Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.558
Filtrar
1.
Vet Res ; 55(1): 106, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227887

RESUMO

Frequent RNA virus mutations raise concerns about evolving virulent variants. The purpose of this study was to investigate genetic variation in salmonid alphavirus-3 (SAV3) over the course of an experimental infection in Atlantic salmon and brown trout. Atlantic salmon and brown trout parr were infected using a cohabitation challenge, and heart samples were collected for analysis of the SAV3 genome at 2-, 4- and 8-weeks post-challenge. PCR was used to amplify eight overlapping amplicons covering 98.8% of the SAV3 genome. The amplicons were subsequently sequenced using the Nanopore platform. Nanopore sequencing identified a multitude of single nucleotide variants (SNVs) and deletions. The variation was widespread across the SAV3 genome in samples from both species. Mostly, specific SNVs were observed in single fish at some sampling time points, but two relatively frequent (i.e., major) SNVs were observed in two out of four fish within the same experimental group. Two other, less frequent (i.e., minor) SNVs only showed an increase in frequency in brown trout. Nanopore reads were de novo clustered using a 99% sequence identity threshold. For each amplicon, a number of variant clusters were observed that were defined by relatively large deletions. Nonmetric multidimensional scaling analysis integrating the cluster data for eight amplicons indicated that late in infection, SAV3 genomes isolated from brown trout had greater variation than those from Atlantic salmon. The sequencing methods and bioinformatics pipeline presented in this study provide an approach to investigate the composition of genetic diversity during viral infections.


Assuntos
Infecções por Alphavirus , Alphavirus , Doenças dos Peixes , Variação Genética , Sequenciamento por Nanoporos , Salmo salar , Truta , Animais , Salmo salar/virologia , Doenças dos Peixes/virologia , Alphavirus/genética , Infecções por Alphavirus/veterinária , Infecções por Alphavirus/virologia , Sequenciamento por Nanoporos/veterinária , Sequenciamento por Nanoporos/métodos , Truta/virologia
2.
Arch Microbiol ; 206(10): 406, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39292269

RESUMO

Mayaro virus (MAYV) is the causative agent of Mayaro fever, which is characterized mainly by acute fever and long-term severe arthralgia, common manifestations of other arbovirus infections, making the correct diagnosis a challenge. Besides, MAYV infections have been reported in South America, especially in Brazil. However, the lack of vaccines or specific antiviral drugs to control these infections makes the search for new antivirals an urgent need. Herein, we evaluated the antiviral potential of synthetic ß-enaminoesters derivatives against MAYV replication and their pharmacokinetic and toxicological (ADMET) properties using in vitro and in silico strategies. For this purpose, Vero cells were infected with MAYV at an MOI of 0.1, treated with compounds (50 µM) for 24 h, and virus titers were quantified by plaque reduction assays. Compounds 2b (83.33%) and 2d (77.53%) exhibited the highest activity with inhibition rates of 83.33% and 77.53%, respectively. The most active compounds 2b (EC50 = 18.92 µM; SI > 52.85), and 2d (EC50 = 14.52 µM; SI > 68.87) exhibited higher potency and selectivity than the control drug suramin (EC50 = 38.97 µM; SI > 25.66). Then, we investigated the mechanism of action of the most active compounds. None of the compounds showed virucidal activity, neither inhibited virus adsorption, but compound 2b inhibited virus entry (62.64%). Also, compounds 2b and 2d inhibited some processes involved with the release of new virus particles. Finally, in silico results indicated good ADMET parameters of the most active compounds and reinforced their promising profile as drug candidates against MAYV.


Assuntos
Alphavirus , Antivirais , Ésteres , Replicação Viral , Antivirais/farmacologia , Antivirais/química , Chlorocebus aethiops , Animais , Células Vero , Ésteres/farmacologia , Ésteres/química , Alphavirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Simulação por Computador , Brasil , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/virologia
3.
Nat Commun ; 15(1): 8106, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39285216

RESUMO

Alphaviruses, such as chikungunya virus (CHIKV), are mosquito-borne viruses that represent a significant threat to human health due to the current context of global warming. Efficient alphavirus infection relies on the activity of the non-structural protein 3 (nsP3), a puzzling multifunctional molecule whose role in infection remains largely unknown. NsP3 is a component of the plasma membrane-bound viral RNA replication complex (vRC) essential for RNA amplification and is also found in large cytoplasmic aggregates of unknown function. Here, we report the cryo-electron microscopy (cryo-EM) structure of the CHIKV nsP3 at 2.35 Å resolution. We show that nsP3 assembles into tubular structures made by a helical arrangement of its alphavirus unique domain (AUD). The nsP3 helical scaffolds are consistent with crown structures found on tomographic reconstructions of the mature viral RCs. In addition, nsP3 helices assemble into cytoplasmic granules organized in a network of tubular structures that contain viral genomic RNA and capsid as well as host factors required for productive infection. Structure-guided mutagenesis identified residues that prevent or disturb nsP3 assemblies, resulting in impaired viral replication or transcription. Altogether, our results reveal an unexpected nsP3-dependent molecular organization essential for different phases of alphavirus infection.


Assuntos
Vírus Chikungunya , Microscopia Crioeletrônica , Grânulos Citoplasmáticos , RNA Viral , Proteínas não Estruturais Virais , Replicação Viral , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/química , Vírus Chikungunya/genética , Vírus Chikungunya/metabolismo , Vírus Chikungunya/fisiologia , Humanos , Animais , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/ultraestrutura , RNA Viral/metabolismo , RNA Viral/genética , Alphavirus/genética , Alphavirus/metabolismo , Alphavirus/fisiologia , Alphavirus/ultraestrutura , Chlorocebus aethiops , Modelos Moleculares
4.
Viruses ; 16(9)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39339832

RESUMO

Advances in diagnostic techniques coupled with ongoing environmental changes have resulted in intensified surveillance and monitoring of arbovirus circulation in the Amazon. This increased effort has resulted in increased detection of insect-specific viruses among hematophagous arthropods collected in the field. This study aimed to document the first isolation of Agua Salud alphavirus in mosquitoes collected within the Brazilian Amazon. Arthropods belonging to the family Culicidae were collected within a forest fragment located in the Environmental Protection Area of the metropolitan region of Belem. Subsequently, these specimens were meticulously identified to the species level. Afterward, the collected batches were macerated, and the resulting supernatant was then inoculated into C6/36 and Vero cell cultures to facilitate viral isolation. The presence of arboviruses within the inoculated cell cultures was determined through indirect immunofluorescence analysis. Furthermore, positive supernatant samples underwent nucleotide sequencing to precisely identify the viral strains present. Notably, a batch containing Culex (Melanoconion) mosquitoes was identified to be positive for the genus Alphavirus via indirect immunofluorescence. This study is the first report on insect-specific alphavirus isolation in Brazil and the first-ever description of Agua Salud alphavirus isolation within Amazon Forest remnants.


Assuntos
Alphavirus , Culex , Animais , Alphavirus/isolamento & purificação , Alphavirus/genética , Alphavirus/classificação , Brasil , Células Vero , Chlorocebus aethiops , Culex/virologia , Mosquitos Vetores/virologia , Filogenia , Arbovírus/isolamento & purificação , Arbovírus/genética , Arbovírus/classificação
5.
Nucleic Acids Res ; 52(16): 9745-9759, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39106168

RESUMO

Cellular stress pathways that inhibit translation initiation lead to transient formation of cytoplasmic RNA/protein complexes known as stress granules. Many of the proteins found within stress granules and the dynamics of stress granule formation and dissolution are implicated in neurodegenerative disease. Whether stress granule formation is protective or harmful in neurodegenerative conditions is not known. To address this, we took advantage of the alphavirus protein nsP3, which selectively binds dimers of the central stress granule nucleator protein G3BP and markedly reduces stress granule formation without directly impacting the protein translational inhibitory pathways that trigger stress granule formation. In Drosophila and rodent neurons, reducing stress granule formation with nsP3 had modest impacts on lifespan even in the setting of serial stress pathway induction. In contrast, reducing stress granule formation in models of ataxia, amyotrophic lateral sclerosis and frontotemporal dementia largely exacerbated disease phenotypes. These data support a model whereby stress granules mitigate, rather than promote, neurodegenerative cascades.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Doenças Neurodegenerativas , Neurônios , Grânulos de Estresse , Animais , Grânulos de Estresse/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/genética , Humanos , Neurônios/metabolismo , Demência Frontotemporal/metabolismo , Demência Frontotemporal/genética , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/genética , Camundongos , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , RNA Helicases/metabolismo , RNA Helicases/genética , Ataxia/genética , Ataxia/metabolismo , DNA Helicases/metabolismo , DNA Helicases/genética , Alphavirus/genética , Alphavirus/metabolismo , Ratos , Proteínas de Transporte/metabolismo , Drosophila/metabolismo , Grânulos Citoplasmáticos/metabolismo , Estresse Fisiológico , Proteínas de Ligação a DNA
6.
Emerg Infect Dis ; 30(9): 1918-1921, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39174038

RESUMO

Alphavirus infections are transmitted by mosquitoes, but the mode of transmission for Mycobacterium ulcerans, which causes Buruli ulcer, is contested. Using notification data for Victoria, Australia, during 2017-2022, adjusted for incubation period, we show close alignment between alphavirus and Buruli ulcer seasons, supporting the hypothesis of mosquito transmission of M. ulcerans.


Assuntos
Infecções por Alphavirus , Úlcera de Buruli , Mosquitos Vetores , Mycobacterium ulcerans , Úlcera de Buruli/transmissão , Úlcera de Buruli/epidemiologia , Úlcera de Buruli/microbiologia , Mycobacterium ulcerans/isolamento & purificação , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/epidemiologia , Humanos , Animais , Vitória/epidemiologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Alphavirus/isolamento & purificação , Culicidae/microbiologia , Culicidae/virologia , Notificação de Doenças
7.
Viruses ; 16(8)2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39205271

RESUMO

Mayaro virus (MAYV) is an arbovirus first isolated in Trinidad and Tobago in 1954. MAYV is the causative agent of Mayaro fever, which is characterised by high fever, maculopapular rash, myalgia and arthralgia. The potential for chronic arthralgia is of particular clinical concern. Currently, MAYV outbreaks are restricted to South and Central America, with some cases reported in Africa as well as several imported cases in Europe. However, in recent years, MAYV has become a growing global concern due to its potential to emerge into urban transmission cycles. Challenges faced with diagnostics, as well as a lack of specific antivirals or licensed vaccines further exacerbate the potential global health threat posed by MAYV. In this review, we discuss this emerging arboviral threat with a particular focus on the current treatment and vaccine development efforts. Overall, MAYV remains a neglected arbovirus due to its limited area of transmission. However, with the potential of its urbanisation and expanding circulation, the threat MAYV poses to global health cannot be overlooked. Further research into the improvement of current diagnostics, as well as the development of efficacious antivirals and vaccines will be crucial to help prevent and manage potential MAYV outbreaks.


Assuntos
Infecções por Alphavirus , Alphavirus , Humanos , Alphavirus/isolamento & purificação , Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/virologia , Infecções por Alphavirus/transmissão , Animais , América/epidemiologia , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/virologia , Surtos de Doenças , Vacinas Virais/imunologia , Antivirais/uso terapêutico , Saúde Global
8.
PLoS Pathog ; 20(8): e1011836, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39207950

RESUMO

The host interferon pathway upregulates intrinsic restriction factors in response to viral infection. Many of them block a diverse range of viruses, suggesting that their antiviral functions might have been shaped by multiple viral families during evolution. Host-virus conflicts have led to the rapid adaptation of host and viral proteins at their interaction hotspots. Hence, we can use evolutionary genetic analyses to elucidate antiviral mechanisms and domain functions of restriction factors. Zinc finger antiviral protein (ZAP) is a restriction factor against RNA viruses such as alphaviruses, in addition to other RNA, retro-, and DNA viruses, yet its precise antiviral mechanism is not fully characterized. Previously, an analysis of 13 primate ZAP orthologs identified three positively selected residues in the poly(ADP-ribose) polymerase-like domain. However, selective pressure from ancient alphaviruses and others likely drove ZAP adaptation in a wider representation of mammals. We performed positive selection analyses in 261 mammalian ZAP using more robust methods with complementary strengths and identified seven positively selected sites in all domains of the protein. We generated ZAP inducible cell lines in which the positively selected residues of ZAP are mutated and tested their effects on alphavirus replication and known ZAP activities. Interestingly, the mutant in the second WWE domain of ZAP (N658A) is dramatically better than wild-type ZAP at blocking replication of Sindbis virus and other ZAP-sensitive alphaviruses due to enhanced viral translation inhibition. The N658A mutant is adjacent to the previously reported poly(ADP-ribose) (PAR) binding pocket, but surprisingly has reduced binding to PAR. In summary, the second WWE domain is critical for engineering a more potent ZAP and fluctuations in PAR binding modulate ZAP antiviral activity. Our study has the potential to unravel the role of ADP-ribosylation in the host innate immune defense and viral evolutionary strategies that antagonize this post-translational modification.


Assuntos
Infecções por Alphavirus , Alphavirus , Humanos , Animais , Alphavirus/genética , Infecções por Alphavirus/virologia , Infecções por Alphavirus/genética , Domínios Proteicos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Seleção Genética , Evolução Molecular , Proteínas Repressoras
9.
Front Cell Infect Microbiol ; 14: 1421571, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39211797

RESUMO

Arthritogenic alphaviruses pose a significant public health concern due to their ability to cause joint inflammation, with emerging evidence of potential neurological consequences. In this review, we examine the immunopathology and immune evasion strategies employed by these viruses, highlighting their complex mechanisms of pathogenesis and neurological implications. We delve into how these viruses manipulate host immune responses, modulate inflammatory pathways, and potentially establish persistent infections. Further, we explore their ability to breach the blood-brain barrier, triggering neurological complications, and how co-infections exacerbate neurological outcomes. This review synthesizes current research to provide a comprehensive overview of the immunopathological mechanisms driving arthritogenic alphavirus infections and their impact on neurological health. By highlighting knowledge gaps, it underscores the need for research to unravel the complexities of virus-host interactions. This deeper understanding is crucial for developing targeted therapies to address both joint and neurological manifestations of these infections.


Assuntos
Infecções por Alphavirus , Alphavirus , Barreira Hematoencefálica , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Humanos , Alphavirus/patogenicidade , Alphavirus/imunologia , Animais , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Interações Hospedeiro-Patógeno/imunologia , Barreira Hematoencefálica/imunologia , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/virologia
10.
Virology ; 598: 110174, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39029332

RESUMO

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.


Assuntos
Antivirais , Genes Reporter , Proteínas de Fluorescência Verde , Testes de Neutralização , Animais , Antivirais/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Replicação Viral/efeitos dos fármacos , Alphavirus/genética , Alphavirus/efeitos dos fármacos , Suínos , Cricetinae
11.
J Med Virol ; 96(7): e29788, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38982767

RESUMO

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.


Assuntos
Infecções por Alphavirus , Alphavirus , Vírus Chikungunya , Reação em Cadeia da Polimerase em Tempo Real , Sensibilidade e Especificidade , Humanos , Alphavirus/genética , Alphavirus/isolamento & purificação , Infecções por Alphavirus/diagnóstico , Infecções por Alphavirus/virologia , Infecções por Alphavirus/prevenção & controle , Infecções por Alphavirus/epidemiologia , China/epidemiologia , Reação em Cadeia da Polimerase em Tempo Real/métodos , Vírus Chikungunya/genética , Vírus Chikungunya/isolamento & purificação , Estudos Retrospectivos , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/prevenção & controle , Febre de Chikungunya/virologia , Febre de Chikungunya/epidemiologia , Vírus da Encefalite Equina do Leste/genética , Surtos de Doenças/prevenção & controle , Sindbis virus/genética , Vírus da Encefalite Equina do Oeste/genética , Ross River virus/genética , Ross River virus/isolamento & purificação , Vírus da Encefalite Equina Venezuelana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , RNA Viral/genética
12.
ACS Infect Dis ; 10(8): 2507-2524, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-38992989

RESUMO

The Alphavirus genus includes viruses that cause encephalitis due to neuroinvasion and viruses that cause arthritis due to acute and chronic inflammation. There is no approved therapeutic for alphavirus infections, but significant efforts are ongoing, more so in recent years, to develop vaccines and therapeutics for alphavirus infections. This review article highlights some of the major advances made so far to identify small molecules that can selectively target the structural and the nonstructural proteins in alphaviruses with the expectation that persistent investigation of an increasingly expanding chemical space through a variety of structure-based design and high-throughput screening strategies will yield candidate drugs for clinical studies. While most of the works discussed are still in the early discovery to lead optimization stages, promising avenues remain for drug development against this family of viruses.


Assuntos
Alphavirus , Antivirais , Proteínas não Estruturais Virais , Alphavirus/efeitos dos fármacos , Alphavirus/química , Antivirais/farmacologia , Antivirais/química , Humanos , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/virologia , Animais , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/antagonistas & inibidores
13.
mBio ; 15(8): e0099324, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38953633

RESUMO

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.


Assuntos
Infecções por Alphavirus , Alphavirus , Genoma Viral , Proteínas não Estruturais Virais , Replicação Viral , Replicação Viral/genética , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Animais , Alphavirus/genética , Alphavirus/patogenicidade , Camundongos , Infecções por Alphavirus/virologia , Genoma Viral/genética , Linhagem Celular , Humanos , Austrália
14.
J Gen Virol ; 105(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38995674

RESUMO

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.


Assuntos
Infecções por Alphavirus , Alphavirus , Genótipo , Macaca fascicularis , RNA Viral , Viremia , Animais , Macaca fascicularis/virologia , Alphavirus/genética , Alphavirus/patogenicidade , Alphavirus/classificação , Alphavirus/isolamento & purificação , Infecções por Alphavirus/virologia , Infecções por Alphavirus/veterinária , Viremia/virologia , RNA Viral/genética , Anticorpos Antivirais/sangue , Anticorpos Neutralizantes/sangue , Modelos Animais de Doenças , Filogenia , Citocinas/genética , Citocinas/sangue
15.
Virology ; 597: 110152, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38968676

RESUMO

Cobalt-porphyrin phospholipid displays recombinant protein antigens on liposome surfaces via antigen polyhistidine-tag (His-tag), and when combined with monophosphorylated lipid A and QS-21 yields the "CPQ" vaccine adjuvant system. In this proof of principle study, CPQ was used to generate vaccine prototypes that elicited antibodies for two different alphaviruses (AV). Mice were immunized with computationally designed, His-tagged, physicochemical property consensus (PCPcon) protein antigens representing the variable B-domain of the envelope protein 2 (E2) from the serotype specific Venezuelan Equine Encephalitis Virus (VEEVcon) or a broad-spectrum AV-antigen termed EVCcon. The CPQ adjuvant enhanced the antigenicity of both proteins without eliciting detectable anti-His-tag antibodies. Antibodies elicited from mice immunized with antigens admixed with CPQ showed orders-of-magnitude higher levels of antigen-specific IgG compared to alternative control adjuvants. The ELISA results correlated with antiviral activity against VEEV strain TC83 and more weakly to Chikungunya virus 118/25. Thus, display of E.coli-produced His-tagged E2 protein segments on the surface of immunogenic liposomes elicits high levels of antigen-specific and AV neutralizing antibodies in mice with vaccination, while facilitating vaccine preparation and providing dose-sparing potential.


Assuntos
Adjuvantes Imunológicos , Alphavirus , Anticorpos Antivirais , Antígenos Virais , Lipossomos , Proteínas do Envelope Viral , Vacinas Virais , Animais , Anticorpos Antivirais/imunologia , Camundongos , Lipossomos/imunologia , Alphavirus/imunologia , Antígenos Virais/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Vírus da Encefalite Equina Venezuelana/imunologia , Feminino , Anticorpos Neutralizantes/imunologia , Vírus Chikungunya/imunologia , Camundongos Endogâmicos BALB C , Imunoglobulina G/imunologia , Imunoglobulina G/sangue
16.
Vaccine ; 42(25): 126136, 2024 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-39004524

RESUMO

Getah virus (GETV) is an emerging mosquito-borne virus with economic impact on the livestock industry in East Asia. In this study, we successfully produced GETV virus-like particles (VLPs) in insect cells using the baculovirus expression vector system. We show that the GETV envelope glycoproteins were successfully expressed at the surface of the insect cell and were glycosylated. VLPs were isolated from the culture fluid as enveloped particles of 60-80 nm in diameter. Two 1 µg vaccinations with this GETV VLP vaccine, without adjuvant, generated neutralizing antibody responses and protected wild-type C57/BL6 mice against GETV viremia and arthritic disease. The GETV VLP vaccine may find application as a horse and/or pig vaccine in the future.


Assuntos
Alphavirus , Anticorpos Neutralizantes , Anticorpos Antivirais , Artrite , Camundongos Endogâmicos C57BL , Vacinas de Partículas Semelhantes a Vírus , Viremia , Animais , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Viremia/prevenção & controle , Viremia/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Camundongos , Artrite/imunologia , Artrite/prevenção & controle , Alphavirus/imunologia , Alphavirus/genética , Infecções por Alphavirus/prevenção & controle , Infecções por Alphavirus/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Feminino , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Baculoviridae/genética , Baculoviridae/imunologia , Células Sf9
17.
Appl Microbiol Biotechnol ; 108(1): 355, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38822832

RESUMO

Getah virus (GETV) is a re-emerging mosquito-borne alphavirus that is highly pathogenic, mainly to pigs and horses. There are no vaccines or treatments available for GETV in swine in China. Therefore, the development of a simple, rapid, specific, and sensitive serological assay for GETV antibodies is essential for the prevention and control of GETV. Current antibody monitoring methods are time-consuming, expensive, and dependent on specialized instrumentation, and these features are not conducive to rapid detection in clinical samples. To address these problem, we developed immunochromatographic test strips (ICTS) using eukaryotically expressed soluble recombinant p62-E1 protein of GETV as a labelled antigen, which has good detection sensitivity and no cross-reactivity with other common porcine virus-positive sera. The ICTS is highly compatible with IFA and ELISA and can be stored for 1 month at 37 °C and for at least 3 months at room temperature. Hence, p62-E1-based ICTS is a rapid, accurate, and convenient method for rapid on-site detection of GETV antibodies. KEY POINTS: • We established a rapid antibody detection method that can monitor GETV infection • We developed colloidal gold test strips with high sensitivity and specificity • The development of colloidal gold test strips will aid in the field serologic detection of GETV.


Assuntos
Alphavirus , Anticorpos Antivirais , Coloide de Ouro , Sensibilidade e Especificidade , Animais , Coloide de Ouro/química , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Alphavirus/imunologia , Suínos , Cromatografia de Afinidade/métodos , Infecções por Alphavirus/diagnóstico , Infecções por Alphavirus/imunologia , Doenças dos Suínos/diagnóstico , Doenças dos Suínos/virologia , Fitas Reagentes , China , Ensaio de Imunoadsorção Enzimática/métodos
18.
Front Cell Infect Microbiol ; 14: 1388360, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38841111

RESUMO

Background: Alphaviruses are a diverse group of pathogens that have garnered considerable attention due to their impact on human health. By investigating alphavirus receptors, researchers can elucidate viral entry mechanisms and gain important clues for the prevention and treatment of viral diseases. This study presents an in-depth analysis of the research progress made in the field of alphavirus receptors through bibliometric analysis. Methods: This study encompasses various aspects, including historical development, annual publication trends, author and cited-author analysis, institutional affiliations, global distribution of research contributions, reference analysis with strongest citation bursts, keyword analysis, and a detailed exploration of recent discoveries in alphavirus receptor research. Results: The results of this bibliometric analysis highlight key milestones in alphavirus receptor research, demonstrating the progression of knowledge in this field over time. Additionally, the analysis reveals current research hotspots and identifies emerging frontiers, which can guide future investigations and inspire novel therapeutic strategies. Conclusion: This study provides an overview of the state of the art in alphavirus receptor research, consolidating the existing knowledge and paving the way for further advancements. By shedding light on the significant developments and emerging areas of interest, this study serves as a valuable resource for researchers, clinicians, and policymakers engaged in combating alphavirus infections and improving public health.


Assuntos
Alphavirus , Bibliometria , Humanos , Receptores Virais/metabolismo , Animais , Internalização do Vírus , Infecções por Alphavirus/virologia , Pesquisa Biomédica/tendências
19.
Front Immunol ; 15: 1401086, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903507

RESUMO

The mitochondrial anti-viral signaling (MAVS) protein is an intermediary adaptor protein of retinoic acid-inducible gene-1 (RIG-I) like receptor (RLR) signaling, which activates the transcription factor interferon (IFN) regulatory factor 3 (IRF3) and NF-kB to produce type I IFNs. MAVS expression has been reported in different fish species, but few studies have shown its functional role in anti-viral responses to fish viruses. In this study, we used the transcription activator-like effector nuclease (TALEN) as a gene editing tool to disrupt the function of MAVS in Chinook salmon (Oncorhynchus tshawytscha) embryonic cells (CHSE) to understand its role in induction of interferon I responses to infections with the (+) RNA virus salmonid alphavirus subtype 3 (SAV-3), and the dsRNA virus infectious pancreatic necrosis virus (IPNV) infection. A MAVS-disrupted CHSE clone with a 7-aa polypeptide (GVFVSRV) deletion mutation at the N-terminal of the CARD domain infected with SAV-3 resulted in significantly lower expression of IRF3, IFNa, and ISGs and increased viral titer (1.5 log10) compared to wild-type. In contrast, the IPNV titer in MAVS-disrupted cells was not different from the wild-type. Furthermore, overexpression of salmon MAVS in MAVS-disrupted CHSE cells rescued the impaired type I IFN-mediated anti-viral effect against SAV-3.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Infecções por Alphavirus , Alphavirus , Doenças dos Peixes , Vírus da Necrose Pancreática Infecciosa , Transdução de Sinais , Replicação Viral , Animais , Vírus da Necrose Pancreática Infecciosa/fisiologia , Vírus da Necrose Pancreática Infecciosa/imunologia , Alphavirus/imunologia , Alphavirus/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Salmão/virologia , Salmão/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Proteínas de Peixes/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Infecções por Birnaviridae/imunologia , Infecções por Birnaviridae/veterinária , Infecções por Birnaviridae/virologia
20.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38851803

RESUMO

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.


Assuntos
Alphavirus , Antivirais , Receptores de LDL , Internalização do Vírus , Animais , Humanos , Alphavirus/efeitos dos fármacos , Alphavirus/fisiologia , Alphavirus/genética , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/virologia , Antivirais/uso terapêutico , Antivirais/farmacologia , Ligação Proteica , Receptores de LDL/metabolismo , Receptores de LDL/genética , Receptores Virais/metabolismo , Receptores Virais/química , Vírion/metabolismo , Internalização do Vírus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA