Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 589
Filtrar
1.
Viruses ; 16(4)2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38675846

RESUMEN

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.


Asunto(s)
Alphavirus , ARN Viral , Alphavirus/genética , Alphavirus/inmunología , ARN Viral/genética , Animales , Humanos , Vacunas Virales/inmunología , Vacunas Virales/genética , Replicación Viral , Infecciones por Alphavirus/virología , Infecciones por Alphavirus/prevención & control , Infecciones por Alphavirus/inmunología , Desarrollo de Vacunas
3.
Int J Biol Macromol ; 265(Pt 1): 130847, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490381

RESUMEN

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.


Asunto(s)
Alphavirus , Animales , Humanos , Alphavirus/genética , Alphavirus/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , ARN Viral/genética , ARN Guía de Sistemas CRISPR-Cas , Genómica , Virión/genética
4.
Parasit Vectors ; 17(1): 73, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38374048

RESUMEN

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.


Asunto(s)
Aedes , Alphavirus , Coinfección , Virus del Dengue , Dengue , Flavivirus , Animales , Humanos , Temperatura , Mosquitos Vectores , Alphavirus/genética , Flavivirus/genética
5.
Virology ; 591: 109990, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38224661

RESUMEN

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.


Asunto(s)
Alphavirus , Culicidae , Ratones , Animales , Porcinos , Caballos , Alphavirus/genética , Proteínas de la Cápside/genética , Anticuerpos Monoclonales , Epítopos de Linfocito B/genética
6.
Nat Commun ; 15(1): 246, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38172096

RESUMEN

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.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Virus de la Encefalitis Equina del Este , Caballos , Animales , Ratones , Alphavirus/genética , Virus de la Encefalitis Equina del Este/genética , Virus de los Bosques Semliki/genética , Lipoproteínas LDL
7.
Nat Commun ; 15(1): 622, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38245515

RESUMEN

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.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Culicidae , Animales , Humanos , Alphavirus/genética , Internalización del Virus , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Infecciones por Alphavirus/genética
8.
Viruses ; 15(12)2023 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-38140691

RESUMEN

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.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Enfermedades de los Peces , Salmo salar , Animales , Alphavirus/genética , Corazón
9.
Int J Mol Sci ; 24(21)2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37958918

RESUMEN

Infection by arthritogenic alphaviruses (aavs) can lead to reactive arthritis, which is characterized by inflammation and persistence of the virus; however, its mechanisms remain ill-characterized. Intriguingly, it has been shown that viral persistence still takes place in spite of robust innate and adaptive immune responses, characterized notably by the infiltration of macrophages (sources of TNF-alpha) as well as T/NK cells (sources of IFN-gamma) in the infected joint. Aavs are known to target mesenchymal stem cells (MSCs) in the synovium, and we herein tested the hypothesis that the infection of MSCs may promote the expression of immunoregulators to skew the anti-viral cellular immune responses. We compared the regulated expression via human synovial MSCs of pro-inflammatory mediators (e.g., IL-1ß, IL6, CCL2, miR-221-3p) to that of immunoregulators (e.g., IDO, TSG6, GAS6, miR146a-5p). We used human synovial tissue-derived MSCs which were infected with O'Nyong-Nyong alphavirus (ONNV, class II aav) alone, or combined with recombinant human TNF-α or IFN-γ, to mimic the clinical settings. We confirmed via qPCR and immunofluorescence that ONNV infected human synovial tissue-derived MSCs. Interestingly, ONNV alone did not regulate the expression of pro-inflammatory mediators. In contrast, IDO, TSG6, and GAS6 mRNA expression were increased in response to ONNV infection alone, but particularly when combined with both recombinant cytokines. ONNV infection equally decreased miR-146a-5p and miR-221-3p in the untreated cells and abrogated the stimulatory activity of the recombinant TNF-α but not the IFN-gamma. Our study argues for a major immunoregulatory phenotype of MSCs infected with ONNV which may favor virus persistence in the inflamed joint.


Asunto(s)
Alphavirus , Artritis Infecciosa , Células Madre Mesenquimatosas , MicroARNs , Humanos , Alphavirus/genética , Alphavirus/metabolismo , Inmunidad , Mediadores de Inflamación , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Factor de Necrosis Tumoral alfa
10.
J Clin Microbiol ; 61(12): e0015223, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-37982611

RESUMEN

Eastern equine encephalitis virus (EEEV), Madariaga virus (MADV), and Venezuelan equine encephalitis virus complex (VEEV) are New World alphaviruses transmitted by mosquitoes. They cause febrile and sometimes severe neurological diseases in human and equine hosts. Detecting them during the acute phase is hindered by non-specific symptoms and limited diagnostic tools. We designed and clinically assessed real-time reverse transcription polymerase chain reaction assays (rRT-PCRs) for VEEV complex, MADV, and EEEV using whole-genome sequences. Validation involved 15 retrospective serum samples from 2015 to 2017 outbreaks, 150 mosquito pools from 2015, and 118 prospective samples from 2021 to 2022 surveillance in Panama. The rRT-PCRs detected VEEV complex RNA in 10 samples (66.7%) from outbreaks, with one having both VEEV complex and MADV RNAs. VEEV complex RNA was found in five suspected dengue cases from disease surveillance. The rRT-PCR assays identified VEEV complex RNA in three Culex (Melanoconion) vomerifer pools, leading to VEEV isolates in two. Phylogenetic analysis revealed the VEEV ID subtype in positive samples. Notably, 11.9% of dengue-like disease patients showed VEEV infections. Together, our rRT-PCR validation in human and mosquito samples suggests that this method can be incorporated into mosquito and human encephalitic alphavirus surveillance programs in endemic regions.


Asunto(s)
Alphavirus , Culicidae , Dengue , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina Oriental , Encefalomielitis Equina Venezolana , Humanos , Animales , Caballos/genética , Virus de la Encefalitis Equina del Este/genética , Encefalomielitis Equina Venezolana/diagnóstico , Encefalomielitis Equina Venezolana/epidemiología , Culicidae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Filogenia , Estudios Prospectivos , Vigilancia en Salud Pública , Estudios Retrospectivos , Alphavirus/genética , ARN
11.
Int J Mol Sci ; 24(19)2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37834397

RESUMEN

Sindbis alphavirus vectors offer a promising platform for cancer therapy, serving as valuable models for alphavirus-based treatment. This review emphasizes key studies that support the targeted delivery of Sindbis vectors to tumor cells, highlighting their effectiveness in expressing tumor-associated antigens and immunomodulating proteins. Among the various alphavirus vectors developed for cancer therapy, Sindbis-vector-based imaging studies have been particularly extensive. Imaging modalities that enable the in vivo localization of Sindbis vectors within lymph nodes and tumors are discussed. The correlation between laminin receptor expression, tumorigenesis, and Sindbis virus infection is examined. Additionally, we present alternative entry receptors for Sindbis and related alphaviruses, such as Semliki Forest virus and Venezuelan equine encephalitis virus. The review also discusses cancer treatments that are based on the alphavirus vector expression of anti-tumor agents, including tumor-associated antigens, cytokines, checkpoint inhibitors, and costimulatory immune molecules.


Asunto(s)
Alphavirus , Virus de la Encefalitis Equina Venezolana , Neoplasias , Humanos , Alphavirus/genética , Vectores Genéticos/genética , Neoplasias/terapia , Terapia Genética/métodos
12.
Viruses ; 15(10)2023 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-37896867

RESUMEN

As an epizootic causative agent, the Getah virus (GETV) can cause moderate illness in horses, lethal disease in foxes, and reproductive disorders and fetal death in pigs. Due to the wide range of hosts and multiple routes of transmission, GETV has become a growing potential threat to the global livestock industry, and even to public health. More attention and research on GETV are urgently needed. In this study, we successfully isolated a novel GETV strain, named BJ0304, from a commercial live vaccine against porcine reproductive and respiratory syndrome virus (PRRSV) and determined its growth kinetics. Then, genetic and phylogenetic analyses were performed. The results revealed that BJ0304 was clustered into Group III, and it was most related to the GETV-V1 strain based on the complete genome sequence. Furthermore, the pathogenicity of the isolate was assessed and found to be a low virulent strain in mice relative to its closest homolog GETV-V1. Finally, mutation and glycosylation analysis showed that a unique mutation (171 T > I) at one amino acid of E2, which affected the glycosylation of E2, may be associated with viral pathogenicity. In summary, the general characteristic of a novel Group III-classified GETV-BJ0304 isolated from commercial live PRRSV vaccine was defined and then mutation/glycosylation-related potential virulence factor was discussed. This study highlights the complexity of GETV transmission routes in swine and the need for more surveillance on commercial animal vaccines, contributes to the understanding of genetic characterization of clinical isolates, provides possible virulence factors in favor of unveiling the viral pathogenesis, and eventually lays the foundation for the prevention and control of GETV.


Asunto(s)
Alphavirus , Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Vacunas , Animales , Porcinos , Caballos , Ratones , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Alphavirus/genética , Filogenia , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control
13.
J Virol ; 97(11): e0122523, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37877718

RESUMEN

IMPORTANCE: Alphavirus replicons are being developed as self-amplifying RNAs aimed at improving the efficacy of mRNA vaccines. These replicons are convenient for genetic manipulations and can express heterologous genetic information more efficiently and for a longer time than standard mRNAs. However, replicons mimic many aspects of viral replication in terms of induction of innate immune response, modification of cellular transcription and translation, and expression of nonstructural viral genes. Moreover, all replicons used in this study demonstrated expression of heterologous genes in cell- and replicon's origin-specific modes. Thus, many aspects of the interactions between replicons and the host remain insufficiently investigated, and further studies are needed to understand the biology of the replicons and their applicability for designing a new generation of mRNA vaccines. On the other hand, our data show that replicons are very flexible expression systems, and additional modifications may have strong positive impacts on protein expression.


Asunto(s)
Alphavirus , Regulación Viral de la Expresión Génica , Interacciones Microbiota-Huesped , Replicón , Proteínas Virales , Alphavirus/genética , Alphavirus/metabolismo , Vacunas de ARNm/genética , Replicón/genética , Replicación Viral/genética , ARN Viral/biosíntesis , ARN Viral/genética , Interacciones Microbiota-Huesped/genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética
14.
Int Rev Cell Mol Biol ; 379: 143-168, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37541722

RESUMEN

Alphaviruses have frequently been engineered for cancer therapy, cancer immunotherapy, and cancer vaccine development. As members of self-replicating RNA viruses, alphaviruses provide high levels of transgene expression through efficient self-amplifying of their RNA genome in host cells. Alphavirus vectors can be used as recombinant viral particles or oncolytic viruses. Alternatively, either naked or nanoparticle-encapsulated RNA and DNA replicons can be utilized. In the context of cancer prevention and treatment, antitumor, cytotoxic and suicide genes have been expressed from alphavirus vectors to provide tumor regression and tumor eradication. Moreover, immunostimulatory genes such as cytokines and chemokines have been used for cancer immunotherapy approaches. Expression of tumor antigens has been applied for cancer vaccine development. Alphavirus vectors has demonstrated tumor regression and even cure in various preclinical animal models. Immunization has elicited strong immune responses and showed protection against challenges with tumor cells in animal models. Several clinical trials have confirmed good safety and tolerability of alphaviruses in cancer patients although therapeutic efficacy will still require optimization.


Asunto(s)
Alphavirus , Vacunas contra el Cáncer , Neoplasias , Animales , Alphavirus/genética , Vectores Genéticos , Inmunoterapia , ARN , Neoplasias/terapia
16.
Virol Sin ; 38(4): 585-594, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37390870

RESUMEN

Alphaviruses, which contain a variety of mosquito-borne pathogens, are important pathogens of emerging/re-emerging infectious diseases and potential biological weapons. Currently, no specific antiviral drugs are available for the treatment of alphaviruses infection. For most highly pathogenic alphaviruses are classified as risk group-3 agents, the requirement of biosafety level 3 (BSL-3) facilities limits the live virus-based antiviral study. To facilitate the antiviral development of alphaviruses, we developed a high throughput screening (HTS) platform based on a recombinant Semliki Forest virus (SFV) which can be manipulated in BSL-2 laboratory. Using the reverse genetics approach, the recombinant SFV and SFV reporter virus expressing eGFP (SFV-eGFP) were successfully rescued. The SFV-eGFP reporter virus exhibited robust eGFP expression and remained relatively stable after four passages in BHK-21 â€‹cells. Using a broad-spectrum alphavirus inhibitor ribavirin, we demonstrated that the SFV-eGFP can be used as an effective tool for antiviral study. The SFV-eGFP reporter virus-based HTS assay in a 96-well format was then established and optimized with a robust Z' score. A section of reference compounds that inhibit highly pathogenic alphaviruses were used to validate that the SFV-eGFP reporter virus-based HTS assay enables rapid screening of potent broad-spectrum inhibitors of alphaviruses. This assay provides a safe and convenient platform for antiviral study of alphaviruses.


Asunto(s)
Alphavirus , Animales , Alphavirus/genética , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Antivirales/farmacología , Antivirales/metabolismo , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Línea Celular , Replicación Viral
17.
J Ethnopharmacol ; 311: 116436, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37003399

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Mayaro virus (MAYV) is an arbovirus endemic to the Amazon region, which comprises the states of the North and Midwest region of Brazil and encompasses the largest tropical forest in the world, the Amazon Forest. The confirmation of its potential transmission by Aedes aegypti and recent cases in Brazil, mainly in large centers in the northern region, led to the classification of Mayaro fever as an emerging disease. Traditional medicine is commonly used to treat various diseases, mainly by local riverside populations. Some species of the genus Maytenus, which have similar morphologies, are popularly used to treat infections and inflammations. In this context, our research group has studied and confirmed the antiviral activity of several plant-derived compounds. However, several species of this same genus have not been studied and therefore deserve attention. AIM OF THE STUDY: This study aimed to demonstrate the effects of ethyl acetate extracts of leaves (LAE) and branches (TAE) of Maytenus quadrangulata against MAYV. MATERIALS AND METHODS: Mammalian cells (Vero cells) were used to evaluate the cytotoxicity of the extracts. After cell infection by MAYV and the treatment with the extracts, we evaluated the selectivity index (SI), the virucidal effect, viral adsorption and internalization, and the effect on viral gene expression. The antiviral action was confirmed by quantifying the viral genome using RT-qPCR and by analyzing the effect on virus yield in infected cells. The treatment was performed based on the effective concentration protective for 50% of the infected cells (EC50). RESULTS: The leaves (LAE; EC50 12.0 µg/mL) and branches (TAE; EC50 101.0 µg/mL) extracts showed significative selectivity against the virus, with SI values of 79.21 and 9.91, respectively, which were considered safe. Phytochemical analysis revealed that the antiviral action was associated with the presence of catechins, mainly in LAE. This extract was chosen for the subsequent studies since it reduced the viral cytopathic effect and virus production, even at high viral loads [MOI (multiplicity of infection) 1 and 5]. The effects of LAE resulted in a marked reduction in viral gene expression. The viral title was drastically reduced when LAE was added to the virus before infection or during replication stages, reducing virus production up to 5-log units compared to infected and untreated cells. CONCLUSION: Through kinetic replication, MAYV was not detected in Vero cells treated with LAE throughout the viral cycle. The virucidal effect of LAE inactivates the viral particle and can intercept the virus at the end of the cycle when it gains the extracellular environment. Therefore, LAE is a promising source of antiviral agents.


Asunto(s)
Alphavirus , Catequina , Maytenus , Animales , Chlorocebus aethiops , Antivirales/farmacología , Antivirales/química , Catequina/farmacología , Células Vero , Alphavirus/genética , Mamíferos
18.
Parasit Vectors ; 16(1): 133, 2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069603

RESUMEN

BACKGROUND: O'nyong-nyong virus (ONNV) is a mosquito-borne alphavirus causing sporadic outbreaks of febrile illness with rash and polyarthralgia. Up to now, ONNV has been restricted to Africa and only two competent vectors have been found, Anopheles gambiae and An. funestus, which are also known malaria vectors. With globalization and invasive mosquito species migrating to ONNV endemic areas, there is a possible risk of introduction of the virus to other countries and continents. Anopheles stephensi, is closely related to An. gambiae and one of the invasive mosquito species of Asian origin that is now present in the Horn of Africa and spreading further east. We hypothesize that An. stephensi, a known primary urban malaria vector, may also serve as a new possible vector for ONNV. METHODS: One-week-old female adult An. stephensi were exposed to ONNV-infected blood, and the vector competence for ONNV, i.e. infection rates (IRs), dissemination rates (DRs), transmission rates (TRs), dissemination efficiency (DEs) and transmission efficiency (TEs), were evaluated. Infection (IRs), dissemination efficiency (DEs) and transmission efficiency (TEs) were determined. Detection of ONNV RNA was analysed by RT-qPCR in the thorax and abdomen, head, wings, legs and saliva of the infected mosquitoes at four different time points, day 7, 14, 21 and 28 after blood meal. Infectious virus in saliva was assessed by infection of Vero B4 cells. RESULTS: The mean mortality across all sampling times was 27.3% (95 confidence interval [CI] 14.7-44.2%). The mean rate of infection across all sampling periods was 89.5% (95% CI 70.6-95.9). The mean dissemination rate across sampling intervals was 43.4% (95% CI 24.3-64.2%). The mean TR and TE across all mosquito sampling time intervals were 65.3 (95% CI 28.6-93.5) and 74.6 (95% CI 52.1-89.4). The IR was 100%, 79.3%, 78.6% and 100% respectively at 7, 14, 21 and 28 dpi. The DR was the highest at 7 dpi with 76.0%, followed by 28 dpi at 57.1%, 21 dpi at 27.3% and 14 dpi at the lowest DR of 13.04%. DE was 76%, 13.8%, 25%, 57.1% and TR was 79%, 50%, 57.1% and 75% at 7, 14, 21 and 28 dpi respectively. The TE was the highest at 28 dpi, with a proportion of 85.7%. For 7, 14 and 21 dpi the transmission efficiency was 72.0%, 65.5% and 75.0% respectively. CONCLUSION: Anopheles stephensi is a competent vector for ONNV and being an invasive species spreading to different parts of the world will likely spread the virus to other regions.


Asunto(s)
Alphavirus , Anopheles , Malaria , Animales , Femenino , Humanos , Virus O'nyong-nyong , Anopheles/genética , Mosquitos Vectores , Malaria/epidemiología , Alphavirus/genética
19.
Vet Microbiol ; 281: 109742, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37075664

RESUMEN

Getah virus (GETV), is an often neglected and re-emerging mosquito-borne RNA virus. GETV can cause illness accompanied with high fever, rash, incapacitating arthralgia and chronic arthritis or encephalitic disease in affected animals. Currently, there is no specific treatment or vaccine against GETV infection. In this study, we developed three recombinant viruses by inserting different reporter protein genes between the Cap and pE2 genes. The reporter viruses exhibited high replication capacity similar to the parental virus. The rGECiLOV and rGECGFP viruses were genetically stable within at least ten rounds of passages in BHK-21 cells. We confirmed that the reporter virus, rGECGFP, facilitated the antiviral assays against GETV by testing it with the known inhibitor, ribavirin. It was also found that the compound, doxycycline, showed an inhibitory effect on GETV replication. In addition, rGECGFP was found to be an authentic mimic of the parental virus infection in 3-day-old mice, but with milder pathogenicity. The reporter viruses will contribute to the assessment of viral replication and proliferation, tracking and elucidating of alphavirus-host interactions. In addition, they will help in the screening of potential antiviral compounds.


Asunto(s)
Alphavirus , Culicidae , Animales , Ratones , Alphavirus/genética , Antivirales/farmacología , Evaluación Preclínica de Medicamentos/veterinaria , Replicación Viral
20.
Viruses ; 15(4)2023 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-37112813

RESUMEN

Certain re-emerging alphaviruses, such as chikungunya virus (CHIKV), cause serious disease and widespread epidemics. To develop virus-specific therapies, it is critical to understand the determinants of alphavirus pathogenesis and virulence. One major determinant is viral evasion of the host interferon response, which upregulates antiviral effectors, including zinc finger antiviral protein (ZAP). Here, we demonstrated that Old World alphaviruses show differential sensitivity to endogenous ZAP in 293T cells: Ross River virus (RRV) and Sindbis virus (SINV) are more sensitive to ZAP than o'nyong'nyong virus (ONNV) and CHIKV. We hypothesized that the more ZAP-resistant alphaviruses evade ZAP binding to their RNA. However, we did not find a correlation between ZAP sensitivity and binding to alphavirus genomic RNA. Using a chimeric virus, we found the ZAP sensitivity determinant lies mainly within the alphavirus non-structural protein (nsP) gene region. Surprisingly, we also did not find a correlation between alphavirus ZAP sensitivity and binding to nsP RNA, suggesting ZAP targeting of specific regions in the nsP RNA. Since ZAP can preferentially bind CpG dinucleotides in viral RNA, we identified three 500-bp sequences in the nsP region where CpG content correlates with ZAP sensitivity. Interestingly, ZAP binding to one of these sequences in the nsP2 gene correlated to sensitivity, and we confirmed that this binding is CpG-dependent. Our results demonstrate a potential strategy of alphavirus virulence by localized CpG suppression to evade ZAP recognition.


Asunto(s)
Alphavirus , Virus Chikungunya , Alphavirus/genética , Alphavirus/metabolismo , Antivirales/farmacología , Virus Chikungunya/genética , Virus Chikungunya/metabolismo , ARN Viral/metabolismo , Virus Sindbis/genética , Replicación Viral , Dedos de Zinc , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...