Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
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.
Antiviral Res ; 229: 105977, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089332

ABSTRACT

Rabies is a fatal neurological disorder caused by rabies virus (RABV) infection. Approximately 60,000 patients die from rabies annually, and there are no effective treatments for this disease. Nucleoside analogs are employed as antiviral drugs based on their broad antiviral spectrum, and certain nucleoside analogs have been reported to exhibit anti-RABV activity. The nucleoside analog ß-d-N4-hydroxycytidine (NHC) has antiviral effects against a range of RNA viruses. Molnupiravir (MPV), a prodrug of NHC, is clinically used as an oral antiviral drug for coronavirus infections. Despite its broad-spectrum activity, the antiviral activity of NHC against RABV remains unclear. In this study, we reveal that NHC exhibits comparable in vitro anti-RABV activity as ribavirin and favipiravir (also known as T-705) with a 90% effective concentration of 6 µM in mouse neuroblastoma cells. NHC reduced viral loads in neuronal and nonneuronal cells in a dose-dependent manner. Both laboratory and field RABVs (fixed and street strains, respectively) were susceptible to NHC. However, no increase in survival or reduction in viral titers in the brain was observed in RABV-infected mice treated prophylactically with MPV. These findings highlight the potential and challenges of NHC in the treatment of RABV infection.


Subject(s)
Amides , Antiviral Agents , Cytidine , Rabies virus , Rabies , Viral Load , Animals , Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Cytidine/pharmacology , Rabies virus/drug effects , Mice , Rabies/drug therapy , Rabies/virology , Amides/pharmacology , Viral Load/drug effects , Pyrazines/pharmacology , Ribavirin/pharmacology , Hydroxylamines/pharmacology , Cell Line, Tumor , Cell Line
3.
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
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.
Acta Trop ; 257: 107309, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38955321

ABSTRACT

Bats are the second most diverse order of mammals and play a central role in ecosystem dynamics. They are also important reservoirs of potentially zoonotic microorganisms, of which rabies virus is the most lethal among the bat-transmitted zoonotic pathogens. Importantly, recent outbreaks of human rabies have been reported from the Brazilian Amazon. Here we present a survey of bat species and rabies virus (RABV) circulation in a bat assemblage in the Marajó region, northern Brazil. Using data from mist-net captures and bioacoustic sampling, 56 bat species were recorded along the Jacundá River basin over a 10-day expedition in November 2022. For the investigation of RABV, we used the direct fluorescent antibody test (DFAT) and the rapid fluorescent focus inhibition test (RFFIT). In total, 159 bat individuals from 22 species were investigated for RABV. Five adults of the common vampire bat, Desmodus rotundus, showed RABV-specific antibodies in serum samples. Additionally, we report on local residents with injuries caused by D. rotundus bites and the occurrence of colonies of non-hematophagous bats from different species roosting inside human residences. This scenario raises concerns about the risks of new cases of human rabies and other zoonotic diseases associated with bats in the region and highlights the need for epidemiological surveillance and mitigation measures to prevent outbreaks of emerging infectious diseases.


Subject(s)
Antibodies, Viral , Chiroptera , Disease Outbreaks , Rabies virus , Rabies , Zoonoses , Chiroptera/virology , Animals , Brazil/epidemiology , Rabies virus/immunology , Rabies virus/isolation & purification , Rabies virus/classification , Rabies/epidemiology , Rabies/veterinary , Rabies/virology , Humans , Zoonoses/epidemiology , Zoonoses/virology , Antibodies, Viral/blood , Female , Male , Adult , Middle Aged , Adolescent
6.
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
7.
Front Immunol ; 15: 1392804, 2024.
Article in English | MEDLINE | ID: mdl-38868762

ABSTRACT

Rabies virus (RABV) causes a fatal neurological disease, consisting of unsegmented negative-strand RNA, which encodes five structural proteins (3'-N-P-M-G-L-5'). Apolipoprotein D (ApoD), a lipocalin, is upregulated in the nervous system after injury or pathological changes. Few studies have focused on the role of ApoD during virus infection so far. This study demonstrated that ApoD is upregulated in the mouse brain (in vivo) and C8-D1A cells (in vitro) after RABV infection. By upregulating ApoD expression in C8-D1A cells, we found that ApoD facilitated RABV replication. Additionally, Co-immunoprecipitation demonstrated that ApoD interacted with RABV glycoprotein (G protein). The interaction could promote RABV replication by upregulating the cholesterol level. These findings revealed a novel role of ApoD in promoting RABV replication and provided a potential therapeutic target for rabies.


Subject(s)
Apolipoproteins D , Cholesterol , Rabies virus , Rabies , Virus Replication , Animals , Female , Humans , Male , Mice , Apolipoproteins D/metabolism , Apolipoproteins D/genetics , Brain/virology , Brain/metabolism , Cell Line , Cholesterol/metabolism , HEK293 Cells , Rabies/metabolism , Rabies/virology , Rabies virus/physiology , Up-Regulation
8.
Viruses ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38932209

ABSTRACT

A proteomics analysis of purified rabies virus (RABV) revealed 47 entrapped host proteins within the viral particles. Out of these, 11 proteins were highly disordered. Our study was particularly focused on five of the RABV-entrapped mouse proteins with the highest levels of disorder: Neuromodulin, Chmp4b, DnaJB6, Vps37B, and Wasl. We extensively utilized bioinformatics tools, such as FuzDrop, D2P2, UniProt, RIDAO, STRING, AlphaFold, and ELM, for a comprehensive analysis of the intrinsic disorder propensity of these proteins. Our analysis suggested that these disordered host proteins might play a significant role in facilitating the rabies virus pathogenicity, immune system evasion, and the development of antiviral drug resistance. Our study highlighted the complex interaction of the virus with its host, with a focus on how the intrinsic disorder can play a crucial role in virus pathogenic processes, and suggested that these intrinsically disordered proteins (IDPs) and disorder-related host interactions can also be a potential target for therapeutic strategies.


Subject(s)
Intrinsically Disordered Proteins , Rabies virus , Virion , Rabies virus/physiology , Animals , Mice , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Virion/metabolism , Proteomics , Host-Pathogen Interactions , Rabies/virology , Computational Biology/methods , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry
9.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891803

ABSTRACT

Rabies virus (RABV) is a neurotropic virus that causes fatal neurological disease, raising serious public health issues and attracting extensive attention in society. To elucidate the molecular mechanism of RABV-induced neuronal damage, we used hematoxylin-eosin staining, transmission electron microscopy, transcriptomics analysis, and immune response factor testing to investigate RABV-infected neurons. We successfully isolated the neurons from murine brains. The specificity of the isolated neurons was identified by a monoclonal antibody, and the viability of the neurons was 83.53-95.0%. We confirmed that RABV infection induced serious damage to the neurons according to histochemistry and transmission electron microscope (TEM) scanning. In addition, the transcriptomics analysis suggested that multiple genes related to the pyroptosis pathway were significantly upregulated, including gasdermin D (Gsdmd), Nlrp3, caspase-1, and IL-1ß, as well as the chemokine genes Ccl2, Ccl3, Ccl4, Ccl5, Ccl7, Ccl12, and Cxcl10. We next verified this finding in the brains of mice infected with the rRC-HL, GX074, and challenge virus standard strain-24 (CVS-24) strains of RABV. Importantly, we found that the expression level of the Gsdmd protein was significantly upregulated in the neurons infected with different RABV strains and ranged from 691.1 to 5764.96 pg/mL, while the basal level of mock-infected neurons was less than 100 pg/mL. Taken together, our findings suggest that Gsdmd-induced pyroptosis is involved in the neuron damage caused by RABV infection.


Subject(s)
Neurons , Phosphate-Binding Proteins , Pyroptosis , Rabies virus , Rabies , Animals , Neurons/virology , Neurons/metabolism , Neurons/pathology , Rabies virus/pathogenicity , Rabies virus/physiology , Rabies/virology , Rabies/pathology , Rabies/metabolism , Mice , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Brain/virology , Brain/pathology , Brain/metabolism , Gasdermins
10.
Front Cell Infect Microbiol ; 14: 1394713, 2024.
Article in English | MEDLINE | ID: mdl-38836054

ABSTRACT

The rabies virus enters the nervous system by interacting with several molecular targets on host cells to modify behavior and trigger receptor-mediated endocytosis of the virion by poorly understood mechanisms. The rabies virus glycoprotein (RVG) interacts with the muscle acetylcholine receptor and the neuronal α4ß2 subtype of the nicotinic acetylcholine receptor (nAChR) family by the putative neurotoxin-like motif. Given that the neurotoxin-like motif is highly homologous to the α7 nAChR subtype selective snake toxin α-bungarotoxin (αBTX), other nAChR subtypes are likely involved. The purpose of this study is to determine the activity of the RVG neurotoxin-like motif on nAChR subtypes that are expressed in brain regions involved in rabid animal behavior. nAChRs were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to collect concentration-response data to measure the functional effects. The RVG peptide preferentially and completely inhibits α7 nAChR ACh-induced currents by a competitive antagonist mechanism. Tested heteromeric nAChRs are also inhibited, but to a lesser extent than the α7 subtype. Residues of the RVG peptide with high sequence homology to αBTX and other neurotoxins were substituted with alanine. Altered RVG neurotoxin-like peptides showed that residues phenylalanine 192, arginine 196, and arginine 199 are important determinants of RVG peptide apparent potency on α7 nAChRs, while serine 195 is not. The evaluation of the rabies ectodomain reaffirmed the observations made with the RVG peptide, illustrating a significant inhibitory impact on α7 nAChR with potency in the nanomolar range. In a mammalian cell culture model of neurons, we confirm that the RVG peptide binds preferentially to cells expressing the α7 nAChR. Defining the activity of the RVG peptide on nAChRs expands our understanding of basic mechanisms in host-pathogen interactions that result in neurological disorders.


Subject(s)
Glycoproteins , Rabies virus , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Rabies virus/physiology , Rabies virus/metabolism , Humans , Glycoproteins/metabolism , Glycoproteins/genetics , Oocytes/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Host-Pathogen Interactions , Protein Binding , Rabies/metabolism , Rabies/virology , Acetylcholine/metabolism , Acetylcholine/pharmacology , Neurotoxins/metabolism , Neurotoxins/pharmacology
11.
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
12.
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
13.
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
14.
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
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.
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
17.
J Virol ; 98(7): e0060624, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38809020

ABSTRACT

Rabies virus (RABV) is highly lethal and triggers severe neurological symptoms. The neuropathogenic mechanism remains poorly understood. Ras-related C3 botulinum toxin substrate 1 (Rac1) is a Rho-GTPase that is involved in actin remodeling and has been reported to be closely associated with neuronal dysfunction. In this study, by means of a combination of pharmacological inhibitors, small interfering RNA, and specific dominant-negatives, we characterize the crucial roles of dynamic actin and the regulatory function of Rac1 in RABV infection, dominantly in the viral entry phase. The data show that the RABV phosphoprotein interacts with Rac1. RABV phosphoprotein suppress Rac1 activity and impedes downstream Pak1-Limk1-Cofilin1 signaling, leading to the disruption of F-actin-based structure formation. In early viral infection, the EGFR-Rac1-signaling pathway undergoes a biphasic change, which is first upregulated and subsequently downregulated, corresponding to the RABV entry-induced remodeling pattern of F-actin. Taken together, our findings demonstrate for the first time the role played by the Rac1 signaling pathway in RABV infection and may provide a clue for an explanation for the etiology of rabies neurological pathogenesis.IMPORTANCEThough neuronal dysfunction is predominant in fatal rabies, the detailed mechanism by which rabies virus (RABV) infection causes neurological symptoms remains in question. The actin cytoskeleton is involved in numerous viruses infection and plays a crucial role in maintaining neurological function. The cytoskeletal disruption is closely associated with abnormal nervous symptoms and induces neurogenic diseases. In this study, we show that RABV infection led to the rearrangement of the cytoskeleton as well as the biphasic kinetics of the Rac1 signal transduction. These results help elucidate the mechanism that causes the aberrant neuronal processes by RABV infection and may shed light on therapeutic development aimed at ameliorating neurological disorders.


Subject(s)
Actin Cytoskeleton , Actins , Rabies virus , Signal Transduction , rac1 GTP-Binding Protein , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Humans , Actin Cytoskeleton/metabolism , Animals , Rabies virus/physiology , Actins/metabolism , ErbB Receptors/metabolism , ErbB Receptors/genetics , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Lim Kinases/metabolism , Lim Kinases/genetics , Virus Internalization , Rabies/metabolism , Rabies/virology , Cell Line , Phosphoproteins/metabolism , Phosphoproteins/genetics , Actin Depolymerizing Factors/metabolism
18.
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
19.
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
20.
Rev. cuba. med. trop ; 75(1)abr. 2023.
Article in Spanish | LILACS, CUMED | ID: biblio-1550871

ABSTRACT

Introducción: La rabia es una enfermedad zoonótica asociada al virus RABV, el cual tiene características neurotrópicas. El virus se transmite por el contacto con saliva de animales infectados; la mordedura de un perro es la causa más común. Es un virus que causa la muerte de miles de personas cada año. Objetivo: Describir a profundidad los principios moleculares de la infección por rabia, así como su patogenia, diagnóstico y tratamiento. Métodos: Se realizó una búsqueda de bibliografía en PubMed, SciELO, Scopus, Researchgate; se consultaron 163 referencias y se seleccionaron 51 fuentes que contenían la información más relevante para cumplir con el objetivo del trabajo. Conclusión: Actualmente es posible entender de mejor manera los mecanismos de transmisión y propagación del virus en el organismo; existe nuevo conocimiento sobre los receptores involucrados, así como la función de estos en la replicación viral. Sin embargo, el objetivo de la erradicación de la rabia a corto plazo es complejo. La invasión de territorios selváticos vuelve a la rabia un posible patógeno reemergente; la vacunación de especies transmisoras es el medio ideal para conseguir el control de la enfermedad.


Introduction: Rabies is a zoonotic disease associated with the RABV virus, which has neurotropic characteristics. The virus is transmitted by contact with saliva from infected animals; a dog's bite is the most common cause. This virus causes the death of thousands of people every year. Objective: To describe in depth the molecular principles of rabies infection, as well as its pathogenesis, diagnosis and treatment. Methods: A literature search was conducted in PubMed, SciELO, Scopus, and Researchgate. A total of 163 references were consulted, and 51 sources containing the most relevant information were selected to fulfill the objective of the work. Conclusions: It is currently possible to better understand the mechanisms of transmission and spread of the virus in the organism; there is new knowledge about the receptors involved, as well as their function in viral replication. However, the goal of eradicating rabies in the short term is complex. The invasion of wild territories makes rabies a possible re-emerging pathogen; vaccination of transmitting species is the ideal means to achieve disease control.


Subject(s)
Humans , Rabies/epidemiology , Rabies/virology
SELECTION OF CITATIONS
SEARCH DETAIL