Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Med Virol ; 96(7): e29788, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982767

ABSTRACT

Molecular surveillance is vital for monitoring arboviruses, often employing genus-specific quantitative reverse-transcription polymerase chain reaction (RT-qPCR). Despite this, an overlooked chikungunya fever outbreak occurred in Yunnan province, China, in 2019 and false negatives are commonly encountered during alphaviruses screening practice, highlighting the need for improved detection methods. In this study, we developed an improved alphaviruses-specific RT-qPCR capable of detecting chikungunya virus, eastern equine encephalitis virus, western equine encephalitis virus, Venezuelan equine encephalitis virus, Sindbis virus, Mayaro virus, and Ross River virus with high sensitivity and specificity. The assay identified three chikungunya virus-positive cases out of 188 sera retrospectively. Later genetic characterization suggested that imported cases from neighboring countries may be responsible for the neglected chikungunya fever outbreak of 2019 in Yunnan. Our findings underscore the value of improved alphaviruses-specific RT-qPCR in bolstering alphaviruses surveillance and informing preventive strategies.


Subject(s)
Alphavirus Infections , Alphavirus , Chikungunya virus , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Humans , Alphavirus/genetics , Alphavirus/isolation & purification , Alphavirus Infections/diagnosis , Alphavirus Infections/virology , Alphavirus Infections/prevention & control , Alphavirus Infections/epidemiology , China/epidemiology , Real-Time Polymerase Chain Reaction/methods , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Retrospective Studies , Chikungunya Fever/diagnosis , Chikungunya Fever/prevention & control , Chikungunya Fever/virology , Chikungunya Fever/epidemiology , Encephalitis Virus, Eastern Equine/genetics , Disease Outbreaks/prevention & control , Sindbis Virus/genetics , Encephalitis Virus, Western Equine/genetics , Ross River virus/genetics , Ross River virus/isolation & purification , Encephalitis Virus, Venezuelan Equine/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , RNA, Viral/genetics
2.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38995674

ABSTRACT

Mayaro virus (MAYV), a mosquito-borne alphavirus, is considered an emerging threat to public health with epidemic potential. Phylogenetic studies show the existence of three MAYV genotypes. In this study, we provide a preliminary analysis of the pathogenesis of all three MAYV genotypes in cynomolgus macaques (Macaca facicularis, Mauritian origin). Significant MAYV-specific RNAemia and viremia were detected during acute infection in animals challenged intravenously with the three MAYV genotypes, and strong neutralizing antibody responses were observed. MAYV RNA was detected at high levels in lymphoid tissues, joint muscle and synovia over 1 month after infection, suggesting that this model could serve as a promising tool in studying MAYV-induced chronic arthralgia, which can persist for years. Significant leucopenia was observed across all MAYV genotypes, peaking with RNAemia. Notable differences in the severity of acute RNAemia and composition of cytokine responses were observed among the three MAYV genotypes. Our model showed no outward signs of clinical disease, but several major endpoints for future MAYV pathology and intervention studies are described. Disruptions to normal blood cell counts and cytokine responses were markedly distinct from those observed in macaque models of CHIKV infection, underlining the importance of developing non-human primate models specific to MAYV infection.


Subject(s)
Alphavirus Infections , Alphavirus , Genotype , Macaca fascicularis , RNA, Viral , Viremia , Animals , Macaca fascicularis/virology , Alphavirus/genetics , Alphavirus/pathogenicity , Alphavirus/classification , Alphavirus/isolation & purification , Alphavirus Infections/virology , Alphavirus Infections/veterinary , Viremia/virology , RNA, Viral/genetics , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Disease Models, Animal , Phylogeny , Cytokines/genetics , Cytokines/blood
3.
Virology ; 598: 110174, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39029332

ABSTRACT

Getah virus (GETV) is a re-emerging mosquito-borne RNA virus that induces fever, hind limb edema, swollen submandibular lymph nodes, and urticaria in horses. In pigs, the virus often results in stillbirths among pregnant sows, and neurological symptoms leading to death in piglets. Currently, there are no specific treatments or drugs available for GETV infection. The use of reporter viruses to monitor viral replication and spread in real-time within infected cells and animals provides a powerful tool for targeting antiviral drugs throughout the viral life cycle. Their fluorescence-tracked characteristics greatly facilitate virus neutralization tests (VNTs). In this study, we engineered two recombinant viruses by inserting different reporter protein genes at the 3' end of the structural protein gene, an unreported location that can accommodate exogenous genes. The rGEEiLOV and rGEEGFP viruses demonstrated genetic stability for at least five passages and replicated at a rate similar to that of the parental virus in BHK-21 cells. The rGEEGFP virus facilitated viral neutralization testing. Additionally, we used the reporter virus rGEEGFP to confirm ivermectin, a broad-spectrum antiparasitic agent, as a potential inhibitor of GETV in vitro. Ivermectin appears to inhibit the early replication stages of the virus and can block cell-to-cell viral transmission. In conclusion, rGEEGFP holds significant potential for antiviral screening to identify specific inhibitors against GETV and for use in viral neutralization tests.


Subject(s)
Antiviral Agents , Genes, Reporter , Green Fluorescent Proteins , Neutralization Tests , Animals , Antiviral Agents/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Cell Line , Drug Evaluation, Preclinical/methods , Virus Replication/drug effects , Alphavirus/genetics , Alphavirus/drug effects , Swine , Cricetinae
4.
mBio ; 15(8): e0099324, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38953633

ABSTRACT

Barmah Forest virus (BFV) is a mosquito-borne virus that causes arthralgia with accompanying rash, fever, and myalgia in humans. The virus is mainly found in Australia and has caused outbreaks associated with significant health concerns. As the sole representative of the Barmah Forest complex within the genus Alphavirus, BFV is not closely related genetically to other alphaviruses. Notably, basic knowledge of BFV molecular virology has not been well studied due to a lack of critical investigative tools such as an infectious clone. Here we describe the construction of an infectious BFV cDNA clone based on Genbank sequence and demonstrate that the clone-derived virus has in vitro and in vivo properties similar to naturally occurring virus, BFV field isolate 2193 (BFV2193-FI). A substitution in nsP4, V1911D, which was identified in the Genbank reference sequence, was found to inhibit virus rescue and replication. T1325P substitution in nsP2 selected during virus passaging was shown to be an adaptive mutation, compensating for the inhibitory effect of nsP4-V1911D. The two mutations were associated with changes in viral non-structural polyprotein processing and type I interferon (IFN) induction. Interestingly, a nuclear localization signal, active in mammalian but not mosquito cells, was identified in nsP3. A point mutation abolishing nsP3 nuclear localization attenuated BFV replication. This effect was more prominent in the presence of type I interferon signaling, suggesting nsP3 nuclear localization might be associated with IFN antagonism. Furthermore, abolishing nsP3 nuclear localization reduced virus replication in mice but did not significantly affect disease.IMPORTANCEBarmah Forest virus (BFV) is Australia's second most prevalent arbovirus, with approximately 1,000 cases reported annually. The clinical symptoms of BFV infection include rash, polyarthritis, arthralgia, and myalgia. As BFV is not closely related to other pathogenic alphaviruses or well-studied model viruses, our understanding of its molecular virology and mechanisms of pathogenesis is limited. There is also a lack of molecular tools essential for corresponding studies. Here we describe the construction of an infectious clone of BFV, variants harboring point mutations, and sequences encoding marker protein. In infected mammalian cells, nsP3 of BFV was located in the nuclei. This finding extends our understanding of the diverse mechanisms used by alphavirus replicase proteins to interact with host cells. Our novel observations highlight the complex synergy through which the viral replication machinery evolves to correct mutation errors within the viral genome.


Subject(s)
Alphavirus Infections , Alphavirus , Genome, Viral , Viral Nonstructural Proteins , Virus Replication , Virus Replication/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Animals , Alphavirus/genetics , Alphavirus/pathogenicity , Mice , Alphavirus Infections/virology , Genome, Viral/genetics , Cell Line , Humans , Australia
5.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932175

ABSTRACT

Newly emerging viruses, primarily zoonotic or vector-borne, pose a persistent threat to public health and have led to outbreaks of global concern [...].


Subject(s)
Alphavirus Infections , Alphavirus , Flavivirus Infections , Flavivirus , Alphavirus/physiology , Alphavirus/genetics , Humans , Animals , Flavivirus/genetics , Flavivirus/physiology , Alphavirus Infections/virology , Alphavirus Infections/epidemiology , Flavivirus Infections/virology , Flavivirus Infections/epidemiology
6.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851803

ABSTRACT

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Subject(s)
Alphavirus , Antiviral Agents , Receptors, LDL , Virus Internalization , Animals , Humans , Alphavirus/drug effects , Alphavirus/physiology , Alphavirus/genetics , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Protein Binding , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Virion/metabolism , Virus Internalization/drug effects
7.
Microbiol Spectr ; 12(8): e0048324, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38916356

ABSTRACT

From May to July of 2023, one pig farm in Heyuan city, Guangdong Province of China, suffered severe piglet death and sow reproductive disorders. The common pig viruses and bacteria tested negative. To uncover the possible cause of the disease, a metagenomic analysis was performed in the pooled small intestine samples from three 8-day-old diseased piglets. The results showed that Getah virus (GETV), an RNA virus, might be the potential pathogen that affects pig health. Subsequently, GETV nucleotide was detected in all of the 15 samples collected from three diseased piglets using quantitative reverse transcription PCR, suggesting GETV as the main pathogen of the disease. A GETV strain, designated as GDHYLC23, was successfully isolated using the swine testicle cell line. Sequence analysis showed that the epidemic strain had a unique 32-nucleotide repeat insertion in the 3' noncoding region. Phylogenetic analysis showed that GDHYLC23 belonged to the pandemic group III. The identification of GETV with new variations implies the continuous evolution of the virus, which poses potential threats to the swine industry.IMPORTANCEPig farms are faced with emerging and re-emerging viruses that may cause substantial economic loss. The identification of potentially pathogenic viruses helps to prevent and control the spread of diseases. In this study, by using metagenomic analysis, we found that a neglected virus, GETV with a unique insertion in the genome, was the main pathogen in one pig farm that suffered severe piglet death and sow reproductive disorders. Although the potential impact of such an insertion on viral pathogenicity is unknown, the surveillance of the continuing evolution of GETV in pig farms cannot be ignored.


Subject(s)
Phylogeny , Swine Diseases , Animals , Swine , Swine Diseases/virology , Swine Diseases/epidemiology , China/epidemiology , Metagenomics , Alphavirus/genetics , Alphavirus/isolation & purification , Alphavirus/classification , Genome, Viral/genetics , Farms
8.
Mol Ther ; 32(8): 2519-2534, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38894543

ABSTRACT

Self-amplifying mRNA (SAM) vaccines can be rapidly deployed in the event of disease outbreaks. A legitimate safety concern is the potential for recombination between alphavirus-based SAM vaccines and circulating viruses. This theoretical risk needs to be assessed in the regulatory process for SAM vaccine approval. Herein, we undertake extensive in vitro and in vivo assessments to explore recombination between SAM vaccine and a wide selection of alphaviruses and a coronavirus. SAM vaccines were found to effectively limit alphavirus co-infection through superinfection exclusion, although some co-replication was still possible. Using sensitive cell-based assays, replication-competent alphavirus chimeras were generated in vitro as a result of rare, but reproducible, RNA recombination events. The chimeras displayed no increased fitness in cell culture. Viable alphavirus chimeras were not detected in vivo in C57BL/6J, Rag1-/- and Ifnar-/- mice, in which high levels of SAM vaccine and alphavirus co-replicated in the same tissue. Furthermore, recombination between a SAM-spike vaccine and a swine coronavirus was not observed. In conclusion we state that although the ability of SAM vaccines to recombine with alphaviruses might be viewed as an environmental safety concern, several key factors substantially mitigate against in vivo emergence of chimeric viruses from SAM vaccine recipients.


Subject(s)
Alphavirus , Recombination, Genetic , mRNA Vaccines , Animals , Mice , Alphavirus/genetics , Alphavirus/immunology , Mice, Inbred C57BL , Humans , Receptor, Interferon alpha-beta/genetics , Virus Replication , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/adverse effects , Mice, Knockout , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/adverse effects
9.
Viruses ; 16(5)2024 05 20.
Article in English | MEDLINE | ID: mdl-38793690

ABSTRACT

The Mayaro virus (MAYV) is an arbovirus with emerging potential, though with a limited understanding of its epidemiology and evolution due to the lack of studies and surveillance. Here, we investigated 71 MAYV genome sequences from the Americas available at GenBank and characterized the phylogenetic relationship among virus strains. A phylogenetic analysis showed that sequences were grouped according to the genotypes L, D, and N. Genotype D sequences were closely related to sequences collected in adjacent years and from their respective countries, suggesting that isolates may have originated from circulating lineages. The coalescent analysis demonstrated similar results, indicating the continuous circulation of the virus between countries as well. An unidentified sequence from the USA was grouped with genotype D, suggesting the insertion of this genotype in the country. Furthermore, the recombination analysis detected homologous and three heterologous hybrids which presented an insertion into the nsP3 protein. Amino acid substitutions among sequences indicated selective pressure sites, suggesting viral adaptability. This also impacted the binding affinity between the E1-E2 protein complex and the Mxra8 receptor, associated with MAYV entry into human cells. These results provide information for a better understanding of genotypes circulating in the Americas.


Subject(s)
Evolution, Molecular , Genetic Variation , Genome, Viral , Genotype , Phylogeny , Americas/epidemiology , Humans , Alphavirus/genetics , Alphavirus/classification , Alphavirus/isolation & purification , Animals , Recombination, Genetic , Alphavirus Infections/virology , Alphavirus Infections/epidemiology
11.
Viruses ; 16(4)2024 03 25.
Article in English | MEDLINE | ID: mdl-38675846

ABSTRACT

Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the presence of RNA-amplifying enzymes known as replicases. We recently demonstrated that this property could enhance immune responses with minimal injected RNA amounts. In saRNA-based vaccines, replicase and antigens are encoded on the same mRNA molecule, resulting in very long RNA sequences, which poses significant challenges in production, delivery, and stability. In taRNA-based vaccines, these challenges can be overcome by splitting the replication system into two parts: one that encodes replicase and the other that encodes a short antigen-encoding RNA called transreplicon. Here, we review the identification and use of transreplicon RNA in alphavirus research, with a focus on the development of novel taRNA technology as a state-of-the art vaccine platform. Additionally, we discuss remaining challenges essential to the clinical application and highlight the potential benefits related to the unique properties of this future vaccine platform.


Subject(s)
Alphavirus , RNA, Viral , Vaccine Development , Animals , Humans , Alphavirus/genetics , Alphavirus/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , RNA, Viral/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Virus Replication
12.
Int J Biol Macromol ; 265(Pt 1): 130847, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490381

ABSTRACT

Getah virus (GETV) belongs to the Alphavirus genus in the Togaviridae family and is a zoonotic arbovirus causing disease in both humans and animals. The capsid protein (CP) of GETV regulates the viral core assembly, but the mechanism underlying this process is poorly understood. In this study, we demonstrate that CP undergoes liquid-liquid phase separation (LLPS) with the GETV genome RNA (gRNA) in vitro and forms cytoplasmic puncta in cells. Two regions of GETV gRNA (nucleotides 1-4000 and 5000-8000) enhance CP droplet formation in vitro and the lysine-rich Link region of CP is essential for its phase separation. CP(K/R) mutant with all lysines in the Link region replaced by arginines exhibits improved LLPS versus wild type (WT) CP, but CP(K/E) mutant with lysines substituted by glutamic acids virtually loses condensation ability. Consistently, recombinant virus mutant with CP(K/R) possesses significantly higher gRNA binding affinity, virion assembly efficiency and infectivity than the virus with WT-CP. Overall, our findings provide new insights into the understanding of GETV assembly and development of new antiviral drugs against alphaviruses.


Subject(s)
Alphavirus , Animals , Humans , Alphavirus/genetics , Alphavirus/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , RNA, Viral/genetics , RNA, Guide, CRISPR-Cas Systems , Genomics , Virion/genetics
13.
Parasit Vectors ; 17(1): 73, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38374048

ABSTRACT

BACKGROUND: Increasing global temperatures and unpredictable climatic extremes have contributed to the spread of vector-borne diseases. The mosquito Aedes aegypti is the main vector of multiple arboviruses that negatively impact human health, mostly in low socioeconomic areas of the world. Co-circulation and co-infection of these viruses in humans have been increasingly reported; however, how vectors contribute to this alarming trend remains unclear. METHODS: Here, we examine single and co-infection of Mayaro virus (D strain, Alphavirus) and dengue virus (serotype 2, Flavivirus) in Ae. aegypti adults and cell lines at two constant temperatures, moderate (27 °C) and hot (32 °C), to quantify vector competence and the effect of temperature on infection, dissemination and transmission, including on the degree of interaction between the two viruses. RESULTS: Both viruses were primarily affected by temperature but there was a partial interaction with co-infection. Dengue virus quickly replicates in adult mosquitoes with a tendency for higher titers in co-infected mosquitoes at both temperatures, and mosquito mortality was more severe at higher temperatures in all conditions. For dengue, and to a lesser extent Mayaro, vector competence and vectorial capacity were higher at hotter temperature in co- vs. single infections and was more evident at earlier time points (7 vs. 14 days post infection) for Mayaro. The temperature-dependent phenotype was confirmed in vitro by faster cellular infection and initial replication at higher temperatures for dengue but not for Mayaro virus. CONCLUSIONS: Our study suggests that contrasting kinetics of the two viruses could be related to their intrinsic thermal requirements, where alphaviruses thrive better at lower temperatures compared to flaviviruses. However, more studies are necessary to clarify the role of co-infection at different temperature regimes, including under more natural temperature settings.


Subject(s)
Aedes , Alphavirus , Coinfection , Dengue Virus , Dengue , Flavivirus , Animals , Humans , Temperature , Mosquito Vectors , Alphavirus/genetics , Flavivirus/genetics
14.
Nat Commun ; 15(1): 246, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38172096

ABSTRACT

Members of the low-density lipoprotein receptor (LDLR) family, including LDLRAD3, VLDLR, and ApoER2, were recently described as entry factors for different alphaviruses. However, based on studies with gene edited cells and knockout mice, blockade or abrogation of these receptors does not fully inhibit alphavirus infection, indicating the existence of additional uncharacterized entry factors. Here, we perform a CRISPR-Cas9 genome-wide loss-of-function screen in mouse neuronal cells with a chimeric alphavirus expressing the Eastern equine encephalitis virus (EEEV) structural proteins and identify LDLR as a candidate receptor. Expression of LDLR on the surface of neuronal or non-neuronal cells facilitates binding and infection of EEEV, Western equine encephalitis virus, and Semliki Forest virus. Domain mapping and binding studies reveal a low-affinity interaction with LA domain 3 (LA3) that can be enhanced by concatenation of LA3 repeats. Soluble decoy proteins with multiple LA3 repeats inhibit EEEV infection in cell culture and in mice. Our results establish LDLR as a low-affinity receptor for multiple alphaviruses and highlight a possible path for developing inhibitors that could mitigate infection and disease.


Subject(s)
Alphavirus Infections , Alphavirus , Encephalitis Virus, Eastern Equine , Horses , Animals , Mice , Alphavirus/genetics , Encephalitis Virus, Eastern Equine/genetics , Semliki forest virus/genetics , Lipoproteins, LDL
15.
Virology ; 591: 109990, 2024 03.
Article in English | MEDLINE | ID: mdl-38224661

ABSTRACT

Getah virus (GETV) is an emerging mosquito-borne alphavirus that can infect horses, pigs and other animals. Given the public health threat posed by GETV, research on its pathogenesis, diagnosis and prevention is urgently needed. In the current study, prokaryotic expression systems were used to express the capsid protein of GETV. This protein was then used to immunize BALB/c mice in order to generate monoclonal antibodies (mAbs). Subsequently, hybridoma cells secreting a mAb (2B11-4) against the capsid protein were obtained using the hybridoma technique. A B cell linear epitope, 18-PAYRPWR-24, located at the capsid protein's N-terminal region was identified using western blotting analysis with the produced mAb, 2B11-4. Sequence alignment indicated that this epitope was highly conserved in group III (GIII) strains of GETV, but varied among the other genotypes. Western blotting showed that mAb 2B11-4 could discriminate Group III GETVs from other genotypes. This study describes the preparation of a mAb against the GETV capsid protein and the identification of the specific localization of B-cell epitopes, and will contribute towards a better understanding of the biological importance of the GETV capsid protein. It will also pave the way for developing immunological detection methods and genotype diagnosis for GETVs.


Subject(s)
Alphavirus , Culicidae , Mice , Animals , Swine , Horses , Alphavirus/genetics , Capsid Proteins/genetics , Antibodies, Monoclonal , Epitopes, B-Lymphocyte/genetics
16.
Nat Commun ; 15(1): 622, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38245515

ABSTRACT

Alphaviruses are arboviruses transmitted by mosquitoes and are pathogenic to humans and livestock, causing a substantial public health burden. So far, several receptors have been identified for alphavirus entry; however, they cannot explain the broad host range and tissue tropism of certain alphaviruses, such as Getah virus (GETV), indicating the existence of additional receptors. Here we identify the evolutionarily conserved low-density lipoprotein receptor (LDLR) as a new cell entry factor for GETV, Semliki Forest virus (SFV), Ross River virus (RRV) and Bebaru virus (BEBV). Ectopic expression of LDLR facilitates cellular binding and internalization of GETV, which is mediated by the interaction between the E2-E1 spike of GETV and the ligand-binding domain (LBD) of LDLR. Antibodies against LBD block GETV infection in cultured cells. In addition, the GST-LBD fusion protein inhibits GETV infection both in vitro and in vivo. Notably, we identify the key amino acids in LDLR-LBD that played a crucial role in viral entry; specific mutations in the CR4 and CR5 domain of LDLR-LBD reduce viral entry to cells by more than 20-fold. These findings suggest that targeting the LDLR-LBD could be a potential strategy for the development of antivirals against multiple alphaviruses.


Subject(s)
Alphavirus Infections , Alphavirus , Culicidae , Animals , Humans , Alphavirus/genetics , Virus Internalization , Semliki forest virus/genetics , Semliki forest virus/metabolism , Alphavirus Infections/genetics
17.
Viruses ; 15(12)2023 12 17.
Article in English | MEDLINE | ID: mdl-38140691

ABSTRACT

Salmonid alphavirus strain 3 is responsible for outbreaks of pancreas disease in salmon and rainbow trout in Norway. Although the extensive amount of research on SAV3 focused mainly on the heart and pancreas (of clinical importance), tropism and pathogenesis studies of the virus in other salmon tissues are limited. Here, we used a combination of RT-qPCR (Q_nsp1 gene) and in situ hybridization (RNAscope®) to demonstrate the tropism of SAV3 in situ in tissues of Atlantic salmon, employing a challenge model (by cohabitation). In addition, as previous results suggested that the pseudobranch may harbor the virus, the change in the expression of different immune genes upon SAV3 infection (RT-qPCR) was focused on the pseudobranch in this study. In situ hybridization detected SAV3 in different tissues of Atlantic salmon during the acute phase of the infection, with the heart ventricle showing the most extensive infection. Furthermore, the detection of the virus in different adipose tissues associated with the internal organs of the salmon suggests a specific affinity of SAV3 to adipocyte components. The inconsistent immune response to SAV3 in the pseudobranch after infection did not mitigate the infection in that tissue and is probably responsible for the persistent low infection at 4 weeks post-challenge. The early detection of SAV3 in the pseudobranch after infection, along with the persistent low infection over the experimental infection course, suggests a pivotal role of the pseudobranch in SAV3 pathogenesis in Atlantic salmon.


Subject(s)
Alphavirus Infections , Alphavirus , Fish Diseases , Salmo salar , Animals , Alphavirus/genetics , Heart
19.
BMC health serv. res. (Online) ; 20(226): 1-16, 2020. tab, mapas, ilus
Article in English | RSDM | ID: biblio-1358131

ABSTRACT

Background: Mosquito-borne diseases involving arboviruses represent expanding threats to sub-Saharan Africa imposing as considerable burden to human and veterinary public health. In Mozambique over one hundred species of potential arbovirus mosquito vectors have been identi-fied, although their precise role in maintaining such viruses in circulation in the country remains to be elucidated. The aim of this study was to screen for the presence of flaviviruses, alphaviruses and bunyaviruses in mosquitoes from different regions of Mozambique. Results: Our survey analyzed 14,519 mosquitoes, and the results obtained revealed genetically distinct insectspecific flaviviruses, detected in multiple species of mosquitoes from different genera. In addition, smaller flaviviruslike NS5 sequences, frequently detected in Mansonia seemed to correspond to defective viral sequences, present as viral DNA forms. Furthermore, three lineages of putative members of the Phenuiviridae family were also detected, two of which apparently corresponding to novel viral genetic lineages. Conclusion: This study reports for the first-time novel insect-specific flaviviruses and novel phenuiviruses, as well as frequent flavivirus-like viral DNA forms in several widely known vector species. This unique work represents recent investigation of virus screening conducted in mosquitoes from Mozambique and an important contribution to inform the establishment of a vector control program for arbovirus in the country and in the region.


Subject(s)
Animals , Bunyaviridae/genetics , DNA, Viral/genetics , Alphavirus/genetics , Flavivirus/genetics , Mosquito Vectors/virology , Culicidae/virology , Mozambique
20.
Mem. Inst. Oswaldo Cruz ; 112(7): 510-513, July 2017. tab, graf
Article in English | LILACS | ID: biblio-841812

ABSTRACT

ABSTRACT We describe a sensitive method for simultaneous detection of Oropouche and Oropouche-like viruses carrying the Oropouche S segment, as well as the Mayaro virus, using a multiplexed one-step reverse transcription real-time polymerase chain reaction (RT-qPCR). A chimeric plasmid containing both Mayaro and Oropouche targets was designed and evaluated for the in vitro production of transcribed RNA, which could be easily used as a non-infectious external control. To track false-negative results due to PCR inhibition or equipment malfunction, the MS2 bacteriophage was also included in the multiplex assay as an internal positive control. The specificity of the multiplex assay was evaluated by Primer-Blast analysis against the entire GenBank database, and further against a panel of 17 RNA arboviruses. The results indicated an accurate and highly sensitive assay with amplification efficiency greater than 98% for both targets, and a limit of detection between two and 20 copies per reaction. We believe that the assay described here will provide a tool for Mayaro and Oropouche virus detection, especially in areas where differential diagnosis of Dengue, Zika and Chikungunya viruses should be performed.


Subject(s)
Humans , Orthobunyavirus/classification , Orthobunyavirus/genetics , Bunyaviridae Infections/diagnosis , Bunyaviridae Infections/virology , Alphavirus Infections/diagnosis , Alphavirus Infections/virology , Alphavirus/classification , Alphavirus/genetics , Reverse Transcriptase Polymerase Chain Reaction , Multiplex Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL