Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78.381
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
J Ethnopharmacol ; 336: 118719, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39179057

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Morus alba L. are widely used as ethnomedicine and functional food in China, Japan, Korea and other Asian countries. Morus alba L. have a variety of pharmacological activity such as antiviral, antioxidation, anti-cholesterol, anticancer, hypoglycemia, and neuroprotection. Morus alba L. has demonstrated antiviral efficacy against influenza viruses, SARS-CoV-2 and so on, but its potential activity against pseudorabies virus (PRV) remains uncertain. AIM OF THE STUDY: This study endeavors to delve into the anti-pseudorabies virus (PRV) potential of the ethanol extract of Morus alba L. leaves (MLE), while simultaneously elucidating its underlying mechanism of action. MATERIALS AND METHODS: The anti-PRV activities of Morus alba L. extracts at different concentrations were evaluated by qPCR and immunoblotting. The inhibitory effects of MLE on PRV replication in three distinct treatment modes (pretreatment, co-treatment, and post-treatment) were detected by qPCR and indirect immunofluorescence assays. qPCR was used to investigate the effects of MLE on PRV attachment, entrance, and cytokine expression in PRV-infected cells. The chemical components in MLE were analyzed by UPLC-MS/MS. RESULTS: MLE significantly inhibits PRV replication and protein expression in a dose-dependent manner. MLE displays inhibitory effects against PRV at three different modes of treatment. The most significant inhibitory effect of MLE was observed when used in co-treatment mode, resulting in an inhibition rate of 99.42%. MLE inhibits PRV infection in the early stage. MLE inhibits PRV infection by affecting viral attachment and viral entry. Furthermore, MLE exerts its inhibition on PRV replication by mitigating the heightened expression of cytokines (TNF-α and IFN-α) triggered by PRV. Analysis of its chemical composition highlights phenolic acids and flavonoids as the principal constituents of MLE. CONCLUSION: The results illustrate that MLE effectively impedes PRV infection by suppressing viral adsorption and entry, while also curbing the expression of antiviral cytokines. Therefore, MLE may be a potential resource for creating new medications to treat human and animal PRV infections.


Assuntos
Antivirais , Herpesvirus Suídeo 1 , Morus , Extratos Vegetais , Folhas de Planta , Replicação Viral , Herpesvirus Suídeo 1/efeitos dos fármacos , Morus/química , Antivirais/farmacologia , Antivirais/isolamento & purificação , Extratos Vegetais/farmacologia , Animais , Replicação Viral/efeitos dos fármacos , Folhas de Planta/química , Citocinas/metabolismo , Cães , Células Madin Darby de Rim Canino , Internalização do Vírus/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos
2.
J Ethnopharmacol ; 336: 118704, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39182703

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Viral pneumonia is the leading cause of death after SARS-CoV-2 infection. Despite effective at early stage, long-term treatment with glucocorticoids can lead to a variety of adverse effects and limited benefits. The Chinese traditional herb Pogostemonis Herba is the aerial part of Pogostemon Cablin (Blanco) Benth., which has potent antiviral, antibacterial, anti-inflammatory, and anticancer effects. It was used widely for treating various throat and respiratory diseases, including COVID-19, viral infection, cough, allergic asthma, acute lung injury and lung cancer. AIM OF THE STUDY: To investigate the antiviral and anti-inflammatory effects of chemical compounds from Pogostemonis Herba in SARS-CoV-2-infected hACE2-overexpressing mouse macrophage RAW264.7 cells and hACE2 transgenic mice. MATERIALS AND METHODS: The hACE2-overexpressing RAW264.7 cells were exposed with SARS-CoV-2. The cell viability was detected by CCK8 assay and cell apoptotic rate was by flow cytometric assay. The expressions of macrophage M1 phenotype markers (TNF-α and IL-6) and M2 markers (IL-10 and Arg-1) as well as the viral loads were detected by qPCR. The mice were inoculated intranasally with SARS-CoV-2 omicron variant to induce viral pneumonia. The levels of macrophages, neutrophils, and T cells in the lung tissues of infected mice were analyzed by full spectrum flow cytometry. The expressions of key proteins were detected by Western blot assay. RESULTS: Diosmetin-7-O-ß-D-glucopyranoside (DG) presented the strongest anti-SARS-CoV-2 activity. Intervention with DG at the concentrations of 0.625-2.5 µM not only reduced the viral replication, cell apoptosis, and the productions of inflammatory cytokines (IL-6 and TNF-α) in SARS-CoV-2-infected RAW264.7 cells, but also reversed macrophage polarity from M1 to M2 phenotype. Furthermore, treatment with DG (25-100 mg/kg) alleviated acute lung injury, and reduced macrophage infiltration in SARS-COV-2-infected mice. Mechanistically, DG inhibited SARS-COV-2 gene expression and HK3 translation via targeting YTHDF1, resulting in the inactivation of glycolysis-mediated NF-κB pathway. CONCLUSIONS: DG exerted the potent antiviral and anti-inflammatory activities. It reduced pneumonia in SARS-COV-2-infected mice via inhibiting the viral replication and accelerating M2 macrophage polarization via targeting YTHDF1, indicating its potential for COVID-19 treatment.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , COVID-19 , Macrófagos , SARS-CoV-2 , Replicação Viral , Animais , Camundongos , Células RAW 264.7 , Replicação Viral/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/virologia , SARS-CoV-2/efeitos dos fármacos , Antivirais/farmacologia , Camundongos Transgênicos , Pogostemon/química , Citocinas/metabolismo , Apoptose/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/virologia , Pulmão/patologia , Glucosídeos/farmacologia , Glucosídeos/isolamento & purificação , Flavonoides/farmacologia , Flavonoides/isolamento & purificação , Flavonoides/uso terapêutico , Enzima de Conversão de Angiotensina 2/metabolismo , Anti-Inflamatórios/farmacologia , Masculino , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/virologia , Humanos
3.
J Ethnopharmacol ; 336: 118720, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39197802

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Jinye Baidu granules (JYBD) have been used to treat acute respiratory tract infections and demonstrated clinical efficacy for the treatment of emerging or epidemic respiratory viruses such as SARS-CoV-2 and influenza virus. AIM OF THE STUDY: This study is to investigate the antiviral effect of JYBD against influenza A viruses (IAV) in vitro and in vivo and elucidate its underlying mechanism. MATERIALS AND METHODS: Ultra-high-performance liquid chromatography connected with Orbitrap mass spectrometer (UHPLC-Orbitrap MS) was employed to describe the chemical profile of JYBD. The potential pathways and targets involved in JYBD against IAV infection were predicted by network pharmacology. The efficacy and mechanism of JYBD were validated through both in vivo and in vitro experiments. Moreover, combination therapy with JYBD and the classic anti-influenza drugs was also investigated. RESULTS: A total of 126 compounds were identified by UHPLC-Orbitrap MS, of which 9 compounds were unambiguously confirmed with reference standards. JYBD could significantly inhibit the replication of multiple strains of IAV, especially oseltamivir-resistant strains. The results of qRT-PCR and WB demonstrated that JYBD could inhibit the excessive induction of pro-inflammatory cytokines induced by IAV infection and regulate inflammatory response through inhibiting JAK/STAT, NF-κB and MAPK pathways. Moreover, both JYBD monotherapy or in combination with oseltamivir could alleviate IAV-induced severe lung injury in mice. CONCLUSIONS: JYBD could inhibit IAV replication and mitigate virus-induced excessive inflammatory response. Combinations of JYBD and neuraminidase inhibitors conferred synergistic suppression of IAV both in vitro and in vivo. It might provide a scientific basis for clinical applications of JYBD against influenza virus infected diseases.


Assuntos
Antivirais , Medicamentos de Ervas Chinesas , Vírus da Influenza A , Farmacologia em Rede , Infecções por Orthomyxoviridae , Antivirais/farmacologia , Animais , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Vírus da Influenza A/efeitos dos fármacos , Cães , Camundongos , Humanos , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , Células Madin Darby de Rim Canino , Replicação Viral/efeitos dos fármacos , Células A549 , Camundongos Endogâmicos BALB C , Masculino , Feminino , Cromatografia Líquida de Alta Pressão
4.
Math Biosci Eng ; 21(9): 7165-7193, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39483079

RESUMO

Multiscale modelling is a promising quantitative approach for studying infectious disease dynamics. This approach garners attention from both individuals who model diseases and those who plan for public health because it has great potential to contribute in expanding the understanding necessary for managing, reducing, and potentially exterminating infectious diseases. In this article, we developed a nested multiscale model of hepatitis B virus (HBV) that integrates the within-cell scale and the between-cell scale at cell level of organization of this disease system. The between-cell scale is linked to the within-cell scale by a once off inflow of initial viral infective inoculum dose from the between-cell scale to the within-cell scale through the process of infection; the within-cell scale is linked to the between-cell scale through the outflow of the virus from the within-cell scale to the between-cell scale through the process of viral shedding or excretion. The resulting multiple scales model is bidirectionally coupled in such a way that the within-cell scale and between-cell scale sub-models mutually affect each other, creating a reciprocal relationship. The computed reproductive number from the multiscale model confirms that the within-host scale and the between-host scale influence each other in a reciprocal manner. Numerical simulations are presented that also confirm the theoretical results and support the initial assumption that the within-cell scale and the between-cell scale influence each other in a reciprocal manner. This multiple scales modeling approach serves as a valuable tool for assessing the impact and success of health strategies aimed at controlling hepatitis B virus disease system.


Assuntos
Número Básico de Reprodução , Simulação por Computador , Vírus da Hepatite B , Hepatite B , Modelos Biológicos , Replicação Viral , Vírus da Hepatite B/fisiologia , Humanos , Hepatite B/virologia , Eliminação de Partículas Virais , Hepatócitos/virologia , Algoritmos
5.
Front Cell Infect Microbiol ; 14: 1399761, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39359939

RESUMO

Introduction: Cytomegaloviruses (CMVs) extensively reorganize the membrane system of the cell and establish a new structure as large as the cell nucleus called the assembly compartment (AC). Our previous studies on murine CMV (MCMV)-infected fibroblasts indicated that the inner part of the AC contains rearranged early endosomes, recycling endosomes, endosomal recycling compartments and trans-Golgi membrane structures that are extensively tubulated, including the expansion and retention of tubular Rab10 elements. An essential process that initiates Rab10-associated tubulation is cargo sorting and retrieval mediated by SNX27, Retromer, and ESCPE-1 (endosomal SNX-BAR sorting complex for promoting exit 1) complexes. Objective: The aim of this study was to investigate the role of SNX27:Retromer:ESCPE-1 complexes in the biogenesis of pre-AC in MCMV-infected cells and subsequently their role in secondary envelopment and release of infectious virions. Results: Here we show that SNX27:Retromer:ESCPE1-mediated tubulation is essential for the establishment of a Rab10-decorated subset of membranes within the pre-AC, a function that requires an intact F3 subdomain of the SNX27 FERM domain. Suppression of SNX27-mediated functions resulted in an almost tenfold decrease in the release of infectious virions. However, these effects cannot be directly linked to the contribution of SNX27:Retromer:ESCPE-1-dependent tubulation to the secondary envelopment, as suppression of these components, including the F3-FERM domain, led to a decrease in MCMV protein expression and inhibited the progression of the replication cycle. Conclusion: This study demonstrates a novel and important function of membrane tubulation within the pre-AC associated with the control of viral protein expression.


Assuntos
Endossomos , Nexinas de Classificação , Replicação Viral , Endossomos/metabolismo , Endossomos/virologia , Animais , Camundongos , Humanos , Nexinas de Classificação/metabolismo , Nexinas de Classificação/genética , Fibroblastos/virologia , Fibroblastos/metabolismo , Muromegalovirus/fisiologia , Muromegalovirus/genética , Linhagem Celular , Montagem de Vírus , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/genética , Citomegalovirus/fisiologia , Citomegalovirus/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética
6.
J Med Virol ; 96(10): e29942, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39360648

RESUMO

Approximately 22% of moderately to severely affected atopic dermatitis (AD) patients have a history of eczema herpeticum, a disseminated rash primarily caused by herpes simplex virus type 1 (HSV-1). Reduced activity of antimicrobial peptides may contribute to the increased susceptibility of AD patients to HSV-1. We previously demonstrated that the antimicrobial protein RNase 7 limits HSV-1 infection of human keratinocytes by promoting self-DNA sensing. Here, we addressed whether RNase 7 has any effect on HSV-1 infection when infecting keratinocytes without exogenously added costimulatory DNA, and which step(s) of the infection cycle RNase 7 interferes with. We quantified viral gene expression by RT-qPCR and flow cytometry, viral genome replication by qPCR, virucidal effects by plaque titration, and plaque formation and the subcellular localization of incoming HSV-1 particles by microscopy. Recombinant RNase 7 restricted HSV-1 gene expression, genome replication, and plaque formation in human keratinocytes. It decreased HSV-1 immediate-early transcripts independently of the induction of interferon-stimulated genes. Its main effect was on intracellular infection processes and not on extracellular virions or virus binding to cells. RNase 7 reduced the amount of cell-associated capsids and the HSV-1 envelope glycoprotein D at 3 but not at 0.5 h postinfection. Our data show that RNase 7 directly restricts HSV-1 infection of human keratinocytes, possibly by promoting the degradation of incoming HSV-1 particles. This suggests that RNase 7 may limit HSV-1 spread in the skin and that mechanisms that reduce its activity in the lesional skin of AD patients may increase their susceptibility to eczema herpeticum.


Assuntos
Herpesvirus Humano 1 , Queratinócitos , Ribonucleases , Replicação Viral , Humanos , Queratinócitos/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Ribonucleases/metabolismo , Ribonucleases/genética , Ensaio de Placa Viral , Células Cultivadas
7.
Vopr Virusol ; 69(4): 309-319, 2024 Sep 26.
Artigo em Russo | MEDLINE | ID: mdl-39361925

RESUMO

INTRODUCTION: The persistence of immune dysfunction during therapy has serious consequences for the health of HIV-infected people. Therefore, an important direction is the search for drugs that can reduce the inflammatory potential of the immune system and serve as an additional component of antiviral therapy. Aim ‒ to study the effect of the immunomodulatory drug Sodium deoxyribonucleate with iron complex (DNA-Na-Fe) on the expression of activation markers in MT-4 cells infected with HIV-1. MATERIALS AND METHODS: Expression levels of CD4, CD28, CD38, CD62L and HLA-DR proteins on the plasma membrane were measured in cells. To assess viral activity, the p24 protein was quantified by ELISA. RESULTS AND DISCUSSION: The two cell variants with different replicative activity were analyzed. Control cells, cells with DNA-Na-Fe, infected cells and infected cells with DNA-Na-Fe were tested. Based on the results obtained, it can be concluded that antiviral activity of the drug in MT-4 cells infected with HIV-1 is associated with immunomodulatory activity that enhances the expression of membrane proteins CD4, CD28, CD38 and CD62L. Diversity in the effect of DNA-Na-Fe on the studied surface proteins expression in two cell lines indicates that they depend on the characteristics of the combined molecular biological processes occurring in cells. And the increased effects observed in a system with changes in replicative activity assumes its active participation in virus replication at the stages of virus penetration and budding. CONCLUSION: Studies have shown that DNA-Na-Fe has antiviral and immunomodulatory activity.


Assuntos
HIV-1 , Ferro , Humanos , HIV-1/efeitos dos fármacos , HIV-1/genética , Ferro/metabolismo , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Infecções por HIV/virologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Antígenos CD28/metabolismo , Antígenos CD28/genética , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/metabolismo , Antígenos CD4/metabolismo , Antígenos CD4/genética
8.
J Med Virol ; 96(10): e29958, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39370884

RESUMO

Kaposi's Sarcoma Herpesvirus (KSHV) is the causative agent of several human diseases. There are no cures for KSHV infection. KSHV establishes biphasic lifelong infections. During the lytic phase, new genomes are replicated by seven viral DNA replication proteins. The processivity factor's (PF-8) functions to tether DNA polymerase to DNA, so new viral genomes are efficiently synthesized. PF-8 self-associates, interacts with KSHV DNA replication proteins and the viral DNA. Inhibition of viral DNA replication would diminish the infection within a host and reduce transmission to new individuals. In this review we summarize PF-8 molecular and structural studies, detail the essential protein-protein and nucleic acid interactions needed for efficient lytic DNA replication, identify future areas for investigation and propose PF-8 as a promising antiviral target. Additionally, we discuss similarities that the processivity factor from Epstein-Barr virus shares with PF-8, which could promote a pan-herpesvirus antiviral therapeutic targeting strategy.


Assuntos
Herpesvirus Humano 8 , Proteínas Virais , Replicação Viral , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Replicação Viral/efeitos dos fármacos , Humanos , Proteínas Virais/metabolismo , Proteínas Virais/genética , Replicação do DNA , Antivirais/farmacologia , DNA Viral/genética
9.
Cell Host Microbe ; 32(10): 1639-1641, 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39389024

RESUMO

Anti-phage defenses must rapidly sense and respond to diverse viruses. A recent pair of papers in Nature reveal via structural and functional assays how the PARIS defense system, a recently discovered toxin-antitoxin system, senses phage-associated molecular patterns (PhAMPs), thereby activating an endonuclease toxin that cleaves tRNA to block phage replication.


Assuntos
Bacteriófagos , RNA de Transferência , RNA de Transferência/metabolismo , Bacteriófagos/genética , Bacteriófagos/metabolismo , Bactérias/virologia , Bactérias/metabolismo , Bactérias/genética , Sistemas Toxina-Antitoxina , Replicação Viral , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Biossíntese de Proteínas
10.
Methods Enzymol ; 705: 81-109, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39389674

RESUMO

There is growing recognition that viral RNA genomes possess enzymatically incorporated modified nucleosides. These small chemical changes are analogous to epigenomic modifications in DNA and have the potential to be similarly important modulators of viral transcription and evolution. However, the molecular level consequences of individual sites of modification remain to be broadly explored. Here we describe an in vitro assay to examine the impact of nucleoside modifications on the rate and fidelity of SARS-CoV-2 RNA transcription. Establishing the role of modified nucleotides in SARS-CoV-2 is of interest both for advancing fundamental knowledge of RNA modifications in viruses, and because modulating the modification-landscape of SARS-CoV-2 may represent a therapeutic strategy to interfere with viral RNA replication. Our approach can be used to assess the influence both of modifications present in a template RNA, as well nucleotide analog inhibitors. These methods provide a reproducible guide for generating active SARS-CoV-2 replication/transcription complexes capable of establishing how RNA modifications influence the pre-steady state rate constants of nucleotide addition by RNA-dependent RNA polymerases.


Assuntos
Nucleosídeos , RNA Viral , SARS-CoV-2 , Replicação Viral , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Nucleosídeos/metabolismo , Nucleosídeos/química , Humanos , Replicação Viral/genética , Transcrição Viral/genética , COVID-19/virologia , COVID-19/metabolismo , Transcrição Gênica
11.
Nat Commun ; 15(1): 8822, 2024 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-39394194

RESUMO

Zika virus (ZIKV), an emerging mosquito-borne flavivirus, is associated with congenital neurological complications. Here, we investigate potential pathological correlates of virus gene expression in representative ZIKV strains through RNA sequencing and ribosome profiling. In addition to the single long polyprotein found in all flaviviruses, we identify the translation of unrecognised upstream open reading frames (uORFs) in the genomic 5' region. In Asian/American strains, ribosomes translate uORF1 and uORF2, whereas in African strains, the two uORFs are fused into one (African uORF). We use reverse genetics to examine the impact on ZIKV fitness of different uORFs mutant viruses. We find that expression of the African uORF and the Asian/American uORF1 modulates virus growth and tropism in human cortical neurons and cerebral organoids, suggesting a potential role in neurotropism. Although the uORFs are expressed in mosquito cells, we do not see a measurable effect on transmission by the mosquito vector in vivo. The discovery of ZIKV uORFs sheds new light on the infection of the human brain cells by this virus and raises the question of their existence in other neurotropic flaviviruses.


Assuntos
Encéfalo , Neurônios , Fases de Leitura Aberta , Infecção por Zika virus , Zika virus , Zika virus/genética , Zika virus/fisiologia , Humanos , Fases de Leitura Aberta/genética , Infecção por Zika virus/virologia , Animais , Encéfalo/virologia , Neurônios/virologia , Neurônios/metabolismo , Replicação Viral , Organoides/virologia , Chlorocebus aethiops , Tropismo Viral , Células Vero , Mosquitos Vetores/virologia , Ribossomos/metabolismo
12.
PLoS Pathog ; 20(10): e1012612, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39352890

RESUMO

Published studies have generated compelling results indicating that type I IFN modulates function of HSV-1 latency-associated transcript (LAT). One member of type I IFN is IFNα2A also called Roferon-A). IFNα2A has been used in monotherapy or in combination therapy with other drugs to treat viral infections and different kinds of cancer in humans. The goal of this study was to determine whether the absence of IFNα2A affects primary and latent infections in ocularly infected mice. Therefore, we generated a mouse strain lacking IFNα2A expression (IFNα2A-/-). Ocular HSV-1 replication, IFN and immune cell expressions on days 3 and 5 post infection (PI), as well as eye disease, survival, latency-reactivation, and T cell exhaustion were evaluated in ocularly infected IFNα2A-/- and wild type (WT) control mice. Absence of IFNα2A did not affect other members of the IFNα family but it affected IFNß and IFNγ expressions as well as some immune cells on day 5 PI compared to WT mice. Viral replication in the eye, eye disease, and survival amongst ocularly infected IFNα2A-/- mice were similar to that of WT infected mice. The absence of IFNα2A significantly reduced the levels of latency and T cell exhaustion but not time of reactivation compared with control mice. Our results suggest that blocking IFNα2A expression may be a useful tool in reducing latency and the subsequent side effects associated with higher levels of latency.


Assuntos
Herpesvirus Humano 1 , Interferon-alfa , Camundongos Knockout , Linfócitos T , Latência Viral , Animais , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/fisiologia , Camundongos , Interferon-alfa/metabolismo , Interferon-alfa/imunologia , Linfócitos T/imunologia , Camundongos Endogâmicos C57BL , Interferon alfa-2/farmacologia , Herpes Simples/imunologia , Herpes Simples/virologia , Replicação Viral , Exaustão das Células T
13.
Vet Res ; 55(1): 135, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39390558

RESUMO

In this study, equine intestinal enteroids (EIEs) were generated from the duodenum, jejunum, and ileum and inoculated with equine coronavirus (ECoV) to investigate their suitability as in vitro models with which to study ECoV infection. Immunohistochemistry revealed that the EIEs were composed of various cell types expressed in vivo in the intestinal epithelium. Quantitative reverse-transcription PCR (qRT-PCR) and virus titration showed that ECoV had infected and replicated in the EIEs. These results were corroborated by electron microscopy. This study suggests that EIEs can be novel in vitro tools for studying the interaction between equine intestinal epithelium and ECoV.


Assuntos
Doenças dos Cavalos , Animais , Cavalos , Doenças dos Cavalos/virologia , Replicação Viral , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Mucosa Intestinal/virologia , Betacoronavirus 1/fisiologia
14.
Physiol Plant ; 176(5): e14540, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39357999

RESUMO

Various members of the viral genera Furovirus and Bymovirus are damaging pathogens of a range of crop species. Infection of the soil-borne plasmodiophorid Polymyxa graminis transmits both Japanese soil-borne wheat mosaic virus (JSBWMV) and the barley yellow mosaic virus (BaYMV) to barley, but their interaction during an episode of their co-infection has not been characterized to date. Here, we present an analysis of the titer of JSBWMV and BaYMV in plants of winter barley growing over a five-month period from late fall until mid-spring. Although JSBWMV was detectable in the plants' roots four weeks earlier than BaYMV, the translocation of both viruses from the root to the leaves occurred nearly simultaneously. Both viruses were co-localized in the roots, leaf sheathes, and leaf blades; however, in some stripes of leaf veins where infection by JSBWMV was prominent, BaYMV was not detectable. A substantial titer of both viruses persisted until early spring, after which JSBWMV became more prominent, being in a range of 10 to 100 times abundant of BaYMV. However, JSBWMV was only able to infect a single wheat accession (cv. Norin 61), whereas all of the wheat entries assayed appeared to be immune to BaYMV infection. Overall, our findings highlight the importance of resistance mechanisms against soil-borne viruses in cereal crops, expanding our understanding of plant-virus interactions and potentially informing strategies for crop protection against viral pathogens.


Assuntos
Hordeum , Doenças das Plantas , Folhas de Planta , Raízes de Plantas , Potyviridae , Coinfecção/virologia , Hordeum/virologia , Vírus do Mosaico/fisiologia , Vírus do Mosaico/patogenicidade , Doenças das Plantas/virologia , Folhas de Planta/virologia , Raízes de Plantas/virologia , Potyviridae/fisiologia , Potyviridae/patogenicidade , Solo , Microbiologia do Solo , Triticum/virologia , Replicação Viral
15.
Avian Dis ; 68(3): 254-258, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39400220

RESUMO

Plant essential oils (EOs) possess established antimicrobial properties; however, research on their antiviral activity, particularly against avian coronaviruses, remains limited. EOs offer a promising plant-based alternative for viral control, especially in scenarios where conventional chemical use is restricted. This study aimed to evaluate the antiviral effects of six different essential oils derived from Syzygium aromaticum, Origanum vulgare, Cymbopogon martinii, Cymbopogon citratus, Mentha piperita, and Mentha spicata against avian coronavirus (AvCov) at 0.1% and 1% dilutions. The antiviral effects of the EOs were assessed via virus isolation from embryonated chicken eggs, and the ability of the EOs to inhibit AvCoV replication was evaluated. The EOs from Syzygium aromaticum, Origanum vulgare, Cymbopogon martinii, and Cymbopogon citratus completely inhibited AvCov replication at a 1% dilution. Conversely, absent to partial inhibitory effect was observed at the 0.1% dilution for all tested EOs, with O. vulgare derived EO exhibiting the greatest inhibitory effect (over 70%). Notably, EOs from Mentha piperita and Mentha spicata were unable to completely inhibit AvCov at either concentration. Our findings highlight the potent antiviral activity of the EOs from Syzygium aromaticum, Origanum vulgare, Cymbopogon martinii, and Cymbopogon citratus against AvCov at a 1% dilution. The complete inhibition observed for these EOs suggests their potential for AvCov control. However, further research is necessary to elucidate the mechanisms of action, optimize formulations, and evaluate the efficacy against other coronaviruses, including those relevant to human health.


Nota de investigación- Efecto antiviral de diferentes aceites esenciales sobre el coronavirus aviar. Los aceites esenciales vegetales poseen propiedades antimicrobianas establecidas; sin embargo, la investigación sobre su actividad antiviral, particularmente contra los coronavirus aviares, sigue siendo limitada. Los aceites esenciales ofrecen una alternativa vegetal prometedora para el control viral, especialmente en escenarios donde el uso de químicos convencionales está restringido. Este estudio tuvo como objetivo evaluar los efectos antivirales de seis aceites esenciales diferentes derivados de Syzygium aromaticum, Origanum vulgare, Cymbopogon martinii, Cymbopogon citratus, Mentha piperita y Mentha spicata contra el coronavirus aviar (AvCov) en diluciones de 0.1% y 1%. Los efectos antivirales de los aceites esenciales se evaluaron mediante el aislamiento del virus a partir de huevos embrionados de pollo y se evaluó la capacidad de los aceites esenciales para inhibir la replicación del coronavirus aviar. Los aceites esenciales de Syzygium aromaticum, Origanum vulgare, Cymbopogon martinii y Cymbopogon citratus inhibieron completamente la replicación del coronavirus aviar en una dilución del 1%. Por el contrario, se observó un efecto inhibidor nulo o parcial con la dilución del 0.1% para todos los aceites esenciales analizados, siendo el aceite esencial derivado de O. vulgare el que exhibió el mayor efecto inhibidor (más del 70%). En particular, los aceites esenciales de Mentha piperita y Mentha spicata no pudieron inhibir completamente al coronavirus aviar en ninguna de las concentraciones. Nuestros hallazgos resaltan la potente actividad antiviral de los aceites esenciales de Syzygium aromaticum, Origanum vulgare, Cymbopogon martinii y Cymbopogon citratus contra coronavirus en una dilución del 1%. La inhibición completa observada para estos aceites esenciales sugiere su potencial para el control de coronavirus. Sin embargo, se necesitan más investigaciones para dilucidar los mecanismos de acción, optimizar las formulaciones y evaluar la eficacia contra otros coronavirus, incluidos los relevantes para la salud humana.


Assuntos
Antivirais , Óleos Voláteis , Óleos Voláteis/farmacologia , Animais , Antivirais/farmacologia , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/tratamento farmacológico , Embrião de Galinha , Óleos de Plantas/farmacologia , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Infecções por Coronavirus/tratamento farmacológico , Replicação Viral/efeitos dos fármacos , Cymbopogon/química
16.
PLoS Pathog ; 20(10): e1012607, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39356716

RESUMO

The RNA genome of orthoflaviviruses encodes a methyltransferase within the non-structural protein NS5, which is involved in 2'-O-methylation of the 5'-terminal nucleotide of the viral genome resulting in a cap1 structure. While a 2'-O-unmethylated cap0 structure is recognized in vertebrates by the RNA sensor RIG-I, the cap1 structure allows orthoflaviviruses to evade the vertebrate innate immune system. Here, we analyzed whether the cap0 structure is also recognized in mosquitoes. Replication analyses of 2'-O-methyltransferase deficient yellow fever virus mutants (YFV NS5-E218A) of the vaccine 17D and the wild-type Asibi strain in mosquito cells revealed a distinct downregulation of the cap0 viruses. Interestingly, the level of inhibition differed for various mosquito cells. The most striking difference was found in Aedes albopictus-derived C6/36 cells with YFV-17D cap0 replication being completely blocked. Replication of YFV-Asibi cap0 was also suppressed in mosquito cells but to a lower extent. Analyses using chimeras between YFV-17D and YFV-Asibi suggest that a synergistic effect of several mutations across the viral genome accompanied by a faster initial growth rate of YFV-Asibi cap1 correlates with the lower level of YFV-Asibi cap0 attenuation. Viral growth analyses in Dicer-2 knockout cells demonstrated that Dicer-2 is entirely dispensable for attenuating the YFV cap0 viruses. Translation of a replication-incompetent cap0 reporter YFV-17D genome was reduced in mosquito cells, indicating a cap0 sensing translation regulation mechanism. Further, oral infection of Aedes aegypti mosquitoes resulted in lower infection rates for YFV-Asibi cap0. The latter is related to lower viral loads found in the midguts, which largely diminished dissemination to secondary tissues. After intrathoracic infection, YFV-Asibi cap0 replicated slower and to decreased amounts in secondary tissues compared to YFV-Asibi cap1. These results suggest the existence of an ubiquitously expressed innate antiviral protein recognizing 5'-terminal RNA cap-modifications in mosquitoes, both in the midgut as well as in secondary tissues.


Assuntos
Aedes , Metiltransferases , Replicação Viral , Febre Amarela , Vírus da Febre Amarela , Animais , Aedes/virologia , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/fisiologia , Replicação Viral/fisiologia , Metiltransferases/metabolismo , Metiltransferases/genética , Febre Amarela/virologia , Febre Amarela/transmissão , Mosquitos Vetores/virologia , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , RNA Viral/genética
17.
Int J Mol Sci ; 25(19)2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39408931

RESUMO

Antimicrobial peptides represent a promising alternative to traditional drugs in relation to cost, toxicity, and, primarily, the growing problem of drug resistance. Here, we report on the activity against HSV-1 and HSV-2 of a previously described wide-spectrum synthetic decapeptide, Killer Peptide (KP). As determined by plaque reduction assays, treatment with KP at 100 µg/mL resulted in a reduction in the viral yield titer of 3.5 Logs for HSV-1 and 4.1 Logs for HSV-2. Further evaluation of KP antiviral activity focused on the early stages of the virus replicative cycle, including the determination of the residual infectivity of viral suspensions treated with KP. A direct effect of the peptide on viral particles impairing virus absorption and penetration was shown. The toxicity profile proved to be extremely good, with a selectivity index of 29.6 for HSV-1 and 156 for HSV-2. KP was also active against acyclovir (ACV)-resistant HSV isolates, while HSV subcultures in the presence of sub-inhibitory doses of KP did not lead to the emergence of resistant strains. Finally, the antiviral action of KP proved to be synergistic with that of ACV. Overall, these results demonstrate that KP could represent an interesting addition/alternative to acyclovir for antiviral treatment.


Assuntos
Aciclovir , Antivirais , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Antivirais/farmacologia , Antivirais/química , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Chlorocebus aethiops , Células Vero , Animais , Aciclovir/farmacologia , Humanos , Replicação Viral/efeitos dos fármacos , Farmacorresistência Viral/efeitos dos fármacos , Herpes Simples/tratamento farmacológico , Herpes Simples/virologia , Peptídeos Antimicrobianos/farmacologia , Peptídeos Antimicrobianos/química
18.
Mol Plant Pathol ; 25(10): e70017, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39412487

RESUMO

Positive-sense RNA viruses remodel cellular cytoplasmic membranes as the membranous sources for the formation of viral replication organelles (VROs) for viral genome replication. In plants, they traffic through plasmodesmata (PD), plasma membrane-lined pores enabling cytoplasmic connections between cells for intercellular movement and systemic infection. In this study, we employed turnip mosaic virus (TuMV), a plant RNA virus to investigate the involvement of RTNLB3 and RTNLB6, two ER (endoplasmic reticulum) membrane-bending, PD-located reticulon-like (RTNL) non-metazoan group B proteins (RTNLBs) in viral infection. We show that RTNLB3 interacts with TuMV 6K2 integral membrane protein and RTNLB6 binds to TuMV coat protein (CP). Knockdown of RTNLB3 promoted viral infection, whereas downregulation of RTNLB6 restricted viral infection, suggesting that these two RTNLs play contrasting roles in TuMV infection. We further demonstrate that RTNLB3 targets the α-helix motif 42LRKSM46 of 6K2 to interrupt 6K2 self-interactions and compromise 6K2-induced VRO formation. Moreover, overexpression of AtRTNLB3 apparently promoted the selective degradation of the ER and ER-associated protein calnexin, but not 6K2. Intriguingly, mutation of the α-helix motif of 6K2 that is required for induction of VROs severely affected 6K2 stability and abolished TuMV infection. Thus, RTNLB3 attenuates TuMV replication, probably through the suppression of 6K2 function. We also show that RTNLB6 promotes viral intercellular movement but does not affect viral replication. Therefore, the proviral role of RTNLB6 is probably by enhancing viral cell-to-cell trafficking. Taken together, our data demonstrate that RTNL family proteins may play diverse complex, even opposite, roles in viral infection in plants.


Assuntos
Nicotiana , Doenças das Plantas , Potyvirus , Potyvirus/fisiologia , Potyvirus/patogenicidade , Nicotiana/virologia , Doenças das Plantas/virologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Arabidopsis/virologia , Arabidopsis/metabolismo , Arabidopsis/genética , Replicação Viral , Proteínas de Membrana/metabolismo , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Proteínas Virais/metabolismo
19.
Int J Mol Sci ; 25(19)2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39408667

RESUMO

Human exploration of the solar system will expose crew members to galactic cosmic radiation (GCR), with a potential for adverse health effects. GCR particles (protons and ions) move at nearly the speed of light and easily penetrate space station walls, as well as the human body. Previously, we have shown reactivation of latent herpesviruses, including herpes simplex virus, Varicella zoster virus, Epstein-Barr virus, and cytomegalovirus (CMV), during stays at the International Space Station. Given the prevalence of latent CMV and the known propensity of space radiation to cause alterations in many cellular processes, we undertook this study to understand the role of GCR in reactivating latent CMV. Latently infected Kasumi cells with CMV were irradiated with 137Cs gamma rays, 150 MeV protons, 600 MeV/n carbon ions, 600 MeV/n iron ions, proton ions, and simulated GCR. The CMV copy number increased significantly in the cells exposed to radiation as compared with the non-irradiated controls. Viral genome sequencing did not reveal significant nucleotide differences among the compared groups. However, transcriptome analysis showed the upregulation of transcription of the UL49 ORF, implicating it in the switch from latent to lytic replication. These findings support our hypothesis that GCR may be a strong contributor to the reactivation of CMV infection seen in ISS crew members.


Assuntos
Radiação Cósmica , Citomegalovirus , Ativação Viral , Replicação Viral , Radiação Cósmica/efeitos adversos , Citomegalovirus/fisiologia , Citomegalovirus/efeitos da radiação , Humanos , Replicação Viral/efeitos da radiação , Ativação Viral/efeitos da radiação , Latência Viral/efeitos da radiação , Genoma Viral , Raios gama , Infecções por Citomegalovirus/virologia , Linhagem Celular
20.
Cell Syst ; 15(10): 911-929.e8, 2024 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-39368468

RESUMO

The abundance of a protein is defined by its continuous synthesis and degradation, a process known as protein turnover. Here, we systematically profiled the turnover of proteins in influenza A virus (IAV)-infected cells using a pulse-chase stable isotope labeling by amino acids in cell culture (SILAC)-based approach combined with downstream statistical modeling. We identified 1,798 virus-affected proteins with turnover changes (tVAPs) out of 7,739 detected proteins (data available at pulsechase.innatelab.org). In particular, the affected proteins were involved in RNA transcription, splicing and nuclear transport, protein translation and stability, and energy metabolism. Many tVAPs appeared to be known IAV-interacting proteins that regulate virus propagation, such as KPNA6, PPP6C, and POLR2A. Notably, our analysis identified additional IAV host and restriction factors, such as the splicing factor GPKOW, that exhibit significant turnover rate changes while their total abundance is minimally affected. Overall, we show that protein turnover is a critical factor both for virus replication and antiviral defense.


Assuntos
Vírus da Influenza A , Influenza Humana , Replicação Viral , Humanos , Influenza Humana/metabolismo , Influenza Humana/virologia , Marcação por Isótopo , Interações Hospedeiro-Patógeno , Células A549 , Células Madin Darby de Rim Canino , Células HEK293
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA