Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.292
Filter
Add more filters

Publication year range
1.
PLoS Biol ; 20(4): e3001607, 2022 04.
Article in English | MEDLINE | ID: mdl-35442969

ABSTRACT

A recent study in PLOS Biology shows that a betaherpesvirus circulating with the vampire bat, Desmodus rotundus, could serve as an effective vector for a transmissible vaccine capable of reducing the risk of rabies virus spillover in Peru.


Subject(s)
Chiroptera , Rabies virus , Rabies , Vaccines , Animals , Chiroptera/virology , Disease Vectors , Rabies/immunology , Rabies/prevention & control , Rabies/transmission , Rabies virus/genetics , Rabies virus/immunology
2.
Med Microbiol Immunol ; 213(1): 7, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38761268

ABSTRACT

The incidence of rabies in Thailand reached its peak in 2018 with 18 human deaths. Preexposure prophylaxis (PrEP) vaccination is thus recommended for high-risk populations. WHO has recently recommended that patients who are exposed to a suspected rabid animal and have already been immunized against rabies should receive a 1-site intradermal (ID) injection of 0.1 mL on days 0 and 3 as postexposure prophylaxis (PEP). In Thailand, village health and livestock volunteers tasked with annual dog vaccination typically receive only a single lifetime PrEP dose and subsequent boosters solely upon confirmed animal bites. However, the adequacy of a single PrEP dose for priming and maintaining immunity in this high-risk group has not been evaluated. Therefore, our study was designed to address two key questions: (1) sufficiency of single-dose PrEP-to determine whether a single ID PrEP dose provides adequate long-term immune protection for high-risk individuals exposed to numerous dogs during their vaccination duties. (2) Booster efficacy for immune maturation-to investigate whether one or two additional ID booster doses effectively stimulate a mature and sustained antibody response in this population. The level and persistence of the rabies antibody were determined by comparing the immunogenicity and booster efficacy among the vaccination groups. Our study demonstrated that rabies antibodies persisted for more than 180 days after cost-effective ID PrEP or the 1st or the 2nd single ID booster dose, and adequate antibody levels were detected in more than 95% of participants by CEE-cELISA and 100% by indirect ELISA. Moreover, the avidity maturation of rabies-specific antibodies occurred after the 1st single ID booster dose. This smaller ID booster regimen was sufficient for producing a sufficient immune response and enhancing the maturation of anti-rabies antibodies. This safe and effective PrEP regimen and a single visit involving a one-dose ID booster are recommended, and at least one one-dose ID booster regimen could be equitably implemented in at-risk people in Thailand and other developing countries. However, an adequate antibody level should be monitored before the booster is administered.


Subject(s)
Antibodies, Viral , Immunization, Secondary , Rabies Vaccines , Rabies , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Rabies/prevention & control , Rabies/immunology , Antibodies, Viral/blood , Thailand , Humans , Injections, Intradermal , Animals , Female , Adult , Male , Young Adult , Antibody Affinity , Middle Aged , Dogs , Pre-Exposure Prophylaxis/methods , Adolescent , Post-Exposure Prophylaxis/methods , Antibody Formation/immunology
3.
Biochemistry (Mosc) ; 89(3): 574-582, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648774

ABSTRACT

Rabies is a zoonotic disease with high lethality. Most human deaths are associated with the bites received from dogs and cats. Vaccination is the most effective method of preventing rabies disease in both animals and humans. In this study, the ability of an adjuvant based on recombinant Salmonella typhimurium flagellin to increase protective activity of the inactivated rabies vaccine in mice was evaluated. A series of inactivated dry culture vaccine for dogs and cats "Rabikan" (strain Shchelkovo-51) with addition of an adjuvant at various dilutions were used. The control preparation was a similar series of inactivated dry culture vaccine without an adjuvant. Protective activity of the vaccine preparations was evaluated by the NIH potency test, which is the most widely used and internationally recommended method for testing effectiveness of the inactivated rabies vaccines. The value of specific activity of the tested rabies vaccine when co-administered with the adjuvant was significantly higher (48.69 IU/ml) than that of the vaccine without the adjuvant (3.75 IU/ml). Thus, recombinant flagellin could be considered as an effective adjuvant in the composition of future vaccine preparations against rabies virus.


Subject(s)
Adjuvants, Immunologic , Flagellin , Rabies Vaccines , Rabies , Vaccines, Inactivated , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Animals , Flagellin/immunology , Mice , Rabies/prevention & control , Rabies/immunology , Vaccines, Inactivated/immunology , Dogs , Rabies virus/immunology , Salmonella typhimurium/immunology , Female , Cats
4.
J Biol Chem ; 296: 100096, 2021.
Article in English | MEDLINE | ID: mdl-33208464

ABSTRACT

Rabies virus (RABV) matrix protein (M) plays crucial roles in viral transcription, replication, assembly, and budding; however, its function during the early stage of virus replication remains unknown. Here, we mapped the protein interactome between RABV M and human host factors using a proteomic approach, finding a link to the V-type proton ATPase catalytic subunit A (ATP6V1A), which is located in the endosomes where RABV first enters. By downregulating or upregulating ATP6V1A expression in HEK293T cells, we found that ATP6V1A facilitated RABV replication. We further found that ATP6V1A was involved in the dissociation of incoming viral M proteins during viral uncoating. Coimmunoprecipitation demonstrated that M interacted with the full length or middle domain of ATP6V1A, which was dependent on the lysine residue at position 256 and the glutamic acid residue at position 279. RABV growth and uncoating in ATP6V1A-depleted cells was restored by trans-complementation with the full length or interaction domain of ATP6V1A. Moreover, stably overexpressed ATP6V1A enhanced RABV growth in Vero cells, which are used for the production of rabies vaccine. Our findings identify a new partner for RABV M proteins and establish a new role of ATP6V1A by promoting virion uncoating during RABV replication.


Subject(s)
Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cell Survival/genetics , Cell Survival/physiology , Chlorocebus aethiops , HEK293 Cells , Humans , Immunoprecipitation , Mass Spectrometry , Plasmids/genetics , Proteomics , RNA Interference , Rabies/immunology , Rabies/prevention & control , Rabies Vaccines/immunology , Rabies Vaccines/therapeutic use , Rabies virus/immunology , Rabies virus/pathogenicity , Vacuolar Proton-Translocating ATPases/genetics , Vero Cells , Virus Replication/genetics , Virus Replication/physiology
5.
J Virol ; 95(24): e0082921, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34613801

ABSTRACT

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.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation/immunology , Immunity, Humoral/immunology , Lymphoid Tissue/immunology , Rabies virus/immunology , Toll-Like Receptor 4/genetics , Animals , Antibodies, Viral/blood , Female , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Rabies/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/immunology , Toll-Like Receptor 4/immunology
6.
J Virol ; 95(20): e0079021, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34346765

ABSTRACT

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.


Subject(s)
Rabies Vaccines/immunology , Rabies/immunology , Vaccination/methods , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , Disease Models, Animal , Female , Genetic Engineering/methods , Germinal Center/immunology , Glycoproteins/genetics , Immunization/methods , Male , Mice , Mice, Inbred ICR , Rabies/metabolism , Rabies Vaccines/metabolism , Rabies Vaccines/pharmacology , Rabies virus/immunology , Semliki forest virus/immunology , Vesiculovirus/genetics , Viral Proteins/genetics
7.
J Virol ; 95(23): e0141421, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34495701

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic , Manganese/pharmacology , Rabies Vaccines/immunology , Animals , Antibodies, Viral/blood , Antibody-Producing Cells/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes , Cats , Dendritic Cells/immunology , Disease Models, Animal , Dogs , Female , Germinal Center/immunology , Immunity, Humoral , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Plasma Cells/immunology , Rabies/immunology , Rabies virus/immunology , Vaccination , Vaccine Development
8.
Semin Immunol ; 39: 44-51, 2018 10.
Article in English | MEDLINE | ID: mdl-30033150

ABSTRACT

Recent technological advances have provided deeper insights into the role of small molecules in biological processes. Metabolic profiling has thus entered the arena of -omics studies and rapidly proven its value both as stand-alone and as complement to other more advanced approaches, notably transcriptomics. Here we describe the potential of metabolic profiling for vaccinology embedded in the context of infection and immunity. This discussion is preceded by a description of the relevant technical and analytical tools for biological interpretation of metabolic data. Although not as widely applied as other -omics technologies, we believe that metabolic profiling can make important contributions to the better understanding of mechanisms underlying vaccine-induced responses and their effects on the prevention of infection or disease.


Subject(s)
Dengue/metabolism , Onchocerciasis/metabolism , Pneumonia/metabolism , Rabies/metabolism , Sepsis/metabolism , Tuberculosis/metabolism , Vaccines/metabolism , Dengue/immunology , Dengue/prevention & control , Humans , Metabolome , Metabolomics/methods , Onchocerciasis/immunology , Onchocerciasis/prevention & control , Pneumonia/immunology , Pneumonia/prevention & control , Principal Component Analysis , Rabies/immunology , Rabies/prevention & control , Sepsis/immunology , Sepsis/prevention & control , Systems Biology/methods , Tuberculosis/immunology , Tuberculosis/prevention & control , Vaccination , Vaccines/administration & dosage , Vaccines/chemical synthesis
9.
Circulation ; 142(6): 556-574, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32441115

ABSTRACT

BACKGROUND: Stroke is a leading cause of adult disability that can severely compromise the quality of life of patients, yet no effective medication currently exists to accelerate rehabilitation. A variety of circular RNA (circRNA) molecules are known to function in ischemic brain injury. Lentivirus-based expression systems have been widely used in basic studies of circRNAs, but safety issues with such delivery systems have limited exploration of the potential therapeutic roles for circRNAs. METHODS: Circular RNA SCMH1 (circSCMH1) was screened from the plasma of patients with acute ischemic stroke by using circRNA microarrays. Engineered rabies virus glycoprotein-circSCMH1-extracellular vesicles were generated to selectively deliver circSCMH1 to the brain. Nissl staining was used to examine infarct size. Behavioral tasks were performed to evaluate motor functions in both rodent and nonhuman primate ischemic stroke models. Golgi staining and immunostaining were used to examine neuroplasticity and glial activation. Proteomic assays and RNA-sequencing data combined with transcriptional profiling were used to identify downstream targets of circSCMH1. RESULTS: CircSCMH1 levels were significantly decreased in the plasma of patients with acute ischemic stroke, offering significant power in predicting stroke outcomes. The decreased levels of circSCMH1 were further confirmed in the plasma and peri-infarct cortex of photothrombotic stroke mice. Beyond demonstrating proof-of-concept for an RNA drug delivery technology, we observed that circSCMH1 treatment improved functional recovery after stroke in both mice and monkeys, and we discovered that circSCMH1 enhanced the neuronal plasticity and inhibited glial activation and peripheral immune cell infiltration. CircSCMH1 binds mechanistically to the transcription factor MeCP2 (methyl-CpG binding protein 2), thereby releasing repression of MeCP2 target gene transcription. CONCLUSIONS: Rabies virus glycoprotein-circSCMH1-extracellular vesicles afford protection by promoting functional recovery in the rodent and the nonhuman primate ischemic stroke models. Our study presents a potentially widely applicable nucleotide drug delivery technology and demonstrates the basic mechanism of how circRNAs can be therapeutically exploited to improve poststroke outcomes.


Subject(s)
Brain/pathology , Ischemic Stroke/rehabilitation , Lentivirus/genetics , RNA, Circular/genetics , Rabies Vaccines/immunology , Rabies virus/physiology , Rabies/immunology , Animals , Disease Models, Animal , Drug Delivery Systems , Extracellular Vesicles , Gene Expression Regulation , Genetic Vectors , Humans , Ischemic Stroke/genetics , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Rats , Recovery of Function
10.
J Gen Virol ; 102(10)2021 10.
Article in English | MEDLINE | ID: mdl-34661517

ABSTRACT

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.


Subject(s)
Interferon Regulatory Factor-7/physiology , Rabies virus/immunology , Rabies virus/pathogenicity , Rabies/immunology , Rabies/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antibody-Producing Cells/immunology , B-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Disease Models, Animal , Female , Immunity, Humoral , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferons/analysis , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Rabies Vaccines/immunology , Th1 Cells/immunology , Viral Load
11.
J Gen Virol ; 102(7)2021 07.
Article in English | MEDLINE | ID: mdl-34269675

ABSTRACT

Rabies virus (RABV) infection can initiate the host immune defence response and induce an antiviral state characterized by the expression of interferon (IFN)-stimulated genes (ISGs), among which the family of genes of IFN-induced protein with tetratricopeptide repeats (Ifits) are prominent representatives. Herein, we demonstrated that the mRNA and protein levels of Ifit1, Ifit2 and Ifit3 were highly increased in cultured cells and mouse brains after RABV infection. Recombinant RABV expressing Ifit3, designated rRABV-Ifit3, displayed a lower pathogenicity than the parent RABV in C57BL/6 mice after intramuscular administration, and Ifit3-deficient mice exhibited higher susceptibility to RABV infection and higher mortality during RABV infection. Moreover, compared with their individual expressions, co-expression of Ifit2 and Ifit3 could more effectively inhibit RABV replication in vitro. These results indicate that murine Ifit3 plays an essential role in restricting the replication and reducing the pathogenicity of RABV. Ifit3 acts synergistically with Ifit2 to inhibit RABV replication, providing further insight into the function and complexity of the Ifit family.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Rabies virus/physiology , Rabies/virology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain/metabolism , Brain/virology , Cell Line , Female , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rabies/immunology , Rabies virus/pathogenicity , Transcriptome , Viral Load , Virus Replication
12.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32102880

ABSTRACT

Rabies, caused by rabies virus (RABV), is a fatal encephalitis in humans and other mammals, which continues to present a public health threat in most parts of the world. Our previous study demonstrated that Toll-like receptor 7 (TLR7) is essential in the induction of anti-RABV antibodies via the facilitation of germinal center formation. In the present study, we investigated the role of TLR7 in the pathogenicity of RABV in a mouse model. Using isolated plasmacytoid dendritic cells (pDCs), we demonstrated that TLR7 is an innate recognition receptor for RABV. When RABV invaded from the periphery, TLR7 detected viral single-stranded RNA and triggered immune responses that limited the virus's entry into the central nervous system (CNS). When RABV had invaded the CNS, its detection by TLR7 led to the production of cytokines and chemokines and an increase the permeability of the blood-brain barrier. Consequently, peripheral immune cells, including pDCs, macrophages, neutrophils, and B cells infiltrated the CNS. While this immune response, triggered by TLR7, helped to clear viruses, it also increased neuroinflammation and caused immunopathology in the mouse brain. Our results demonstrate that TLR7 is an innate recognition receptor for RABV, which restricts RABV invasion into the CNS in the early stage of viral infection but also contributes to immunopathology by inducing neuroinflammation.IMPORTANCE Developing targeted treatment for RABV requires understanding the innate immune response to the virus because early virus clearance is essential for preventing the fatality when the infection has progressed to the CNS. Previous studies have revealed that TLR7 is involved in the immune response to RABV. Here, we establish that TLR7 recognizes RABV and facilitates the production of some interferon-stimulated genes. We also demonstrated that when RABV invades into the CNS, TLR7 enhances the production of inflammatory cytokines which contribute to immunopathology in the mouse brain. Taken together, our findings suggest that treatments for RABV must consider the balance between the beneficial and harmful effects of TLR7-triggered immune responses.


Subject(s)
Rabies virus/metabolism , Rabies/pathology , Toll-Like Receptor 7/metabolism , Animals , Antibodies, Viral , B-Lymphocytes/immunology , Blood-Brain Barrier/metabolism , Brain/virology , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Immunity, Innate/immunology , Interferons , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability/drug effects , Rabies/immunology , Rabies virus/immunology , Rabies virus/pathogenicity , Toll-Like Receptor 7/immunology
13.
PLoS Pathog ; 15(6): e1007799, 2019 06.
Article in English | MEDLINE | ID: mdl-31220188

ABSTRACT

The development of high-throughput genome sequencing enables accurate measurements of levels of sub-consensus intra-host virus genetic diversity and analysis of the role played by natural selection during cross-species transmission. We analysed the natural and experimental evolution of rabies virus (RABV), an important example of a virus that is able to make multiple host jumps. In particular, we (i) analyzed RABV evolution during experimental host switching with the goal of identifying possible genetic markers of host adaptation, (ii) compared the mutational changes observed during passage with those observed in natura, and (iii) determined whether the colonization of new hosts or tissues requires adaptive evolution in the virus. To address these aims, animal infection models (dog and fox) and primary cell culture models (embryo brain cells of dog and fox) were developed and viral variation was studied in detail through deep genome sequencing. Our analysis revealed a strong unidirectional host evolutionary effect, as dog-adapted rabies virus was able to replicate in fox and fox cells relatively easily, while dogs or neuronal dog cells were not easily susceptible to fox adapted-RABV. This suggests that dog RABV may be able to adapt to some hosts more easily than other host variants, or that when RABV switched from dogs to red foxes it lost its ability to adapt easily to other species. Although no difference in patterns of mutation variation between different host organs was observed, mutations were common following both in vitro and in vivo passage. However, only a small number of these mutations also appeared in natura, suggesting that adaptation during successful cross-species virus transmission is a complex, multifactorial evolutionary process.


Subject(s)
Dog Diseases , Evolution, Molecular , Host-Parasite Interactions/immunology , Rabies virus/physiology , Rabies , Animals , Cell Line , Dog Diseases/genetics , Dog Diseases/immunology , Dogs , Female , Foxes/genetics , Foxes/immunology , Foxes/virology , High-Throughput Nucleotide Sequencing , Host-Parasite Interactions/genetics , Male , Mutation , Rabies/genetics , Rabies/immunology
14.
Microb Pathog ; 157: 104971, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34029660

ABSTRACT

Rabies is always fatal, when post-exposure prophylaxis is administered after the onset of clinical symptoms. To date, there is no effective treatment of rabies once clinical symptoms has initiated. Therefore, we aimed to provide evidences which indicate the promising effects of combination treatment with TLR agonists following rabies infection. Four groups of rabies infected-mice (10-mice/group) were treated with PolyI:C 50 µg (a TLR3 agonist), Imiquimod50 µg (a TLR7 agonist), (Poly + Imi)25 µg and (Poly + Imi)50 µg respectively. The immune responses in each experimental groups were investigated in the brain through evaluation of GFAP, MAP2, CD4, HSP70, TLR3, TLR7 and apoptotic cell expression as well as determination of IFN-γ, TNF-α and IL-4, levels. The treatment with combination of agonists (Poly + Imi)50 µg/mouse resulted a 75% decrease of mortality rate and better extended survival time following street rabies virus infection. Higher number of CD4+T cells, TLR3 and TLR7 expression in the brain parenchyma observed in the groups receiving both combined agonist therapies at the levels of 25 µg and 50 µg. In spite of decreased number of neuronal cell, significant higher number of astrocytes was shown in the group given (Poly + Imi)25 µg. The obtained results also pointed to the dramatic decrease of HSP70 expression in all groups of infected mice whereas higher number of apoptotic cells and Caspase 8 expression were recorded in (Poly + Imi)25 µg treated group. Furthermore, the cytokine profile consisting the increased levels of TNF-α, IFN-γ and IL-4 revealed that both humoral and cellular responses were highly modulated in combination therapy of 50 µg of Imiquimod and Poly I:C. Reduced viral load as quantified by real-time PCR of rabies N gene expression in the brain also correlated with the better survival of agonist-treated groups of mice. Based on obtained results, we have presented evidences of beneficial utilization of combined agonist therapy composed of TLR3/TLR7 ligands. This treatment regimen extended survival of infected mice and decreased significantly their mortality rate. We believe that the results of synergy-inducing protection of both TLR3/TLR7 agonists lead to the enhancement of innate immune responses cells residing in the CNS which warrant the studies to further understanding of crosstalk mechanisms in cellular immunity against rabies in the future.


Subject(s)
Rabies , Toll-Like Receptor 3/agonists , Toll-Like Receptor 7/agonists , Animals , Immunity, Innate , Mice , Rabies/drug therapy , Rabies/immunology , Rabies virus
15.
J Virol ; 93(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30567978

ABSTRACT

Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease.IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Glycoproteins/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Rabies virus/immunology , Rabies/immunology , Animals , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rabies Vaccines/immunology , Vaccination/methods , Vero Cells , Viral Vaccines/immunology
16.
J Neurovirol ; 26(5): 764-768, 2020 10.
Article in English | MEDLINE | ID: mdl-32725420

ABSTRACT

Rabies is a neurological disease with 100% lethality. Some of the rare human patients who survived after multiple drug treatment had severe sequelae. The present study showed that after 48 h of RABV inoculation, mice injected intracerebrally with anti-RABV F (ab')2 plus Bioporter® showed 70% survival compared to the control group, suggesting that transfection of anti-RABV antibodies to the brain may prevent or delay the spread of RABV at an early stage of infection. This result may provide important protocol results in intracellular antibody delivery to prevent the fatal outcome of the disease.


Subject(s)
Antibodies, Viral/administration & dosage , Drug Delivery Systems/methods , Immunoglobulin Fab Fragments/administration & dosage , Rabies Vaccines/administration & dosage , Rabies virus/drug effects , Rabies/prevention & control , Vaccination/methods , Animals , Brain/drug effects , Brain/virology , Disease Models, Animal , Female , Fluorescein-5-isothiocyanate/chemistry , Fluorescent Dyes/chemistry , Humans , Injections, Intraventricular , Mice , Rabies/immunology , Rabies/mortality , Rabies/virology , Rabies virus/immunology , Rabies virus/pathogenicity , Survival Analysis , Transfection/methods
17.
Virol J ; 17(1): 157, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33081802

ABSTRACT

BACKGROUND: Street rabies virus (RABV) usually infects hosts at peripheral sites and migrates from motor or sensory nerves to the central nervous system. Several studies have found that inflammation is mild in a mouse model of street RABV infection. However, the pathogenetic mechanisms of street RABV in naturally infected dogs or humans are not well understood. METHODS: Brain tissues collected from 3 dogs and 3 humans were used; these tissue samples were collected under the natural condition of rabies-induced death. The inflammatory response and pathway activation in the brain tissue samples of dogs and humans were evaluated by HE, IHC, ARY006, WB and ELISA. The clinical isolate street RABV strains CGS-17 and CXZ-15 from 30 six-week-old ICR mice were used to construct the mouse infection model presented here. RESULTS: Neuronal degeneration and increased lymphocyte infiltration in the cerebral cortex, especially marked activation of microglia, formation of glial nodules, and neuronophagy, were observed in the dogs and humans infected with the street RABV strains. The various levels of proinflammatory chemokines, particularly CXCL1, CXCL12, CCL2, and CCL5, were increased significantly in the context of infection with street RABV strains in dogs and humans in relation to healthy controls, and the levels of MAPK and NF-κB phosphorylation were also increased in dogs and humans with natural infection. We also found that the degrees of pathological change, inflammatory response, MAPK and NF-κB signaling pathway activation were obviously increased during natural infection in dogs and humans compared with artificial model infection in mice. CONCLUSION: The data obtained here provide direct evidence for the RABV-induced activation of the inflammatory response in a dog infection model, which is a relatively accurate reflection of the pathogenic mechanism of human street RABV infection. These observations provide insight into the precise roles of underlying mechanisms in fatal natural RABV infection.


Subject(s)
Brain/virology , Inflammation/physiopathology , Inflammation/virology , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Rabies virus/genetics , Rabies/physiopathology , Rabies/veterinary , Animals , Brain/pathology , Disease Models, Animal , Dogs/virology , Gene Expression Regulation , Humans , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinase Kinases/immunology , NF-kappa B/immunology , Rabies/immunology , Rabies/mortality , Rabies virus/immunology , Rabies virus/pathogenicity , Signal Transduction
18.
Appl Microbiol Biotechnol ; 104(9): 4127-4139, 2020 May.
Article in English | MEDLINE | ID: mdl-32170383

ABSTRACT

We developed a fast, rabies virus-free, in vitro method, based on a blocking ELISA (bELISA), to detect and accurately quantify anti-rabies glycoprotein antibodies in serum of several animal species. In this method, purified rabies virus-like particles (VLPs) are used as antigen to coat the plates, while the presence of specific rabies immunoglobulins is revealed through blocking the recognition of these VLPs by a biotinylated monoclonal antibody. A quality by design approach was carried out in order to optimize the method performance, improving the sensitivity and, thereby, reducing the limit of detection of this assay. After the method validation, we confirmed that the bELISA method is able to detect a concentration of 0.06 IU/mL rabies immunoglobulins, titer lower than the 0.5 IU/mL cutoff value established as indication for correct vaccination. Further, we assessed the correlation between bELISA, the MNT, and the Platelia methods, confirming the accuracy of this new assay. On the other hand, precision was evaluated, obtaining acceptable repeatability and intermediate precision values, showing that this bELISA could be proposed as a potential alternative method, replacing the gold standard techniques in vaccination schemes and becoming a routine control technique within regional rabies surveillance programs.


Subject(s)
Antibodies, Blocking/immunology , Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay/methods , Rabies/blood , Rabies/diagnosis , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/blood , Cats , Cattle , Dogs , Humans , Limit of Detection , Panthera , Rabies/immunology , Rabies virus/immunology , Reproducibility of Results , Sensitivity and Specificity
19.
Biologicals ; 68: 54-59, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32868168

ABSTRACT

Several studies have investigated the use of simple in vitro tests for the assessment of rabies antibody titers in serum samples from vaccinated human subjects, which would allow the effectiveness of rabies vaccination to be conveniently evaluated. To this end, a novel time-resolved fluoroimmunoassay (TRFIA) for the assessment of rabies antibody titers was established in this study for evaluating the effectiveness of protection against rabies. The TRFIA had a satisfactory limit of detection value (0.035 IU/mL) under optimal conditions. Additionally, the application of the TRFIA was demonstrated in 68 serum samples with satisfactory results. The coefficient variations (CVs) were all <10%, and the recoveries were in the range of 90-110%. The correlation coefficient of titer values obtained using the present TRFIA and the rapid fluorescent focus inhibition test (RFFIT) was 0.733, with a coincidence rate regarding the evaluation results (protected or not protected by vaccination) of 100%. The preliminary results confirmed that the TRFIA had a higher performance than an enzyme-linked immunosorbent assay (ELISA), and could potentially replace the ELISA. Based on these results, the novel TRFIA appears to be a convenient tool for the evaluation of rabies vaccination results based on serum samples from vaccinated human subjects.


Subject(s)
Antibodies, Viral/immunology , Fluoroimmunoassay/methods , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/immunology , Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay/methods , Humans , Rabies/diagnosis , Rabies/virology , Rabies Vaccines/administration & dosage , Rabies virus/physiology , Reproducibility of Results , Sensitivity and Specificity , Time Factors , Vaccination/methods
20.
Biologicals ; 68: 74-78, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32859463

ABSTRACT

The direct rapid immunohistochemical test (dRIT) has been recommended for laboratorial diagnosis of rabies, especially in developing countries. The absence of commercial primary antibodies, however, still represents a major limitation to its wider use in testing. We describe here the development of a biotinylated polyclonal antibody against Rabies lyssavirus (RABV) ribonucleoprotein (RNP) and its use as a primary reagent in dRIT. Anti-RNP polyclonal horse IgG was purified by ionic exchange chromatography followed by immunoaffinity column chromatography, and its affinity, diagnostic sensitivity, and specificity were evaluated. CNS samples (120) of suspected rabies cases in different animal species were tested by dRIT, with the positive (n = 14) and negative (n = 106) results confirmed by direct fluorescence antibody testing (dFAT). Comparing the results of dRIT and dFAT, we found that the biotinylated anti-RNP IgG delivered 100% diagnostic specificity and sensibility for rabies diagnosis. Our findings show that the biotinylated anti-RNP polyclonal IgG can be produced with the quality required for application in dRIT. This work represents an important step in efforts to diagnose rabies in developing countries.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Viral/immunology , Immunoglobulin G/immunology , Rabies virus/immunology , Rabies/immunology , Ribonucleoproteins/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Biotinylation , Brain/immunology , Brain/virology , Cats , Cattle , Chiroptera , Dogs , Fluorescent Antibody Technique, Direct/methods , Horses , Immunoglobulin G/metabolism , Immunohistochemistry/methods , Primates , Rabies/diagnosis , Rabies/virology , Sensitivity and Specificity , Species Specificity , Swine
SELECTION OF CITATIONS
SEARCH DETAIL