, ultimately compromising VSV and RABV entry into host cells. These findings provide the first evidence that brain cholesterol oxidation products may play a critical role in viral infection.


Assuntos
Internalização do Vírus , Animais , Camundongos , Colesterol/metabolismo , COVID-19/metabolismo , COVID-19/virologia , Homeostase , SARS-CoV-2/metabolismo , Colesterol 24-Hidroxilase/metabolismo
10.
Emerg Microbes Infect ; 12(2): 2270081, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37819147

RESUMO

The persistence and clinical consequences of rabies virus (RABV) infection have prompted global efforts to develop a safe and effective vaccines against rabies. mRNA vaccines represent a promising option against emerging and re-emerging infectious diseases, gaining particular interest since the outbreak of COVID-19. Herein, we report the development of a highly efficacious rabies mRNA vaccine composed of sequence-modified mRNA encoding RABV glycoprotein (RABV-G) packaged in core-shell structured lipopolyplex (LPP) nanoparticles, named LPP-mRNA-G. The bilayer structure of LPP improves protection and delivery of RABV-G mRNA and allows gradual release of mRNA molecules as the polymer degrades. The unique core-shell structured nanoparticle of LPP-mRNA-G facilitates vaccine uptake and demonstrates a desirable biodistribution pattern with low liver targeting upon intramuscular immunization. Single administration of low-dose LPP-mRNA-G in mice elicited potent humoral immune response and provided complete protection against intracerebral challenge with lethal RABV. Similarly, single immunization of low-dose LPP-mRNA-G induced high levels of virus-neutralizing antibody titers in dogs. Collectively, our data demonstrate the potential of LPP-mRNA-G as a promising next-generation rabies vaccine used in human and companion animals.


Assuntos
Vacina Antirrábica , Vírus da Raiva , Raiva , Cães , Animais , Camundongos , Humanos , Raiva/prevenção & controle , Imunidade Humoral , Distribuição Tecidual , Anticorpos Antivirais , Vacinas de mRNA , Vírus da Raiva/genética , Imunização , RNA Mensageiro/genética
11.
Biosens Bioelectron ; 204: 114078, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35180689

RESUMO

Rabies is caused by the infection of Rabies virus, it leads to fatal encephalitis, developing a highly sensitive and specific detection method for Rabies virus remains a challenge. Herein, we report an electrochemiluminescence (ECL) biosensor for Rabies viral RNA based on dual-signal amplification and DNA nanotweezers (DTs). Dual-signal amplification process includes target binding induced isothermal amplification and CRISPR-based amplification. In the presence of target RNA, two assisted probes simultaneously hybridized with it to trigger isothermal amplification with the help of polymerase and nicking enzyme. This process generated a large amount of single-stranded DNA (ssDNA) as products. The products hybridized with CRISPR RNA to activate the trans-cleavage activity of Cas12a to indiscriminately cleave predesigned single-stranded trigger (ST) strands. After mixing the cleavage products with DTs and hemin molecules, DTs cannot be closed by cleaved ST strands to capture hemin to the electrode to quench the ECL signal. Therefore, the higher concentration of the target, the stronger intensity of the ECL signal. The detection limit is as low as 2.8 pM and the detection range is from 5 pM to 5 nM with excellent specificity and stability. The proposed method provides a promising strategy for Rabies detection, and can be easily adapted to other analytes via reasonable design as a valuable and versatile tool in bioanalysis.


Assuntos
Técnicas Biossensoriais , Raiva , Técnicas Biossensoriais/métodos , Sistemas CRISPR-Cas/genética , DNA/genética , Humanos , RNA Viral/genética , Raiva/genética
12.
Cell Insight ; 1(5): 100057, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37193556

RESUMO

Tripartite motif-containing protein 25 (Trim25) is an E3 ubiquitin ligase that activates retinoid acid-inducible gene I (RIG-I) and promotes the antiviral interferon response. Recent studies have shown that Trim25 can bind and degrade viral proteins, suggesting a different mechanism of Trim25 on its antiviral effects. In this study, Trim25 expression was upregulated in cells and mouse brains after rabies virus (RABV) infection. Moreover, expression of Trim25 limited RABV replication in cultured cells. Overexpression of Trim25 caused attenuated viral pathogenicity in a mouse model that was intramuscularly injected with RABV. Further experiments confirmed that Trim25 inhibited RABV replication via two different mechanisms: an E3 ubiquitin ligase-dependent mechanism and an E3 ubiquitin ligase-independent mechanism. Specifically, the CCD domain of Trim25 interacted with RABV phosphoprotein (RABV-P) at amino acid (AA) position at 72 and impaired the stability of RABV-P via complete autophagy. This study reveals a novel mechanism by which Trim25 restricts RABV replication by destabilizing RABV-P, which is independent of its E3 ubiquitin ligase activity.

13.
Viruses ; 11(12)2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31816996

RESUMO

Rabies, as one of the most threatening zoonoses in the world, causes a fatal central nervous system (CNS) disease. So far, vaccination with rabies vaccines has been the most effective measure to prevent and control this disease. At present, inactivated rabies vaccines are widely used in humans and domestic animals. However, humoral immune responses induced by inactivated rabies vaccines are relatively low and multiple shots are required to achieve protective immunity. Supplementation with an adjuvant is a practical way to improve the immunogenicity of inactivated rabies vaccines. In this study, we found that monophosphoryl-lipid A (MPLA), a well-known TLR4 agonist, could significantly promote the maturation of bone marrow-derived dendritic cells (BMDC) through a TLR4-dependent pathway in vitro and the maturation of conventional DCs (cDCs) in vivo. We also found that MPLA, serving as an adjuvant for inactivated rabies vaccines, could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, and plasma cells (PCs), consequently enhancing the production of RABV-specific total-IgG, IgG2a, IgG2b, and the virus-neutralizing antibodies (VNAs). Furthermore, MPLA could increase the survival ratio of mice challenged with virulent RABV. In conclusion, our results demonstrate that MPLA serving as an adjuvant enhances the intensity of humoral immune responses by activating the cDC-Tfh-GC B axis. Our findings will contribute to the improvement of the efficiency of traditional rabies vaccines.


Assuntos
Adjuvantes Imunológicos , Lipídeo A/análogos & derivados , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Vacinas de Produtos Inativados/imunologia , Anticorpos Antivirais/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Humanos , Imunidade Humoral , Imunização , Imunoglobulina G/imunologia , Imunofenotipagem , Lipídeo A/imunologia , Plasmócitos/imunologia , Raiva/imunologia , Raiva/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA