Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.400
Filter
1.
Sci Rep ; 14(1): 18509, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122768

ABSTRACT

Rabies virus (RABV) is the causative agent of rabies, a lethal neurological disease in mammals. RABV strains can be classified into fixed strains (laboratory strains) and street strains (field/clinical strains), which have different properties including cell tropism and neuroinvasiveness. RABV Toyohashi strain is a street strain isolated in Japan from an imported case which had been bitten by rabid dog in the Philippines. In order to facilitate molecular studies of RABV, we established a reverse genetics (RG) system for the study of the Toyohashi strain. The recombinant virus was obtained from a cDNA clone of Toyohashi strain and exhibited similar growth efficiency as the original virus in cultured cell lines. Both the original and recombinant strains showed similar pathogenicity with high neuroinvasiveness in mice, and the infected mice developed a long and inconsistent incubation period, which is characteristic of street strains. We also generated a recombinant Toyohashi strain expressing viral phosphoprotein (P protein) fused with the fluorescent protein mCherry, and tracked the intracellular dynamics of the viral P protein using live-cell imaging. The presented reverse genetics system for Toyohashi strain will be a useful tool to explore the fundamental molecular mechanisms of the replication of RABV street strains.


Subject(s)
Rabies virus , Rabies , Reverse Genetics , Rabies virus/genetics , Rabies virus/pathogenicity , Animals , Reverse Genetics/methods , Mice , Rabies/virology , Dogs , Humans , Cell Line , Virus Replication/genetics , Philippines
2.
Emerg Microbes Infect ; 13(1): 2389115, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39129566

ABSTRACT

Rabies is a lethal zoonotic disease that threatens human health. As the only viral surface protein, the rabies virus (RABV) glycoprotein (G) induces main neutralizing antibody (Nab) responses; however, Nab titre is closely correlated with the conformation of G. Virus-like particles (VLP) formed by the co-expression of RABV G and matrix protein (M) improve retention and antigen presentation, inducing broad, durable immune responses. RABV nucleoprotein (N) can elicit humoral and cellular immune responses. Hence, we developed a series of nucleoside-modified RABV mRNA vaccines encoding wild-type G, soluble trimeric RABV G formed by an artificial trimer motif (tG-MTQ), membrane-anchored prefusion-stabilized G (preG). Furthermore, we also developed RABV VLP mRNA vaccine co-expressing preG and M to generate VLPs, and VLP/N mRNA vaccine co-expressing preG, M, and N. The RABV mRNA vaccines induced higher humoral and cellular responses than inactivated rabies vaccine, and completely protected mice against intracerebral challenge. Additionally, the IgG and Nab titres in RABV preG, VLP and VLP/N mRNA groups were significantly higher than those in G and tG-MTQ groups. A single administration of VLP or VLP/N mRNA vaccines elicited protective Nab responses, the Nab titres were significantly higher than that in inactivated rabies vaccine group at day 7. Moreover, RABV VLP and VLP/N mRNA vaccines showed superior capacities to elicit potent germinal centre, long-lived plasma cell and memory B cell responses, which linked to high titre and durable Nab responses. In summary, our data demonstrated that RABV VLP and VLP/N mRNA vaccines could be promising candidates against rabies.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Immunity, Cellular , Immunity, Humoral , Rabies Vaccines , Rabies virus , Rabies , Vaccines, Virus-Like Particle , Animals , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies/prevention & control , Rabies/immunology , Rabies virus/immunology , Rabies virus/genetics , Mice , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Female , mRNA Vaccines/immunology , Mice, Inbred BALB C , Nucleosides/immunology , Glycoproteins/immunology , Glycoproteins/genetics , Humans , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Antigens, Viral/immunology , Antigens, Viral/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology
3.
NPJ Biofilms Microbiomes ; 10(1): 68, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117662

ABSTRACT

Shrews being insectivores, serve as natural reservoirs for a wide array of zoonotic viruses, including the recently discovered Langya henipavirus (LayV) in China in 2018. It is crucial to understand the shrew-associated virome, viral diversity, and new viruses. In the current study, we conducted high-throughput sequencing on lung samples obtained from 398 shrews captured along the eastern coast of China, and characterized the high-depth virome of 6 common shrew species (Anourosorex squamipes, Crocidura lasiura, Crocidura shantungensis, Crocidura tanakae, Sorex caecutiens, and Suncus murinus). Our analysis revealed numerous shrew-associated viruses comprising 54 known viruses and 72 new viruses that significantly enhance our understanding of mammalian viruses. Notably, 34 identified viruses possess spillover-risk potential and six were human pathogenic viruses: LayV, influenza A virus (H5N6), rotavirus A, rabies virus, avian paramyxovirus 1, and rat hepatitis E virus. Moreover, ten previously unreported viruses in China were discovered, six among them have spillover-risk potential. Additionally, all 54 known viruses and 12 new viruses had the ability to cross species boundaries. Our data underscore the diversity of shrew-associated viruses and provide a foundation for further studies into tracing and predicting emerging infectious diseases originated from shrews.


Subject(s)
High-Throughput Nucleotide Sequencing , Lung , Shrews , Virome , Animals , Shrews/virology , China , Lung/virology , Virome/genetics , Phylogeny , RNA Viruses/genetics , RNA Viruses/classification , RNA Viruses/isolation & purification , RNA, Viral/genetics , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Rabies virus/genetics , Rabies virus/classification , Rabies virus/isolation & purification , Disease Reservoirs/virology
4.
Viruses ; 16(7)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39066269

ABSTRACT

In addition to the rabies virus (RABV), 16 more lyssavirus species have been identified worldwide, causing a disease similar to RABV. Non-rabies-related human deaths have been described, but the number of cases is unknown, and the potential of such lyssaviruses causing human disease is unpredictable. The current rabies vaccine does not protect against divergent lyssaviruses such as Mokola virus (MOKV) or Lagos bat virus (LBV). Thus, a more broad pan-lyssavirus vaccine is needed. Here, we evaluate a novel lyssavirus vaccine with an attenuated RABV vector harboring a chimeric RABV glycoprotein (G) in which the antigenic site I of MOKV replaces the authentic site of rabies virus (RABVG-cAS1). The recombinant vaccine was utilized to immunize mice and analyze the immune response compared to homologous vaccines. Our findings indicate that the vaccine RABVG-cAS1 was immunogenic and induced high antibody titers against both RABVG and MOKVG. Challenge studies with different lyssaviruses showed that replacing a single antigenic site of RABV G with the corresponding site of MOKV G provides a significant improvement over the homologous RABV vaccine and protects against RABV, Irkut virus (IRKV), and MOKV. This strategy of epitope chimerization paves the way towards a pan-lyssavirus vaccine to safely combat the diseases caused by these viruses.


Subject(s)
Antibodies, Viral , Lyssavirus , Rabies Vaccines , Rabies virus , Rabies , Animals , Lyssavirus/immunology , Lyssavirus/genetics , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Rabies virus/immunology , Rabies virus/genetics , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Rabies/prevention & control , Rabies/immunology , Rabies/virology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/virology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Female , Viral Vaccines/immunology , Glycoproteins/immunology , Glycoproteins/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccine Development , Humans , Antigens, Viral/immunology , Mice, Inbred BALB C
5.
Virol J ; 21(1): 154, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978059

ABSTRACT

BACKGROUND: Rabies is a fatal zoonotic disease whose pathogenesis has not been fully elucidated, and vaccination is the only effective method for protecting against rabies virus infection. Most inactivated vaccines are produced using Vero cells, which are African green monkey kidney cells, to achieve large-scale production. However, there is a potential carcinogenic risk due to nonhuman DNA contamination. Thus, replacing Vero cells with human diploid cells may be a safer strategy. In this study, we developed a novel 2BS cell-adapted rabies virus strain and analysed its sequence, virulence and immunogenicity to determine its application potential as a human diploid cell inactivated vaccine. METHODS AND RESULTS: The 2BS cell-adapted rabies virus strain 2aG4-B40 was established by passage for 40 generations and selection of plaques in 2BS cells. RNA sequence analysis revealed that mutations in 2BS cell-adapted strains were not located at key sites that regulate the production of neutralizing antibodies or virulence in the aG strain (GQ412744.1). The gradual increase in virulence (remaining above 7.0 logLD50/ml from the 40th to 55th generation) and antigen further indicated that these mutations may increase the affinity of the adapted strains for human diploid cells. Identification tests revealed that the 2BS cell-adapted virus strain was neutralized by anti-rabies serum, with a neutralization index of 19,952. PrEP and PEP vaccination and the NIH test further indicated that the vaccine prepared with the 2aG4-B40 strain had high neutralizing antibody levels (2.24 to 46.67 IU/ml), immunogenicity (protection index 270) and potency (average 11.6 IU/ml). CONCLUSIONS: In this study, a 2BS cell-adapted strain of the 2aG4 rabies virus was obtained by passage for 40 generations. The results of sequencing analysis and titre determination of the adapted strain showed that the mutations in the adaptive process are not located at key sequence regions of the virus, and these mutations may enhance the affinity of the adapted strain for human diploid cells. Moreover, vaccines made from the adapted strain 2aG4-B40 had high potency and immunogenicity and could be an ideal candidate rabies virus strain for inactivated vaccine preparation.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Rabies Vaccines , Rabies virus , Rabies , Rabies virus/immunology , Rabies virus/genetics , Rabies virus/pathogenicity , Animals , Rabies Vaccines/immunology , Rabies Vaccines/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Rabies/prevention & control , Rabies/immunology , Rabies/virology , Humans , Antibodies, Viral/immunology , Antibodies, Viral/blood , Chlorocebus aethiops , Virulence , Vaccines, Inactivated/immunology , Vero Cells , China , Mice , Cell Line , Mutation , Female , Immunogenicity, Vaccine
6.
Hum Vaccin Immunother ; 20(1): 2382499, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39069645

ABSTRACT

Rabies, primarily transmitted to humans by dogs (accounting for 99% of cases). Once rabies occurs, its mortality rate is approximately 100%. Post-exposure prophylaxis (PEP) is critical for preventing the onset of rabies after exposure to rabid animals, and vaccination is a pivotal element of PEP. However, high costs and complex immunization protocols have led to poor adherence to rabies vaccinations. Consequently, there is an urgent need to develop new rabies vaccines that are safe, highly immunogenic, and cost-effective to improve compliance and effectively prevent rabies. In recent years, mRNA vaccines have made significant progress in the structural modification and optimization of delivery systems. Various mRNA vaccines are currently undergoing clinical trials, positioning them as viable alternatives to the traditional rabies vaccines. In this article, we discuss a novel mRNA rabies vaccine currently undergoing clinical and preclinical testing, and evaluate its potential to replace existing vaccines.


Subject(s)
Post-Exposure Prophylaxis , Rabies Vaccines , Rabies , mRNA Vaccines , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies/prevention & control , Animals , Humans , Post-Exposure Prophylaxis/methods , Rabies virus/immunology , Rabies virus/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccine Development , Dogs , Clinical Trials as Topic , RNA, Messenger/genetics , RNA, Messenger/immunology
7.
Emerg Infect Dis ; 30(8): 1642-1650, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39043404

ABSTRACT

Rabies, a viral disease that causes lethal encephalitis, kills ≈59,000 persons worldwide annually, despite availability of effective countermeasures. Rabies is endemic in Kenya and is mainly transmitted to humans through bites from rabid domestic dogs. We analyzed 164 brain stems collected from rabid animals in western and eastern Kenya and evaluated the phylogenetic relationships of rabies virus (RABV) from the 2 regions. We also analyzed RABV genomes for potential amino acid changes in the vaccine antigenic sites of nucleoprotein and glycoprotein compared with RABV vaccine strains commonly used in Kenya. We found that RABV genomes from eastern Kenya overwhelmingly clustered with the Africa-1b subclade and RABV from western Kenya clustered with Africa-1a. We noted minimal amino acid variances between the wild and vaccine virus strains. These data confirm minimal viral migration between the 2 regions and that rabies endemicity is the result of limited vaccine coverage rather than limited efficacy.


Subject(s)
Genome, Viral , Phylogeny , Rabies Vaccines , Rabies virus , Rabies , Rabies virus/genetics , Rabies virus/immunology , Rabies virus/classification , Animals , Kenya/epidemiology , Rabies/epidemiology , Rabies/veterinary , Rabies/virology , Rabies/prevention & control , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Dogs , Sequence Alignment , Humans , Phylogeography
8.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932168

ABSTRACT

Seroprevalence of lyssaviruses in certain bat species has been proven in the Republic of Croatia, but there have been no confirmed positive bat brain isolates or human fatalities associated with bat injuries/bites. The study included a retrospective analysis of bat injuries/bites, post-exposure prophylaxis (PEP) and geographic distribution of bat injuries in persons examined at the Zagreb Antirabies Clinic, the Croatian Reference Centre for Rabies. In the period 1995-2020, we examined a total of 21,910 patients due to animal injuries, of which 71 cases were bat-related (0.32%). Of the above number of patients, 4574 received rabies PEP (20.87%). However, for bat injuries, the proportion of patients receiving PEP was significantly higher: 66 out of 71 patients (92.95%). Of these, 33 received only the rabies vaccine, while the other 33 patients received the vaccine with human rabies immunoglobulin (HRIG). In five cases, PEP was not administered, as there was no indication for treatment. Thirty-five of the injured patients were biologists or biology students (49.29%). The bat species was confirmed in only one of the exposure cases. This was a serotine bat (Eptesicus serotinus), a known carrier of Lyssavirus hamburg. The results showed that the bat bites were rather sporadic compared to other human injuries caused by animal bites. All bat injuries should be treated as if they were caused by a rabid animal, and according to WHO recommendations. People who come into contact with bats should be strongly advised to be vaccinated against rabies. Entering bat habitats should be done with caution and in accordance with current recommendations, and nationwide surveillance should be carried out by competent institutions and in close collaboration between bat experts, epidemiologists and rabies experts.


Subject(s)
Bites and Stings , Chiroptera , Post-Exposure Prophylaxis , Rabies Vaccines , Rabies , Rabies/epidemiology , Rabies/prevention & control , Chiroptera/virology , Humans , Animals , Croatia/epidemiology , Female , Bites and Stings/epidemiology , Adult , Male , Retrospective Studies , Middle Aged , Young Adult , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Adolescent , Child , Rabies virus/immunology , Rabies virus/genetics , Aged , Child, Preschool , Seroepidemiologic Studies , Lyssavirus/immunology , Lyssavirus/genetics
9.
Vet Microbiol ; 295: 110159, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38941768

ABSTRACT

Rabies, caused by lyssavirus rabies (Rabies lyssavirus, RABV), is a fatal disease among humans and almost all warm-blooded animals. In this study, we found that RABV infection induces the up-regulation of receptor transporter protein 4 (RTP4) in mouse brains and different cells of nervous tissue. Over-expression of RTP4 reduces the viral titer of RABV in different neuronal cells. Furthermore, a recombinant RABV expressing RTP4, named rRABV-RTP4, was constructed and displayed a lower viral titer in different neuronal cells due to the expression of RTP4. Moreover, the survival rates of mice infected with rRABV-RTP4 were significantly higher than those of mice infected with parent virus rRABV or control virus rRABV-RTP4(-). In terms of mechanism, RTP4 could bind viral genomic RNA (vRNA) of RABV, and suppress the whole viral genome amplification. In addition, we found that the zinc finger domain (ZFD) of RTP4 exerts the antiviral function by truncation analysis, and an important amino acids site (C95) in the RTP4 3CxxC motif which is essential for its antiviral function was identified by mutation analysis. This study contributes to our understanding of how RTP4 or other RTP proteins play a role in defense against the invasion of RABV or other viruses.


Subject(s)
RNA, Viral , Rabies virus , Rabies , Animals , Humans , Mice , Brain/virology , Cell Line , Genome, Viral , Lyssavirus/genetics , Neurons/virology , Rabies/virology , Rabies virus/genetics , Rabies virus/physiology , Rabies virus/pathogenicity , RNA, Viral/genetics , Virus Replication
10.
J Virol Methods ; 327: 114948, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718900

ABSTRACT

Rabies, a fatal zoonotic viral disease affecting mammals, including humans, remains a significant global health concern, particularly in low-income countries. The disease, primarily transmitted through infected animal saliva, prompts urgent diagnosis for timely post-exposure prophylaxis (PEP). The gold standard diagnostic test, direct fluorescent antibody test (dFAT), while sensitive, suffers from limitations such as subjective interpretation and high costs. As a confirmatory technique, the LN34 Pan-Lyssavirus RT-qPCR assay has emerged as a promising tool for universal Lyssavirus detection. This study evaluated its performance using 130 rabies virus isolates representing eleven Brazilian variants and 303 clinical samples from surveillance operations. The LN34 assay demonstrated 100% sensitivity and 98% specificity compared to dFAT. Additionally, it detected all samples, including those missed by dFAT, indicating superior sensitivity. The assay's specificity was confirmed through Sanger nucleotide sequencing, with only a minimal false-positive rate. Comparative analysis revealed higher accuracy and concordance with dFAT than traditional rabies tissue culture infection tests (RTCIT). False-negative RTCIT results were attributed to low viral load or suboptimal sampling. These findings underscore the LN34 assay's utility as a confirmatory technique, enhancing rabies surveillance and control in Brazil. Its widespread adoption could significantly improve diagnostic sensitivity, crucial for effective PEP and public health interventions.


Subject(s)
Rabies virus , Rabies , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Rabies/diagnosis , Rabies/veterinary , Rabies/virology , Brazil , Rabies virus/genetics , Rabies virus/isolation & purification , Rabies virus/classification , Humans , Animals , Real-Time Polymerase Chain Reaction/methods , Lyssavirus/genetics , Lyssavirus/isolation & purification , Lyssavirus/classification , RNA, Viral/genetics , Viral Load
11.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731834

ABSTRACT

Tripartite motif (TRIM) proteins are a multifunctional E3 ubiquitin ligase family that participates in various cellular processes. Recent studies have shown that TRIM proteins play important roles in regulating host-virus interactions through specific pathways, but their involvement in response to rabies virus (RABV) infection remains poorly understood. Here, we identified that several TRIM proteins are upregulated in mouse neuroblastoma cells (NA) after infection with the rabies virus using RNA-seq sequencing. Among them, TRIM44 was found to regulate RABV replication. This is supported by the observations that downregulation of TRIM44 inhibits RABV replication, while overexpression of TRIM44 promotes RABV replication. Mechanistically, TRIM44-induced RABV replication is brought about by activating autophagy, as inhibition of autophagy with 3-MA attenuates TRIM44-induced RABV replication. Additionally, we found that inhibition of autophagy with rapamycin reverses the TRIM44-knockdown-induced decrease in LC3B expression and autophagosome formation as well as RABV replication. The results suggest that TRIM44 promotes RABV replication by an autophagy-dependent mechanism. Our work identifies TRIM44 as a key host factor for RABV replication, and targeting TRIM44 expression may represent an effective therapeutic strategy.


Subject(s)
Autophagy , Rabies virus , Tripartite Motif Proteins , Virus Replication , Animals , Humans , Mice , Autophagy/genetics , Cell Line, Tumor , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Rabies/virology , Rabies/metabolism , Rabies virus/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics
12.
Antiviral Res ; 227: 105905, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740191

ABSTRACT

The rapid emergence of Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2) variants, coupled with severe immune evasion and imprinting, has jeopardized the vaccine efficacy, necessitating urgent development of broad protective vaccines. Here, we propose a strategy employing recombinant rabies viruses (RABV) to create a universal SARS-CoV-2 vaccine expressing heterologous tandem receptor-binding domain (RBD) trimer from the SARS-CoV-2 Prototype, Delta, and Omicron strains (SRV-PDO). The results of mouse immunization indicated that SRV-PDO effectively induced cellular and humoral immune responses, and demonstrated higher immunogenicity and broader SARS-CoV-2 neutralization compared to the recombinant RABVs that only expressed RBD monomers. Moreover, SRV-PDO exhibited full protection against SARS-CoV-2 in the challenge assay. This study demonstrates that recombinant RABV expressing tandem RBD-heterotrimer as a multivalent immunogen could elicit a broad-spectrum immune response and potent protection against SARS-CoV-2, making it a promising candidate for future human or veterinary vaccines and offering a novel perspective in other vaccine design.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Mice, Inbred BALB C , Rabies virus , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Rabies virus/immunology , Rabies virus/genetics , COVID-19 Vaccines/immunology , Mice , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Female , Humans , Immunity, Humoral , Genetic Vectors , Vaccine Efficacy , Vaccines, Synthetic/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/administration & dosage
13.
Sci Rep ; 14(1): 12559, 2024 05 31.
Article in English | MEDLINE | ID: mdl-38822013

ABSTRACT

Rabies virus (RABV) causes fatal neurological disease. Pre-exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP) using inactivated-virus vaccines are the most effective measures to prevent rabies. In Japan, HEP-Flury, the viral strain, used as a human rabies vaccine, has historically been propagated in primary fibroblast cells derived from chicken embryos. In the present study, to reduce the cost and labor of vaccine production, we sought to adapt the original HEP-Flury (HEP) to Vero cells. HEP was repeatedly passaged in Vero cells to generate ten- (HEP-10V) and thirty-passaged (HEP-30V) strains. Both HEP-10V and HEP-30V grew significantly better than HEP in Vero cells, with virulence and antigenicity similar to HEP. Comparison of the complete genomes with HEP revealed three non-synonymous mutations in HEP-10V and four additional non-synonymous mutations in HEP-30V. Comparison among 18 recombinant HEP strains constructed by reverse genetics and vesicular stomatitis viruses pseudotyped with RABV glycoproteins indicated that the substitution P(L115H) in the phosphoprotein and G(S15R) in the glycoprotein improved viral propagation in HEP-10V, while in HEP-30V, G(V164E), G(L183P), and G(A286V) in the glycoprotein enhanced entry into Vero cells. The obtained recombinant RABV strain, rHEP-PG4 strain, with these five substitutions, is a strong candidate for production of human rabies vaccine.


Subject(s)
Amino Acid Substitution , Rabies Vaccines , Rabies virus , Animals , Vero Cells , Chlorocebus aethiops , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies virus/genetics , Rabies virus/immunology , Humans , Rabies/prevention & control , Rabies/virology , Genome, Viral
14.
Viruses ; 16(5)2024 04 28.
Article in English | MEDLINE | ID: mdl-38793581

ABSTRACT

Rabies is a fatal encephalitic infectious disease caused by the rabies virus (RABV). RABV is highly neurotropic and replicates in neuronal cell lines in vitro. The RABV fixed strain, HEP-Flury, was produced via passaging in primary chicken embryonic fibroblast cells. HEP-Flury showed rapid adaptation when propagated in mouse neuroblastoma (MNA) cells. In this study, we compared the growth of our previously constructed recombinant HEP (rHEP) strain-based on the sequence of the HEP (HEP-Flury) strain-with that of the original HEP strain. The original HEP strain exhibited higher titer than rHEP and a single substitution at position 80 in the matrix (M) protein M(D80N) after incubation in MNA cells, which was absent in rHEP. In vivo, intracerebral inoculation of the rHEP-M(D80N) strain with this substitution resulted in enhanced viral growth in the mouse brain and a significant loss of body weight in the adult mice. The number of viral antigen-positive cells in the brains of adult mice inoculated with the rHEP-M(D80N) strain was significantly higher than that with the rHEP strain at 5 days post-inoculation. Our findings demonstrate that a single amino acid substitution in the M protein M(D80N) is associated with neurovirulence in mice owing to adaptation to mouse neuronal cells.


Subject(s)
Amino Acid Substitution , Rabies virus , Rabies , Viral Matrix Proteins , Virulence , Animals , Mice , Brain/virology , Cell Line , Neurons/virology , Neurons/pathology , Rabies/virology , Rabies virus/genetics , Rabies virus/pathogenicity , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virulence/genetics , Virus Replication
15.
Vector Borne Zoonotic Dis ; 24(8): 552-562, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38775097

ABSTRACT

Background: Throughout the Americas, Lyssavirus rabies (RV) perpetuates as multiple variants among bat and mesocarnivore species. Interspecific RV spillover occurs on occasion, but clusters and viral host shifts are rare. The spillover and host shift of a big brown bat (Eptesicus fuscus) RV variant Ef-W1 into mesocarnivores was reported previously on several occasions during 2001-2009 in Flagstaff, Arizona, USA, and controlled through rabies vaccination of target wildlife. During autumn 2021, a new cluster of Ef-W1 RV cases infecting striped skunks (Mephitis mephitis) was detected from United States Department of Agriculture enhanced rabies surveillance in Flagstaff. The number of Ef-W1 RV spillover cases within a short timeframe suggested the potential for transmission between skunks and an emerging host shift. Materials and Methods: Whole and partial RV genomic sequencing was performed to evaluate the phylogenetic relationships of the 2021-2023 Ef-W1 cases infecting striped skunks with earlier outbreaks. Additionally, real-time reverse-transcriptase PCR (rtRT-PCR) was used to opportunistically compare viral RNA loads in brain and salivary gland tissues of naturally infected skunks. Results: Genomic RV sequencing revealed that the origin of the 2021-2023 epizootic of Ef-W1 RV was distinct from the multiple outbreaks detected from 2001-2009. Naturally infected skunks with the Ef-W1 RV showed greater viral RNA loads in the brain, but equivalent viral RNA loads in the mandibular salivary glands, compared to an opportunistic sample of skunks naturally infected with a South-Central skunk RV from northern Colorado, USA. Conclusion: Considering a high risk for onward transmission and spread of the Ef-W1 RV in Flagstaff, public outreach, enhanced rabies surveillance, and control efforts, focused on education, sample characterization, and vaccination, have been ongoing since 2021 to mitigate and prevent the spread and establishment of Ef-W1 RV in mesocarnivores.


Subject(s)
Chiroptera , Mephitidae , Phylogeny , Rabies , Animals , Arizona/epidemiology , Mephitidae/virology , Rabies/epidemiology , Rabies/veterinary , Rabies/virology , Chiroptera/virology , Rabies virus/genetics , Rabies virus/classification , Rabies virus/isolation & purification , Lyssavirus/genetics , Lyssavirus/classification , Lyssavirus/isolation & purification , Communicable Diseases, Emerging/virology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/veterinary , Genome, Viral
16.
J Vet Diagn Invest ; 36(4): 522-528, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38653733

ABSTRACT

Rabies virus (RABV; Lyssavirus rabies) is a neurotropic virus that can be transmitted to mammals by the hematophagous bat Desmodus rotundus. An accurate, accessible method for the detection of RABV in cattle is necessary in Paraguay; thus, we evaluated the detection of RABV using 4 techniques: fluorescent antibody test (FAT), immunochromatography rapid detection test (RDT; Anigen Rapid Rabies Ag test kit; Bionote), a reverse-transcription PCR (RT-PCR) assay, and histologic lesions in different portions of the CNS of 49 Paraguayan cattle to determine the most sensitive and specific technique. By FAT and RDT, 15 of 49 (31%) samples were positive. By RT-PCR amplification of N and G genes, 13 of 49 (27%) and 12 of 49 (25%) were positive, respectively. RDT had high agreement with FAT (kappa = 1); sensitivity was 100% (95% CI: 97-100%) and specificity was 100% (95% CI: 99-100%). The amplification of the N and G genes resulted in substantial agreement (kappa of 0.9 and 0.8, respectively) compared with FAT, and the sensitivity and specificity of the N gene were 87% (95% CI: 66-100%) and 100% (95% CI: 98-100%), respectively, and those of the G gene were 80% (95% CI: 56-100%) and 100% (95% CI: 98-100%), respectively. Histologic lesions observed were lymphoplasmacytic meningoencephalitis, gliosis, and neuronophagia. The agreement observed between the FAT and RDT tests suggests that RDT is an accurate tool for the detection of RABV. Histopathology can be used to confirm lesions caused by RABV and to rule out other conditions; the RT-PCR assay is useful for molecular epidemiology studies.


Subject(s)
Cattle Diseases , Rabies virus , Rabies , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Animals , Rabies/veterinary , Rabies/diagnosis , Rabies/virology , Cattle , Paraguay , Rabies virus/isolation & purification , Rabies virus/genetics , Cattle Diseases/virology , Cattle Diseases/diagnosis , Cattle Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Fluorescent Antibody Technique/veterinary
17.
J Wildl Dis ; 60(3): 745-752, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38685759

ABSTRACT

Wildlife translocation and cross-species transmission can impede control and elimination of emerging zoonotic diseases. Tracking the geographic origin of both host and virus (i.e., translocation versus local infection) may help determine the most effective response when high-risk cases of emerging pathogens are identified in wildlife. In May 2022, a coyote (Canis latrans) infected with the raccoon (Procyon lotor) rabies virus variant (RRV) was collected in Lewis County, West Virginia, USA, an area free from RRV. We applied host population genomics and RRV phylogenetic analyses to determine the most likely geographic origin of the rabid coyote. Coyote genomic analyses included animals from multiple eastern states bordering West Virginia, with the probable origin of the rabid coyote being the county of collection. The RRV phylogenetic analyses included cases detected from West Virginia and neighboring states, with most similar RRV sequences collected in a county 80 km to the northeast, within the oral rabies vaccination zone. The combined results suggest that the coyote was infected in an RRV management area and carried the RRV to Lewis County, a pattern consistent with coyote local movement ecology. Distant cross-species transmission and subsequent host movement presents a low risk for onward transmission in raccoon populations. This information helped with emergency response decision-making, thereby saving time and resources.


Subject(s)
Coyotes , Phylogeny , Rabies virus , Rabies , Animals , Coyotes/virology , West Virginia/epidemiology , Rabies/veterinary , Rabies/epidemiology , Rabies virus/genetics , Rabies virus/isolation & purification , Rabies virus/classification , Raccoons/virology , Animals, Wild
18.
Vet J ; 304: 106096, 2024 04.
Article in English | MEDLINE | ID: mdl-38503385

ABSTRACT

Feline viral rhinotracheitis (FVR) is caused by the feline herpesvirus-1 (FHV-1), which commonly results in upper respiratory symptoms, and can result in death in the kittens and weak cats. Rabies is an infectious disease with zoonotic characteristics highly relevant to public health and also poses a serious threat to cats. Vaccines are the most effective method to control the spread of both FHV-1 and RABV and have the advantage that they produce long-term specific immune responses. In this study, we constructed a bivalent vaccine against FHV-1 and rabies virus (RABV) simultaneously. The vaccine was constructed by cloning FHV-1 gB into a RABV based vector, and the recombinant RABV (SRV9-FHV-gB) expressing the FHV-1 gB protein was rescued. The growth characteristics of SRV9-FHV-gB were analyzed on NA and BSR cells. To assess the immunogenicity of the vaccine, mice and cats were immunized with SRV9-FHV-gB supplemented with Gel02 adjuvant. The SRV9-FHV-gB exhibited the same growth characteristics as the parent virus SRV9 in both BSR cells and NA cells. The safety of SRV9-FHV-gB was evaluated using 5-day-old and 14-day-old suckling mice. The results showed that mice infected with the SRV9-FHV-gB survived for longer than those in the SRV9 group. Mice immunized with inactivated SRV9-FHV-gB produced high titers of specific antibodies against FHV-1 and neutralizing antibodies against RABV. Cats that received three immunizations with SRV9-FHV-gB also produced neutralizing antibodies against both FHV-1 and RABV. This study represents the first time that a bivalent vaccine targeting FHV-1 and RABV has been constructed, laying the foundations and providing inspiration for the development of other multivalent vaccines.


Subject(s)
Cat Diseases , Rabies Vaccines , Rabies virus , Rabies , Rodent Diseases , Varicellovirus , Cats , Animals , Female , Mice , Rabies/prevention & control , Rabies/veterinary , Rabies virus/genetics , Vaccines, Combined , Vaccines, Synthetic , Antibodies, Neutralizing , Antibodies, Viral , Cat Diseases/prevention & control
19.
Adv Sci (Weinh) ; 11(21): e2309305, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38509833

ABSTRACT

Spinal cord injury (SCI) has no effective treatment modalities. It faces a significant global therapeutical challenge, given its features of poor axon regeneration, progressive local inflammation, and inefficient systemic drug delivery due to the blood-spinal cord barrier (BSCB). To address these challenges, a new nano complex that achieves targeted drug delivery to the damaged spinal cord is proposed, which contains a mesoporous silica nanoparticle core loaded with microRNA and a cloaking layer of human umbilical cord mesenchymal stem cell membrane modified with rabies virus glycoprotein (RVG). The nano complex more readily crosses the damaged BSCB with its exosome-resembling properties, including appropriate size and a low-immunogenic cell membrane disguise and accumulates in the injury center because of RVG, where it releases abundant microRNAs to elicit axon sprouting and rehabilitate the inflammatory microenvironment. Culturing with nano complexes promotes axonal growth in neurons and M2 polarization in microglia. Furthermore, it showed that SCI mice treated with this nano complex by tail vein injection display significant improvement in axon regrowth, microenvironment regulation, and functional restoration. The efficacy and biocompatibility of the targeted delivery of microRNA by nano complexes demonstrate their immense potential as a noninvasive treatment for SCI.


Subject(s)
Disease Models, Animal , MicroRNAs , Rabies virus , Silicon Dioxide , Spinal Cord Injuries , Animals , MicroRNAs/genetics , MicroRNAs/administration & dosage , Spinal Cord Injuries/therapy , Mice , Silicon Dioxide/chemistry , Rabies virus/genetics , Glycoproteins/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Cell Membrane/metabolism , Drug Delivery Systems/methods , Nanoparticles/chemistry
20.
Microbes Infect ; 26(4): 105321, 2024.
Article in English | MEDLINE | ID: mdl-38461968

ABSTRACT

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Subject(s)
DEAD Box Protein 58 , Rabies virus , Humans , Cell Line , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Interferon Type I/metabolism , Interferon Type I/immunology , Rabies/immunology , Rabies/virology , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies virus/genetics , Rabies virus/pathogenicity , Receptors, Immunologic/metabolism , RNA, Viral/genetics , Signal Transduction , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL